ÌåäÓíèâåð - MedUniver.com Âñå ðàçäåëû ñàéòà Âèäåî ïî ìåäèöèíå Êíèãè ïî ìåäèöèíå Ôîðóì êîíñóëüòàöèé âðà÷åé  
Ðåêîìåíäóåì:
Ïåäèàòðèÿ:
Ïåäèàòðèÿ
Ãåíåòèêà â ïåäèàòðèè
Äåòñêàÿ àëëåðãîëîãèÿ è èììóíîëîãèÿ
Äåòñêàÿ ãàñòðîýíòåðîëîãèÿ
Äåòñêàÿ ãåìàòîëîãèÿ
Äåòñêàÿ ãèíåêîëîãèÿ
Äåòñêàÿ äåðìàòîëîãèÿ
Äåòñêàÿ è ïîäðîñòêîâàÿ ñòîìàòîëîãèÿ
Äåòñêàÿ êàðäèîëîãèÿ
Äåòñêàÿ íåâðîëîãèÿ
Äåòñêàÿ íåôðîëîãèÿ
Äåòñêàÿ îíêîëîãèÿ
Äåòñêàÿ îðòîïåäèÿ
Äåòñêàÿ îòîðèíîëàðèíãîëîãèÿ
Äåòñêàÿ îôòàëüìîëîãèÿ
Äåòñêàÿ ïóëüìîíîëîãèÿ
Äåòñêàÿ ðåàáèëèòàöèÿ
Äåòñêàÿ ðåâìàòîëîãèÿ
Äåòñêàÿ óðîëîãèÿ
Äåòñêàÿ ôàðìàêîëîãèÿ
Äåòñêàÿ ýíäîêðèíîëîãèÿ
Èíôåêöèîííûå áîëåçíè ó äåòåé
Íåîíàòîëîãèÿ
Íåîòëîæíûå ñîñòîÿíèÿ äåòåé
Ïîäðîñòêîâàÿ ìåäèöèíà
Ðîñò è ðàçâèòèå ðåáåíêà
Îðãàíèçàöèÿ ïåäèàòðè÷åñêîé ïîìîùè
Ôîðóì
 

Àâòîðû òðóäîâ (ñòàòåé) èñïîëüçîâàííûå ïðè ïîäãîòîâêå ìàòåðèàëîâ ïî äåòñêîé íåâðîëîãèè â ïåäèàòðèè

 ñòàòüÿõ íà ñàéòå èñïîëüçîâàíû ñëåäóþùèå ñîêðàùåíèÿ:

  • °F — òåìïåðàòóðà ïî Ôàðåíãåéòó
  • °C — òåìïåðàòóðà ïî Öåëüñèþ
  • 5-ÍÒ — 5-ãèäðîêñèèíäîëóêñóñíàÿ êèñëîòà ìî÷è
  • ÀÁ — àíòèáèîòèê(è)
  • ÀÁËÀ — àëëåðãè÷åñêèé áðîíõîëåãî÷íûé àñïåðãèëëåç
  • ÀÁÒ — àíòèáàêòåðèàëüíàÿ òåðàïèÿ
  • ÀÂ — àòðèîâåíòðèêóëÿðíûé
  • ÀÃ — àðòåðèàëüíàÿ ãèïåðòåíçèÿ
  • ÀÃí — àíòèãåí
  • ÀÄ — àðòåðèàëüíîå äàâëåíèå
  • ÀÄà — àíòèäèóðåòè÷åñêèé ãîðìîí
  • ÀÄÑ — àíàòîêñèí äèôòåðèéíî-ñòîëáíÿ÷íûé
  • ÀÄÑ-Ì — àíàòîêñèí äèôòåðèéíî-ñòîëáíÿ÷íûé (ìàëûå äîçû)
  • ÀÄÔ — àäåíîçèíäèôîñôîðíàÿ êèñëîòà, àäåíîçèíäèôîñôàò
  • ÀÊÄÑ — àíàòîêñèí êîêëþøíî-äèôòåðèéíî-ñòîëáíÿ÷íûé
  • ÀÊÒÃ — àäðåíîêîðòèêîòðîïíûé ãîðìîí
  • ÀÊØ — àîðòîêîðîíàðíîå øóíòèðîâàíèå
  • ÀËÒ — àëàíèíàìèíîòðàíñôåðàçà
  • àìåð. — àìåðèêàíñêèé
  • ÀÌÊ — àçîò ìî÷åâèíû êðîâè
  • ÀÌÔ — àäåíîçèíìîíîôîñôîðíàÿ êèñëîòà, àäåíîçèíìîíîôîñôàò
  • ÀÍÀ — àíòèíóêëåàðíûå àíòèòåëà
  • àíàò. — àíàòîìè÷åñêèé
  • àíãë. — àíãëèéñêèé
  • ÀÏÔ — àíãèîòåíçèí-ïðåâðàùàþùèé ôåðìåíò
  • ÀÐÂÒ — àíòèðåòðîâèðóñíàÿ òåðàïèÿ
  • ÀÐÏ — àêòèâíîñòü ðåíèíà â ïëàçìå êðîâè
  • ACT — àñïàðòàòàìèíîòðàíñôåðàçà
  • ÀÒë — àíòèòåëî
  • ÀÒÔ — àäåíîçèíòðèôîñôîðíàÿ êèñëîòà, àäåíîçèíòðèôîñôàò
  • ÀóÄ — àóòîñîìíî-äîìèíàíòíûé
  • ÀóÐ — àóòîñîìíî-ðåöåññèâíûé
  • ÀÔÏ — α-ôåòîïðîòåèí
  • ÀÔÑ — àíòèôîñôîëèïèäíûé ñèíäðîì
  • ÀÕÝ — àöåòèëõîëèíýñòåðàçà
  • ÀÖÕ — àöåòèëõîëèí
  • À×Ò — àêòèâèðîâàííîå ÷àñòè÷íîå òðîìáîïëàñòèíîâîå âðåìÿ
  • ÁÀ — áðîíõèàëüíàÿ àñòìà
  • ÁÀÄ — áèîëîãè÷åñêè àêòèâíàÿ äîáàâêà
  • ÁÀË — áðîíõîàëüâåîëÿðíûé ëàâàæ
  • ÁÀÐ — áèïîëÿðíîå àôôåêòèâíîå ðàññòðîéñòâî (I èëè II òèïà)
  • ÁÊÊ — áîëüøîé êðóã êðîâîîáðàùåíèÿ
  • ÁËÄ — áðîíõîëåãî÷íàÿ äèñïëàçèÿ
  • ÁËÍÏà — áëîêàäà ëåâîé íîæêè ïó÷êà Ãèñà
  • ÁÌÑÝ — áþðî ìåäèêî-ñîöèàëüíîé ýêñïåðòèçû
  • ÁÏ — áðþøíàÿ ïîëîñòü
  • ÁÏÍÏà — áëîêàäà ïðàâîé íîæêè ïó÷êà Ãèñà
  • ÁÐÌ — áàçîâûå ðåàíèìàöèîííûå ìåðîïðèÿòèÿ
  • ÁÐÍÑ — áûñòðî ðàçðåøèâøååñÿ íåîáúÿñíèìîå ñîáûòèå
  • ÁÖÆ — áàöèëëà Êàëüìåòòà-Ãåðåíà â ò.÷. — â òîì ÷èñëå
  • ÁÔÏ - áèîôèçè÷åñêèé ïðîôèëü ïëîäà
  • â. — âåê
  • â/ — âíóòðè... â ñëîæíîñîñòàâíûõ ñëîâàõ (íàïð. â/ñîñóäèñòûé, â/÷åðåïíîé è ò.ï.)
  • â/à — âíóòðèàðòåðèàëüíî
  • â/â — âíóòðèâåííî
  • â/ê — âíóòðèêîæíî
  • â/ì — âíóòðèìûøå÷íî
  • ââ. — âåêà
  • â-âî — âåùåñòâî
  • ÂÂÝ — âîåííî-âðà÷åáíàÿ ýêñïåðòèçà
  • ÂÃÄ — âíóòðèãëàçíîå äàâëåíèå
  • ÂÃ× — âèðóñ ãåðïåñà ÷åëîâåêà
  • ÂÄÏ — âåðõíèå äûõàòåëüíûå ïóòè
  • ÂÆÊ — âíóòðèæåëóäî÷êîâîå êðîâîèçëèÿíèå
  • ÂÇÊ — âîñïàëèòåëüíûå çàáîëåâàíèÿ êèøå÷íèêà
  • ÂÇÎÌÒ—âîñïàëèòåëüíûå çàáîëåâàíèÿ îðãàíîâ ìàëîãî òàçà
  • ÂÈÏ — âàçîàêòèâíûé èíòåñòèíàëüíûé ïåïòèä
  • ÂÈ× — âèðóñ èììóíîäåôèöèòà ÷åëîâåêà
  • ÂÌÏ — âûñîêîòåõíîëîãè÷íàÿ ìåäèöèíñêàÿ ïîìîùü
  • ÂÌÑ — âíóòðèìàòî÷íîå ñðåäñòâî (ñïèðàëü)
  • ÂÍ — âèðóñíàÿ íàãðóçêà
  • ÂÍÑ — âåãåòàòèâíàÿ íåðâíàÿ ñèñòåìà
  • ÂΠ— Âåëèêàÿ Îòå÷åñòâåííàÿ âîéíà
  • ÂÎÃÌ — âûñîêîãîðíûé îòåê ãîëîâíîãî ìîçãà
  • ÂÎÇ — Âñåìèðíàÿ îðãàíèçàöèÿ çäðàâîîõðàíåíèÿ
  • ÂÎË — âûñîêîãîðíûé îòåê ëåãêèõ
  • ÂÎÌÊ — âðà÷, îñóùåñòâëÿþùèé ìåäèöèíñêèé êîíòðîëü
  • ÂÎÏ — âðà÷ îáùåé ïðàêòèêè
  • ÂÏÂ — âåðõíÿÿ ïîëàÿ âåíà
  • ÂÏÃ — âèðóñ ïðîñòîãî ãåðïåñà
  • ÂÏÐ — âðîæäåííûå ïîðîêè ðàçâèòèÿ
  • ÂÏÑ — âðîæäåííûé ïîðîê ñåðäöà
  • ÂÏ× — âèðóñ ïàïèëëîìû ÷åëîâåêà
  • ÂÐÒ — âñïîìîãàòåëüíûå ðåïðîäóêòèâíûå òåõíîëîãèè
  • ÂÑÎ — âîäíî-ñîëåâîé îáìåí
  • Â×Ä — âíóòðè÷åðåïíîå äàâëåíèå
  • Â×ÈÂË — âûñîêî÷àñòîòíàÿ èñêóññòâåííàÿ âåíòèëÿöèÿ ëåãêèõ (HFV)
  • ÂÝÁ — âèðóñ Ýïøòåéíà-Áàðð
  • ã — ãðàìì
  • ã. — ãîä
  • Ã-6-ÔÄ — ãëþêîçî-6-ôîñôàòäåãèäðîãåíàçà
  • ÃÀÌÊ — ãàììà-àìèíîìàñëÿíàÿ êèñëîòà
  • ÃÁ — ãèïåðòîíè÷åñêàÿ áîëåçíü
  • ÃÁÍ — ãåìîëèòè÷åñêàÿ áîëåçíü íîâîðîæäåííîãî
  • ÃÁÎ — ãèïåðáàðè÷åñêàÿ îêñèãåíàöèÿ
  • ÃÇÒ — ãèïåð÷óâñòâèòåëüíîñòü çàìåäëåííîãî òèïà
  • ãèñò. — ãèñòîëîãè÷åñêèé
  • ÃÊ — ãðóäíàÿ êëåòêà
  • ÃÊÌÏ — ãèïåðòðîôè÷åñêàÿ êàðäèîìèîïàòèÿ
  • ÃÊÑ — ãëþêîêîðòèêîñòåðîèä(û)
  • ÃËÃ — ãåìîôàãîöèòàðíûé ëèìôîãèñòèîöèòîç
  • ÃËÏÑ — ãåìîððàãè÷åñêàÿ ëèõîðàäêà ñ ïî÷å÷íûì ñèíäðîìîì
  • ÃÌ — ãîëîâíîé ìîçã
  • ÃÌ-ÊÑÔ — ãðàíóëîöèòàðíî-ìàêðîôàãàëüíûé êîëîíèåñòèìóëèðóþùèé ôàêòîð
  • ÃÌÔ — ãóàíîçèíìîíîôîñôàò
  • ÃíÐà — ãîíàäîòðîïèí-ðèëèçèíã-ãîðìîí
  • ÃÍÒ — ãèïåð÷óâñòâèòåëüíîñòü íåìåäëåííîãî òèïà
  • ÃÏÎÄ — ãðûæà ïèùåâîäíîãî îòâåðñòèÿ äèàôðàãìû
  • ÃÐÄÑ — ãèïîêñè÷åñêàÿ ðåàêöèÿ äûõàòåëüíîé ñèñòåìû ãðå÷. — ãðå÷åñêèé
  • ÃÒÔ — ãóàíîçèíòðèôîñôàò
  • ÃÓÑ — ãåìîëèòèêî-óðåìè÷åñêèé ñèíäðîì
  • Ãö — ãåðö
  • ÃÝÁ — ãåìàòîýíöåôàëè÷åñêèé áàðüåð
  • ÃÝÐ — ãàñòðîýçîôàãåàëüíûé ðåôëþêñ
  • ÃÝÐÁ — ãàñòðîýçîôàãåàëüíàÿ ðåôëþêñíàÿ áîëåçíü
  • äÁ — äåöèáåë
  • ÄÂÑ — äèññåìèíèðîâàííîå âíóòðèñîñóäèñòîå ñâåðòûâàíèå
  • ÄÃÒ — äèãèäðîòåñòîñòåðîí
  • ÄÄ — äèôôåðåíöèàëüíàÿ äèàãíîñòèêà, äèôôåðåíöèàëüíî-äèàãíîñòè÷åñêèé
  • ÄÄÁÀ — äëèòåëüíîäåéñòâóþùèå β2-àãîíèñòû
  • ÄÄÊ — äèôôåðåíöèàëüíî-äèàãíîñòè÷åñêèé êðèòåðèé (êðèòåðèè)
  • ÄÄÐÌ - äîïóñòèìûé äèàïàçîí ðàñïðåäåëåíèÿ ìàêðîíóòðèåíòîâ
  • ÄÈ — äîâåðèòåëüíûé èíòåðâàë
  • ÄÊ — äèàãíîñòè÷åñêèé êðèòåðèé
  • ÄÊÀ — äèàáåòè÷åñêèé êåòîàöèäîç
  • ÄÊÌÏ — äèëàòàöèîííàÿ êàðäèîìèîïàòèÿ
  • ÄÌÆÏ — äåôåêò ìåææåëóäî÷êîâîé ïåðåãîðîäêè
  • ÄÌÏÏ — äåôåêò ìåæïðåäñåðäíîé ïåðåãîðîäêè
  • ÄÌÑ — äîáðîâîëüíîå ìåäèöèíñêîå ñòðàõîâàíèå
  • ÄÍ — äûõàòåëüíàÿ íåäîñòàòî÷íîñòü
  • ÄÍÊ — äåçîêñèðèáîíóêëåèíîâàÿ êèñëîòà
  • ÄÍÒ — äåôåêòû íåðâíîé òðóáêè
  • ÄÎ - äûõàòåëüíûé îáúåì
  • ÄÎÔÀ — äèãèäðîêñèôåíèëàëàíèí
  • ÄÏ — äûõàòåëüíûå ïóòè
  • ÄÏÊ — äâåíàäöàòèïåðñòíàÿ êèøêà
  • ÄÏÌ — äàòà ïîñëåäíåé ìåíñòðóàöèè
  • ÄÏÏ - äîïóñòèìûé ïðåäåë ïîòðåáëåíèÿ
  • äð. — äðóãèå
  • ÄÒÏ — äîðîæíî-òðàíñïîðòíîå ïðîèñøåñòâèå
  • ÄÖÏ — äåòñêèé öåðåáðàëüíûé ïàðàëè÷
  • ÅÄ — åäèíèöà äåéñòâèÿ, åäèíèöà
  • ÆÂÏ — æåë÷åâûâîäÿùèå ïóòè
  • ÆÄÀ — æåëåçîäåôèöèòíàÿ àíåìèÿ
  • ÆÅË — æèçíåííàÿ åìêîñòü ëåãêèõ
  • ÆÊÁ — æåë÷åêàìåííàÿ áîëåçíü
  • ÆÊÊ — æåëóäî÷íî-êèøå÷íîå êðîâîòå÷åíèå
  • ÆÊÒ — æåëóäî÷íî-êèøå÷íûé òðàêò
  • ÆÍÂËÏ — æèçíåííî íåîáõîäèìûå è âàæíûå ëåêàðñòâåííûå ïðåïàðàòû (ñïèñîê, ïåðå÷åíü)
  • ÇÂÓÐ — çàäåðæêà âíóòðèóòðîáíîãî ðàçâèòèÿ
  • ÇË — çäîðîâûå ëþäè (ëèöà)
  • ÇÍÎ — çëîêà÷åñòâåííîå íîâîîáðàçîâàíèå
  • ÇÎ — çäðàâîîõðàíåíèå
  • ÇÎÆ — çäîðîâûé îáðàç æèçíè
  • ÇÑÍ — çàñòîéíàÿ ñåðäå÷íàÿ íåäîñòàòî÷íîñòü
  • èÀÏÔ — èíãèáèòîðû àíãèîòåíçèí-ïðåâðàùàþùåãî ôåðìåíòà
  • ÈÁÑ — èøåìè÷åñêàÿ áîëåçíü ñåðäöà
  • ÈÂË — èñêóññòâåííàÿ âåíòèëÿöèÿ ëåãêèõ
  • ÈÃÊÑ — èíãàëÿöèîííûå ãëþêîêîðòèêîñòåðîèäû
  • ÈÇË — èíòåðñòèöèàëüíûå çàáîëåâàíèÿ ëåãêèõ
  • ÈÊÏ — èììóíîêîìïðîìåòèðîâàííûå ïàöèåíòû
  • ÈË — èíòåðëåéêèí
  • ÈËÄÁ — èíòåãðèðîâàííîå ëå÷åíèå äåòñêèõ áîëåçíåé
  • ÈÌ — èíôàðêò ìèîêàðäà
  • ÈÌÀÎ — èíãèáèòîð(û) ìîíîàìèíîêñèäàçû
  • ÈÌáïÁÒ — èíôàðêò ìèîêàðäà áåç ïîäúåìà ñåãìåíòà ST
  • ÈÌÏ — èíôåêöèÿ ìî÷åâûõ (ìî÷åâûâîäÿùèõ) ïóòåé
  • ÈÌïÇÒ — èíôàðêò ìèîêàðäà ñ ïîäúåìîì ñåãìåíòà
  • ST ÈÌÒ — èíäåêñ ìàññû òåëà
  • ÈÎË — èíòðàîêóëÿðíàÿ ëèíçà
  • ÈÏÏ — èíãèáèòîðû ïðîòîííîé ïîìïû (ïðîòîííîãî íàñîñà)
  • ÈÏÏÏ — èíôåêöèè, ïåðåäàâàåìûå ïîëîâûì ïóòåì
  • ÈÑÑ — èíäèâèäóàëüíûå ñïàñàòåëüíûå ñðåäñòâà èñòîð. — èñòîðè÷åñêîå èòàë. — èòàëüÿíñêèé
  • ÈÒÏ — èäèîïàòè÷åñêàÿ òðîìáîöèòîïåíè÷åñêàÿ ïóðïóðà
  • ÈÒØ — èíôåêöèîííî-òîêñè÷åñêèé øîê ÈÔÀ — èììóíîôåðìåíòíûé àíàëèç
  • ÊÀ — êîðîíàðíàÿ(ûå) àðòåðèÿ(è)
  • ê — êèëîâîëüò
  • êã — êèëîãðàìì
  • ÊÄÁÀ — êîðîòêîäåéñòâóþùèå β2-àãîíèñòû
  • ÊÄÄ — êîíå÷íî-äèàñòîëè÷åñêîå äàâëåíèå
  • ÊÄÎ — êîíå÷íî-äèàñòîëè÷åñêèé îáúåì
  • ÊÄÐ — êîíå÷íî-äèàñòîëè÷åñêèé ðàçìåð
  • êèò. — êèòàéñêèé
  • êêàë — êèëîêàëîðèÿ
  • êë. — êëåòêè, êëåòîê
  • ÊÌÏ — êà÷åñòâî ìåäèöèíñêîé ïîìîùè
  • ÊîÀ — êîýíçèì À, êîôåðìåíò À
  • ÊÎÅ — êîëîíèåîáðàçóþùàÿ åäèíèöà
  • ÊÎÊ — êîìáèíèðîâàííûå îðàëüíûå êîíòðàöåïòèâû
  • ÊÏÒ — êîãíèòèâíî-ïîâåäåí÷åñêàÿ òåðàïèÿ
  • ÊÐ — êëèíè÷åñêèå ðåêîìåíäàöèè
  • ÊÑÄ — êîíå÷íî-ñèñòîëè÷åñêîå äàâëåíèå
  • ÊÑÎ — êîíå÷íî-ñèñòîëè÷åñêèé îáúåì
  • ÊÑÐ — êîíå÷íî-ñèñòîëè÷åñêèé ðàçìåð
  • ÊÒ — êîìïüþòåðíàÿ òîìîãðàôèÿ
  • ÊÒÂÐ — êîìïüþòåðíàÿ òîìîãðàôèÿ âûñîêîãî ðàçðåøåíèÿ
  • ÊÒÃ — êàðäèîòîêîãðàôèÿ
  • ÊÔÊ — êðåàòèíôîñôîêèíàçà
  • ÊÙÑ — êèñëîòíî-ùåëî÷íîå ñîñòîÿíèå
  • ë — ëèòð
  • ë/î — ëîæíîîòðèöàòåëüíûé
  • ë/ï — ëîæíîïîëîæèòåëüíûé
  • ËÀ — ëåãî÷íàÿ àðòåðèÿ
  • ëàò. — ëàòèíñêèé
  • ËÃ — ëþòåèíèçèðóþùèé ãîðìîí
  • ËÄÃ — ëàêòàòäåãèäðîãåíàçà
  • ËÆ — ëåâûé æåëóäî÷åê
  • ËÎÐ — èìåþùèé îòíîøåíèå ê îòîðèíîëàðèíãîëîãèè (ËÎÐ-âðà÷, ËÎÐ-îðãàíû)
  • ËÏ — ëåêàðñòâåííûé ïðåïàðàò
  • ËÏÂÏ — ëèïîïðîòåèíû âûñîêîé ïëîòíîñòè
  • ËÏÍÏ — ëèïîïðîòåèíû íèçêîé ïëîòíîñòè
  • ËÏÎÍÏ — ëèïîïðîòåèíû î÷åíü íèçêîé ïëîòíîñòè
  • ËÏÓ — ëå÷åáíî-ïðîôèëàêòè÷åñêîå ó÷ðåæäåíèå
  • ËÑ — ëåêàðñòâåííîå ñðåäñòâî
  • ËÓ — ëèìôàòè÷åñêèé óçåë, ëèìôîóçëû
  • ËÔÊ — ëå÷åáíàÿ ôèçêóëüòóðà
  • ì.á. — ìîæåò (ìîã, ìîãóò, ìîãëè) áûòü
  • Ì/Æ — ñîîòíîøåíèå ìóæ÷èí è æåíùèí
  • ìàêñÄ — ìàêñèìàëüíàÿ äîçà
  • ìàêñÍÄ — ìàêñèìàëüíàÿ íà÷àëüíàÿ äîçà
  • ìàêñÐÄ — ìàêñèìàëüíàÿ ðàçîâàÿ äîçà
  • ìàêñÑÄ — ìàêñèìàëüíàÿ ñóòî÷íàÿ äîçà
  • ÌÀÍÊ — ìåòîä àìïëèôèêàöèè íóêëåèíîâûõ êèñëîò
  • ÌÀÎ — ìîíîàìèíîêñèäàçà
  • ÌÂÏ — ìî÷åâûâîäÿùèå ïóòè ìã — ìèëëèãðàìì
  • ÌÄÁ — ìûøå÷íàÿ äèñòðîôèÿ Áåêêåðà
  • ÌÄÄ — ìûøå÷íàÿ äèñòðîôèÿ Äþøåííà
  • ÌÄÌÀ — ìåòèëåíäèîêñèìåòàìôåòàìèí
  • ME — ìåæäóíàðîäíàÿ åäèíèöà
  • ìåä. — ìåäèöèíñêèé
  • ìåñ — ìåñÿö
  • ÌÆ — ìîëî÷íàÿ æåëåçà
  • ÌÆÏ — ìåææåëóäî÷êîâàÿ ïåðåãîðîäêà
  • ÌÇ — ìèíèñòåðñòâî çäðàâîîõðàíåíèÿ
  • ìèí — ìèíóò(à)
  • ìèíÄ — ìèíèìàëüíàÿ äîçà
  • ìèíÑÄ — ìèíèìàëüíàÿ ñóòî÷íàÿ äîçà
  • ÌÊÁ — ìî÷åêàìåííàÿ áîëåçíü
  • ÌÊÁ-10 — Ìåæäóíàðîäíàÿ êëàññèôèêàöèÿ áîëåçíåé 10-ãî ïåðåñìîòðà
  • ÌÊÁ-11 — Ìåæäóíàðîäíàÿ êëàññèôèêàöèÿ áîëåçíåé 11-ãî ïåðåñìîòðà
  • ìê — ìèêðîâîëüò
  • ìêã — ìèêðîãðàìì
  • ÌÊÊ — ìàëûé êðóã êðîâîîáðàùåíèÿ
  • ìêë — ìèêðîëèòð
  • ìë — ìèëëèëèòð
  • ìëí — ìèëëèîí
  • ìëðä — ìèëëèàðä
  • ìì — ìèëëèìåòð
  • ìì ðò.ñò. — ìèëëèìåòð ðòóòíîãî ñòîëáà ììîëü — ìèëëèìîëü
  • ÌÍÍ — ìåæäóíàðîäíîå íåïàòåíòîâàííîå íàèìåíîâàíèå
  • ÌÍÎ — ìåæäóíàðîäíîå íîðìàëèçîâàííîå îòíîøåíèå
  • ÌÎ — ìåäèöèíñêàÿ îðãàíèçàöèÿ
  • ÌÏ — ìåäèöèíñêàÿ ïîìîùü
  • ÌÏÑ — ìî÷åïîëîâàÿ ñèñòåìà
  • ÌÐ — ìàãíèòíî-ðåçîíàíñíûé
  • ÌÐ-âåíîãðàôèÿ — ÌÐÒ âåí è ñèíóñîâ ãîëîâíîãî ìîçãà
  • ÌÐÒ — ìàãíèòíî-ðåçîíàíñíàÿ òîìîãðàôèÿ
  • ìñ — ìèëëèñåêóíäà
  • ÌÑÊÒ — ìóëüòèñïèðàëüíàÿ êîìïüþòåðíàÿ òîìîãðàôèÿ
  • ÌÑÝ — ìåäèêî-ñîöèàëüíàÿ ýêñïåðòèçà
  • ÌÒ — ìàññà òåëà
  • ìòÄÍÊ — ìèòîõîíäðèàëüíàÿ äåçîêñèðèáîíóêëåèíîâàÿ êèñëîòà
  • ÌÒÐ — ìàññà òåëà ïðè ðîæäåíèè
  • ÌÝ - ìèàëãè÷åñêèé ýíöåôàëîìèåëèò
  • í.ý. — íàøåé ýðû
  • ÍÀ — íåðâíàÿ àíîðåêñèÿ
  • ÍÀÄ — íèêîòèíàìèäàäåíèíäèíóêëåîòèä
  • ÍÀÄÍ — íèêîòèíàìèäàäåíèíäèíóêëåîòèä (âîññòàíîâëåííûé)
  • ÍÀÄÔ — íèêîòèíàìèäàäåíèíäèíóêëåîòèäôîñôàò
  • ÍÀÆÁÏ — íåàëêîãîëüíàÿ æèðîâàÿ áîëåçíü ïå÷åíè íàïð. — íàïðèìåð
  • ÍÁ — íåðâíàÿ áóëèìèÿ
  • ÍÄÎ — íåãàòèâíûé äåòñêèé îïûò
  • íåä — íåäåëè
  • íåì. — íåìåöêèé
  • íì — íàíîìåòð
  • ÍÌÈÖ — íàöèîíàëüíûé ìåäèöèíñêèé èññëåäîâàòåëüñêèé öåíòð
  • ÍÌÏ — íåîòëîæíàÿ (ìåäèöèíñêàÿ) ïîìîùü
  • ÍÌÒ — íèçêàÿ ìàññà òåëà
  • ÍÏÀ — íîðìàòèâíî-ïðàâîâîé àêò
  • ÍÏÂ — íèæíÿÿ ïîëàÿ âåíà
  • ÍÏÂÑ — íåñòåðîèäíûå ïðîòèâîâîñïàëèòåëüíûå ñðåäñòâà
  • ÍÑ — íåðâíàÿ ñèñòåìà
  • ÍÑÒ - íåñòðåññîâûé òåñò
  • ÍÐÏ — íîðìû ðàöèîíà ïèòàíèÿ
  • ÍÑÃ — íåéðîñîíîãðàôèÿ
  • ÍÝÊ — íåêðîòè÷åñêèé ýíòåðîêîëèò
  • ÍßÊ — íåñïåöèôè÷åñêèé ÿçâåííûé êîëèò
  • ÎÀÊ — îáùèé àíàëèç êðîâè
  • ÎÀÌ — îáùèé àíàëèç ìî÷è
  • ÎÀÏ — îòêðûòûé àðòåðèàëüíûé ïðîòîê
  • ÎÁÏ — îðãàíû áðþøíîé ïîëîñòè
  • ÎÃÁ — îñòðàÿ ãîðíàÿ áîëåçíü
  • ÎÃÌ — îòåê ãîëîâíîãî ìîçãà
  • ÎÄÍ — îñòðàÿ äûõàòåëüíàÿ íåäîñòàòî÷íîñòü
  • ÎÆÑÑ — îáùàÿ æåëåçîñâÿçûâàþùàÿ ñïîñîáíîñòü ñûâîðîòêè
  • ÎÇ — îðãàíèçàöèÿ çäðàâîîõðàíåíèÿ
  • îê. — îêîëî
  • ÎÊÐ — îáñåññèâíî-êîìïóëüñèâíîå ðàññòðîéñòâî
  • ÎÊÑ — îñòðûé êîðîíàðíûé ñèíäðîì
  • ÎËË — îñòðûé ëèìôîáëàñòíûé ëåéêîç
  • ÎÌÑ — îáÿçàòåëüíîå ìåäèöèíñêîå ñòðàõîâàíèå
  • ÎÌÒ — îðãàíû ìàëîãî òàçà
  • ÎÍÌÊ — îñòðîå íàðóøåíèå ìîçãîâîãî êðîâîîáðàùåíèÿ
  • ÎÍÌÏ — îòäåëåíèå íåîòëîæíîé (ìåäèöèíñêîé) ïîìîùè
  • ÎÍÌÒ — î÷åíü íèçêàÿ ìàññà òåëà
  • ÎÎÇ — îðãàíû (îðãàíèçàöèè/óïðàâëåíèÿ) çäðàâîîõðàíåíèÿ
  • ÎÎÍ — Îðãàíèçàöèÿ Îáúåäèíåííûõ Íàöèé
  • ÎÏÆ — îæèäàåìàÿ ïðîäîëæèòåëüíîñòü æèçíè
  • ÎÏÍ — îñòðàÿ ïî÷å÷íàÿ íåäîñòàòî÷íîñòü
  • ÎÏÑÑ - îáùåå ïåðèôåðè÷åñêîå ñîñóäèñòîå ñîïðîòèâëåíèå
  • ÎÐÂÈ — îñòðàÿ ðåñïèðàòîðíàÿ âèðóñíàÿ èíôåêöèÿ
  • ÎÐÄÑ — îñòðûé ðåñïèðàòîðíûé äèñòðåññ-ñèíäðîì
  • ÎÐÇ — îñòðîå ðåñïèðàòîðíîå çàáîëåâàíèå
  • ÎÐÈÒ — îòäåëåíèå ðåàíèìàöèè è èíòåíñèâíîé òåðàïèè (ICU)
  • ÎÑÂÎ — îáùåå ñîäåðæàíèå âîäû â îðãàíèçìå
  • ÎÔÂ — îáúåì ôîðñèðîâàííîãî âûäîõà
  • ÎÔÂ1 — îáúåì ôîðñèðîâàííîãî âûäîõà çà 1-þ ñåêóíäó
  • ÎÖÊ — îáúåì öèðêóëèðóþùåé êðîâè
  • ï.ç. — ïîëå çðåíèÿ (ìèêðîñêîïà)
  • ï/ê — ïîäêîæíî
  • ÏÀÂ — ïñèõîàêòèâíûå âåùåñòâà
  • ÏÂ — ïðîòðîìáèíîâîå âðåìÿ
  • ÏÂÊ — ïåðèôåðè÷åñêèé âåíîçíûé êàòåòåð
  • ÏÂË — ïåðèâåíòðèêóëÿðíàÿ ëåéêîìàëÿöèÿ
  • Ïã — ïðîñòàãëàíäèí
  • ÏÃÃ — ïðîãðàììà ãîñóäàðñòâåííûõ ãàðàíòèé îêàçàíèÿ ãðàæäàíàì ÐÔ áåñïëàòíîé ìåäèöèíñêîé ïîìîùè
  • ÏÄÊÂ — ïîëîæèòåëüíîå äàâëåíèå êîíöà âûäîõà (PEEP)
  • ÏÆ — ïðàâûé æåëóäî÷åê
  • ÏÆÆ — ïîäæåëóäî÷íàÿ æåëåçà
  • ÏÆÊ — ïîäêîæíî-æèðîâàÿ êëåò÷àòêà (ãèïîäåðìà)
  • ÏÊÀ — ïî÷å÷íûé êàíàëüöåâûé àöèäîç
  • ÏÊÌÄ — ïîÿñíî-êîíå÷íîñòíàÿ ìûøå÷íàÿ äèñòðîôèÿ
  • ÏËÑÃ — ïÿòíèñòàÿ ëèõîðàäêà Ñêàëèñòûõ ãîð
  • ÏÌÄÐ — ïðåäìåíñòðóàëüíîå äèñôîðè÷åñêîå ðàññòðîéñòâî
  • ÏÌÊ — ïðîëàïñ ìèòðàëüíîãî êëàïàíà
  • ÏÌÑ — ïðåäìåíñòðóàëüíûé ñèíäðîì
  • ÏÌÑÏ — ïåðâè÷íàÿ ìåäèêî-ñàíèòàðíàÿ ïîìîùü
  • ÏÍÆÊ — ïîëèíåíàñûùåííûå æèðíûå êèñëîòû
  • ÏÎÐÈÒ— ïåäèàòðè÷åñêîå îòäåëåíèå ðåàíèìàöèè è èíòåíñèâíîé òåðàïèè
  • ÏÎÒ — ïîñòóðàëüíàÿ îðòîñòàòè÷åñêàÿ òàõèêàðäèÿ
  • ÏÏÂ — ïíåâìîêîêêîâàÿ ïîëèñàõàðèäíàÿ âàêöèíà
  • ÏÏÎÀÊ — ïðÿìûå ïåðîðàëüíûå àíòèêîàãóëÿíòû
  • ÏÏÒ — ïëîùàäü ïîâåðõíîñòè òåëà
  • ÏÑÀ — ïðîñòàòîñïåöèôè÷åñêèé àíòèãåí
  • ÏÑÂ — ïèêîâàÿ ñêîðîñòü âûäîõà
  • ÏÑÂÕ — ïðîãðåññèðóþùèé ñåìåéíûé âíóòðèïå÷åíî÷-íûé õîëåñòàç
  • ÏÒ — ïñèõîòåðàïèÿ
  • ÏÒÂ — ïðîòðîìáèíîâîå âðåìÿ
  • ÏÒÃ — ïàðàòèðåîèäíûé ãîðìîí
  • ÏÒÈ — ïðîòðîìáèíîâûé èíäåêñ
  • ÏÒÑÐ — ïîñòòðàâìàòè÷åñêîå ñòðåññîâîå ðàññòðîéñòâî
  • ÏÔÝ — ïèùåâîé ôîëàòíûé ýêâèâàëåíò
  • ÏÖÐ — ïîëèìåðàçíàÿ öåïíàÿ ðåàêöèÿ
  • ÏØÃ — ïóðïóðà Øåíëåéíà-Ãåíîõà
  • ÏÙÆ — ïàðàùèòîâèäíûå æåëåçû
  • ÏÝÒ — ïîçèòðîííî-ýìèññèîííàÿ òîìîãðàôèÿ ð/ — ðàç â ... (ñ, ìèí, ÷, ñóò è ò.ï.)
  • ÐÀ — ðåâìàòîèäíûé àðòðèò
  • ÐÀÀÑ — ðåíèí-àíãèîòåíçèí-àëüäîñòåðîíîâàÿ ñèñòåìà ðàçã. — ðàçãîâîðíîå ðàçë. — ðàçëè÷íîå
  • ÐÀÑ — ðàññòðîéñòâî àóòèñòè÷åñêîãî ñïåêòðà
  • ÐÄÑ — ðåñïèðàòîðíûé äèñòðåññ-ñèíäðîì ðèì. — ðèìñêèé
  • ÐÈÔ — ðåàêöèÿ èììóíîôëþîðåñöåíöèè
  • ÐÊÈ — ðàíäîìèçèðîâàííûå êëèíè÷åñêèå èññëåäîâàíèÿ
  • ÐÊÌÏ — ðåñòðèêòèâíàÿ êàðäèîìèîïàòèÿ
  • ÐÈÃÀ — ðåàêöèÿ íåïðÿìîé ãåìàããëþòèíàöèè
  • ÐÍ - ðåòèíîïàòèÿ íåäîíîøåííûõ
  • ÐÍÊ — ðèáîíóêëåèíîâàÿ êèñëîòà
  • ÐÍÏ - ðåêîìåíäóåìóþ íîðìó ïîòðåáëåíèÿ
  • ÐÎÃÊ — ðåíòãåíîãðàôèÿ (ðåíòãåíîãðàììà) îðãàíîâ ãðóäíîé êëåòêè
  • ÐÎËÑÍÑ — ðåêîìåíäàöèè ïî îöåíêå è ëå÷åíèþ ïðè ñîðòèðîâêå â íåîòëîæíûõ ñèòóàöèÿõ
  • ÐÏÃÀ — ðåàêöèÿ ïðÿìîé ãåìàããëþòèíàöèè
  • ð-ð — ðàñòâîð
  • ð-ðèòåëü — ðàñòâîðèòåëü
  • ÐÐÊ — ðåöåïòîðû ðåòèíîåâîé êèñëîòû
  • PPM — ðàñøèðåííûå ðåàíèìàöèîííûå ìåðîïðèÿòèÿ
  • ÐÑÂ — ðåñïèðàòîðíî-ñèíöèòèàëüíûé âèðóñ
  • ÐÑÄ — ðàññòðîéñòâî ñòåðåîòèïíûõ äâèæåíèé
  • ÐÑÊ — ðåàêöèÿ ñâÿçûâàíèÿ êîìïëåìåíòà
  • ÐÑÏ - ðàñ÷åòíàÿ ñðåäíÿÿ ïîòðåáíîñòü
  • ÐÒÃÀ — ðåàêöèÿ òîðìîæåíèÿ ãåìàããëþòèíàöèè
  • ÐÒÍÃÀ — ðåàêöèÿ òîðìîæåíèÿ íåïðÿìîé ãåìàããëþòèíàöèè
  • ÐÔ — Ðîññèéñêàÿ Ôåäåðàöèÿ
  • ÐÕÐ — ðåòèíîåâûå Õ-ðåöåïòîðû
  • Ð×À — ðàäèî÷àñòîòíàÿ àáëÿöèÿ
  • ÐÝÑ — ðåòèêóëîýíäîòåëèàëüíàÿ ñèñòåìà
  • ñ — ñåêóíäà
  • ñâ-âî — ñâîéñòâî
  • ÑÀÌ - ñèíäðîì àñïèðàöèè ìåêîíèÿ
  • ÑÂÄÑ — ñèíäðîì âíåçàïíîé äåòñêîé ñìåðòè
  • ÑÃß — ñèíäðîì ãèïåðñòèìóëÿöèè ÿè÷íèêîâ
  • ÑÄ — ñàõàðíûé äèàáåò
  • ÑÄ-1 — ñàõàðíûé äèàáåò 1-ãî òèïà
  • ÑÄ-2 — ñàõàðíûé äèàáåò 2-ãî òèïà
  • ÑÄÂÃ — ñèíäðîì äåôèöèòà âíèìàíèÿ è ãèïåðàêòèâíîñòè
  • ÑÅ — ñóáúåäèíèöà
  • ÑÆÊ — ñâîáîäíûå æèðíûå êèñëîòû
  • ÑÇÄ - ñëóæáà çàùèòû äåòåé
  • ÑÇÑÒ — ñèñòåìíîå(ûå) çàáîëåâàíèå(ÿ) ñîåäèíèòåëüíîé òêàíè
  • ÑÈÇ — ñðåäñòâà èíäèâèäóàëüíîé çàùèòû
  • ÑÈÎÇÑ— ñåëåêòèâíûé èíãèáèòîð îáðàòíîãî çàõâàòà ñåðîòîíèíà
  • ÑÈÎÇÑÍ — ñåëåêòèâíûå èíãèáèòîðû îáðàòíîãî çàõâàòà ñåðîòîíèíà è íîðýïèíåôðèíà
  • ÑÊÀ — ñåðïîâèäíî-êëåòî÷íàÿ àíåìèÿ
  • ÑÊ — ñåðïîâèäíî-êëåòî÷íàÿ áîëåçíü
  • ÑÊ — ñèñòåìíàÿ êðàñíàÿ âîë÷àíêà
  • ÑÊÔ — ñêîðîñòü êëóáî÷êîâîé ôèëüòðàöèè
  • ÑËÐ — ñåðäå÷íî-ëåãî÷íàÿ ðåàíèìàöèÿ
  • ñì — ñàíòèìåòð
  • ñì âîä.ñò. — ñàíòèìåòð âîäÿíîãî ñòîëáà
  • ÑÌÆ — ñïèííîìîçãîâàÿ æèäêîñòü
  • ÑÌÈ — ñðåäñòâà ìàññîâîé èíôîðìàöèè
  • ÑÌÎ — ñòðàõîâàÿ ìåäèöèíñêàÿ îðãàíèçàöèÿ
  • ÑÌÏ — ñêîðàÿ ìåäèöèíñêàÿ ïîìîùü
  • ÑÌÝ — ñóäåáíî-ìåäèöèíñêàÿ ýêñïåðòèçà
  • ÑÍ — ñåðäå÷íàÿ íåäîñòàòî÷íîñòü
  • ÑÍÑÀÄÃ — ñèíäðîì íåàäåêâàòíîé ñåêðåöèè ÀÄÃ
  • ÑÎÀÑ — ñèíäðîì îáñòðóêòèâíîãî àïíîý ñíà (âî ñíå) ñîâð. — ñîâðåìåííûé
  • ÑÎÝ — ñêîðîñòü îñåäàíèÿ ýðèòðîöèòîâ
  • ÑÏÈÄ — ñèíäðîì ïðèîáðåòåííîãî èììóíîäåôèöèòà
  • ÑÏÊß — ñèíäðîì ïîëèêèñòîçíûõ ÿè÷íèêîâ
  • ÑÏÎÍ — ñèíäðîì ïîëèîðãàííîé íåäîñòàòî÷íîñòè
  • ÑÐÂ — Ñ-ðåàêòèâíûé áåëîê
  • ÑÐÊ — ñèíäðîì ðàçäðàæåííîãî êèøå÷íèêà
  • ÑðÑÄ — ñðåäíÿÿ ñóòî÷íàÿ äîçà
  • ÑÑÂ — ñèíäðîì Ñòåðäæà-Âåáåðà
  • ÑÑÂÐ — ñèíäðîì ñèñòåìíîé âîñïàëèòåëüíîé ðåàêöèè
  • ÑÑÇ — ñåðäå÷íî-ñîñóäèñòûå çàáîëåâàíèÿ
  • ÑÑÑ — ñåðäå÷íî-ñîñóäèñòàÿ ñèñòåìà
  • ÑÑÑÓ — ñèíäðîì ñëàáîñòè ñèíóñîâîãî óçëà
  • ÑÒ — Ñèíäðîì Òóðåòòà
  • ÑÒÃ — ñîìàòîòðîïíûé ãîðìîí
  • ÑÒÐ — ñòîéêîå (õðîíè÷åñêîå) ìîòîðíîå èëè âîêàëüíîå òèêîçíîå ðàññòðîéñòâî
  • ñóò — ñóòêè
  • ÑÕÓ — ñèíäðîì õðîíè÷åñêîé óñòàëîñòè
  • ò.ä. — òàê äàëåå
  • ò.å. — òî åñòü
  • ò.ê. — òàê êàê
  • ò.î. — òàêèì îáðàçîì
  • ò.ï. — òîìó ïîäîáíîå
  • Ò3 — òðèéîäòèðîíèí
  • Ò4 — òèðîêñèí
  • ÒÀÍÊ — òåñò àìïëèôèêàöèè íóêëåèíîâûõ êèñëîò
  • ÒÁÑ — òàçîáåäðåííûé ñóñòàâ
  • ÒÃÂ — òðîìáîç ãëóáîêèõ âåí
  • ÒÃÑÊ — òðàíñïëàíòàöèÿ ãåìîïîýòè÷åñêèõ ñòâîëîâûõ êëåòîê
  • ÒÈÀ — òðàíçèòîðíàÿ èøåìè÷åñêàÿ àòàêà
  • ÒÈÀÁ — òîíêîèãîëüíàÿ àñïèðàöèîííàÿ áèîïñèÿ
  • ÒÌÎ — òâåðäàÿ ìîçãîâàÿ îáîëî÷êà
  • TH — òîðãîâîå íàèìåíîâàíèå ëåêàðñòâåííûõ ñðåäñòâ
  • ÒÏÃÃ — òåððèòîðèàëüíàÿ ïðîãðàììà ãîñóäàðñòâåííûõ ãàðàíòèé îêàçàíèÿ ãðàæäàíàì ÐÔ áåñïëàòíîé ìåäèöèíñêîé ïîìîùè
  • ÒÏÌ — òðàâìàòè÷åñêîå ïîâðåæäåíèå ìîçãà
  • ÒÒ — òåìïåðàòóðà òåëà
  • ÒÒÃ — òèðåîòðîïíûé ãîðìîí
  • ÒÔÐ — òðàíñôîðìèðóþùèé ôàêòîð ðîñòà
  • ÒÖÀ — òðèöèêëè÷åñêèå àíòèäåïðåññàíòû
  • òûñ. — òûñÿ÷à
  • ÒÝÄ —òðàíçèòîðíàÿ ýðèòðîáëàñòîïåíèÿ äåòñêîãî âîçðàñòà
  • ÒÝËÀ — òðîìáîýìáîëèÿ ëåãî÷íîé àðòåðèè
  • ÒÝÎ — òðîìáîýìáîëè÷åñêèå îñëîæíåíèÿ
  • ÓÇÄÑ — óëüòðàçâóêîâîå äóïëåêñíîå ñêàíèðîâàíèå
  • ÓÇÈ — óëüòðàçâóêîâîå èññëåäîâàíèå
  • óñòàð. — óñòàðåâøåå
  • ÓÔ — óëüòðàôèîëåòîâûé
  • ÓÔÎ — óëüòðàôèîëåòîâîå îáëó÷åíèå
  • ÔÀÏ — ôåëüäøåðñêî-àêóøåðñêèé ïóíêò
  • ÔÂ — ôðàêöèÿ âûáðîñà
  • ÔÂÄ — ôóíêöèè âíåøíåãî äûõàíèÿ
  • ÔÆÅË — ôîðñèðîâàííàÿ æèçíåííàÿ åìêîñòü ëåãêèõ
  • ÔÇ — Ôåäåðàëüíûé çàêîí
  • ôèçèîë. — ôèçèîëîãè÷åñêèé
  • ÔÊ — ôóíêöèîíàëüíûé êëàññ
  • ÔÊÑ — ôèáðîêîëîíîñêîïèÿ
  • ÔÊÓ — ôåíèëêåòîíóðèÿ
  • ÔÍ — ôèçè÷åñêàÿ íàãðóçêà
  • ÔÈÎ — ôàêòîð íåêðîçà îïóõîëè
  • ÔÎÌÑ — ôåäåðàëüíûé ôîíä îáÿçàòåëüíîãî ìåäèöèíñêîãî ñòðàõîâàíèÿ
  • ÔÎÑ — ôîñôîðîðãàíè÷åñêèå ñîåäèíåíèÿ
  • ÔÏ — ôèáðèëëÿöèÿ ïðåäñåðäèé
  • ôð. — ôðàíöóçñêèé
  • ÔÑÃ — ôîëëèêóëîñòèìóëèðóþùèé ãîðìîí
  • ÔÝÃÄÑ — ôèáðîýçîôàãîãàñòðîäóîäåíîñêîïèÿ
  • ÕÁÏ — õðîíè÷åñêàÿ áîëåçíü ïî÷åê
  • ÕÃ× — õîðèîíè÷åñêèé ãîíàäîòðîïèí ÷åëîâåêà
  • ÕÄÍ — õðîíè÷åñêàÿ äûõàòåëüíàÿ íåäîñòàòî÷íîñòü
  • õèì. — õèìè÷åñêàÿ
  • ÕÍÇË —õðîíè÷åñêèå íåñïåöèôè÷åñêèå çàáîëåâàíèÿ ëåãêèõ
  • ÕÎÁË — õðîíè÷åñêàÿ îáñòðóêòèâíàÿ áîëåçíü ëåãêèõ
  • ÕÏÍ — õðîíè÷åñêàÿ ïî÷å÷íàÿ íåäîñòàòî÷íîñòü
  • ÕÑ — õîëåñòåðèí
  • ÕÑÍ — õðîíè÷åñêàÿ ñåðäå÷íàÿ íåäîñòàòî÷íîñòü
  • XT — õèìèîòåðàïèÿ
  • ÖÂÄ — öåíòðàëüíîå âåíîçíîå äàâëåíèå
  • ÖÂÊ — öåíòðàëüíûé âåíîçíûé êàòåòåð
  • ÖÈÊ — öèðêóëèðóþùèå èììóííûå êîìïëåêñû
  • ÖÌÂ — öèòîìåãàëîâèðóñ
  • ÖÍÑ — öåíòðàëüíàÿ íåðâíàÿ ñèñòåìà
  • ÖÎÃ — öèêëîîêñèãåíàçà
  • ÖÏÄ — öåðåáðàëüíîå ïåðôóçèîííîå äàâëåíèå
  • ÖÑÒÑ — öåðåáðàëüíûé ñîëüòåðÿþùèé ñèíäðîì ÷ — ÷àñ
  • ×Ä — ÷àñòîòà äûõàíèÿ
  • ×ÄÄ — ÷àñòîòà äûõàòåëüíûõ äâèæåíèé
  • ×Ê — ÷ðåñêîæíîå êîðîíàðíîå âìåøàòåëüñòâî
  • ×ÌÍ — ÷åðåïíî-ìîçãîâûå íåðâû
  • ×ÌÒ — ÷åðåïíî-ìîçãîâàÿ òðàâìà
  • ×Í — ÷åðåïíûå íåðâû
  • ×Ñ — ÷óâñòâèòåëüíîñòü/ñïåöèôè÷íîñòü (×Ñ 97%/87%)
  • ×ÑÑ — ÷àñòîòà ñåðäå÷íûõ ñîêðàùåíèé
  • ×ÑÑÏ - ÷àñòîòà ñåðäå÷íûõ ñîêðàùåíèé ïëîäà
  • ×Ò — ÷àñòè÷íîå òðîìáîïëàñòèíîâîå âðåìÿ
  • ØÊÃ — øêàëà êîìû Ãëàçãî
  • ØÎÏ — øåéíûé îòäåë ïîçâîíî÷íèêà
  • ÙÆ — ùèòîâèäíàÿ æåëåçà
  • ÃÖÔ — ùåëî÷íàÿ ôîñôàòàçà
  • ÝÀÐ — ýêâèâàëåíò àêòèâíîñòè ðåòèíîëà
  • ÝÄÒÀ — ýòèëåíäèàìèíòåòðàóêñóñíàÿ êèñëîòà (ýòèëåíäèàìèíòåòðààöåòàò)
  • ÝÊÃ — ýëåêòðîêàðäèîãðàôèÿ
  • ÝÊÌÎ — ýêñòðàêîðïîðàëüíàÿ ìåìáðàííàÿ îêñèãåíàöèÿ
  • ÝÊÎ — ýêñòðàêîðïîðàëüíîå îïëîäîòâîðåíèå
  • ÝÊÑ — ýëåêòðîêàðäèîñòèìóëÿòîð, ýëåêãðîêàðäèîñòèìóëÿöèÿ
  • ÝÌÃ — ýëåêòðîìèîãðàôèÿ
  • ÝÌÊ — ýëåêòðîííàÿ ìåäèöèíñêàÿ êàðòà
  • ÝÌÏ — ýêñòðåííàÿ ìåäèöèíñêàÿ ïîìîùü
  • ÝÍÌÏ — ýêñòðåííàÿ è íåîòëîæíàÿ ìåäèöèíñêàÿ ïîìîùü
  • ÝÍÌÒ — ýêñòðåìàëüíî íèçêàÿ ìàññà òåëà
  • ÝÒÒ — ýíäîòðàõåàëüíàÿ òðóáêà
  • ÝõîÊà — ýõîêàðäèîãðàôèÿ
  • ÝÝÃ — ýëåêòðîýíöåôàëîãðàôèÿ
  • ÞÄÌ — þâåíèëüíûé äåðìàòîìèîçèò
  • ÞÈÀ — þâåíèëüíûé èäèîïàòè÷åñêèé àðòðèò
  • ßÁ — ÿçâåííàÿ áîëåçíü
  • ÀÀ — àìèëîèä A (amyloid À)
  • ÀÀÐ — Àìåðèêàíñêàÿ àêàäåìèÿ ïåäèàòðèè (American Academy of Pediatrics)
  • ABC — àëãîðèòì íåîòëîæíîé ïîìîùè «äûõàòåëüíûå ïóòè-äûõàíèå-êðîâîîáðàùåíèå» (Airways-Breathing-Circulation)
  • ÀÂÑÀ3 — áåëêîâûé ÷ëåí ÀÒÔ-ñâÿçûâàþùåé êàññåòû À3
  • ÀÂÑÂ11— ãåí ÀÒÔ-ñâÿçûâàþùåé êàññåòû, 11-é ÷ëåí ïîäñåìåéñòâà  (ATP-binding cassette, sub-family  member 11)
  • ÀÂÑÂ4 — ãåí ÀÒÔ-ñâÿçûâàþùåé êàññåòû, 4-é ÷ëåí ïîäñåìåéñòâà  (ATP-binding cassette 4 gene)
  • ÀÂÑÑ2 — ãåí, êîäèðóþùèé 2-é ÷ëåí ïîäñåìåéñòâà Ñ ÀÒÔ-ñâÿçûâàþùèõ êàññåò (ATP-binding cassette sub-family Ñ member 2)
  • ABCDE — àëãîðèòì íåîòëîæíîé ïîìîùè «äûõàòåëüíûå ïóòè-äûõàíèå-êðîâîîáðàùåíèå-íåâðîëîãè÷åñêèé ñòàòóñ-âíåøíèé âèä» (Airways-Breathing-Circulation-Disability- Exposure)
  • ABCG5/G8 — ãåòåðîäèìåð ïåðåíîñ÷èêà ÀÒÔ-ñâÿçûâàþùåé êàññåòû ABCG5 è ABCG8 (The heterodimer of ATP-binding cassette transporter ABCG5 and ABCG8)
  • AC — äî åäû (ïðè íàçíà÷åíèÿõ) — ante cibum
  • ÀÑÅÐ — Àìåðèêàíñêàÿ êîëëåãèÿ âðà÷åé íåîòëîæíîé ïîìîùè (American College of Emergency Physicians)
  • ACOG — Àìåðèêàíñêàÿ êîëëåãèÿ àêóøåðîâ è ãèíåêîëîãîâ (American College of Obstetricians and Gynecologists)
  • AHA — Àìåðèêàíñêàÿ êàðäèîëîãè÷åñêàÿ àññîöèàöèÿ (American Heart Association)
  • ALTE — î÷åâèäíîå îïàñíîå äëÿ æèçíè ñîáûòèå (apparent life-threatening event)
  • APLS — êóðñû ðàñøèðåííûõ ðåàíèìàöèîííûõ ìåðîïðèÿòèé â ïåäèàòðèè, ñïîíñèðóåìûå Àìåðèêàíñêîé àêàäåìèåé ïåäèàòðèè è Àìåðèêàíñêîé êîëëåãèåé âðà÷åé íåîòëîæíîé ïîìîùè (Advanced Pediatric Life Support)
  • ARPKD — àóòîñîìíî-ðåöåññèâíûé ïîëèêèñòîç ïî÷åê
  • ASCVD-ðèñê — ðèñê ðàçâèòèÿ àòåðîñêëåðîòè÷åñêîãî ñåðäå÷íî-ñîñóäèñòîãî çàáîëåâàíèÿ (ArterioSclerotic Cardiovascular Disease, ASCVD)
  • AVPU — øêàëà äëÿ îöåíêè óðîâíÿ ñîçíàíèÿ («â ÿñíîì ñîçíàíèè»; «ðåàêöèÿ íà âåðáàëüíûå ðàçäðàæèòåëè»; «ðåàêöèÿ íà áîëü»; «áåç ñîçíàíèÿ») (alert, verbal, pain, unresponsive)
  • BCS — ìèòîõîíäðèàëüíûé øàïåðîí BCS1 (mitochondrial chaperone BCS1)
  • BIN — áèíîìèíàëüíàÿ íîìåíêëàòóðà «æèâîé» ïðèðîäû
  • BiPAP — ðåæèì èñêóññòâåííîé âåíòèëÿöèè ëåãêèõ ñ äâóìÿ óðîâíÿìè ïîëîæèòåëüíîãî äàâëåíèÿ (bilevel positive airway pressure) = ÂÐÀÐ
  • BNP — íàòðèéóðåòè÷åñêèé ïåïòèä Â-òèïà — ìîçãîâîé ( (brain)-type natriuretic peptide)
  • BRUE — áûñòðî ðàçðåøèâøèåñÿ íåîáúÿñíèìûå ñîáûòèÿ (Brief resolved unexplained events)
  • BSEP — íàñîñ âûâåäåíèÿ ñîëåé æåë÷íûõ êèñëîò
  • CADASIL — öåðåáðàëüíàÿ àóòîñîìíî-äîìèíàíòíàÿ àðòåðèîïàòèÿ ñ ïîäêîðêîâûìè èíôàðêòàìè è ëåéêîýíöåôàëîïàòèåé (cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy)
  • CAPS — êðèîïèðèí-àññîöèèðîâàííûé ïåðèîäè÷åñêèé ñèíäðîì (cryopyrin-associated periodic syndrome)
  • CBIT — êîìïëåêñíîå ïîâåäåí÷åñêîå âîçäåéñòâèå ïðè òèêàõ (Comprehensive behavioral intervention for tics)
  • CD — êëàñòåðû äèôôåðåíöèðîâêè (clusters of differentiation)
  • CDC — Öåíòð ïî êîíòðîëþ è ïðîôèëàêòèêå çàáîëåâàíèé, ÑØÀ (Center for Disease Control and Prevention)
  • CDG — âðîæäåííûå íàðóøåíèÿ ãëèêîçèëèðîâàíèÿ (Congenital Disorder of Glycosylation)
  • CFTR — òðàíñìåìáðàííûé ðåãóëÿòîð ìóêîâèñöèäîçà (cystic fibrosis transmembrane regulator)
  • CH — ãåìîëèòè÷åñêèé êîìïëåìåíò (hemolytic complement)
  • CLLS — øêàëà îöåíêè îñòðîé ãîðíîé áîëåçíè îçåðà Ëóèç ó äåòåé (Childrens Lake Louise Score)
  • CMT — Øàðêî-Ìàðè-Òóòà áîëåçíü (Charcot-Marie-Tooth disease)
  • ÑÐÀÐ — ïîñòîÿííîå ïîëîæèòåëüíîå äàâëåíèå â äûõàòåëüíûõ ïóòÿõ (Constant Positive Airway Pressure)
  • CYP — öèòîõðîì îáùèé
  • DAF — ôàêòîð óñêîðåíèÿ ðàñïàäà (decay-accelerating factor)
  • DGAT1 —äèàöèëãëèöåðèí-1-àöèëòðàíñôåðàçà (diacylglycerol acyltransferase 1)
  • DHR — äèãèäðîðîäàìèí (dihydrorhodamine)
  • DSM — Äèàãíîñòè÷åñêîå è ñòàòèñòè÷åñêîå ðóêîâîäñòâî ïî ïñèõè÷åñêèì ðàññòðîéñòâàì (Diagnostic and Statistical Manual of Mental Disorders)
  • EAST syndrome — ýïèëåïñèÿ, àòàêñèÿ, ñåíñîíåâðàëüíàÿ òóãîóõîñòü è òóáóëîïàòèÿ (epilepsy, ataxia, sensorineural hearing loss, and tubulopath)
  • EBM — äîêàçàòåëüíàÿ ìåäèöèíà (Evidence based medicine)
  • EPCAM— ìîëåêóëà àäãåçèè ýïèòåëèàëüíûõ êëåòîê (epithelial cell adhesion molecule)
  • ESC — Åâðîïåéñêîå îáùåñòâî êàðäèîëîãèè (European Society of Cardiology)
  • EXIT — ëå÷åíèå âíå ìàòêè âî âðåìÿ ðîäîâ (Ex utero intrapartum treatment)
  • FAST — ñôîêóñèðîâàííàÿ ñîíîãðàôèÿ áðþøíîé ïîëîñòè ïðè òðàâìå (focused assessment with sonography in trauma)
  • FDA — Êîìèòåò ïî êîíòðîëþ çà ëåêàðñòâåííûìè âåùåñòâàìè è ïèùåâûìè äîáàâêàìè, ÑØÀ (Food and Drug Administration)
  • FFR — ôðàêöèîííûé ðåçåðâ êðîâîòîêà (Fractional Flow Reserve)
  • FGF-23 — ôàêòîð ðîñòà ôèáðîáëàñòîâ-23 (Fibroblast growth factor-23)
  • FIC 1 —ñåìåéíûé âíóòðèïå÷åíî÷íûé õîëåñòàç 1-ãî òèïà, áîëåçíü Áàéëåðà (familial intrahepatic cholestasis)
  • FiO2 — ôðàêöèÿ êèñëîðîäà (âî âäûõàåìîì âîçäóõå, ãàçîâîé ñìåñè)
  • FISH — ôëóîðåñöåíòíàÿ in situ ãèáðèäèçàöèÿ (fluorescence in situ hybridization)
  • FLAIR — âîññòàíîâëåíèå èíâåðñèè ñ îñëàáëåíèåì æèäêîñòè (fluid-attenuated inversion recovery)
  • FMF — ñåìåéíàÿ ñðåäèçåìíîìîðñêàÿ ëèõîðàäêà (Familial Mediterranean fever)
  • GATA — gata-ñâÿçûâàþùèé áåëîê
  • HADH — ãèäðîêñèëàöèë-ÊîÀ-äåãèäðîãåíàçà (Hydro-xyacyl-Coenzyme A dehydrogenase)
  • HAV — âèðóñ ãåïàòèòà Â (hepatitis A virus)
  • Hb — ãåìîãëîáèí
  • HBcAg — ñåðäöåâèííûé àíòèãåí âèðóñà ãåïàòèòà Â
  • HBeAg — àíòèãåí âèðóñà ãåïàòèòà Â
  • HBsAg — ïîâåðõíîñòíûé àíòèãåí âèðóñà ãåïàòèòà Â
  • HBV — âèðóñ ãåïàòèòà  (hepatitis  virus)
  • HCV — âèðóñ ãåïàòèòà Ñ (hepatitis Ñ virus)
  • HDV — âèðóñ ãåïàòèòà D (hepatitis D virus)
  • HFNC — íàçàëüíûå êàíþëè âûñîêîãî ïîòîêà (heated, high-flow nasal cannula)
  • Hib — ãåìîôèëüíàÿ ïàëî÷êà òèïà  (Haemophilus influenzae  type)
  • HLA — ëåéêîöèòàðíûå àíòèãåíû (ãëàâíîãî êîìïëåêñà ãèñòîñîâìåñòèìîñòè) ÷åëîâåêà (human leukocyte antigens)
  • HR — îòíîøåíèå ðèñêîâ (hazard ratio)
  • HRT — òåðàïèÿ îòìåíû ïðèâû÷êè (Habit reversal therapy)
  • Ht — ãåìàòîêðèò
  • HTLV — Ò-ëèìôîòðîïíûé âèðóñ ÷åëîâåêà (human T-lymphotropic virus)
  • Ig — èììóíîãëîáóëèí
  • IgA — èììóíîãëîáóëèí A
  • IgE — èììóíîãëîáóëèí E
  • IgG — èììóíîãëîáóëèí G
  • IgM — èììóíîãëîáóëèí M
  • IL — èíòåðëåéêèí
  • IPEX — Õ-ñöåïëåííûé ñèíäðîì èììóííîé äèñðå-ãóëÿöèè, ïîëèýíäîêðèíîïàòèè è ýíòåðîïàòèè (Immunedysregulation polyendocrinopathy enteropathy, X-linked)
  • IQ — êîýôôèöèåíò óìñòâåííîãî ðàçâèòèÿ (intelligence quotient)
  • JAK — ÿíóñ-êèíàçà (Janus kinase)
  • LFA — àíòèãåí, àêòèâèðóþùèé ôóíêöèþ ëåéêîöèòîâ (Lymphocyte function-associated antigen)
  • LT — ëåéêîòðèåí
  • MALT — ëèìôîèäíàÿ òêàíü ñëèçèñòîé îáîëî÷êè (mucosa-associated lymphoid tissue)
  • MASP — ÌÑË-àññîöèèðîâàííàÿ ñåðèíîâàÿ ïðîòåàçà (MBL-associated serine protease)
  • MBL — ìàííîçî-ñâÿçûâàþùèé ëåêòèí (mannose-
  • binding lectin)
  • MCP — ìåìáðàííûé áåëîê-êîôàêòîð (membrane cofactor protein)
  • MDR3 — áåëîê ìíîæåñòâåííîé ëåêàðñòâåííîé óñòîé÷èâîñòè 3 (multidrug resistance protein 3)
  • MELAS — ìèòîõîíäðèàëüíàÿ ýíöåôàëîïàòèÿ, ëàêòîàöèäîç è èíñóëüòîïîäîáíûå ýïèçîäû (mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes)
  • MERRF — ìèîêëîíè÷åñêàÿ ýïèëåïñèÿ ñ ðâàíûìè êðàñíûìè âîëîêíàìè (Myoclonic epilepsy with ragged red fibers)
  • Mr — êàæóùàÿñÿ ìîëåêóëÿðíàÿ ìàññà
  • MRP — áåëîê ñ ìíîæåñòâåííîé ëåêàðñòâåííîé óñòîé÷èâîñòüþ (multidrug-resistant protein)
  • MRSA — ìåòèöèëëèíðåçèñòåíòíûé çîëîòèñòûé ñòàôèëîêîêê
  • MSSA — ìåòèöèëëèí÷óâñòâèòåëüíûé çîëîòèñòûé ñòàôèëîêîêê
  • NB! — âàæíî, îáðàòèòü âíèìàíèå (Nota bene)
  • NICE — Íàöèîíàëüíûé èíñòèòóò çäîðîâüÿ è êëèíè÷åñêîãî ñîâåðøåíñòâîâàíèÿ (êà÷åñòâà ìåäèöèíñêîé ïîìîùè) Âåëèêîáðèòàíèè (National Institute for Health and Clinical Excellence)
  • NK — åñòåñòâåííûå êëåòêè-êèëëåðû (natural killer)
  • NMDA — N-ìåòèë-D-àñïàðòàò (N-methyl-D-aspartate)
  • NMDA — N-ìåòèë-D-àñïàðòàòíûå ðåöåïòîðû
  • NNT — ÷èñëî áîëüíûõ, êîòîðûõ íåîáõîäèìî ïðîëå÷èòü, ÷òîáû äîñòè÷ü êàêîãî-òî óêàçàííîãî ýôôåêòà (Number Need to Treatment)
  • OR — îòíîøåíèå øàíñîâ (odds ratio)
  • paCO2 — ïàðöèàëüíîå äàâëåíèå óãëåêèñëîãî ãàçà â àðòåðèàëüíîé êðîâè
  • PALS — êóðñû ðàñøèðåííûõ ðåàíèìàöèîííûõ ìåðîïðèÿòèé â ïåäèàòðèè (Pediatric Advanced Life Support)
  • PANDAS — àóòîèììóííîå íåéðîïñèõèàòðè÷åñêîå ðàññòðîéñòâî â äåòñêîì âîçðàñòå, àññîöèèðîâàííîå ñî ñòðåïòîêîêêîâîé èíôåêöèåé (Pediatric autoimmune neuropsychiatric disorder associated with streptococcal infection)
  • PANS — îñòðûé íåéðîïñèõèàòðè÷åñêèé ñèíäðîì â äåòñêîì âîçðàñòå (Pediatric acute-onset neuropsychiatric syndrome)
  • paO2 — ïàðöèàëüíîå äàâëåíèå êèñëîðîäà â àðòåðèàëüíîé êðîâè
  • PAS — ôóêñèíñåðíèñòàÿ êèñëîòà, ðåàêòèâ Øèôôà (periodic acid-Shiff)
  • PC — ïîñëå åäû (ïðè íàçíà÷åíèÿõ) — post cibum ðÑO2 — ïàðöèàëüíîå äàâëåíèå óãëåêèñëîãî ãàçà
  • PCSK-9 — ïðîïðîòåèíîâàÿ êîíâåðòàçà ñóáòèëèçèí-êåêñè-íîâîãî òèïà 9 (proprotein convertase subtilisin/ kexin type 9)
  • PFAPA — ïåðèîäè÷åñêàÿ ëèõîðàäêà ñ àôòîçíûì ñòîìàòèòîì, ôàðèíãèòîì è ëèìôàäåíèòîì (Periodic Fevers with Aphthous stomatitis, Pharyngitis and Adenitis)
  • PFIC — ñåìåéíûé âíóòðèïå÷åíî÷íûé õîëåñòàç (familial intrahepatic cholestasis)
  • Pg — ïðîñòàãëàíäèí
  • pH — âîäîðîäíûé ïîêàçàòåëü
  • PIM — ïîêàçàòåëü ëåòàëüíîãî èñõîäà ó äåòåé (Pediatric Index of Mortality)
  • piO2 — ïàðöèàëüíîå äàâëåíèå êèñëîðîäà âî âäûõàåìîì
  • âîçäóõå
  • ðO2 — ïàðöèàëüíîå äàâëåíèå êèñëîðîäà
  • POLG — ñóáúåäèíèöà ÄÍÊ-ïîëèìåðàçû ó
  • PRISA II — îöåíêà ðèñêà ãîñïèòàëèçàöèè ó äåòåé II (Pediatric Risk of Admission)
  • PRISM — ðèñê ëåòàëüíîãî èñõîäà ó äåòåé (Pediatric Risk of Mortality)
  • PRN — ïðè (ïî) íåîáõîäèìîñòè (Pro re nata — ïðè âîçíèêíîâåíèè îáñòîÿòåëüñòâ)
  • PRSS — ãåí ñåðèíîâîé ïðîòåàçû
  • PS. — ïðèìå÷àíèå (Post scriptum)
  • PUVA-òåðàïèÿ — ïñîðàëåí-óëüòðàôèîëåò À-òåðàïèÿ (psoralen and ultraviolet À)
  • Q#H — êàæäûå # ÷àñîâ (ïðè íàçíà÷åíèÿõ) — quaque ... hora
  • QAM — êàæäîå óòðî (ïðè íàçíà÷åíèÿõ) — quaque ante meridiem
  • QPM — êàæäûé âå÷åð (ïðè íàçíà÷åíèÿõ) — quaque post meridiem
  • RAG — ãåí, àêòèâèðóþùèé ðåêîìáèíàçó
  • RePEAT— ïåðåñìîòðåííûé èíñòðóìåíò äëÿ îöåíêè ïåäèàòðè÷åñêîé íåîòëîæíîé ìåäèöèíñêîé ïîìîùè (Revised Pediatric Emergency Assessment Tool)
  • RF — ðåâìàòîèäíûé ôàêòîð
  • Rh — ðåçóñ(-ôàêòîð)
  • Rh«-» — ðåçóñ-îòðèöàòåëüí(ûé)
  • Rh«+» — ðåçóñ-ïîëîæèòåëüí(ûé)
  • ROHHAD — áûñòðî ðàçâèâàþùååñÿ îæèðåíèå ñ äèñôóíêöèåé ãèïîòàëàìóñà, ãèïîâåíòèëÿöèåé è ñïîíòàííîé äèñðåãóëÿöèåé (rapid-onset obesity with hypothalamic dysfunction, hypoventilation and autonomic dysregulation)
  • RR — îòíîñèòåëüíûé ðèñê (relative risk èëè risk ratio) RW — ðåàêöèÿ Âàññåðìàíà (reaction of Wassermann)
  • SaO2 — ñàòóðàöèÿ àðòåðèàëüíîé êðîâè êèñëîðîäîì SatO2 — íàñûùåíèå êðîâè êèñëîðîäîì
  • ScvO2 — íàñûùåíèå êèñëîðîäîì öåíòðàëüíîé âåíîçíîé êðîâè (Central venous oxygen saturation)
  • SD — ñòàíäàðòíîå îòêëîíåíèå (standart deviation) SFTPA — ãåí ñóðôàêòàíòíîãî áåëêà A
  • SFTPB — ãåí ñóðôàêòàíòíîãî áåëêà Â
  • SFTPC — ãåí ñóðôàêòàíòíîãî áåëêà Ñ
  • SPINK — èíãèáèòîð ñåðèíîâîé ïðîòåàçû
  • SpO2 — íàñûùåíèå (ñàòóðàöèÿ) ãåìîãëîáèíà êèñëîðîäîì spp. — âèäû (ïðè ðîäîâîì èìåíè ìèêðîîðãàíèçìîâ)
  • Src. — èñòî÷íèê èíôîðìàöèè, áèáëèîãðàôè÷åñêàÿ ññûëêà (source)
  • STAT1 — ñèãíàëüíûé ïðåîáðàçîâàòåëü è àêòèâàòîð òðàíñêðèïöèè 1 (signal transducer and activator of transcription)
  • TA — ìåæäóíàðîäíàÿ àíàòîìè÷åñêàÿ òåðìèíîëîãèÿ TCR — T-êëåòî÷íûé ðåöåïòîð (T-cell receptor)
  • TLRs — Òîëë-ïîäîáíûå ðåöåïòîðû (Toll-like receptors) TNF — ôàêòîð íåêðîçà îïóõîëè (tumor necrosis factor)
  • TORCH — òîêñîïëàçìîç, êðàñíóõà, öèòîìåãàëîâèðóñíàÿ èíôåêöèÿ, ãåðïåñ è äðóãèå èíôåêöèè (Toxoplasmosis, Other infections, Rubella, Cytomegalovirus Herpes simplex)
  • TPM — ìóòàöèÿ òðîïîìèîçèíà (mutation of the tropomyosin)
  • TRAP — ñèíäðîì îáðàòíîé àðòåðèàëüíîé ïåðôóçèè áëèçíåöîâ (twin reversed arterial perfusion)
  • TRAPS — ïåðèîäè÷åñêèé ñèíäðîì, àññîöèèðîâàííûé ñ ðåöåïòîðîì ôàêòîðà íåêðîçà îïóõîëè (Tumor Necrosis Factor Receptor-Associated Periodic Syndrome)
  • ™ — òîðãîâàÿ ìàðêà
  • URL — èíòåðíåò-ññûëêà, àäðåñ èíòåðíåò-ðåñóðñà (Uniform Resource Locator)
  • VEGF — ôàêòîð ðîñòà ýíäîòåëèÿ ñîñóäîâ (Vascular Endothelial Growth Factor)
  • WPW — ñèíäðîì Âîëüôà-Ïàðêèíñîíà-Óàéòà (Wolff-Parkinson-White)
  • XLA — Õ-ñöåïëåííàÿ àãàììàãëîáóëèíåìèÿ (X-linked agammaglobulinemia)
  • aDG-RD — äèñòðîôèè, ñâÿçàííûå ñ a-äèñòðîãëèêàíàìè (alpha dystroglycan-related dystrophies)
  • β-ÕÃ× — β-ñóáúåäèíèöà õîðèîíè÷åñêîãî ãîíàäîòðîïèíà ÷åëîâåêà

 õîäå ïîäãîòîâêè ñòàòåé ïî äåòñêîé íåâðîëîãèè â ïåäèàòðèè äëÿ ïîëüçîâàòåëåé ñàéòà ÌåäÓíèâåð èñïîëüçîâàíû òðóäû ñëåäóþùèõ àâòîðîâ:

  1. Avery RA, Shah SS, Licht DJ, et al: Reference range for cerebrospinal fluid opening pressure in children, N Engl J Med 363:891–893, 2010.
  2. Barkovich JA, Raybaud C: Pediatric neuroimaging, ed 5 rev, Philadelphia, 2011, Lippincott Williams & Wilkins.
  3. Beitzke D, Simbrunner J, Riccabona M: MRI in paediatric hypoxic-ischemic disease, metabolic disorders, and malformations—a review, Eur J Radiol 68:199–213, 2008.
  4. Campbell WW: DeJong’s the neurologic examination, ed 7, Philadelphia, 2012, Lippincott Williams & Wilkins.
  5. Chiappa KH: Evoked potentials in clinical medicine, ed 3, Philadelphia, 1997, Lippincott–Raven.
  6. DeRoos ST, Chillag KL, Keeler M, et al: Effects of sleep deprivation on the pediatric electroencephalogram, Pediatrics 123:703–708, 2009.
  7. Jan MMS: Neurological examination of difficult and poorly cooperative children, J Child Neurol 22:1209–1213, 2007.
  8. Moeschler JB, Shevell M: Committee on Genetics: comprehensive evaluation of the child with intellectual disability or global developmental delays, Pediatrics 134:e903–e918, 2014.
  9. Portnoy JM, Olson LC: Normal cerebrospinal fluid values in children: another look, Pediatrics 75:484–487, 1985.
  10. Aradhya S, Manning MA, Splendore A, et al: Whole-genome array-CGH identifies novel contiguous gene deletions and duplications associated with developmental delay, mental retardation, and dysmorphic features, Am J Med Genet 143A:1431–1441, 2007.
  11. Stothard J, Tennant PW, Bell R, et al: Maternal overweight and obesity and the risk of congenital anomalies: a systematic review and meta-analysis, JAMA 301:636–650, 2009.
  12. Adzick NS, Thom EA, Spong CY, et al: A randomized trial of prenatal versus postnatal repair of myelomeningocele, N Engl J Med 364:993–1004, 2011.
  13. Arth A, Kancherla V, Pachon H, et al: A 2015 global update on folic acid-preventable spina bifida and anencephaly, Birth Defects Res A Clin Mol Teratol 106(7):520–529, 2016.
  14. Bauer SB: Neurogenic bladder: etiology and assessment, Pediatr Nephrol 23:541–551, 2008.
  15. Belfort MA, Whitehead WE, Shamshirsaz AA, et al: Fetoscopic open neural tube defect repair, Obstet Gynecol 129(4):734–743, 2017.
  16. Bitsko RH, Reefhuis J, Romitti PA, et al: Periconceptional consumption of vitamins containing folic acid and risk for multiple congenital anomalies, Am J Med Genet 143A:2397–2405, 2007.
  17. Cameron M, Moran P: Prenatal screening and diagnosis of neural tube defects, Prenat Diagn 29:402–411, 2009.
  18. Clarke R, Bennett D: Folate and prevention of neural tube defects, BMJ 349:g4810, 2014.
  19. Cochrane DD: Cord untethering for lipomyelomeningocele: expectation after surgery, Neurosurg Focus 23:1–7, 2007.
  20. Dicianno BE, Kurowski BG, Yang JM, et al: Rehabilitation and medical management of the adult with spina bifida, Am J Phys Med Rehabil 87:1027–1050, 2008.
  21. Guggisberg D, Hadj-Rabia S, Viney C, et al: Skin markers of occult spinal dysraphism in children, Arch Dermatol 140:1109–1115, 2004.
  22. Heuer GG, Moldenhauer JS, Scott Adzick N: Prenatal surgery for myelomeningocele: review of the literature and future directions, Childs Nerv Syst 33(7):1149–1155, 2017.
  23. Ickowicz V, Ewin D, Maugay-Laulom B, et al: Meckel-gruber syndrome, sonography and pathology, Ultrasound Obstet Gynecol 27:296–300, 2006.
  24. Kahn L, Biro EE, Smith RD, et al: Spina bifida occulta and aperta: a review of current treatment paradigms, J Neurosurg Sci 59(1):79–90, 2015.
  25. Kancherla V, Walani SR, Weakland AP, et al: Scorecard for spina bifida research, prevention, and policy—a development process, Prev Med 99:13–20, 2017.
  26. Madden-Fuentes RJ, McNamara ER, Lloyd JC, et al: Variation in definitions of urinary tract infections in spina bifida patients: a systematic review, Pediatrics 132:132–139, 2013.
  27. Nagaraj UD, Bierbrauer KS, Zhang B, et al: Hindbrain herniation in chiari II malformation on fetal and postnatal MRI, AJNR Am J Neuroradiol 38(5):1031–1036, 2017.
  28. O’Neill BR, Gallegos D, Herron A, et al: Use of magnetic resonance imaging to detect occult spinal dysraphism in infants, J Neurosurg Pediatr 19(2):217–226, 2017.
  29. Peranteau WH, Adzick S: Prenatal surgery for myelomeningocele, Curr Opin Obstet Gynecol 28:111–118, 2016.
  30. Scales CD, Wiener JS: Evaluating outcomes of enterocystoplasty in patients with spina bifida: a review of the literature, J Urol 180:2323–2329, 2008.
  31. Sepulveda W, Wong AE, Sepulveda F, et al: Prenatal diagnosis of spina bifida: from intracranial translucency to intrauterine surgery, Childs Nerv Syst 33(7):1083–1099, 2017.
  32. Shin M, Kucik JE, Siffel C, et al: Improved survival among children with spina bifida in the United States, J Pediatr 161:1132–1137, 2012.
  33. Tubbs RS, Bui CJ, Loukas M, et al: The horizontal sacrum as an indicator of the tethered spinal cord in spina bifida aperta and occulta, Neurosurg Focus 23:1–4, 2007.
  34. U.S. Preventive Services Task Force: Recommendation statement: folic acid for the prevention of neural tube defects, Ann Intern Med 150:626–631, 2009.
  35. Urrutia J, Cuellar J, Zamora T: Spondylolysis and spina bifida occulta in pediatric patients: prevalence study using computed tomography as a screening method, Eur Spine J 25(2):590–595, 2016.
  36. Yu J, Maheshwari M, Foy AB, et al: Neonatal lumbosacral ulceration masking lumbosacral and intraspinal hemangiomas associated with occult spinal dysraphism, J Pediat 175:211–215, 2016.
  37. Ware AL, Kulesz PA, Juranek J, et al: Cognitive control and associated neural correlates in adults with spina bifida myelomeningocele, Neuropsychology 31(4):411–423, 2017.
  38. Williams H: Spinal sinuses, dimples, pits and patches: what lies beneath? Arch Dis Child 91:ep75–ep80, 2006.
  39. France D, Alonso N, Ruas R, et al: Transsphenoidal meningoencephalocele associated with cleft lip and palate: challenges for diagnosis and surgical treatment, Childs Nerv Syst 25:1455–1458, 2009.
  40. Joe JG, Papp Z, Berkes E, et al: Non-syndromic encephalocele: a 26-year experience, Dev Med Child Neurol 50:958–960, 2008.
  41. Leitch CC, Zaghloul NA, Davis EE, et al: Hypomorphic mutations in syndromic encephalocele genes are associated with Bardet-biedl syndrome, Nat Genet 40: 443–448, 2008.
  42. Ramdurg SR, Sukanya M, Maitra J: Pediatric encephaloceles: a series of 20 cases over a period of 3 years, J Pediatr Neurosci. 10(4):317–320, 2015.
  43. Yucetas SC, U?ler N: A retrospective analysis of neonatal encephalocele predisposing factors and outcomes, Pediatr Neurosurg 52(2):73–76, 2017.
  44. Abdel Razek AAK, Kandell AY, Elsorogy LG, et al: Disorders of cortical formation: MR imaging features, AJNR Am J Neuroradiol 30:4–11, 2009.
  45. Almgren M, Schalling M, Lavebratt C: Idiopathic megalencephaly—possible cause and treatment opportunities: from patient to lab, Eur J Paediatr Neurol 12:438–445, 2008.
  46. Breedveld G, de Coo IF, Lequin MH, et al: Novel mutations in three families confirm a major role of COL4A1 in hereditary porencephaly, J Med Genet 43:490–495, 2006.
  47. Cossu M, Pelliccia V, Gozzo F, et al: Surgical treatment of polymicrogyria-related epilepsy, Epilepsia 57(12):2001–2010, 2016.
  48. Gould DB, Phalan FC, Breedveld GJ, et al: Mutations in col4a1 cause perinatal cerebral hemorrhage and porencephaly, Science 308:1167–1171, 2005.
  49. Guerrini R, Dobyns WB, Barkovich AJ: Abnormal development of the human cerebral cortex: genetics, functional consequences and treatment options, Trends Neurosci 31:154–162, 2008.
  50. Hayashi N, Tsutsumi Y, Barkovich AJ: Polymicrogyria without porencephaly/schizencephaly. MRI analysis of the spectrum and the prevalence of macroscopic findings in the clinical population, Neuroradiology 44:647–655, 2002.
  51. Jamuar SS, Lam ATN, Kircher M, et al: Somatic mutations in cerebral cortical malformations, N Engl J Med 371:733–742, 2014.
  52. Kerjan G, Gleeson JG: Genetic mechanisms underlying abnormal neuronal migration in classical lissencephaly, Trends Genet 23:623–630, 2007.
  53. McGovern M, Flanagan O, Lynch B, et al: Novel COL4A2 variant in a large pedigree: consequences and dilemmas, Clin Genet 92(4):447–448, 2017.
  54. Meuwissen ME, Halley DJ, Smit LS, et al: The expanding phenotype of COL4A1 and COL4A2 mutations: clinical data on 13 newly identified families and a review of the literature, Genet Med 17(11):843–853, 2015.
  55. Parrini E, Conti V, Dobyns WB, Guerrini R: Genetic basis of brain malformations, Mol Syndromol. 7(4):220–233, 2016.
  56. Ruggieri M, Pratic? AD: Mosaic neurocutaneous disorders and their causes, Semin Pediatr Neurol 22(4):207–233, 2015.
  57. Sharif U, Kuban K: Prenatal intracranial hemorrhage and neurologic complications in alloimmune thrombocytopenia, J Child Neurol 16:838–842, 2001.
  58. Spalice A, Parisi P, Nicita F, et al: Neuronal migration disorders: clinical, neuroradiologic and genetics aspects, Acta Paediatr 98:421–433, 2009.
  59. Stutterd CA, Leventer RJ: Polymicrogyria: a common and heterogeneous malformation of cortical development, Am J Med Genet (Semin Med Genet) 166C:227–239, 2014.
  60. van der Knaap MS, Smit LME, Barkhof F, et al: Neonatal porencephaly and adult stroke related to mutations in collagen IV A1, Ann Neurol 59:504–511, 2006.
  61. Wynshaw-Boris A: Lissencephaly and LIS1: insights into the molecular mechanisms of neuronal migration and development, Clin Genet 72:296–304, 2007.
  62. Bedeschi MF, Bonaglia MC, Grasso R, et al: Agenesis of the corpus callosum: clinical and genetic study in 63 young patients, Pediatr Neurol 34:186–193, 2006.
  63. Bendavid C, Dubourg C, Gicquel I, et al: Molecular evaluation of foetuses with holoprosencephaly shows high incidence of microdeletions in the HPE genes, Hum Genet 119:1–8, 2006.
  64. Dubourg C, Lazaro L, Pasquier L, et al: Molecular screening of SHH, ZIC2, SIX3, and TGIF genes in patients with features of holoprosencephaly spectrum: mutation review and genotype-phenotype correlations, Hum Mutat 24:43–51, 2004.
  65. Glass HC, Shaw GM, Ma C, et al: Agenesis of the corpus callosum in California 1983–2003: a population-based study, Am J Med Genet 146A:2495–2500, 2008.
  66. Goetzinger KR, Stamilio DM, Dicke JM, et al: Evaluating the incidence and likelihood ratios for chromosomal abnormalities in fetuses with common central nervous system malformations, Am J Obstet Gynecol 199(285):e1–e6, 2008.
  67. Griffiths PD, Batty R, Connolly DA, et al: Effects of failed commissuration on the septum pellucidum and fornix: implications for fetal imaging, Neuroradiology 51:347–356, 2009.
  68. Heide S, Keren B, Billette de Villemeur T, et al: Copy number variations found in patients with a corpus callosum abnormality and intellectual disability, J Pediatr 185:160–166, 2017.
  69. Herman-Sucharska I, Bekiesinska-Figatowska M, Urbanik A: Fetal central nervous system malformations on MR images, Brain Dev 31:185–199, 2009.
  70. Houtmeyers R, Tchouate Gainkam O, Glanville-Jones HA, et al: Zic2 mutation causes holoprosencephaly via disruption of NODAL signaling, Hum Mol Genet 25(18):3946–3959, 2016.
  71. Kerjan G, Gleeson JG: Genetic mechanisms underlying abnormal neuronal migration in classical lissencephaly, Trends Genet 23:623–630, 2007.
  72. Miller SP, Shevell MI, Patenaude Y, et al: Septo-optic dysplasia plus: a spectrum of malformations of cortical development, Neurology 54:1701–1703, 2000.
  73. Moes P, Schilmoeller K, Schilmoeller G: Physical, motor, sensory and developmental features associated with agenesis of the corpus callosum, Child Care Health Dev 35:656–672, 2009.
  74. Mouden C, Dubourg C, Carr? W, et al: Complex mode of inheritance in holoprosencephaly revealed by whole exome sequencing, Clin Genet 89(6):659–668, 2016.
  75. Passos-Bueno MR, et al: Genetics of craniosynostosis: genes, syndromes, mutations and genotype-phenotype correlations, Front Oral Biol 12:107–143, 2008.
  76. Polizzi A, Pavone P, Iannetti P, et al: Septo-optic dysplasia complex: a heterogeneous malformation syndrome, Pediatr Neurol 34:66–71, 2006.
  77. Romaniello R, Marelli S, Giorda R, et al: Clinical characterization, genetics, and long-term follow-up of a large cohort of patients with agenesis of the corpus callosum, J Child Neurol 32(1):60–71, 2017.
  78. Saba L, Anzidel M, Raz E, et al: MR and CT of Brain’s cava, J Neuroimaging 23:326–335, 2013.
  79. Schell-Apacik CC, Wagner K, Bihler M, et al: Agenesis and dysgenesis of the corpus callosum: clinical, genetic and neuroimaging findings in a series of 41 patients, Am J Med Genet 146A:2501–2511, 2008.
  80. Smith T, Tekes A, Boltshauser E, et al: Commissural malformations: beyond the corpus callosum, J Neuroradiol 35:301–303, 2008.
  81. Wegiel J, Flory M, Kaczmarski W, et al: Partial agenesis and hypoplasia of the corpus callosum in idiopathic autism, J Neuropathol Exp Neurol 76(3):225–237, 2017.
  82. Bolduc M, Limperopoulos C: Neurodevelopmental outcomes in children with cerebellar malformations: a systematic review, Dev Med Child Neurol 51:256–267, 2009.
  83. Brancati F, Dallapiccola B, Valente EM: Joubert syndrome and related disorders, Orphanet J Rare Dis 5(20):1–10, 2010.
  84. Huang H, Hwang CW, Lai PH, et al: M?bius syndrome as a syndrome of rhombencephalic maldevelopment: a case report, Pediatr Neonatol 50:36–38, 2009.
  85. Hwang JY, Yoon HK, Lee JH, et al: Cranial nerve disorders in children: MR imaging findings, Radiographics 36(4):1178–1194, 2016.
  86. Jaspan T: New concepts on posterior fossa malformations, Pediatr Radiol 38:S409–S414, 2008.
  87. Khalsa SSS, Siu A, DeFreitas TA, et al: Comparison of posterior fossa volumes and clinical outcomes after decompression of chiari malformation type I, J Neurosurg Pediatr 19(5):511–517, 2017.
  88. Long A, Moran P, Robson S: Outcome of fetal cerebral posterior fossa anomalies, Prenat Diagn 26:707–710, 2006.
  89. Martinez-Lage JF, Guillen-Navarro E, Lopez-Guerrero A, et al: Chiari type 1 anomaly in pseudohypoparathyroidism type ia: pathogenetic hypothesis, Childs Nerv Syst 27:2035–2039, 2011.
  90. Maso AF, Poca MA, de la Calzada MD, et al: Sleep disturbance: a forgotten syndrome in patients with chiari I malformation, Neurologia 29(5):294–304, 2014.
  91. Milhorat TH, Bolognese PA, Nishikawa M, et al: Syndrome of occipitoatlantoaxial hypermobility, cranial settling, and chiari malformation type I in patients with hereditary disorders of connective tissue, J Neurosurg Spine 7:601–609, 2007.
  92. Poretti A, Ashmawy R, Garzon-Muvdi T, et al: Chiari type 1 deformity in children: pathogenetic, Clinical, Neuroimaging, and management aspects, Neuropediatrics 47(5):293–307, 2016.
  93. Rao KI, Hesselink J, Trauner DA: Chiari I malformation in nephropathic cystinosis, J Pediatr 167:1126–1129, 2015.
  94. Raza-Knight S, Mankad K, Prabhakar P, et al: Headache outcomes in children undergoing foramen magnum decompression for chiari I malformation, Arch Dis Child 102(3):238–243, 2017.
  95. Romani M, Micalizzi A, Valente EM: Joubert syndrome: congenital cerebellar ataxia with the molar tooth, Lancet Neurol 12:894–905, 2013.
  96. Sasaki-Adams D, Elbabaa SK, Jewells V, et al: The Dandy-walker variant: a case series of 24 pediatric patients and evaluation of associated anomalies, incidence of hydrocephalus, and developmental outcomes, J Neurosurg Pediatr 2:194–199, 2008.
  97. Traboulsi EI: Congenital abnormalities of cranial nerve development: overview, molecular mechanisms, and further evidence of heterogeneity and complexity of syndromes with congenital limitation of eye movements, Trans Am Ophthalmol Soc 102:373–389, 2004.
  98. Vandertop WP: Syringomyelia, Neuropediatrics 45:3–9, 2013.
  99. Abuelo D: Microcephaly syndromes, Semin Pediatr Neurol 14:118–127, 2007.
  100. Auger N, Quach C, Healy-Profitos J, et al: Congenital microencephaly in Quebec: baseline prevalence, risk factors and outcomes in a large cohort of neonates, Arch Dis Child Fetal Neonatal Ed 103(3):F167–F172, 2017.
  101. Barkovich AJ, Kuzniecky RI, Jackson MD, et al: Classification system for malformations of cortical development, Neurology 57:2168–2178, 2001.
  102. Clark GD: The classification of cortical dysplasias through molecular genetics, Brain Dev 26:351–362, 2004.
  103. Denis D, Chateil JF, Brun M, et al: Schizencephaly: clinical and imaging features in 30 infantile cases, Brain Dev 22:475–483, 2000.
  104. D’Orsi G, Tinuper P, Bisulli F, et al: Clinical features and long term outcome of epilepsy in periventricular nodular heterotopia. Simple compared with plus forms, J Neurol Neurosurg Psychiatry 75:873–878, 2004.
  105. Hayashi N, Tsutsumi Y, Barkovich AJ: Morphological features and associated anomalies of schizencephaly in the clinical population: detailed analysis of MR images, Neuroradiology 44:418–427, 2002.
  106. Kinsman SL, Plawner LL, Hahn JS: Holoprosencephaly: recent advances and insights, Curr Opin Neurol 13:127–132, 2000.
  107. Parrish ML, Roessmann U, Levinsohn MW: Agenesis of the corpus callosum: a study of the frequency of associated malformations, Ann Neurol 6:349–354, 1979.
  108. Richards LJ, Plachez C, Ren T: Mechanisms regulating the development of the corpus callosum and its agenesis in mouse and human, Clin Genet 66:276–289, 2004.
  109. Silva AA, Barbieri MA, Alves MT, et al: Prevalence and risk factors for microcephaly at birth in Brazil in, Pediatrics 141(2):e20170589–e20172018, 2010.
  110. Woods CG, Bond J, Enard W: Autosomal recessive primary microcephaly (MCPH): a review of clinical, molecular, and evolutionary findings, Am J Hum Genet 76:717–728, 2005.
  111. Acakpo-Satchivi L, Shannon CN, Tubbs RS, et al: Death in shunted hydrocephalic children: a follow-up study, Childs Nerv Syst 24:197–201, 2008.
  112. Chern JJ, Macias CG, Jea A, et al: Effectiveness of a clinical pathway for patients with cerebrospinal fluid shunt malfunction, J Neurosurg Pediatr 6:318–324, 2010.
  113. Drake JM: The surgical management of pediatric hydrocephalus, Neurosurgery 62(Suppl 2):633–640, 2008.
  114. Greene M, Benacerraf B, Crawford J: Hydranencephaly: US appearance during in utero evolution, Radiology 156:779–780, 1985.
  115. Halperin JJ, Kurlan R, Schwalb JM, et al: Practice guideline: idiopathic normal pressure hydrocephalus: response to shunting and predictors of response, Neurology 85:2063–2071, 2015.
  116. Kahle KT, Kulkarni AV, Limbick DD Jr, Warf BC: Hydrocephalus in children, Lancet 387:788–798, 2016.
  117. Kandasamy J, Jenkinson MD, Mallucci CL: Contemporary management and recent advances in paediatric hydrocephalus, BMJ 343:d4191, 2011.
  118. Novegno F, Caldarelli M, Massa A, et al: The natural history of the chiari type I anomaly, J Neurosurg Pediatr 2:179–187, 2008.
  119. Olney AH: Macrocephaly syndromes, Semin Pediatr Neurol 14:128–135, 2007.
  120. Parisi MA, Dobyns WB: Human malformations of the midbrain and hindbrain: review and proposed classification scheme, Mol Genet Metab 80:36–53, 2003.
  121. Pershad J, Taylor A, Hall K, et al: Imaging strategies for suspected acute cranial shunt failure: a cost-effectiveness analysis, Pediatrics 140(2):e20164263, 2017.
  122. Persson E, Anderson S, Wiklund L, et al: Hydrocephalus in children born in 1999–2002: epidemiology, outcome and ophthalmological findings, Childs Nerv Syst 23:1111–1118, 2007.
  123. Sacko O, Boetto S, Lauwers-Cances V, et al: Endoscopic third ventriculostomy: overcome analysis in 368 procedures, J Neurosurg Pediatr 5:68–74, 2010.
  124. Stevenson KL: Chiari type II malformation: past, present, and future, Neurosurg Focus 16:E5, 2004.
  125. Tully HM, Dobyns WB: Infantile hydrocephalus: a review of epidemiology, classification and causes, Eur J Med genetics 57:359–368, 2014.
  126. Tzekov C, Naydenov E, Kalev O: Ependymoma of the cauda equina starting with communicating hydrocephalus: a case report, Pediatr Neurosurg 43:399–402, 2007.
  127. Van Landingham M, Nguyen TV, Roberts A, et al: Risk factors of congenital hydrocephalus: a 10-year retrospective study, J Neurol Neurosurg Psychiatry 80:213–217, 2009.
  128. Wilson MP, Aronyk KE, Yeo T, et al: Communicating hydrocephalus caused by an unruptured perimedullary arteriovenous fistula in the lumbar region of an infant, J Neurosurg Pediatr 11:346–349, 2013.
  129. Wright Z, Larrew TW, Eskandari R: Pediatric hydrocephalus: current state of diagnosis and treatment, Pediatr Rev 37(11):478–490, 2016.
  130. Boulet SL, Rasmussen SA, Honein MA: A population-based study of craniosynostosis in metropolitan Atlanta, 1989–2003, Am J Med Genet A 146:984–991, 2008.
  131. Bristol RE, Lekovic GP, Rekate HL: The effects of craniosynostosis on the brain with respect to intracranial pressure, Semin Pediatr Neurol 11:262–267, 2004.
  132. Losee JE, Mason AC: Deformational plagiocephaly: diagnosis, prevention, and treatment, Clin Plast Surg 32:53–64, 2005.
  133. McKinney CM, Cunningham ML, Holt VL, et al: Characteristics of 2733 cases diagnosed with deformational plagiocephaly and changes in risk factors over time, Cleft Palate Craniofac J 45:208–216, 2008.
  134. Passos-Bueno MR, Serti Eacute AE, Jehee FS, et al: Genetics of craniosynostosis: genes, syndromes, mutations and genotype-phenotype correlations, Front Oral Biol 12:107–143, 2008.
  135. Rasmussen SA, Yazdy MM, Fr?as JL, et al: Priorities for public health research on craniosynostosis: summary and recommendations from a centers for disease control and Prevention–sponsored meeting, Am J Med Genet A 146:149–158, 2008.
  136. Speltz ML, Kapp-Simon KA, Cunningham M, et al: Single-suture craniosynostosis: a review of neurobehavioral research and theory, J Pediatr Psychol 29:651–668, 2004.
  137. Starr JR, Collett BR, Gaither R, et al: Multicenter study of neurodevelopment in 3-year-old children with and without single-suture craniosynostosis, Arch Pediatr Adolesc Med 166:536–542, 2012.
  138. Van der Meulen J, van der Hulst R, van Adrichem L, et al: The increase of metopic synostosis: a pan-European observation, J Craniofac Surg 20:283–286, 2009.
  139. Wilkie AOM, Johnson D, Wall SA: Clinical genetics of craniosynostosis, Curr Opin Pediatr 29:622–628, 2017.
  140. American Academy of Pediatrics: American Academy of pediatrics task force on infant positioning and SIDS: positioning and SIDS, Pediatrics 89(6 Pt 1):1120–1126, 1992.
  141. Baird LC, et al: Guidelines: congress of neurological surgeons systematic review and Evidence-based guideline for the management of patients with positional plagiocephaly: the role of physical therapy, Neurosurgery 79:E630–E631, 2016.
  142. Bialocerkowski AE, Vladusic SL, Wei Ng C: Prevalence, risk factors, and natural history of positional plagiocephaly: a systematic review, Dev Med Child Neurol 50(8):577–586, 2008.
  143. Collett BR, Gray KE, Starr JR, et al: Development at age 36 months in children with deformational plagiocephaly, Pediatrics 131:e109–e113, 2013.
  144. Collett BR: Helmet therapy for positional plagiocephaly and brachycephaly, BMJ 348:g2906, 2014.
  145. Cummings C: Positional plagiocephaly, Paediatr Child Health 16(8):493–494, 2011.
  146. Flannery AM, Tamber MS, Mazzola C, et al: Congress of neurological surgeons systematic review and evidence-based guidelines for the management of patients with positional plagiocephaly: executive summary, Neurosurgery 79(5):623–624, 2016.
  147. Gatrad AR, Solanki GA, Sheikh A: Baby with an abnormal head, BMJ 348:f7609, 2014.
  148. Glasgow TS, Siddiqi F, Hoff C, et al: Deformational plagiocephaly: development of an objective measure and determination of its prevalence in primary care, J Craniofac Surg 18(1):85–92, 2007.
  149. Huang MH, Gruss JS, Clarren SK, et al: The differential diagnosis of posterior plagiocephaly: true lambdoid synostosis versus positional molding, Plast Reconstr Surg 98(5):765–774, 1996.
  150. Klimo P Jr, et al: Congress of neurological surgeons systematic review and Evidencebased guideline on the management of patients with positional plagiocephaly: the role of repositioning, Neurosurgery 79:E627–E629, 2016.
  151. Kluba S, Kraut W, Renert S, et al: What is the optimal time to start helmet therapy in positional plagiocephaly?, Plast Reconstr Surg 128(2):492–498, 2011.
  152. Laughlin J, Lueerssen TG, Dias MS, et al: Prevention and management of positional skull deformities in infants, Pediatrics 128:1236–1241, 2011.
  153. Lee RP, Teichgraeber JF, Baumgartner JE, et al: Long-term treatment effectiveness of molding helmet therapy in the correction of posterior deformational plagiocephaly: a five-year follow-up, Cleft Palate Craniofac J 45(3):240–245, 2008.
  154. Lipira AB, Gordon S, Darvann TA, et al: Helmet versus active repositioning for plagiocephaly: a three-dimensional analysis, Pediatrics 126(4):e936–e945, 2010.
  155. Looman WS, Flannery AB: Evidence-based care of the child with deformational plagiocephaly, part i: assessment and diagnosis, J Pediatr Health Care 26(4):242–250, 2012.
  156. Lutterodt CG, Sadri A, Eccles S: Effectiveness of conservative therapy and helmet therapy for positional cranial deformation, Plast Reconstr Surg 136:852e–853e, 2015.
  157. Mawji A, Vollman AR, Hatfield J, et al: The incidence of positional plagiocephaly: a cohort study, Pediatrics 132:298–304, 2013.
  158. Mortenson P, Steinbok P, Smith D: Deformational plagiocephaly and orthotic treatment: indications and limitations, Childs Nerv Syst 28(9):1407–1412, 2012.
  159. Nuysink J, Eijsermans MJC, van Haastert IC, et al: Clinical course of asymmetric motor performance and deformational plagiocephaly in very preterm infants, J Pediatr 163:658–665, 2013.
  160. Rogers GF, Oh AK, Mulliken JB: The role of congenital muscular torticollis in the development of deformational plagiocephaly, Plast Reconstr Surg 123(2):643–652, 2009.
  161. Schaaf H, Malik CY, Streckbein P, et al: Three-dimensional photographic analysis of outcome after helmet treatment of a nonsynostotic cranial deformity, J Craniofac Surg 21(6):1677–1682, 2010.
  162. Shamji MF, Fric-Shamji EC, Merchant P, et al: Cosmetic and cognitive outcomes of positional plagiocephaly treatment, Clin Invest Med 35(5):E266, 2012.
  163. Tamber MS, et al: Congress of neurological surgeons systematic review and Evidencebased guideline on the role of cranial molding orthosis (Helmet) therapy for patients with positional plagiocephaly, Neurosurgery 79:E632–E633, 2016.
  164. van Vlimmeren LA, et al: The course of skull deformation from birth to 5 years of age: a prospective cohort study, Eur J Pediatr 176:11–21, 2017.
  165. Van Wijk RM, van Vlimmerem LA, Groothuis-Oudshoorn CGM, et al: Helmet therapy in infants with positional deformation: randomized controlled trial, BMJ 348:g2741, 2014.
  166. Weissler EH, Sherif RD, Taub PJ: An Evidence-based approach to nonsynostotic plagiocephaly, Plast Reconstr Surg 138:682e–689e, 2016.
  167. White N, Warner RM, Noons P, et al: Changing referral patterns to a designated craniofacial centre over a four-year period, J Plast Reconstr Aesthet Surg 63(6):921–925, 2010.
  168. Wilbrand JF, Schmidtberg K, Bierther U, et al: Clinical classification of infant nonsynostotic cranial deformity, J Pediatr 161:1120–1125, 2012.
  169. Wilbrand JF, Seidl M, Wilbran M, et al: A prospective randomized trial on preventative methods for positional head deformity: physiotherapy versus a positioning pillow, J Pediatr 162:1215–1221, 2013.
  170. Yoo H, Rah DK, Kim YO: Outcome analysis of cranial molding therapy in nonsynostotic plagiocephaly, Arch Plast Surg 39(4):338–344, 2012.
  171. American Academy of Pediatrics: Clinical practice guideline for the long-term management of the child with simple febrile seizures, Pediatrics 121:1281–1286, 2008.
  172. American Academy of Pediatrics: Clinical practice guideline—febrile seizures: guideline for the neurodiagnostic evaluation of the child with a simple febrile seizure, Pediatrics 127:389, 2011.
  173. Chen SY, Tsai CN, Lai MW, et al: Norovirus infection as a cause of diarrhea-associated benign infantile seizures, Clin Infect Dis 48:849–854, 2009.
  174. Dayan PS, Lillis K, Bennett J, et al: Prevalence of and risk factors for intracranial abnormalities in unprovoked seizures, Pediatrics 136:e351–e360, 2015.
  175. Fisher RS, Acevedo C, Arzimanoglou A, et al: A practical clinical definition of epilepsy, Epilepsia 55(4):475–482, 2014.
  176. Fisher RS, Cross JH, French JA, et al: Operational classification of seizure types by the international league against epilepsy: position paper of the ILAE commission for classification and terminology, Epilepsia 58(4):522–530, 2017.
  177. Gallentine WB, Shinnar S, Hesdorffer DC, et al: Plasma cytokines associated with febrile status epilepticus in children: a potential biomarker for acute hippocampal injury, Epilepsia 58(6):1102–1111, 2017.
  178. Graves RC, Oehler K, Tingle LE: Febrile seizures: risks, evaluation, and prognosis, Am Fam Physician 85(2):149–153, 2012.
  179. Howell KB, Katanyuwong K, Mackay MT: Long-term follow-up of febrile infectionrelated epilepsy syndrome, Epilepsia 53(1):101–110, 2012.
  180. Lewis DV, Shinnar S, Hesdorffer DC, et al: Hippocampal sclerosis after febrile status epilepticus: the FEBSTAT study, Ann Neurol 75(2):178–185, 2014.
  181. McTague A, Howell KB, Cross JH, et al: The genetic landscape of the epileptic encephalopathies of infancy and childhood, Lancet 15:304–316, 2016.
  182. Offringa M, Newton R, Cozijnsen MA, Nevitt SJ: Prophylactic drug management for febrile seizures in children, Cochrane Database Syst Rev (2):CD003031, 2017.
  183. Oluwabusi T, Sood SK: Update on the management of simple febrile seizures: emphasis on minimal intervention, Curr Opin Pediatr 24:259–265, 2012.
  184. Patel AD, Vidaurre J: Complex febrile seizures: a practical guide to evaluation and treatment, J Child Neurol 28:762–767, 2013.
  185. Scheffer IE, Berkovic S, Capovilla G, et al: ILAE classification of the epilepsies: position paper of the ILAE commission for classification and terminology, Epilepsia 58(4):512–521, 2017.
  186. Whelan H, Harmelink M, Chou E, et al: Complex febrile seizures—a systematic review, Disease Monthly 63(1):5–23, 2017.
  187. Wilden JA, Cohen-Gadol AA: Evaluation of first nonfebrile seizures, Am Fam Physician 86(4):334–340, 2012.
  188. AAN summary of evidence-based guideline for clinicians: evaluating an apparent unprovoked first seizure in adults, Continuum (Minneap Minn) 16(3):255–256, 2010.
  189. Alam S, Lux AL: Epilepsies in infancy, Arch Dis Child 97:985–992, 2012.
  190. Hess CP, Barkovich AJ: Seizures: emergency neuroimaging, Neuroimaging Clin North Am 20:619–637, 2010.
  191. Kullmann DM: Neurological channelopathies, Annu Rev Neurosci 33:151–172, 2010.
  192. Kurian MA, Gissen P, Smith M, et al: The monoamine neurotransmitter disorders: an expanding range of neurological syndromes, Lancet Neurol 10:721–733, 2011.
  193. Michelucci R, Pasini E, Riguzzi P, et al: Genetics of epilepsy and relevance to current practice, Curr Neurol Neurosci Rep 12(4):445–455, 2012.
  194. Moseley BD, Wirrell EC, Nickels K, et al: Electrocardiographic and oximetric changes during partial complex and generalized seizures, Epilepsy Res 95:237–245, 2011.
  195. Muthugovindan D, Hartman AL: Pediatric epilepsy syndromes, Neurologist 16:223–237, 2010.
  196. Ong B, Bergin P, Heffernan T, Stuckey S: Transient seizure-related MRI abnormalities, J Neuroimaging 19:301–310, 2009.
  197. Russ SA, Larson K, Halfon N: A national profile of childhood epilepsy and seizure disorder, Pediatrics 129:256–264, 2012.
  198. Sharma S, Prasad AN: Genetic testing of epileptic encephalopathies of infancy: an approach, Can J Neurol Sci 40(1):10–16, 2013.
  199. Blumcke I, Spreafico R, Haaker G, et al: Histopathological findings in brain tissue obtained during epilepsy surgery, N Engl J Med 377(17):1648–1656, 2017.
  200. Sha Z, Sha L, Li W, et al: Exome sequencing identifies SUCO mutations in mesial temporal lobe epilepsy, Neurosci Lett 591:149–154, 2–15, 2015.
  201. Adcock JE, Panayiotopoulos CP: Occipital lobe seizures and epilepsies, J Clin Neurophysiol 29:397–407, 2012.
  202. Crino PB: mTOR: a pathogenic signaling pathway in developmental brain malformations, Trends Mol Med 17(12):734–742, 2011.
  203. Nabbout R: Autoimmune and inflammatory epilepsies, Epilepsia 53(Suppl 4):58–62, 2012.
  204. Parsley LK, Thomas JA: The patient with infantile seizures, Curr Opin Pediatr 23:693–699, 2011.
  205. Pearl PL: Inherited metabolic epilepsies, New York, 2013, Demos Medical Publishing, LLC. Rahman S, Footitt EJ, Varadkar S, et al: Inborn errors of metabolism causing epilepsy, Dev Med Child Neurol 55(1):23–36, 2013.
  206. Pearl PL: Amenable treatable severe pediatric epilepsies, Semin Pediatr Neurol 23(2):158–166, 2016.
  207. Sejvar JJ, Kakooza AM, Foltz JL, et al: Clinical, neurological, and electrophysiological features of nodding syndrome in kitgum, Uganda: an observational case series, Lancet Neurol 12(2):166–174, 2013.
  208. Shao L, Stafstrom CE: Pediatric epileptic encephalopathies: pathophysiology and animal models, Semin Pediatr Neurol 23(2):98–107, 2016.
  209. Shbaorou R, Mikati MA: The expanding clinical spectrum of genetic pediatric epileptic encephalopathies, Semin Pediatr Neurol 23(2):134–142, 2016.
  210. Wright S, Vincent A: Pediatric autoimmune epileptic encephalopathies, J Child Neurol 32(4):418–428, 2017.
  211. Brigo F, Igwe SC: Ethosuximide, sodium valproate or lamotrigine for absence in children and adolescents (review), Cochrane Database Syst Rev (2):CD003032, 2017.
  212. Brigo F, Igwe SC: Antiepileptic drugs for the treatment of infants with severe myoclonic epilepsy (review), Cochrane Database Syst Rev (10):CD010483, 2015.
  213. Brown JWL, Lawn ND, Lee J, et al: When is it safe to return to driving following first-ever seizure?, J Neurol Neurosurg Psychiatry 86:60–64, 2015.
  214. Capovilla G, Kaufman KR, Perucca E, et al: Epilepsy, seizures, physical exercise, and sports: a report from the ILAE task force on sports and epilepsy, Epilepsia 57(1):6–12, 2016.
  215. Devinsky O, Cross JH, Laux L, et al: Trial of cannabidiol for drug-resistant seizures in the dravet syndrome, N Engl J Med 376(21):2011–2020, 2017.
  216. Devinsky O, Patel AD, Cross H, et al: Effect of cannabidiol on drop seizures in the Lennox-gastaut syndrome, N Engl J Med 378(20):1888–1896, 2018.
  217. Dwivedi R, Ramanujam B, Chandra PS, et al: Surgery for drug-resistant epilepsy in children, N Engl J Med 377(17):1639–1646, 2017.
  218. Federal Drug Administration (Safety Alert): Lamictal (lamotrigine): drug safety communication—serious immune system reaction. https://www.fda.gov/safety/ medwatch/safetyinformation/safetyalertsforhumanmedicalproducts/ucm605628.htm.
  219. Glauser T, Ben-Menachem E, Bourgeois B, et al: ILAE subcommission on AED guidelines. Updated ILAE evidence review of antiepileptic drug efficacy and effectiveness as initial monotherapy for epileptic seizures and syndromes, Epilepsia 54(3):551–563, 2013.
  220. Hani AJ, Mikati MA: Current and emerging therapies of severe epileptic encephalopathies, Semin Pediatr Neurol 23(2):180–186, 2016.
  221. Holt RL, Arehart E, Hunanyan A, et al: Pediatric sudden unexpected death in epilepsy: what have we learned from animal and human studies, and can we prevent it?, Semin Pediatr Neurol 23(2):127–133, 2016.
  222. Jobst BC, Cascino GD: Resective epilepsy surgery for drug-resistant focal epilepsy—a review, JAMA 313(3):285–293, 2015.
  223. Kanner AM, Ashman E, Gloss D, et al: Practice guideline update summary: efficacy and tolerability of the new antiepileptic drugs II: treatment-resistant epilepsy, Neurology 91(2):82–90, 2018.
  224. Levy RG, Cooper PN, Giri P: Ketogenic diet and other dietary treatments for epilepsy, Cochrane Database Syst Rev (3):CD001903, 2012.
  225. Maguire MJ, Jackson CF, Marson AG, et al: Treatments for the prevention of sudden unexpected death in epilepsy (SUDEP), Cochrane Database Syst Rev (7):CD011792, 2016.
  226. The Medical Letter: Brivaracetam (Briviact) for epilepsy, Med Lett 58(1499):95–96, 2016.
  227. The Medical Letter: Drugs for epilepsy, Med Lett 59:121–130, 2017.
  228. Mudigoudar B, Weatherspoon S, Wheeless JW: Emerging antiepileptic drugs for severe pediatric epilepsies, Semin Pediatr Neurol 23(2):167–179, 2016.
  229. Muh CR: Current and emerging surgical therapies for severe pediatric epilepsies, Semin Pediatr Neurol 23(2):143–150, 2016.
  230. Nariai H, Duberstein S, Shinner S: Treatment of epileptic encephalopathies: current state of the art, J Clin Neurol 33(1):41–54, 2018.
  231. Nevitt SJ, Marson AG, Weston J, Tudur Smith C: Carbamazepine versus phenytoin monotherapy for epilepsy: an individual participant data review (review), Cochrane Database Sys Rev (2):CD001911, 2017.
  232. Pearl PL: Amenable treatable severe pediatric epilepsies, Semin Pediatr Neurol 23(2):158–166, 2016.
  233. Pellock JM, Carman WJ, Thyagarajan V, et al: Efficacy of antiepileptic drugs in adults predicts efficacy in children: a systematic review, Neurology 79(14):1482, 2012.
  234. Thiele EA, Marsh ED, French JA, et al: Cannabidiol in patients with seizures associated with Lennox-gastaut syndrome (GWPCARE4): a randomized, double-blind, placebo-controlled phase 3 trial, Lancet 391:1085–1096, 2018.
  235. Tomson T, Battino D, Bonizzoni E, et al: Comparative risk of major congenital malformations with eight different antiepileptic drugs: a prospective cohort study of the EURAP registry, Lancet Neurol 17(6):530–538, 2018.
  236. Ulate-Campos A, Coughlin F, Ga?nza-Lein M, et al: Automated seizure detection systems and their effectiveness for each type of seizure, Seizure 40:88–101, 2016.
  237. Zelano J, Kumlien E: Levetiracetam as alternative stage two antiepileptic drug in status epilepticus: a systematic review, Seizure 21:233–236, 2012.
  238. Abend NS, Wusthoff CJ: Neonatal seizures and status epilepticus, J Clin Neurophysiol 29(5):441–448, 2012.
  239. Collins M, Young M: Benign neonatal shudders, shivers, jitteriness, or tremors: early signs of vitamin D deficiency, Pediatrics 140(2):e20160719, 2017.
  240. Donovan MD, Griffin BT, Kharoshankaya L, et al: Pharmacotherapy for neonatal seizures: current knowledge and future perspectives, Drugs 76(6):647–661, 2016.
  241. Ficicioglu C, Bearden D: Isolated neonatal seizures: when to suspect inborn errors of metabolism, Pediatric Neurol 45:283–291, 2011.
  242. Glass HC, Shellhaas RA, Wustoff CJ, et al: Contemporary profile of seizures in neonates: a prospective cohort study, J Pediatr 174:98–103, 2016.
  243. Glass HC: Neonatal seizures: advances in mechanisms and management, Clin Perinatol 41(1):177–190, 2014.
  244. Pisani F, Piccolo B, Cantalupo G, et al: Neonatal seizures and postneonatal epilepsy: a 7-y follow-up study, Pediatr Res 72:186–192, 2011.
  245. Rakshasbhuvankar A, Paul S, Nagarajan L, et al: Amplitude-integrated EEG for detection of neonatal seizures: a systematic review, Seizure 33:90–98, 2015.
  246. Sands TT, McDonough TL: Recent advances in neonatal seizures, Curr Neurol Neurosci Rep 16:92, 2016.
  247. Shetty J: Neonatal seizures in hypoxic–ischaemic encephalopathy—risks and benefits of anticonvulsant therapy, Dev Med Child Neurol 57:40–43, 2015.
  248. World Health Organization: Guidelines on neonatal seizures, Geneva, 2011, WHO.
  249. Appavu BB, Vanatta L, Condie J, et al: Ketogenic diet treatment for pediatric superrefractory status epilepticus, Seizure 41:62–65, 2016.
  250. Brophy GM, Bell R, Claassen J, et al: Neurocritical care society status epilepticus guideline writing committee: guidelines for the evaluation and management of status epilepticus, Neurocrit Care 17(1):3–23, 2012.
  251. Fernandez IS, Abend NS, Arndt DH, et al: Electrographic seizures after convulsive status epilepticus in children and young adults: a retrospective study, J Pediatr 164:339–346, 2014.
  252. Frank LM, Shinnar S, Hesdorffer DC, et al: Cerebrospinal fluid findings in children with fever-associated status epilepticus: results of the febrile status epilepticus (FEBSTAT) study, J Pediatr 161(6):1169–1171, 2012.
  253. Gaspard N, Foreman BP, Alvarez V, et al: New-onset refractory status epilepticus, Neurology 85:1604–1613, 2015.
  254. Glauser T, Shinnar S, Gloss D, et al: Evidence-based guideline: treatment of convulsive status epilepticus in children and adults: report of the guideline committee of the American epilepsy society, Epilepsy Curr 16:48–61, 2016.
  255. Glauser T, Shinnar S, Gloss D, et al: Evidence-based guideline: treatment of convulsive status epilepticus in children and adults: report of the guideline committee of the American epilepsy society, Epilepsy Curr 16(1):48–61, 2016.
  256. Greiner HM, Holland K, Leach JL, et al: Nonconvulsive status epilepticus: the encephalopathic pediatric patient, Pediatrics 129:e748–e755, 2012.
  257. Martinos MM, Yoong M, Patil S, et al: Early developmental outcomes in children following convulsive status epilepticus: a longitudinal study, Epilepsia 54(6):1012–1019, 2013.
  258. Pujar SS, Martinos MM, Cortina-Borja M, et al: Long-term prognosis after childhood convulsive status epilepticus: a prospective cohort study, Lancet Child Adolescent 2:103–110, 2018.
  259. Shorvon S, Ferlisi M: The treatment of super-refractory status epilepticus: a critical review of available therapies and a clinical treatment protocol, Brain 134(10):2802–2818, 2011.
  260. Smith DM, McGinnis EL, Walleigh DJ, et al: Management of status epilepticus in children, J Clin Med 5(4):47, 2016.
  261. Trinka E, Hesdorffer D, Rossetti AO, et al: A definition and classification of status epilepticus—report of the task force on classification of status epilepticus, Epilepsia 56(10):1515–1523, 2015.
  262. Fisher RS, Harding G, Erba G, et al: Photic- and pattern-induced seizures: a review for the epilepsy foundation of America working group, Epilepsia 46:1426–1441, 2005.
  263. Kasteleijn-Nolst Trenite DGA: Provoked and reflex seizures: surprising or common?, Epilepsia 53:105–115, 2012. Ritaccio AL: Reflex seizures, Neurol Clin 12:57–83, 1994.
  264. Boivin MJ, Kakooza AM, Warf BC, et al: Reducing neurodevelopmental disorders and disability through research and interventions, Nature 527:S155–S160, 2015.
  265. Dowell SF, Sejvar JJ, Riek L, et al: Nodding syndrome, Emerg Infect Dis 19:1374–1384, 2013.
  266. Idro R, Opar B, Wamala J, et al: Is nodding syndrome an Onchocerca volvulus-induced neuroinflammatory disorder? Uganda’s story of research in understanding the disease, Int J Infect Dis 45:112–117, 2016.
  267. Idro R, Opoka RO, Aanyu HT, et al: Nodding syndrome in Ugandan children: clinical features, brain imaging and complications: a case series, BMJ Open 3(5):e002540, 2013.
  268. Iyengar PJ, Wamala J, Ratto J, et al: Prevalence of nodding syndrome–uganda, 2012-2013, MMWR Morb Mortal Wkly Rep 63:603–606, 2014.
  269. Johnson TP, Tyagi R, Lee PR, et al: Nodding syndrome may be an autoimmune reaction to the parasitic worm Onchocerca volvulus, Sci Transl Med 9(377):eaaf6953, 2017.
  270. Sejvar JJ, Kakooza AM, Foltz JL, et al: Clinical, neurological, and electrophysiological features of nodding syndrome in kitgum, Uganda: an observational case series, Lancet Neurol 12:166–174, 2013.
  271. Tumwine JK, Vandemaele K, Chungong S, et al: Clinical and epidemiological characteristics of nodding syndrome in mundri county, southern Sudan, Afr Health Sci 12:242–248, 2012.
  272. Winkler AS, Friedrich K, Konig R, et al: The head nodding syndrome–clinical classification and possible causes, Epilepsia 49:2008–2015, 2008.
  273. Winkler AS, Wallner B, Friedrich K, et al: A longitudinal study on nodding syndrome—a new African epilepsy disorder, Epilepsia 55:86–93, 2014.
  274. Winkler AS, Friedrich K, Velicheti S, et al: MRI findings in people with epilepsy and nodding syndrome in an area endemic for onchocerciasis: an observational study, Afr Health Sci 13:529–540, 2013.
  275. Appavu B, Mangum T, Obeid M: Glucose transporter 1 deficiency: a treatable cause of epileptic myoclonus in infancy with protean presentations, Pediatr Neurol 53(4):364–366, 2015.
  276. Babiker MO, Prasad M: Fifteen-minute consultation: when is a seizure not a seizure? Part 2, the older child, Arch Dis Child Educ Pract Ed 100(6):295–300, 2015.
  277. Gardiner AR, Jaffer F, Dale RC, et al: The clinical and genetic heterogeneity of paroxysmal dyskinesias, Brain 138(Pt 12):3567–3580, 2015.
  278. Masoud M, Prange L, Wuchich J, et al: Diagnosis and treatment of aternating hemiplegia of childhood, Curr Treat Options Neurol 19(2):8, 2017.
  279. Prasad M, Babiker MO: Fifteen-minute consultation: when is a seizure not a seizure? Part 1, the younger child, Arch Dis Child Educ Pract Ed 101(1):15–20, 2016.
  280. Sawchuk T, Buchhalter J: Psychogenic nonepileptic seizures in children: psychological presentation, treatment, and short-term outcomes, Epilepsy Behav 52(Pt A):49–56, 2015.
  281. Tieder JS, Bonkowsky JL, Etzel RA, et al: Subcommittee on apparent Life-threatening events: brief resolved unexplained events (formerly apparent Life-threatening events) and evaluation of Lower-risk infants, Pediatrics 137(5):2016.
  282. Abu-Arafeh I: Flunarizine for the prevention of migraine—a new look at an old drug, Dev Med Child Neurol 54:200–207, 2012.
  283. Afridi SK, Giffin NJ, Kaube H, et al: A randomized controlled trial of intranasal ketamine in migraine with prolonged aura, Neurology 80:642–647, 2013.
  284. Ahmed K, Oas K, Mack KJ, et al: Experience with botulinum toxin type A in medically intractable pediatric chronic daily headache, Pediatr Neurol 43(5):316–319, 2010.
  285. Ahonen K, Hamalainen ML, Rantala H, et al: Nasal sumatriptan is effective in treatment of migraine attacks in children: a randomized trial, Neurology 62(6):883–887, 2004.
  286. Anttila V, Stefansson H, Kallela M, et al: Genome-wide association study of migraine implicates a common susceptibility variant on 8q22.1, Nat Genet 42(10):869–873, 2010.
  287. Armstrong AE, Gillan E, DiMario FJ Jr: SMART syndrome (stroke-like migraine attacks after radiation therapy) in adult and pediatric patients, J Child Neurol 29(3):336–341, 2014.
  288. Arruda MA, Bigal ME: Migraine and migraine subtypes in preadolescent children, Neurology 79:1881–1888, 2012.
  289. Avraham SB, Har-Gil M, Watemberg N: Acute confusional migraine in an adolescent: response to intravenous valproate, Pediatrics 125:e956–e959, 2010.
  290. Baron J, Mulero P, Pedraza MI, et al: HaNDL syndrome: correlation between focal deficits topography and EEG or SPECT abnormalities in a series of 5 new cases, Neurologia 31(5):305–310, 2016.
  291. Berger K, Evers S: Migraine with aura and the risk of increased mortality, BMJ 341:465–466, 2010.
  292. Bond DS, Vithiananthan S, Nash JM, et al: Improvement of migraine headaches in severely obese patients after bariatric surgery, Neurology 76:1135–1138, 2013.
  293. Bruijn J, Duivenvoorden H, Passchier J, et al: Medium-dose riboflavin as a prophylactic agent in children with migraine: a preliminary placebo-controlled, randomized, double-blind, cross-over trial, Cephalalgia 30(12):1426–1434, 2010.
  294. Bruijn J, Locher H, Passchier J, et al: Psychopathology in children and adolescents with migraine in clinical studies: a systematic review, Pediatrics 126:323–332, 2010.
  295. Chasman DI, Sch?rks M, Anttila V, et al: Genome-wide association study reveals three susceptibility loci for common migraine in the general population, Nat Genet 43:695–697, 2011.
  296. Condo M, Posar A, Arbizzania A, et al: Riboflavin prophylaxis in pediatric and adolescent migraine, J Headache Pain 10:361–365, 2009.
  297. Connelly M: Cognitive behavioral therapy for treatment of pediatric chronic migraine, JAMA 310:2617–2618, 2013.
  298. Crawford MJ, Lehman L, Slater S, et al: Menstrual migraine in adolescents, Headache 49:341–347, 2009.
  299. Derosier FJ, Lewis D, Hershey AD, et al: Randomized trial of sumatriptan and naproxen sodium combination in adolescent migraine, Pediatrics 126:e1411–e1420, 2012.
  300. DeSimone R, Ranieri A, Montella S, et al: Intracranial pressure in unresponsive chronic migraine, J Neurol 261(7):1365–1373, 2014.
  301. Dodick DW, Silberstein SD, Reed KL, et al: Safety and efficacy of peripheral nerve stimulation of the occipital nerves for the management of chronic migraine: longterm results from a randomized, multicenter, double-blinded controlled study, Cephalalgia 35(4):344–358, 2015.
  302. Dodick DW, Turkel CC, DeGryse RE, et al: Onabotulinumtoxin A for treatment of chronic migraine: pooled results from the double-blind, randomized, placebocontrolled phases of the PREEMPT clinical program, Headache 50:921–936, 2010.
  303. Dodick DW: Migraine, Lancet 391:1315–1330, 2018.
  304. Edvinsson L, Linde M: New drugs in migraine treatment and prophylaxis: telcagepant and topiramate, Lancet 376:645–654, 2010.
  305. Fernstermacher N, Levin M, Ward T: Pharmacological prevention of migraine, BMJ 342:d583, 2011. Friedman BW, Irizarry E, Solorzano C, et al: Randomized study of IV procholorperazine plus diphenhydramine vs IV hydromorphone for migraine, Neurology 89:2075–2082, 2017.
  306. Fumal A, Vandenheede M, Coppola G, et al: The syndrome of transient headache with neurological deficits and CSF lymphocytosis (HaNDL): electrophysiological findings suggesting a migrainous pathophysiology, Cephalalgia 25:754–758, 2005.
  307. Garg P, Servoss SJ, Wu JC, et al: Lack of association between migraine headache and patent foramen ovale: results of a case-control study, Circulation 121:1406–1412, 2010.
  308. Gelfand AA, Fullerton HJ, Goadsby PJ: Child neurology: migraine with aura in children, Neurology 75:e16–e19, 2010.
  309. Gelfand AA, Goadsbury PJ: Treatment of pediatric migraine in the emergency room, Pediatr Neurol 47:233–241, 2012.
  310. Goadsby PJ, Reuter U, Hallstr?m Y, et al: A controlled trial of erenumab for episodic migraine, N Engl J Med 377(22):2123–2132, 2017.
  311. Graf WD, Kayyali HR, Abdelmoity AT, et al: Incidental neuroimaging findings in nonacute headache, J Child Neurol 25(10):1182–1187, 2010.
  312. Hershey AD, Powers SW, Vockell AL, et al: Coenzyme Q10 deficiency and response to supplementation in pediatric and adolescent migraine, Headache 47:73–80, 2007.
  313. Hershey AD, Powers SW, Vockell AL, et al: PedMIDAS: development of a questionnaire to assess disability of migraines in children, Neurology 57:2034–2039, 2001
  314. Holroyd KA, Bendtsen L: Tricyclic antidepressants for migraine and tension-type headaches, BMJ 341:5250, 2010.
  315. Hougaard A, Amin F, Hauge AW, et al: Provocation of migraine with aura using natural trigger factors, Neurology 80:1–4, 2013.
  316. Jackson JL, Kuriyama A, Hayashino Y: Botulism toxin A for prophylactic treatment of migraine and tension headaches in adults, JAMA 307:1736–1744, 2012.
  317. Kabbouche M, O’Brien H, Hershey AD: OnabotulinumtoxinA in pediatric chronic daily headache, Curr Neurol Neurosci Rep 12(2):114–117, 2012.
  318. Kabbouche MA, Cleves C: Evaluation and management of children and adolescents presenting with an acute setting, Semin Pediatr Neurol 17(2):105–108, 2010.
  319. Karsan N, Prabhakar P, Goadsby PJ: Characterizing the premonitory stage of migraine in children: a clinic-based study of 100 patients in a specialist headache service, J Headache Pain 17:94, 2016.
  320. Kelley SA, Hartman AL, Kossoff EH: Comorbidity of migraine in children presenting with epilepsy to a tertiary care center, Neurology 79:468–473, 2012.
  321. Lateef T, Cui L, Heaton L, et al: Validation of a migraine interview for children and adolescents, Pediatrics 131:e96–e102, 2013.
  322. Lewis D, Winner P, Saper J, et al: Randomized, double-blind, placebo-controlled study to evaluate the efficacy and safety of topiramate for migraine prevention in pediatric subjects 12 to 17 years of age, Pediatrics 123:924–934, 2009.
  323. Lewis DW, Ashwal S, Hershey A, et al: Practice parameter: pharmacological treatment of migraine headache in children and adolescents, Neurology 63:2215–2224, 2004.
  324. Lewis DW: Pediatric migraine, Neurol Clin 27:481–501, 2009.
  325. Loder E: Triptan therapy in migraine, N Engl J Med 363:63–70, 2010.
  326. Mariotti P, Nociti V, Cianfoni A, et al: Migraine-like headache and status migrainosus as attacks of multiple sclerosis in a child, Pediatrics 126:e459–e464, 2010.
  327. McCandless RT, Arrington CB, Nielson DC, et al: Patent foramen ovale in children with migraine headaches, J Pediatr 159:243–247, 2011.
  328. McKeage K: Zolmitriptan nasal spray: a review in acute migraine in pediatric patients aged 12 years and older, Paediatr Drugs 18(1):75–81, 2016.
  329. Mohamed BP, Goadsby PJ, Prabhakar P: Safety and efficacy of flunarizine in childhood migraine: 11 years’ experience, with emphasis on its effect in hemiplegic migraine, Dev Med Child Neurol 54:274–277, 2012.
  330. Oelkers-Ax R, Leins A, Parzer P, et al: Butterbur root extract and music therapy in the prevention of childhood migraine: an explorative study, Eur J Pain 12:301–313, 2008.
  331. Palm-Meinders IH, Koppen H, Terwindt GM, et al: Structural brain changes in migraine, JAMA 308:1889–1896, 2012.
  332. Pascual J, Valle N: Pseudomigraine with lymphocytic pleocytosis, Curr Pain Headache Rep 7:224–228, 2003.
  333. Powers SW, Coffey CS, Chamberlin LA, et al: Trial of amitriptyline, topiramate and placebo for pediatric migraine, N Engl J Med 376:115–124, 2017.
  334. Powers SW, Kahhikar-Zuck S, Allen JR, et al: Cognitive behavioral therapy plus amitriptyline for chronic migraine in children and adolescents: a randomized clinical trial, JAMA 310:2622–2630, 2013.
  335. Pringsheim T, Becker WJ: Triptans for symptomatic treatment of migraine headache, BMJ 348:35–37, 2014.
  336. Rabbie R, Derry S, Moore RA: Ibuprofen with or without an antiemetic for acute migraine headaches in adults, Cochrane Database Syst Rev (4):CD008039, 2013.
  337. Russell MB, Ducros A: Sporadic and familial hemiplegic migraine: pathophysiological mechanisms, clinical characteristics, diagnosis, and management, Lancet Neurol 10:457–470, 2011.
  338. Schoenen J, Vandersmissen B, Jeangette S, et al: Migraine prevention with a supraorbital transcutaneous stimulator, Neurology 80:697–704, 2013.
  339. Silberstein SD, Dodick DW, Bigal ME, et al: Fremanezumab for the preventive treatment of chronic migraine, N Engl J Med 377(22):2113–2122, 2017.
  340. Silberstein SD, Holland S, Freitag F, et al: Evidence-based guideline update: pharmacologic treatment for episodic migraine prevention in adults, Neurology 78:1337–1345, 2012.
  341. Silberstein SD, Rosenberg J: Multispecialty consensus on diagnosis and treatment of headache, Neurology 54:2000, 1553.
  342. Taggert E, Doran S, Kokotillo A, et al: Ketorolac in the treatment of acute migraine: a systematic review, Headache 53:277–287, 2013.
  343. The Medical Letter: A fixed-dose combination of sumatriptan and naproxen for migraine, Med Lett Drugs Ther 50:45–46, 2008.
  344. The Medical Letter: Treatment of migraine, Med Lett 59:27–32, 2017.
  345. The Medical Letter: Warning against the use of valproate for migraine prevention during pregnancy, Med Lett Drugs Ther 55:45, 2013.
  346. Tikka S, Baumann M, Siitonen M, et al: CADASIL and CARASIL, Brain Pathol 24:525–544, 2014.
  347. van Ooserhout WPJ, van der Plas AA, van Zwet EW, et al: Postdural puncture headache in migraineurs and nonheadache subjects, Neurology 80:941–948, 2013.
  348. Visser WH, Winner P, Strohmaier K, et al: Rizatriptan 5 mg for the acute treatment of migraine in adolescents: results from a double-blind, single-attack study and two open-label, multiple-attack studies, Headache 44:89–899, 2004.
  349. Winner P, Rothner D, Saper J, et al: A randomized, double-blind, placebo-controlled study of sumatriptan nasal spray in the treatment of acute migraine in adolescents, Pediatrics 106:9889–9997, 2000.
  350. Arruda MA, Guidetti V, Galli F, et al: Frequent headaches in the preadolescent pediatric population, Neurology 74:903–908, 2010.
  351. Baskin SM, Lipchik GL, Smitherman TA: Mood and anxiety disorders in chronic headache, Headache 46(suppl3):S76–S87, 2006.
  352. Biedron A, Kac?nski M, Skowronek-Bata B: Stabbing headache in an 8-year-old girl: primary or drug induced headache?, Pediatrics 133:e1068–e1071, 2014.
  353. Blume HK, Vavilala MS, Jaffe KM, et al: Headache after pediatric traumatic brain injury: a cohort study, Pediatrics 129(1):e31–e39, 2012.
  354. Cady RK, Schreiber CP: Sinus headache: a clinical conundrum, Otolaryngol Clin North Am 37:267–288, 2004.
  355. Calhoun A: Adjusting estradiol concentrations reduces headache frequency and severity in female migraineurs, Cephalalgia 21:448–449, 2001.
  356. Calhoun AH: A novel specific prophylaxis for menstrual-associated migraine, South Med J 97:819–822, 2004.
  357. Carville S, Padhi S, Reason T, et al: Diagnosis and management of headaches in young people and adults: summary of NICE guidelines, BMJ 345:e5765, 2012.
  358. Cohen AS, Burns B, Goadsby PJ: High-flow oxygen for treatment of cluster headache, JAMA 302:2451–2457, 2009.
  359. Crock C, Orsini F, Lee KJ, et al: Headache after lumbar puncture: randomized crossover trial of 22-gauge versus 25-gauge needles, Arch Dis Child 99:203–207, 2014.
  360. Dafer RM, Jay WM: Headache and the eye, Curr Opin Ophthalmol 20:520–524, 2009.
  361. DeVries A, Young PC, Wall E, et al: CT scan utilization patterns in pediatric patients with recurrent headache, Pediatrics 132:e1–e8, 2013.
  362. Ducros A, Bousser MG: Thunderclap headache, BMJ 345:e8557, 2012.
  363. Friedman BW, Adewunmi V, Campbell C, et al: A randomized trial of intravenous ketorolac versus intravenous metoclopramide plus diphenhydramine for tension-type and all nonmigraine, noncluster recurrent headaches, Ann Emerg Med 62:311–318, 2013.
  364. Friedman DI: The eye and headache, Ophthalmol Clin North Am 17:357–369, 2004.
  365. Furlan JC: Headache attributed to autonomic dysreflexia, Neurology 77:792–798, 2011.
  366. Giza CC, Kutcher JS, Ashwal S, et al: Summary of evidence-based guideline update: evaluation and management of concussion in sports: report of the Guideline Development Subcommittee of the American Academy of Neurology, Neurology 80(24):2250–2257, 2013.
  367. Hagen K, Albretsen C, Vilming ST, et al: Management of medication overuse headache: 1-year randomized multicentre open-label trial, Cephalalgia 29:221–232, 2008.
  368. Hershey AD, Powers SW, Winner P, Kabbouche MA: Pediatric headaches in clinical practice, Oxford, UK, 2009, John Wiley & Sons. Heyer GL, Idris SA: Does analgesic overuse contribute to chronic post-traumatic headaches in adolescent concussion patient?, Pediatr Neurol 50:464–468, 2014.
  369. Kacperski J, Arthur T: Management of post-traumatic headaches in children and adolescents, Headache 56(1):36–48, 2016. Kakisaka Y, Ohara T, Hino-Fukuyo N, et al: Abdominal and lower back pain in pediatric idiopathic stabbing headache, Pediatrics 133:e245–e247, 2014.
  370. Kuczynski A, Crawford S, Bodell L, et al: Characteristics of post-traumatic headaches in children following mild traumatic brain injury and their response to treatment: a prospective cohort, Dev Med Child Neurol 55:636–641, 2013.
  371. Lambru G, Matharu M: Management of trigeminal autonomic cephalalgias in children and adolescents, Curr Pain Headache Rep 17:323, 2013.
  372. Leroux E, Valade D, Taifas I, et al: Suboccipital steroid injections for transitional treatment of patients with more than two cluster headache attacks per day: a randomized, double-blind, placebo-controlled trial, Lancet Neurol 10:891–897, 2011.
  373. MacGregor EA, Victor TW, Hu X, et al: Characteristics of menstrual vs nonmenstrual migraine: a post hoc, within-woman analysis of the usual-care phase of a nonrandomized menstrual migraine clinical trial, Headache 50:528–538, 2010.
  374. Massano D, Julliand S, Kanagarajah L, et al: Headache with focal neurologic signs in children at the emergency department, J Pediatr 165:376–382, 2014.
  375. Murray SC, Muse KN: Effective treatment of severe menstrual migraine headaches with gonadotropin-releasing hormone agonist and «add-back» therapy, Fertil Steril 67:390–393, 1997. Nesbitt AD, Goadsby PJ: Cluster headache, BMJ 344:e2407, 2012.
  376. O’Brien HL, Cohen JM: Young adults with headaches: the transition from adolescents to adults, Headache 55(10):1404–1409, 2015.
  377. Packard R, Ham L: Pathogenesis of posttraumatic headache and migraine: a common headache pathway?, Headache 37:142–152, 1997.
  378. Radat F, Swendsen J: Psychiatric comorbidity in migraine: a review, Cephalalgia 25:165–178, 2005.
  379. Watanabe TK, Bell KR, Walker WC, Schomer K: Systematic review of interventions for post-traumatic headache, PM R 49(2):129–140, 2012.
  380. Zakrzewska JM, Linskey ME: Trigeminal neuralgia, BMJ 348:g474, 2014.
  381. Abu-Arafeh I, Raxak S, Sivaraman B, et al: Prevalence of headache and migraine in children and adolescents; a systemic review of population-based studies, Dev Med Child Neurol 52(12):1088–1097, 2010.
  382. Bonfert M, Straube A, Schroeder AS, et al: Primary headache in children and adolescents: update on pharmacotherapy of migraine and tension-type headache, Neuropediatrics 44(1):3–19, 2013.
  383. Frendrich K, Vennemann M, Pfaffenrath V, et al: Headache prevalence among adolescents—the German DMKG headache study, Cephalalgia 27(4):347–354, 2007.
  384. Harden RN, Cottrill J, Gagnon CM, et al: Botulinum toxin A in the treatment of chronic tension-type headache with cervical myofascial trigger points: a randomized, double-blind, placebo-controlled pilot study, Headache 49:732–743, 2009.
  385. Headache Classification Subcommittee of the International Headache Society: The international classification of headache disorders, Cephalalgia 24(Suppl 1):9–160, 2004.
  386. Kroner-Herwig B, Heinrich M, Morris L: Headache in German children and adolescents; a population-based epidemiological study, Cephalalgia 27(6):519–527, 2007.
  387. Levin M: The international classification of headache disorders, 3rd edition (ICHD III): changes and challenges, Headache 53(8):1383–1395, 2013.
  388. Mack KJ: What incites new daily persistent headache in children?, Pediatr Neurol 31:122–125, 2004.
  389. Wang SJ, Fuh JL, Lu SR: Chronic daily headache in adolescents, Neurology 73:416–422, 2009.
  390. Baser ME, R Evans DG, Gutmann DH: Neurofibromatosis 2, Curr Opin Neurol 16(1):27–33, 2003.
  391. Blakeley JO, Evans DG, Adler J, et al: Consensus recommendations for current treatments and accelerating clinical trials for patients with neurofibromatosis type 2, Am J Med Genet A 158(1):24–41, 2012.
  392. Boyd C, Smith MJ, Kluwe L, et al: Alterations in the SMARCB1 (INI1) tumor suppressor gene in familial schwannomatosis, Clin Genet 74:358–366, 2008.
  393. Brems H, Chmara M, Sahbatou M, et al: Germline loss-of-function mutations in SPRED1 cause a neurofibromatosis 1-like phenotype, Nat Genet 39:1120–1126, 2007.
  394. Cnossen MH, de Goede-Bolder A, van den Broek KM, et al: A prospective 10 year follow up study of patients with neurofibromatosis type 1, Arch Dis Child 78:408–412, 1998.
  395. DeBella K, Szudek J, Friedman JM: Use of the national institutes of health criteria for diagnosis of neurofibromatosis 1 in children, Pediatrics 105:608–614, 2000.
  396. Dombi E, Baldwin A, Marcus LJ, et al: Activity of selumetinib in neurofibromatosis type 1-related plexiform neurofibromas, N Engl J Med 375(26):2550–2560, 2016.
  397. Hersh JH: American Academy of pediatrics committee on genetics: health supervision for children with neurofibromatosis, Pediatrics 121(3):633–642, 2008.
  398. Hyman SL, Shores A, North KN: The nature and frequency of cognitive deficits in children with neurofibromatosis type 1, Neurology 65(7):1037–1044, 2005.
  399. Isaacs H Jr: Perinatal neurofibromatosis: two case reports and review of the literature, Am J Perinatol 27:285–292, 2010.
  400. Koss M, Scott RM, Irons MB, et al: Moyamoya syndrome associated with neurofibromatosis type 1: perioperative and long-term outcome after surgical revascularization, J Neurosurg Pediatr 11(4):417–425, 2013.
  401. Listernick R, Ferner RE, Liu GT, et al: Optic pathway gliomas in neurofibromatosis-1: controversies and recommendations, Ann Neurol 61(3):189–198, 2007.
  402. MacCollin M, Chiocca EA, Evans DG, et al: Diagnostic criteria for schwannomatosis, Neurology 64:1838–1845, 2005. Neurofibromatosis. Conference statement. National institutes of health consensus development Conference, Arch Neurol 45:575–578, 1988.
  403. Rodriguez D, Young Poussaint T: Neuroimaging findings in neurofibromatosis type 1 and 2, Neuroimaging Clin N Am 14(2):149–170, 2004. Ruggieri M, Pratico AD: Mosaic neurocutaneous disorders and their causes, Semin Pediatr Neurol 22:207–233, 2015.
  404. Stumpf DA, Alksne JF, Annegers JF, et al: NIH development conference. Neurofibromatosis: Conference statement, Arch Neurol 45:575–578, 1988.
  405. Bissler JJ, Kingswood JC, Radzikowska E, et al: Everolimus for angiomyolipoma associated with tuberous sclerosis complex or sporadic lymphangioleiomyomatosis (EXIT-2): a multi-centre, randomized, double-blind, placebo-controlled trial, Lancet 381:817–824, 2013.
  406. Cardamore M, Flanagan D, Mowat D, et al: Mammalian target of rapamycin inhibitors for intractable epilepsy and subependymal giant cell astrocytomas in tuberous sclerosis complex, J Pediatr 164:1105–1200, 2014.
  407. Crino PB, Nathanson KL, Henske EP: The tuberous sclerosis complex, N Engl J Med 355:1345–1356, 2006.
  408. Cudzilo CJ, Szczesniak RD, Brody AS, et al: Lymphangioleiomyomatosis screening in women with tuberous sclerosis, Chest 144:578–585, 2013.
  409. Curatolo P, Franz DN, Lawson JA, et al: Adjunctive everolimus for children and adolescents with treatment refractory seizures associated with tuberous sclerosis complex: post-hoc analysis of the phase 3 EXIST-3 trial, Lancet Child Adolesc Health 2:495–504, 2018.
  410. de Vries PJ, Whittemore VH, Leclezio L, et al: Tuberous sclerosis associated neuropsychiatric disorders (TAND) and the TAND checklist, Pediatr Neurol 52:25–35, 2015.
  411. Datta AN, Hahn CD, Sahin M: Clinical presentation and diagnosis of tuberous sclerosis complex in infancy, J Child Neurol 23:268–273, 2008.
  412. Ewalt DH, Diamond N, Rees C, et al: Long-term outcome of transcatheter embolization of renal angiomyolipomas due to tuberous sclerosis complex, J Urol 174(5):1764–1766, 2005.
  413. Franz DN, Belousova E, Sparagana S, et al: Efficacy and safety of everolimus for subependymal giant cell astrocytomas associated with tuberous sclerosis complex (EXIST-1): a multicenter, randomizes, placebo-controlled phase 3 trial, Lancet 381:125–132, 2013.
  414. Kingswood JC, Bissler JJ, Budde K, et al: Review of the tuberous sclerosis renal guidelines from the 2012 consensus Conference: current data and future study, Nephron 134:51–58, 2016.
  415. Lai J, Modi L, Ramai D, Tortora M: Tuberious sclerosis complex and polycystic kidney disease contiguous gene syndrome with moyamoya disease, Pathol Res Pract 213:410–415, 2017.
  416. Siroky BJ, Towbin AJ, Trout AT, et al: Improvement in renal cystic disease of tuberous sclerosis complex after treatment with mammalian target of rapamycin inhibitor, J Pediatr 187:318–322, 2017.
  417. Tworetzky W, McElhinney DB, Margossian R, et al: Association between cardiac tumors and tuberous sclerosis in the fetus and neonate, Am J Cardiol 92(4):487–489, 2003.
  418. Warncke JC, Brodie KE, Grantham EC, et al: Pediatric renal angiomyolipomas in tuberous sclerosis complex, J Urol 197:500–506, 2017.
  419. Comi AM: Advances in Sturge-weber syndrome, Curr Opin Neurol 19(2):124–128, 2006. Dymerska M, Kirkorian AY, Offerman EA, et al: Size of facial port-wine birthmark may predict neurologic outcome in Sturge-weber syndrome, J Pediatr 188:205–209, 2017.
  420. Huang L, Couto JA, Pinto A, et al: Somatic GNAQ mutation is enriched in brain endothelial cells in Sturge-weber syndrome, Pediatr Neurol 67:59–63, 2017.
  421. Kossoff EH, Buck C, Freeman JM: Outcomes of 32 hemispherectomies for Sturge-weber syndrome worldwide, Neurology 59:1735–1738, 2002.
  422. Shirley MD, Tang H, Gallione CJ, et al: Sturge-weber syndrome and port-wine stains caused by somatic mutation in GNAQ, N Engl J Med 368(21):1971–1979, 2013.
  423. Tan OT, Sherwood K, Gilchrest BA: Treatment of children with port-wine stains using the flashlamp-pulsed tunable dye laser, N Engl J Med 320:416–421, 1989.
  424. Latif F, Tory K, Gmarra J, et al: Identification of the von Hippel-lindau disease tumor suppressor gene, Science 260:1317–1320, 1993.
  425. Maher ER, Kaelin WG Jr: Von Hippel-lindau disease, Medicine (Baltimore) 76:381–391, 1997.
  426. Maher ER, Neumann HP, Richard S: Von Hippel-lindau disease: a clinical and scientific review, Eur J Hum Genet 19:617–623, 2011.
  427. Menascu S, Donner EJ: Linear nevus sebaceous syndrome: case reports and review of the literature, Pediatr Neurol 38(3):207–210, 2008.
  428. Drolet BA, Frommelt PC, Chamlin SL, et al: Initiation and use of propranolol for infantile hemangioma: report of a consensus conference, Pediatrics 131:128–140, 2013.
  429. Metry DW, Dowd CF, Barkovich AJ, et al: The many faces of PHACE syndrome, J Pediatr 139:117–123, 2001.
  430. Poetke M, Frommeld T, Berlien HP: PHACE syndrome: new views on diagnostic criteria, Eur J Pediatr Surg 12:366–374, 2002.
  431. Tangtiphaiboontana J, Hess CP, Bayer M, et al: Neurodevelopmental abnormalities in children with PHACE syndrome, J Child Neurol 28:608–614, 2013.
  432. Winter PR, Itinteang T, Leadbitter P, Tan ST: PHACE syndrome–clinical features, aetiology and management, Acta Paediatr 105:145–153, 2016.
  433. Bruckner AL: Incontinentia pigmenti: a window to the role of NF-kappaB function, Semin Cutan Med Surg 23:116–124, 2004.
  434. Meuwissen ME, Mancini GM: Neurological findings in incontinentia pigmenti; a review, Eur J Med Genet 55:323–331, 2012.
  435. Minis S, Trpinac D, Obradovi? M: Systematic review of central nervous system anomalies in incontinentia pigmenti, Orphanet J Rare Dis 8:25, 2013.
  436. Santa-Maria FD, Mariath LM, Poziomczyk CS, et al: Dental anomalies in 14 patients with IP: clinical and radiological analysis and review, Clin Oral Investig 21(5):1845–1852, 2017.
  437. Smahi A, Courtois G, Vabres P, et al: Genomic rearrangement in NEMO impairs NF-kappaB activation and is a cause of incontinentia pigmenti. The international incontinentia pigmenti (IP) consortium, Nature 405:466–472, 2000.
  438. Koy A, Lin JP, Sanger TD, et al: Advances in management of movement disorders in children, Lancet Neurol 15:719–735, 2016.
  439. Sanger TD, et al: Definition and classification of hyperkinetic movements in childhood, Mov Disord 25:1538–1549, 2010.
  440. Singer HS, Mink JW, Gilbert DL, Jankovic J: Movement disorders in childhood, ed 2, London, 2016, Academic Press. Tranchant C, Anheim M: Movement disorders in mitochondrial diseases, Rev Neurol (Paris) 172:524–529, 2016.
  441. Caffarelli M, Kimia AA, Torres AR: Acute ataxia in children: a review of the differential diagnosis and evaluation in the emergency department, Pediatr Neurol 65:14–30, 2016.
  442. de Bot ST, Willemsen MAAP, Vermeer S, et al: Reviewing the genetic causes of spastic-ataxias, Neurology 79:1507–1514, 2012.
  443. Desai J, Mitchell WG: Acute cerebellar ataxia, acute cerebellitis, and opsoclonusmyoclonus syndrome, J Child Neurol 27:1482–1488, 2012.
  444. Jafar-Nejad P, Maricich SM, Zoghbi HY: Swaiman’s pediatric neurology. The cerebellum and the hereditary ataxias, ed 6, Philadelphia, 2017, WB Saunders., pp 689–700. Musselman KE, Stoyanov CT, Marasigan R, et al: Prevalence of ataxia in children: a systematic review, Neurology 82:80–89, 2014.
  445. Rossi M, Perez-Lloret S, Doldan L, et al: Autosomal dominant cerebellar ataxias: a systematic review of clinical features, Eur J Neurol 21:607–615, 2014.
  446. Schiffmann R, Fogli A, Van der Knapp MS, Boespflug-Tanguy O: Childhood ataxia with central nervous system hypomyelination/vanishing white matter, GeneReviews® [Internet], 2003. updated August 9, 2012.
  447. Shakkottai VG, Fogel BL: Clinical neurogenetics—autosomal dominant spinocerebellar ataxia, Neurol Clin 31:987–1007, 2013.
  448. Whelan HT, Aaen GS, Sannagowdara K, DeMara-Hoth MB: Swaiman’s pediatric neurology. Acute cerebellar ataxia, ed 6, Philadelphia, 2017, WB Saunders., pp 701–705.
  449. Albanese A, Bhatia K, Bressman SB, et al: Phenomenology and classification of dystonia: a consensus update, Mov Disord 28(7):863–873, 2013.
  450. Ananth AL, Robichaux-Viehoever A, Kim YM, et al: Clinical course of six children with GNAO1 mutations causing severe and distinctive movement disorder, Pediatr Neurol 59:81–84, 2016.
  451. Dale RC: Immune-mediated extrapyramidal movement disorders, including Sydenham chorea, Handb Clin Neurol 112:1235–1241, 2013.
  452. Garvey MA, Snider LA, Leitman SF, et al: Treatment of Sydenham’s chorea with intravenous immunoglobulin, plasma exchange or prednisone, J Child Neurol 20(5):424–429, 2005.
  453. Hermann A, Walker RH: Diagnosis and treatment of chorea syndromes, Curr Neurol Neurosci Rep 15(1): 2015.
  454. Mencacci NE, Carecchio M: Recent advances in genetics of chorea, Curr Opin Neurol 29(4):486–495, 2016.
  455. Patel NJ, Jankovic J: NKX2-1-related disorders, GeneReviews® [Internet], 2014. updated July 29, 2016.
  456. Sharawat IK, Kasinathan A, Sankhyan N: Infantile tremor syndrome: response to B12 therapy, J Pediatr 196:323, 2018.
  457. Singer HS, Mink JW, Gilbert DL, Jancovic J: Movement disorders in childhood, Philadelphia, 2015, Saunders Elsevier, pp 144–256.
  458. Walker K, Brink A, Lawrenson J, et al: Treatment of syndenham chorea with intravenous immunoglobulin, J Child Neurol 27(2):147–155, 2012.
  459. Desai J, Mitchell WG: Acute cerebellar ataxia, acute cerebellitis, and opsoclonusmyoclonus syndrome, J Child Neurol 27:1482–1488, 2012.
  460. Koy A, Lin JP, Sanger TD, et al: Advances in management of movement disorders in children, Lancet Neurol 15:719–735, 2016.
  461. Pranzatelli MR, Tate ED, Travelstead AL, et al: Rituximab (anti-CD20) adjunctive therapy for opsoclonus-myoclonus syndrome, J Pediatr Hematol Oncol 28(9):585–593, 2006.
  462. Ratner N, Brodeur GM, Dale RC, Schor NF: The «neuro» of neuroblastoma: neuroblastoma as a neurodevelopmental disorder, Ann Neurol 80:13–23, 2016.
  463. Sanger TD, et al: Definition and classification of hyperkinetic movements in childhood, Mov Disord 25:1538–1549, 2010.
  464. Singer HS, Mink JW, Gilbert DL, Jankovic J: Movement disorders in childhood, ed 2, London, 2016, Academic Press.
  465. Albin RL: Dominant ataxias and Friedreich ataxia: an update, Curr Opin Neurol 16:507–514, 2003.
  466. Anheim M, Tranchant C, Koenig M: The autosomal recessive cerebellar ataxias, N Engl J Med 366:636–646, 2012.
  467. Aravamuthan BR, Waugh JL, Stone SS: Deep brain stimulation for monogenic dystonia, Curr Opin Pediatr 29:691–696, 2017.
  468. Avcin T, Benseler SM, Tyrrell PN, et al: A followup study of antiphospholipid antibodies and associated neuropsychiatric manifestations in 137 children with systemic lupus erythematosus, Arthritis Rheum 59(2):206–213, 2008.
  469. Benini R, Ben Amour IM, Shevell MI: Clinical clues to differentiating inherited and noninherited etiologies of childhood ataxias, J Pediatr 160:152–157, 2012.
  470. Bosemani T, Meoded A, Poretti A: Susceptibility-weighted imaging in pantothenate kinase-associated neurodegeneration, J Pediatr 164:212, 2014.
  471. Braddock SR, Henley KM, Maria BL: The face of Joubert syndrome: a study of dysmorphology and anthropometry, Am J Med Genet A 143:3235–3242, 2007.
  472. Cardoso F, Seppi K, Mair KJ, et al: Seminar on choreas, Lancet Neurol 5:589–602, 2006.
  473. Demiroren K, Yavuz H, Cam L, et al: Sydenham’s chorea: a clinical follow-up of 65 patients, J Child Neurol 22:550–554, 2007.
  474. Desai J, Mitchell WG: Acute cerebellar ataxia, acute cerebellitis, and opsoclonusmyoclonus syndrome, J Child Neurol 27:1482–1488, 2012.
  475. Farr AK, Shalev B, Crawford TO, et al: Ocular manifestations of ataxia-telangiectasias, Am J Ophthalmol 134:891–896, 2002.
  476. Fasano A, Nardocci N, Elia AE, et al: Non-DYT1 early-onset primary torsion dystonia: comparison with DYT1 phenotype and review of the literature, Mov Disord 21(9):1411–1418, 2006.
  477. Hogarth P: Neurodegeneration with brain iron accumulation: diagnosis and management, J Mov Disord 8(1):1–13, 2015.
  478. Gibilisco P, Vogel AP: Friedreich ataxia, BMJ 347:f7062, 2013.
  479. Jahn K, Langhagen T, Schroeder AS, et al: Vertigo and dizziness in childhood–update on diagnosis and treatment, Neuropediatrics 42:129–134, 2011.
  480. Keller S, Dure LS: Tremor in childhood, Semin Pediatr Neurol 16:60–70, 2009.
  481. Kim HJ, Jean BS, Yang HJ, et al: In need of something better than sleep, Lancet 381:598, 2013.
  482. Kong MF, Gilbert G, Baleanu F, et al: Progressive cerebellar ataxia and new-onset diabetes, Lancet 383:186, 2014.
  483. Kruer MC, Hoeftberger R, Lim KY, et al: Aggressive course in encephalitis with opsoclonus, ataxia, chorea, and seizures, JAMA Neurol 71:620–622, 2014.
  484. Leblicq C, Duval M, Carmant L, et al: Rising serum thyroxine levels and chorea in Graves’ disease, Pediatrics 131:e616–e619, 2013.
  485. Libri V, Yandim C, Athanasopoulos S, et al: Epigenetic and neurological effects and safety of high-dose nicotinamide in patients with Friedreich’s ataxia: an exploratory, open-label, dose-escalation study, Lancet 384:504–512, 2014.
  486. Lin JP: Advances in pharmacotherapies for movement disorders in children: current limitations and future progress, Curr Opin Pediatr 29:652–664, 2017.
  487. Lumsden DE, King MD, Allen NM: Status dystonicus in childhood, Curr Opin Pediatr 29:674–682, 2017.
  488. Moghimi N, Jabbari B, Szekely AML: Primary dystonias and genetic disorders with dystonia as clinical feature of the disease, Eur J Paediatr Neurol 18:79–105, 2014. Nardocci N, Zorzi G, Barzaghi C, et al: Myoclonus-dystonia syndrome: clinical presentation, disease course, and genetic features in 11 families, Mov Disord 23(1): 28–34, 2008.
  489. Obeso JA, Rodriguez-Oroz MC, Stamelou M, et al: The expanding universe of disorders of the basal ganglia, Lancet 384:523–530, 2014.
  490. Pandolfo M: Friedreich ataxia, Semin Pediatr Neurol 10:163–172, 2003.
  491. Paz JA, Silva CAA, Marques-Dias MJ: Randomized double-blind study with prednisone in Sydenham’s chorea, Pediatr Neurol 34(4):264–269, 2006.
  492. Pearl PL, Taylor JL, Trzcinski S, et al: The pediatric neurotransmitter disorders, J Child Neurol 22(5):606–616, 2007.
  493. Poretti A, Benson JE, Huisman TAGM, et al: Acute ataxia in children: approach to clinical presentation and role of additional investigations, Neuropediatrics 44:127–141, 2013.
  494. Pranzatelli MR, Tate ED, Travelstead AL, et al: Rituximab (anti-CD20) adjunctive therapy for opsoclonus-myoclonus syndrome, J Pediatr Hematol Oncol 28:585–593, 2006.
  495. Rilstone JJ, Alkhater RA, Minassian BA: Brain dopamine-serotonin vesicular transport disease and its treatment, N Engl J Med 368:543–550, 2013.
  496. Rosman NP, Douglass LM, Sharif UM, et al: The neurology of benign paroxysmal torticollis of infancy: report of 10 new cases and review of the literature, J Child Neurol 24(2):155–160, 2009.
  497. Sailer A, Houlden H: Recent advances in the genetics of cerebellar ataxias, Curr Neurol Neurosci Rep 12:227–236, 2012.
  498. Sanger TD, Chen D, Fehlings DL, et al: Definition and classification of hyperkinetic movements in childhood, Mov Disord 25:1538–1549, 2010.
  499. Serrano-Munuera C, Corral-Juan M, Stevanin G, et al: New subtype of spinocerebellar ataxia with altered vertical eye movements mapping to chromosome 1p32, JAMA Neurol 70:764–771, 2013.
  500. Schiffmann R, Fogli A, Van der Knaap MS, et al: Childhood ataxia with central nervous system hypomyelination/vanishing white matter. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews® [Internet]. Seattle (WA), 1993–2014, Seattle, 2003, University of Washington. updated August 9, 2012.
  501. Singer HS, Mink JW, Gilbert DL, et al: Movement disorders in childhood, Philadelphia, 2010, WB Saunders, pp 231–241. Stoessel AJ, Lehericy S, Strafella AP: Imaging insights into basal ganglia function, Parkinson’s disease, and dystonia, Lancet 384:532–542, 2014.
  502. Sweney MT, Silver K, Gerard-Blanluet M, et al: Alternating hemiplegia of childhood: early characteristics and evolution of a neurodevelopmental syndrome, Pediatrics 123(3):e534–e541, 2009. Tabarki B, Al-Sheikh F, Al-Shahwan S, et al: Bilateral external ophthalmoplegia in biotin-responsive basal ganglia disease, J Pediatr 162:1291–1292, 2013.
  503. Taly AB, Meenakshi-Sundaram S, Sinha S, et al: Wilson disease: description of 282 patients evaluated over 3 decades, Medicine (Baltimore) 82(2):112–121, 2007.
  504. The Multiple System Atrophy Research Collaboration: Mutation in COQ2 in familial and sporadic multiple-system atrophy, N Engl J Med 369:233–244, 2013.
  505. Valente EM, Dallapiccola B, Bertini E: Joubert syndrome and related disorders, Handb Clin Neurol 113:1879–1888, 2013.
  506. Van Hove JL, Steyaert J, Matthijs G, et al: Expanded motor and psychiatric phenotype in autosomal dominant Segawa syndrome due to GTP cyclohydrolase deficiency, J Neurol Neurosurg Psychiatry 77(1):18–23, 2006.
  507. Volkman J, Wolters A, Kupsch A, et al: Pallidal deep brain stimulation in patients with primary generalized or segmental dystonia: 5-year follow-up of a randomized trial, Lancet 11:1029–1038, 2012. Waugh JL, Sharma N: Clinical neurogenetics: dystonia from phenotype to genotype, Neurol Clin 31:969–986, 2013.
  508. Albavera-Hernandez C, Rodriguez JM, Idrovo AJ: Safety of botulinum toxin type a among children with spasticity secondary to cerebral palsy: a systemic review of randomized clinical trials, Clin Rehabil 23:394–407, 2009.
  509. Bax M, Tydeman C, Flodmark O: Clinical and MRI correlates of cerebral palsy, JAMA 296:1602–1608, 2006.
  510. Benfer KA, Weir KA, Bell KL, et al: Oropharyngeal dysphagia and gross motor skills in children with cerebral palsy, Pediatrics 131:e1553–e1562, 2013.
  511. Benini R, Dagenais L, Shevell MI, et al: Normal imaging in patients with cerebral palsy: what does it tell us?, J Pediatr 162:369–374, 2013.
  512. Brunstrom JE, Bastion AJ, Wong M: Motor benefit from levodopa in spastic quadriplegic cerebral palsy, Ann Neurol 47:662–665, 2000.
  513. Chang CH, Chen YY, Yeh KK, Chen CL: Gross motor function change after multilevel soft tissue release in children with cerebral palsy, Biomed J. 40(3):163–168, 2017.
  514. Colver A, Fairhurst C, Pharoah POD: Cerebral palsy, Lancet 383:1240–1246, 2014.
  515. Daunter AK, Kratz AL, Hurvitz EA: Long-term impact of childhood selective dorsal rhizotomy on pain, fatigue, and function: a case-control study, Dev Med Child Neurol 59(10):1089–1095, 2017.
  516. Delgado MR, Hirtz D, Aisen M, et al: Practice parameter: pharmacologic treatment of spasticity in children and adolescents with cerebral palsy (an evidence-based review), Neurology 74:336–343, 2010.
  517. Doyle LW, Anderson PJ, Haslam R, et al: School-age outcomes of very preterm infants after antenatal treatment with magnesium sulfate vs placebo, JAMA 321:1105–1112, 2014.
  518. Eek MN: Muscle strength training to improve gait function in children with cerebral palsy, Dev Med Child Neurol 50:759–764, 2008.
  519. Gordon AM, Charles J, Wolf SL: Efficacy of constraint-induced movement therapy on involved upper-extremity use in children with hemiplegic cerebral palsy is not age-dependent, Pediatrics 117:363–373, 2006.
  520. Graham EM, Ruis KA, Hartman AL, et al: A systemic review of the role of intrapartum hypoxia-ischemia in the causation of neonatal encephalopathy, Am J Obstet Gynecol 199:587–595, 2008.
  521. Himmelmann K, McManus V, Hagberg G, et al: Dyskinetic cerebral palsy in Europe: trends in prevalence and severity, Arch Dis Child 94:921–926, 2009.
  522. Hoon AH Jr, Stashinko EE, Nagae LM, et al: Sensory and motor deficits in children with cerebral palsy born preterm correlate with diffusion tensor imaging abnormalities in thalamocortical pathways, Dev Med Child Neurol 51:697–704, 2009.
  523. Huntsman R, Lemire E, Norton J, et al: The differential diagnosis of spastic diplegia, Arch Dis Child 100:500–504, 2015.
  524. Johnston MV, Faemi A, Wilson MA, et al: Treatment advances in neonatal neuroprotection and neurointensive care, Lancet Neurol 10:372–382, 2011.
  525. Kurian MA, Gissen P, Smith M, et al: The monoamine neurotransmitter disorders: an expanding range of neurological syndromes, Lancet Neurol 10:721–731, 2011.
  526. Leach EL, Shevell M, Bowden K, et al: Treatable inborn errors of metabolism presenting as cerebral palsy mimics: systematic literature review, Orphanet J Rare Dis 9:197, 2014.
  527. Lee RW, Poretti A, Cohen JS, et al: A diagnostic approach for cerebral palsy in the genomic era, Neurol Med 16:821–844, 2014.
  528. Mugglestone MA, Eunson P, Murphy MS: Spasticity in children and young people with non-progressive brain disorders: summary of NICE guidelines, BMJ 345:e4845, 2012.
  529. Nagae LM, Hoon AH Jr, Stashinko E, et al: Diffusion tensor imaging in children with periventricular leukomalacia: variability of injuries to white matter tracts, AJNR Am J Neuroradiol 28:1213–1222, 2007.
  530. Nelson KB: Causative factors in cerebral palsy, Clin Obstet Gynecol 51:749–762, 2008.
  531. Novak I, Hines M, Goldsmith S, et al: Clinical prognostic messages from a systematic review on cerebral palsy, Pediatrics 130:e1285–e1312, 2012.
  532. Robertson CM, Watt MJ, Yasul Y: Changes in the prevalence of cerebral palsy for children born very prematurely within a population-based program over 30 years, JAMA 297:2733–2740, 2007.
  533. Robinson MN, Peake LJ, Ditchfield MR, et al: Magnetic resonance imaging findings in a population-based cohort of children with cerebral palsy, Dev Med Child Neurol 51:39–45, 2009.
  534. Rouse DJ, Hirtz DG, Thom E, et al: Eunice kennedy shriver NICHD Maternal-fetal medicine units network: a randomized, controlled trial of magnesium sulfate for the prevention of cerebral palsy, N Engl J Med 359:895–905, 2008.
  535. Sakzewski L, Ziciani J, Boyd R: Systematic review and meta-analysis of therapeutic management of upper-limb dysfunction in children with congenital hemiplegia, Pediatrics 123:e1111–e1122, 2009.
  536. Shevell MI, Dagenais L, Hall N, et al: The relationship of cerebral palsy subtype and functional motor impairment: a population-based study, Dev Med Child Neurol 51:872–877, 2009.
  537. Sterling C, Taub E, Davis D, et al: Structural neuroplastic change after constraint induced movement therapy in children with cerebral palsy, Pediatrics 131:e1664–e1669, 2013.
  538. Taylor CL, de Groot J, Blair EM, et al: The risk of cerebral palsy in survivors of multiple pregnancies with co-fetal loss or death, Am J Obstet Gynecol 201:41–47, 2009.
  539. Wu YW, Croen LA, Torres AR, et al: Interleukin-6 and risk for cerebral palsy in term and near-term infants, Ann Neurol 66:663–670, 2009.
  540. Yeargin-Allsopp M, Van Naarden Barun K, Doernberg N, et al: Prevalence of cerebral palsy in 8-year-old children in three areas of the United States in 2002: a multisite collaboration, Pediatrics 121:547–554, 2008.
  541. Bianchi MC, Sgandurra G, Tosetti M, et al: Brain magnetic resonance in the diagnostic evaluation of mitochondrial encephalopathies, Biosci Rep 27:69–95, 2007.
  542. Chicani CF, Chu ER, Miller G, Comparing EPI: 743 treatment in siblings with Leber’s hereditary optic neuropathy mt14484 mutation, Can J Ophthalmol 48(5):e130–e133, 2013.
  543. Cohen BH, Chinnery PF, Copeland WC: POLG-related disorders. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews [internet], Seattle (WA), 1993-2014, Seattle, March 16, 2010, University of Washington. [updated October 11, 2012].
  544. Crest C, Dupont S, Leguern E, et al: Levetiracetam in progressive myoclonic epilepsy: an exploratory study, Neurology 62:640–643, 2004.
  545. DiMauro S, Schon EA, Carelli V, et al: The clinical maze of mitochondrial neurology, Nat Rev Neurol 9:429–444, 2013.
  546. Donato S: Multisystem manifestations of mitochondrial disorders, J Neurol 256:693–710, 2009. Enns GM, Kinsman SL, Perlman SL, et al: Initial experience in the treatment of inherited mitochondrial disease with EPI-743, Mol Genet Metab 105(1):91–102, 2012.
  547. Eom S, Lee HN, Lee S, et al: Cause of death in children with mitochondrial diseases, Pediatr Neurol 66:82–88, 2017.
  548. Finsterer J, Ahting U: Mitochondrial depletion syndromes in children and adults, Can J Neurol Sci 40:635–644, 2013.
  549. Finsterer J: Leigh and Leigh-like syndrome in children and adults, Pediatr Neurol 39:223–235, 2008.
  550. Finsterer J: Management of mitochondrial stroke-like-episodes, Eur J Neurol 16: 1178–1184, 2009.
  551. Fryer RH, Bain J, De Vivo D: Mitochondrial encephalomyopathy lactic acidosis and stroke-like episodes: a case report and critical reappraisal of treatment options, Pediatr Neurol 56:59–61, 2016.
  552. Horvath R, Kemp JP, Tuppen HAL, et al: Molecular basis of infantile reversible cytochrome c oxidase deficiency myopathy, Brain 132:3165–3174, 2009.
  553. Kitamura M, Yatsuga S, Abe T, et al: L-arginine intervention at hyper-acute phase protects the prolonged MRI abnormality in MELAS, J Neurol 263(8):1666–1668, 2016.
  554. Koga Y, Akita Y, Nishioka J, et al: L-arginine improves the symptoms of stroke-like episodes in MELAS, Neurology 64:710–712, 2005.
  555. Martinelli D, Catteruccia M, Piemonte F, et al: EPI-743 reverses the progression of the pediatric mitochondrial disease–genetically defined leigh syndrome, Mol Genet Metab 107(3):383–388, 2012.
  556. Oldfors A, Tulinius M: Mitochondrial encephalopathies, J Neuropathol Exp Neurol 62:217–227, 2003.
  557. Pfeffer G, Majamaa K, Turnbull DM, et al: Treatment for mitochondrial disorders, Cochrane Database Syst Rev (4):CD004426, 2012.
  558. Scarpelli M, Todeschini A, Volonghi I, et al: Mitochondrial diseases: advances and issues, Appl Clin Genet 10:21–26, 2017.
  559. Taanman JW, Bodnar AG, Cooper JM, et al: Molecular mechanisms in mitochondrial DNA depletion syndrome, Hum Mol Genet 6(6):935–942, 1997.
  560. Tein I: Impact of fatty acid oxidation disorders in child neurology: from reye syndrome to Pandora’s box, Dev Med Child Neurol 57(4):304–306, 2015.
  561. Wani AA, Ahanger SH, Bapat SA, et al: Analysis of mitochondrial DNA sequences in childhood encephalomyopathies reveals new disease-associated variants, PLoS ONE 2:e942, 2007.
  562. Werner KG, Morel CF, Benseler SM, et al: Rolandic mitochondrial encephalomyelopathy and MT-ND3 mutations, Pediatr Neurol 41:27–33, 2009.
  563. Yatsuga S, Povalko N, Nishioka J, et al: MELAS: a nationwide prospective cohort study of 96 patients in Japan, Biochim Biophys Acta 1820:619–624, 2012.
  564. Agarwal A, Kapur G, Altinok D: Childhood posterior reversible encephalopathy syndrome: magnetic resonance imaging findings with emphasis on increased leptomeningeal FLAIR signal, Neuroradiol J. 28(6):638–643, 2015.
  565. Bergamino L, Capra V, Biancheri R, et al: Immunomodulatory therapy in recurrent acute necrotizing encephalopathy ANE1: is it useful?, Brain Dev 34:384–391, 2012.
  566. Britton PN, Dale RC, Blyth CC, et al: ACE study investigators and PAEDS network: Influenza-associated encephalitis/encephalopathy identified by the Australian childhood encephalitis study 2013-2015, Pediatr Infect Dis J 36(11):1021–1026, 2017.
  567. Donmez FY, Guleryuz P, Agildere M: MRI findings in childhood PRES: what is different than the adults?, Clin Neuroradiol 26(2):209–213, 2016.
  568. Gaynon PS, Angiolillo AL, Carroll WL, et al: Long-term results of the children’s cancer group studies for childhood acute lymphoblastic leukemia 1983–2002: a children’s oncology group report, Leukemia 24:285–297, 2010.
  569. Henneke M, Dickmann S, Ohlenbusch A, et al: RNASET2-deficient cystic leukoencephalopathy resembles congenital cytomegalovirus brain infection, Nat Genet 41:773–775, 2009.
  570. Hoshino A, Saitoh M, Oka A, et al: Epidemiology of acute encephalopathy in Japan, with emphasis on the association of viruses and syndromes, Brain Dev 34:337–343, 2012.
  571. Kastrup O, Gerwig M, Frings M, et al: Posterior reversible encephalopathy syndrome (PRES): electroencephalographic findings and seizure patterns, J Neurol 259:1383–1389, 2012.
  572. Lee VH, Wijidicks EFM, Manno EM, et al: Clinical spectrum of reversible posterior leukoencephalopathy syndrome, Arch Neurol 65:205–210, 2008.
  573. Loh NR, Appleton DB: Untreated recurrent acute necrotizing encephalopathy associated with RANBP2 mutation, and normal outcome in a caucasian boy, Eur J Pediatr 169:1299–1302, 2010.
  574. Morris B, Partap S, Yeom K, et al: Cerebrovascular disease in childhood cancer survivors: a Children’s oncology group report, Neurology 73:1906–1913, 2009.
  575. Morris EB, Laninigham FH, Sandlund JT, et al: Posterior reversible encephalopathy syndrome in children with cancer, Pediatr Blood Cancer 48:152–159, 2007.
  576. Neilson DE: The interplay of infection and genetics in acute necrotizing encephalopathy, Curr Opin Pediatr 22:751–757, 2010.
  577. Nielson DE, Adams MD, Orr C, et al: Infection-triggered familial or recurrent cases of acute necrotizing encephalopathy caused by mutation in a component of the nuclear pore, RANBP2, Am J Hum Genet 84:44–51, 2009.
  578. Onder AM, Lopez R, Teomete U, et al: Posterior reversible encephalopathy syndrome in the pediatric renal population, Pediatr Nephrol 22:1921–1929, 2007.
  579. Prasad N, Gulati S, Gupta RK, et al: Spectrum of radiological changes in hypertensive children with reversible posterior leukoencephalopathy, Br J Radiol 80:422–429, 2007.
  580. Wong AM, Simon EM, Zimmerman RA, et al: Acute necrotizing encephalopathy of childhood: correlation of MR findings and clinical outcome, AJNR Am J Neuroradiol 27:1919–1923, 2006.
  581. Yamamoto H, Natsume J, Kidokoro H, et al: Clinical and neuroimaging findings in children with posterior reversible encephalopathy syndrome, Eur J Paediatr Neurol 19(6):672–678, 2015.
  582. Amrom D, Kinay D, Hart Y, et al: Rasmussen encephalitis and comorbid autoimmune diseases, Neurology 83:1049–1055, 2014.
  583. Arino H, Armangu? T, Petit-Pedrol M, et al: Anti-LGI1-associated cognitive impairment: presentation and long-term outcome, Neurology 87:759–765, 2016.
  584. Armangue T, Spatola M, Vlagea A, et al: Frequency, symptoms, risk factors, and outcomes of autoimmune encephalitis after herpes simplex encephalitis: a prospective observational study and retrospective analysis, Lancet Neurol 17:760–772, 2018.
  585. Armangue T, Titulaer MJ, M?laga I, et al: Pediatric anti-N-methyl-D-aspartate receptor encephalitis-clinical analysis and novel findings in a series of 20 patients, J Pediatr 162:850.e2–856.e2, 2012.
  586. Armangue T, Titulaer MJ, Sabater L, et al: A novel treatment-responsive encephalitis with frequent opsoclonus and teratoma, Ann Neurol 75:850–856, 2014.
  587. Bien CG, Granata T, Antozzi C, et al: Pathogenesis, diagnosis and treatment of rasmussen encephalitis: a European consensus statement, Brain 128:454–471, 2005.
  588. Brenton JN, Goodkin HP: Antibody-mediated autoimmune encephalitis in childhood, Pediatr Neurol 60:13–23, 2016.
  589. Crow YJ: Type I interferonopathies: mendelian type I interferon up-regulation, Curr Opin Immunol 32:7–12, 2015.
  590. Dale RC, Brilot F, Duffy LV, et al: Utility and safety of rituximab in pediatric autoimmune and inflammatory CNS disease, Neurology 83:142–150, 2014.
  591. Dale RC, Merheb V, Pillai S, et al: Antibodies to surface dopamine-2 receptor in autoimmune movement and psychiatric disorders, Brain 135:3453–3468, 2012.
  592. Dalmau J, Geis C, Graus F: Autoantibodies to synaptic receptors and neuronal cellsurface proteins in autoimmune diseases of the central nervous system, Physiol Rev 97(2):839–887, 2017.
  593. Dalmau J, Gleichman AJ, Hughes EG, et al: Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies, Lancet Neurol 7:1091–1098, 2008.
  594. Dalmau J, Graus F: Antibody-mediated encephalitis, N Engl J Med 378(9):840–851, 2018.
  595. Dalmau J, Lancaster E, Martinez-Hernandez E, et al: Clinical experience and laboratory investigations in patients with anti-NMDAR encephalitis, Lancet Neurol 10:63–74, 2011.
  596. Deiva K, Mahlaoui N, Beaudonnet F, et al: CNS involvement at the onset of primary hemophagocytic lymphohistiocytosis, Neurology 78:1150–1156, 2012.
  597. Dogan M, Acikgoz E, Acikgoz M, et al: The frequency of hashimoto thyroiditis in children and the relationship between urinary iodine level and hashimoto thyroiditis, J Pediatr Endocrinol Metab 24:75–80, 2011.
  598. Erol I, Saygi S, Alehan F: Hashimoto’s encephalopathy in children and adolescents, Pediatr Neurol 45:420–422, 2011.
  599. Gelfand AA, Gelfand JM, Prabakhar P, et al: Ophthalmoplegic «migraine» or recurrent ophthalmoplegic cranial neuropathy: new cases and a systematic review, J Child Neurol 27:759–766, 2012. Graus F, Titulaer MJ, Balu R, et al: A clinical approach to diagnosis of autoimmune encephalitis, Lancet Neurol 15:391–404, 2016.
  600. Haberlandt E, Bast T, Ebner A, et al: Limbic encephalitis in children and adolescents, Arch Dis Child 96:186–191, 2011.
  601. Hacohen Y, Absoud M, Deiva K, et al: Myelin oligodendrocyte glycoprotein antibodies are associated with a non-MS course in children, Neurol Neuroimmunol Neuroinflamm 2:e81, 2015.
  602. Heine J, Pruss H, Bartsch T, et al: Imaging of autoimmune encephalitis—relevance for clinical practice and hippocampal function, Neurosci 309:68–83, 2015.
  603. Honnorat J, Didelot A, Karantoni E, et al: Autoimmune limbic encephalopathy and anti-hu antibodies in children without cancer, Neurology 80:2226–2232, 2013.
  604. Irani SR, Michell AW, Lang B, et al: Faciobrachial dystonic seizures precede lgi1 antibody limbic encephalitis, Ann Neurol 69:892–900, 2011.
  605. Ize-Ludlow D, Gray JA, Sperling MA, et al: Rapid-onset obesity with hypothalamic dysfunction, hypoventilation, and autonomic dysregulation presenting in childhood, Pediatrics 120:e179–e188, 2007.
  606. Kelley BP, Patel SC, Marin HL, et al: Autoimmune encephalitis: pathophysiology and imaging review of an overlooked diagnosis, AJNR Am J Neuroradiol 38(6):1070–1078, 2017.
  607. Krupp LB, Tardieu M, Amato MP, et al: International pediatric multiple sclerosis study group criteria for pediatric multiple sclerosis and immune-mediated central nervous system demyelinating disorders: revisions to the 2007 definitions, Mult Scler 19:1261–1267, 2013.
  608. Lagarde S, Villeneuve N, Tr?buchon A, et al: Anti-tumor necrosis factor alpha therapy (adalimumab) in Rasmussen’s encephalitis: an open pilot study, Epilepsia 57:956–966, 2016.
  609. Lancaster E, Martinez-Hernandez E, Titulaer MJ, et al: Antibodies to metabotropic glutamate receptor 5 in the ophelia syndrome, Neurology 77:1698–1701, 2011.
  610. Laurent C, Capron J, Quillerou B, et al: Steroid-responsive encephalopathy associated with autoimmune thyroiditis (STREAT): characteristics, treatment and outcome in 251 cases from the literature, Autoimmun Rev 15:1129–1133, 2016.
  611. Lilleker JB, Jones MS, Mohanraj R, et al: The relevance of VGKC positivity in the absence of LGI1 and caspr2 antibodies, Neurology 87:1848–1849, 2016.
  612. Lim M, Hachohen Y, Vincent A: Autoimmune encephalitis, Pediatr Clin North Am 62:667–685, 2015. Longoni G, Levy DM, Yeh EA: The changing landscape of childhood inflammatory central nervous system disorders, J Pediatr 179:24–32, 2016.
  613. McKeon A, Martinez-Hernandez E, Lancaster E, et al: Glycine receptor autoimmune spectrum with stiff-man syndrome phenotype, JAMA Neurol 70:44–50, 2013.
  614. Mitchell WG, Wooten AA, O’Neil SH, et al: Effect of increased immunosuppression on developmental outcome of opsoclonus myoclonus syndrome (OMS), J Child Neurol 30:976–982, 2015.
  615. Nandi-Munshi D, Taplin CE: Thyroid-related neurological disorders and complications in children, Pediatr Neurol 52:373–382, 2015.
  616. Odaka M, Yuki N, Yamada M, et al: Bickerstaff ’s brainstem encephalitis: clinical features of 62 cases and a subgroup associated with Guillain-barr? syndrome, Brain 126:2279–2290, 2003.
  617. Petit-pedrol M, Armangue T, Peng X, et al: Encephalitis with refractory seizures, status epilepticus, and antibodies to the GABA a receptor: a case series, characterisation of the antigen, and analysis of the eff ects of antibodies, Lancet Neurol 13:276–286, 2014.
  618. Pittock SJ, Debruyne J, Krecke KN, et al: Chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids (CLIPPERS), Brain 133:2626–2634, 2010.
  619. Planaguma J, Leypoldt F, Mannara F, et al: Human N-methyl d-aspartate receptor antibodies alter memory and behaviour in mice, Brain 138:94–109, 2015.
  620. Pranzatelli MR, Tate ED, Verhulst SJ, et al: Pediatric dosing of rituximab revisited: serum concentrations in opsoclonus-myoclonus syndrome, J Pediatr Hematol Oncol 32:e167–e172, 2010.
  621. Saiz A, Blanco Y, Sabater L, et al: Spectrum of neurological syndromes associated with glutamic acid decarboxylase antibodies: diagnostic clues for this association, Brain 131:2553–2563, 2008.
  622. Schmitt SE, Pargeon K, Frechette ES, et al: «Extreme delta brush»: a unique EEG pattern in adults with anti-NMDA receptor encephalitis, Neurology 79:1094–1100, 2012.
  623. Schubert D, Bode C, Kenefeck R, et al: Autosomal dominant immune dysregulation syndrome in humans with CTLA4 mutations, Nat Med 20:1410–1416, 2014.
  624. Singh RR, Sedani S, Lim M, et al: RANBP2 mutation and acute necrotizing encephalopathy: 2 cases and a literature review of the expanding clinico-radiological phenotype, Eur J Paediatr Neurol 19:106–113, 2015.
  625. Spatola M, Petit-Pedrol M, Simabukuro MM, et al: Investigations in GABAA receptor antibody-associated encephalitis, Neurology 88(11):1012–1020, 2017.
  626. Symmonds M, Waters PJ, K?ker W, et al: Anti-MOG antibodies with longitudinally extensive transverse myelitis preceded by CLIPPERS, Neurology 84:1177–1179, 2015.
  627. Titulaer MJ, H?ftberger R, Iizuka T, et al: Overlapping demyelinating syndromes and anti-N-methyl-d-aspartate receptor encephalitis, Ann Neurol 75:411–428, 2014.
  628. Titulaer MJ, McCracken L, Gabilondo I, et al: Treatment and prognostic factors for long-term outcome in patients with anti-NMDA receptor encephalitis: an observational cohort study, Lancet Neurol 12:157–165, 2013.
  629. Van BA, Stephani U, Kluger G, et al: FIRES: febrile infection responsive epileptic (FIRE) encephalopathies of school age, Brain Dev 31:91–93, 2009.
  630. Van Sonderen A, Petit-Pedrol M, Dalmau J, Titulaer MJ: The value of LGI1, Caspr2, and voltage-gated potassium channel (VGKC) antibodies in encephalitis, Nat Rev Neurol 13(5):290–301, 2017.
  631. Wingerchuk DM, Banwell B, Bennett JL, et al: International consensus diagnostic criteria for neuromyelitis optica spectrum disorders, Neurology 85:177–189, 2015. Zhou JY, Xu B, Lopes J, et al: Hashimoto encephalopathy: literature review, Acta Neurol Scand 135:285–290, 2017.
  632. Parenti G, Andria G, Ballabio A: Lysosomal storage diseases: from pathophysiology to therapy, Annu Rev Med 66:471–486, 2015.
  633. Platt FM: Sphingolipid lysosomal storage disorders, Nature 510:68–75, 2014.
  634. Rosenberg JB, Kaminsky SM, Aubourg P, et al: Gene therapy for metachromatic leukodystrophy, J Neuro Res 94:1169–1179, 2014.
  635. Sessa M, Lorioli L, Fumagalli F, et al: Lentiviral haemopoietic stem-cell gene therapy in early-onset metachromatic leukodystrophy: an ad-hoc analysis of a non-randomised, open-label, phase 1 /2 trial, Lancet 388:476–486, 2016.
  636. Cooper JD, Tarczyluk MA, Nelvagal HR: Towards a new understanding of NCLpathogenesis, Biochim Biophys Acta 1852:2256–2261, 2015.
  637. Geraets RD, Koh SY, Hastings ML, et al: Moving towards effective therapeutic strategies for neuronal ceroid lipofuscinosis, Orphanet J Rare Dis 11:40, 2016.
  638. Nickel M, Simonati A, Jacoby D, et al: Disease characteristics and progression in patients with late-infantile neuronal ceroid lipofuscionsis type 2 (CLN2) disease: an observational cohort study, Lancet Child Adolesc Health 2:582–590, 2018.
  639. Nita DA, Mole SE, Minassian BA: Neuronal ceroid lipofuscinoses, Epileptic Disord18(Suppl 2):S73–S88, 2016.
  640. Schulz A, Ajayi T, Specchio N, et al: Study of intravenous cerliponase alfa for CLN2 disease, N Engl J Med 378(20):1898–1906, 2018.
  641. Borden EJ, Annuziata I, d’Azzo A: Lysosomal multienzyme complex: pros and cons of working together, Cell Mol Life Sci 71:2017–2032, 2014.
  642. Caciotti A, Di Rocco M, Filocamo M, et al: Type II sialidosis: review of the clinical spectrum and identification of a new splicing defect with chitotriosidase assessment in two patients, J Neurol 256:1911–1915, 2009.
  643. d’Azzo A, Machado E, Annunziata I: Pathogenesis, emerging therapeutic targets and treatment in sialidosis, Expert Opin Orphan Drugs 3:491–504, 2015.
  644. Ramachandran N, Girard JM, Turnbull J, et al: The autosomal recessively inherited progressive myoclonus epilepsies and their genes, Epilepsia 50(Suppl 5):29–36, 2009.
  645. Arber CE, Li A, Houlden H, Wray S: Insights into molecular mechanisms of disease in neurodegeneration with brain iron accumulation:unifying theories, Neuropathol Appl Neurobiol 42:220–241, 2016.
  646. Charzewska A, Wierzba J, Izycka-Swieszewska E, et al: Hypomyelinating leukodystrophies—a molecular insight into the white matter pathology, Clin Genet 90:293–304, 2016.
  647. Helman G, Van Haren K, Escolar ML, Vanderver A: Emerging treatments for pediatric leukodystrophies, Pediatr Clin North Am 62:649–666, 2015.
  648. Kaler SG, et al: ATP7A-related copper transport disorders. In Pagon RA, Adam MP, Ardinger HH, editors: GeneReviews® [internet]. Seattle (WA), Seattle, 2003, University of Washington, pp 1993–2017. [updated August 18, 2016].
  649. Kaufmann WE, Stallworth JL, Everman DB, Skinner SA: Neurobiologically-based treatments in rett syndrome: opportunities and challenges, Expert Opin Orphan Drugs 4:1043–1055, 2016.
  650. Leonard H, Cobb S, Downs J: Clinical and biological progress over 50 years in rett syndrome, Nat Rev Neurol 13:37–51, 2017.
  651. Meyer E, Kurian MA, Hayflick SJ: Neurodegeneration with brain iron accumulation: genetic diversity and pathophysiological mechanisms, Annu Rev Genomics Hum Genet 16:257–279, 2015.
  652. Ma M, Adams HR, Seltzer LE, et al: Phenotype differentiation of FOXG1 and MECP2 disorders: a new method for characterization of developmental encephalopathies, J Pediatr 178:233–240, 2016.
  653. Moller LB, Lenartowicz M, Zabot MT, et al: Clinical expression of menkes disease in females with normal karyotype, Orphanet J Rare Dis 7:6, 2012.
  654. Osorio JM, Rowitch DH, Tesar P, et al: Concise review: stem cell-based treatment of pelizaeus merzbacher disease, Stem Cells 35:311–315, 2017.
  655. Parikh S, Bernard G, Leventer RJ, et al: A clinical approach to the diagnosis of patients with leukodystrophies and genetic leukoencephalopathies, Mol Gen Metab 114:501–515, 2018.
  656. Ramocki MB, Tavyev YJ, Peters SU: The MECP2 duplication syndrome, Am J Med Genet 152A:1079–1088, 2010.
  657. Smpokou P, Samanta M, Berry GT, et al: Menkes disease in affected females: the clinical disease spectrum, Am J Med Genet 167A:417–420, 2014.
  658. Srivastava S, Naidu S, et al: Alexander disease. In Pagon RA, Adam MP, Ardinger HH, editors: GeneReviews® [internet]. Seattle (WA), Seattle, 2002, University of Washington, pp 1993–2017. [updated January 8, 2015].
  659. Tumer Z: An overview and update of ATP7A mutation leading to menkes disease and occipital horn syndrome, Hum Mutat 34:417–429, 2013.
  660. van der Knaap MS, Pronk JC, Scheper GC: Vanishing white matter disease, Lancet Neurol 5:413–423, 2006.
  661. Wortham NC, Proud CG: eIF2B: recent structural and functional insights into a key regulator of translation, Biochem Soc Trans 43:1234–1240, 2015.
  662. Karussis D: The diagnosis of multiple sclerosis and the various related demyelinating syndromes: a critical review, J Autoimmun 48-49:132–142, 2014.
  663. Kitley J, Waters P, Woodhall M, et al: Neuromyelitis optica spectrum disorders with aquaporin-4 and myelin-oligodendrocyte glycoprotein antibodies: a comparative study, JAMA Neurol 71:276–283, 2015.
  664. Krupp LB, Tardieu M, Amato MP, et al: International pediatric multiple sclerosis study group criteria for pediatric multiple sclerosis and immune-mediated central nervous system demyelinating disorders: revision to the 2007 definitions, Mult Scler 19:1261–1267, 2013.
  665. O’Mahony J, Shroff M, Banwell B: Mimics and rare presentations of pediatric demyelination, Neuroimaging Clin North Am 23:321–336, 2013.
  666. Pohl D, Hennemuth I, von Kries R, Hanefield F: Paediatric multiple sclerosis and acute disseminated encephalomyelitis in Germany: results of a nationwide survey, Eur J Pediatr 166:405–412, 2007.
  667. Reindl M, Di Pauli F, Rostasy K, Berger T: The spectrum of MOG autoantibodyassociated demyelinating diseases, Nat Rev Neurol 9:455–461, 2013.
  668. Rostasy K, Bajer-Kornek B: Paediatric multiple sclerosis and other acute demyelinating diseases, Curr Opin Neurol 31:244–248, 2018.
  669. Tardieu M, Deiva K: Rare inflammatory diseases of the white matter and mimics of multiple sclerosis and related disorders, Neuropediatrics 44(6):302–308, 2013.
  670. Thompson AJ, Banwell BL, Barkhof F: Diagnosis of multiple sclerosis: 2017 revisions of the McDonald criteria, Lancet Neurol 17(2):162–173, 2018.
  671. Waldman A, Ghezzi A, Bar-Or A, et al: Multiple sclerosis in children: an update on clinical diagnosis, therapeutic strategies, and research, Lancet Neurol 13(9):936–948, 2014.
  672. Waldman AT, Chahin S, Lavery AM, et al: Binocular low-contrast letter acuity and the symbol digit modalities test improve the ability of the multiple sclerosis functional composite to predict disease in pediatric multiple sclerosis, Mult Scler Relat Disord 10:73–78, 2016.
  673. Duman E, Yorekli VA, Gencpinar P, et al: Unusual clinical cases that mimic acute disseminated encephalomyelitis, Acta Clin Croat 54(3):371–377, 2015.
  674. Krupp LB, et al: International pediatric multiple sclerosis study group criteria for pediatric multiple sclerosis and immune-mediated central nervous system demyelinating disorders: revisions to the 2007 definitions, Mult Scler 19:1261–1267, 2013.
  675. Pohl D, Alper G, Van Haren K, et al: Acute disseminated encephalomyelitis: updates on an inflammatory CNS syndrome, Neurology 87:S38–S45, 2016.
  676. Absoud M, Cummins C, Desai N, et al: Childhood optic neuritis clinical features and outcome, Arch Dis Child 96:860–862, 2011.
  677. Bonhomme GR, Waldman AT, Balcer LJ, et al: Pediatric optic neuritis: brain MRI abnormalities and risk of multiple sclerosis, Neurology 72:881–885, 2009.
  678. Bonhomme GR, Mitchell EB: Treatment of pediatric optic neuritis, Curr Treat Options Neurol 14:93–102, 2012.
  679. Eyre M, Hameed A, Wright S, et al: Retinal nerve fibre layer thinning is associated with worse visual outcome after optic neuritis in children with a relapsing demyelinating syndrome, Dev Med Child Neurol 2018. [Epub ahead of print].
  680. Perez-Cambrodi RJ, Cubillana AGH, et al: Optic neuritis in pediatric population: a review in current tendencies of diagnosis and management, J Optom 7(3):125–130, 2014.
  681. Waldman AT, Stull LB, Galetta SL, et al: Pediatric optic neuritis and risk of multiple sclerosis, J AAPOS 15:441–446, 2011.
  682. Absoud M, et al: Pediatric transverse myelitis, Neurology 87:S46–S52, 2016.
  683. Anderson EW, Kornberg AJ, Freeman JL, et al: Acute flaccid myelitis in childhood: a retrospective cohort study, Eur J Neurol 24(8):1077–1083, 2017.
  684. Banwell B, Dale RC: Understanding risk of relapse and risk of disability after childhood transverse myelitis, Neurology 84:332–334, 2015.
  685. Deiva K, Absoud M, Hemingway C, et al: Acute idiopathic transverse myelitis in children, Neurology 84:341–349, 2015.
  686. Greenberg BM: Treatment of acute transverse myelitis and its early complications, Continuum (Minneap Minn) 17:733–743, 2011.
  687. Mecharles S, Herrmann C, Poullain P, et al: Acute myelitis due to zika virus infection, Lancet 387:1481, 2016.
  688. Messacar K, Schreiner TL, Van Haren K, et al: Acute flaccid myelitis: a clinical review of US cases 2012-2015, Ann Neurol 80:326–338, 2016.
  689. Nelson GR, Bonkowsky JL, Doll E, et al: Recognition and management of acute flaccid myelitis in children, Pediatr Neurol 55:17–21, 2016.
  690. Outteryck O, Baille G, Hodel J, et al: Extensive myelitis associated with anti-NMDA receptor antibodies, BMC Neurol 13:211, 2013.
  691. Sorte DE, Poretti A, Newsome SD, et al: Longitudinally extensive myelopathy in children, Pediatr Radiol 45:244–257, 2015.
  692. Tyler KL: Rationale for the evaluation of fluoxetine in the treatment of enterovirus D68-associated acute flaccid myelitis, JAMA Neurol 72(5):493–494, 2015.
  693. Absoud M, Cummins C, Chong WK, et al: Paediatric UK demyelinating disease longitudinal study (PUDDLS), BMC Pediatr 11:68, 2011.
  694. Aubert-Broche B, Fonov V, Narayanan S, et al: Onset of multiple sclerosis before adulthood leads to failure of age-expected brain growth, Neurology 83:2140–2146, 2014.
  695. Aliaga E, Barkhof F: MRI mimics of multiple sclerosis, Handb Clin Neurol 122:291–316, 2014.
  696. Baumann M, Hennes EM, Schanda K: Children with multiphasic disseminated encephalomyelitis and antibodies to the myelin oligodendrocyte glycoprotein (MOG): extending the spectrum of MOG antibody positive diseases, Mult Scler 22(14):1821–1829, 2016.
  697. Brownlee WJ, Hardy TA, Fazekas F, Miller DH: Multiple sclerosis 1: diagnosis of multiple sclerosis: progress and challenges, Lancet 389:1336–1346, 2017.
  698. Comi G, Radaelli M, S?rensen PS: Evolving concepts in the treatment fo relapsing multiple sclerosis, Lancet 389:1347–1356, 2017.
  699. Fadda G, Brown RA, Longoni G, et al: MRI and laboratory features and the performance of international criteria in the diagnosis of multiple sclerosis in children and adolescents: a prospective cohort study, Lancet Child Adolesc 2:191–204, 2018.
  700. Graves JS, Barcellos LF, Shao X, et al: Genetic predictors of relapse rate in pediatric MS, Mult Scler 22:1528–1535, 2016.
  701. Hacohen Y, Mankad K, Chong WK, et al: Diagnostic algorithm for relapsing acquired demyelinating syndromes in children, Neurology 89(3):269–287, 2017.
  702. Hennes EM, Baumann M, Lechner C, Rostasy K: MOG spectrum disorders and role of MOG-antibodies in clinical practice, Neuropediatrics 49(1):3–11, 2018.
  703. Huppke P, Rostasy K, Karenfort M, et al: Acute disseminated encephalomyelitis followed by recurrent or monophasic optic neuritis in pediatric patients, Mult Scler 19:941–946, 2013.
  704. Kappos L, Bar-Or A, Cree BAC, et al: Siponimod versus placebo in secondary progressive multiple sclerosis (EXPAND): a double-blind, randomised, phase 3 study, Lancet 391:1263–1272, 2018.
  705. Karussis D, Petrou P: The spectrum of post-vaccination inflammatory CNS demyelinating syndromes, Autoimmun Rev 13:215–224, 2014.
  706. Kitley J, Waters P, Woodhall M, et al: Neuromyelitis optica spectrum disorders with aquaporin-4 and myelin-oligodendrocyte glycoprotein antibodies: a comparative study, JAMA Neurol 71:276–283, 2014.
  707. McGinley M, Rossman IT: Bringing the HEET: the argument for High-efficacy early treatment for Pediatric-onset multiple sclerosis, Neurother 14:985–998, 2017.
  708. Olsson T, Barcellos LF, Alfredsson L: Interactions between genetic, lifestyle and environmental risk factors for multiple sclerosis, Nat Rev Neurol 13:25–36, 2017.
  709. Ontaneda D, Thompson AJ, Fox RJ, Cohen JA: Progressive multiple sclerosis: prospects for disease therapy, repair, and restoration of function, Lancet 389:1357–1366, 2017.
  710. Rae-Grant A, Day GS, Marrie RA, et al: Comprehensive systematic review summary: disease-modifying therapies for adults with multiple sclerosis, Neurology 90:789–800, 2018.
  711. Rae-Grant A, Day GS, Marrie RA, et al: Practice guideline recommendations summary: disease-modifying therapies for adults with multiple sclerosis, Neurology 90:777–788, 2018.
  712. Reich DS, Lucchinetti CF, Calabresi PA: Multiple sclerosis, N Engl J Med 378(2):169–180, 2018.
  713. Thompson AJ, Banwell BL, Barkhof F, et al: Diagnosis of multiple sclerosis: 2017 revisions of the McDonald criteria, Lancet 17:162–173, 2018.
  714. Thompson AJ, Baranzini SE, Geurts J, et al: Multiple sclerosis, Lancet 391:1622–1636, 2018.
  715. Alvarenga MP, Fernandez O, Leyva L, et al: The HLA DRB1*03:01 allele is associated with NMO regardless of the NMO-IgG status in Brazilian patients from rio de janeiro, J Neuroimmunol 310:1–7, 2017.
  716. Asgari N, Jarius S, Laustrup H, et al: Aquaporin-4-autoimmunity in patients with systemic lupus erythematosus: a predominantly population-based study, Mult Scler 24(3):331–339, 2018.
  717. Banwell B, Bar-Or A, Arnold DL, et al: Clinical, environmental, and genetic determinants of multiple sclerosis in children with acute demyelination: a prospective national cohort study, Lancet Neurol 10:436–445, 2011.
  718. Dale RC, Brilot F, Duffy LV, et al: Utility and safety of rituximab in pediatric autoimmune and inflammatory CNS disease, Neurology 83:142–150, 2014.
  719. Hacohen Y, Absoud M, Woodhall M, et al: Autoantibody biomarkers in childhoodacquired demyelinating syndromes: results from a national surveillance cohort, J Neurol Neurosurg Psychiatry 85:456–461, 2014.
  720. Hacohen Y, Messina S, Gan HW, et al: Endocrinopathies in paediatric-onset neuromyelitis optica spectrum disorder with aquaporin 4 (AQP4) antibody, Mult Scler 24:679–684, 2018.
  721. Jurynczyk M, Geraldes R, Probert F, et al: Distinct brain imaging characteristics of autoantibody-mediated CNS conditions and multiple sclerosis, Brain 140:617–627, 2017.
  722. Jurynczyk M, Messina S, Woodhall MR, et al: Clinical presentation and prognosis in MOG-antibody disease: a UK study, Brain 140:3128–3138, 2017.
  723. Kim HJ, Paul F, Lana-Peixoto MA, et al: MRI characteristics of neuromyelitis optica spectrum disorder: an international update, Neurology 84:1165–1173, 2015.
  724. Kim SM, Kim SJ, Lee HJ, et al: Differential diagnosis of neuromyelitis optica spectrum disorders, Ther Adv Neurol Disord 10(7):265–289, 2017.
  725. Kitley J, Leite MI, Nakashima I, et al: Prognostic factors and disease course in aquaporin-4 antibody-positive patients with neuromyelitis optica spectrum disorder from the United Kingdom and Japan, Brain 135:1834–1849, 2012.
  726. Kitley J, Palace J: Therapeutic options in neuromyelitis optica spectrum disorders, Expert Rev Neurother 16:319–329, 2016.
  727. Lennon VA, WIngerchuck DM, Kryzer TA, et al: A serum autoantibody marker of neuromyelitis optica: distinction from multiple sclerosis, Lancet 364:2106–2112, 2004.
  728. Liu J, Shi Z, Lian Z, et al: Association of CD58 gene polymorphisms with NMO spectrum disorders in a han Chinese population, J Neuroimmunol 309:23–30, 2017.
  729. Nosadini M, Alper G, RIney CJ, et al: Rituximab monitoring and redosing in pediatric neuromyelitis optica spectrum disorder, Neurol Neuroimmunol Neuroinflamm 3:e188, 2016.
  730. Papadopoulos MC, Verkman AS: Aquaporin water channels in the nervous system, Nat Rev Neurosci 14:265–277, 2013.
  731. Wingerchuk DM, Banwell B, Bennett JL, et al: International consensus diagnostic criteria for neuromyelitis optica spectrum disorders, Neurology 85:177–189, 2015.
  732. Cobo-Calvo A, Ruiz A, Maillart E, et al: Clinical spectrum and prognostic value of CNS MOG autoimmunity in adults: the MOGADOR study, Neurology 90(21):e1858–e1869, 2018.
  733. Duignan S, Wright S, Rossor T, et al: Myelin oligodendrocyte glycoprotein and aquaporin-4 antibodies are highly specific in children with acquired demyelinating syndromes, Dev Med Child Neurol 60(9):958–962, 2018.
  734. Hacohen Y, Rossor T, Mankad K, et al: Leukodystrophy-like’ phenotype in children with myelin oligodendrocyte glycoprotein antibody-associated disease, Dev Med Child Neurol 60(4):417–423, 2018.
  735. Hacohen Y, Vincent A: Autoimmune neurological disorders-does the age matter?, Eur J Paediatr Neurol 22:341–343, 2018.
  736. Hacohen Y, Wong YY, Lechner C, et al: Disease course and treatment responses in children with relapsing myelin oligodendrocyte glycoprotein Antibody-associated disease, JAMA Neurol 75(4):478–487, 2018.
  737. Hennes EM, Baumann M, Schanda K, et al: Prognostic relevance of MOG antibodies in children with an acquired demyelinating syndrome, Neurology 89:900–908, 2017.
  738. Reindl M, Di Pauli F, Rostasy K, Berger T: The spectrum of MOG autoantibodyassociated demyelinating diseases, Nat Rev Neurol 9:455–461, 2013.
  739. AHA/ASA Scientific Statement 2013: Prevention and treatment of thrombosis in pediatric and congenital heart disease, Circulation 128(24):2622–2703, 2013.
  740. American College of Chest Physicians 2012: Antithrombotic therapy in neonates and children: Antithrombotic Therapy and Prevention of Thrombosis, ed 9, AmericanCollege of Chest Physicians Evidence-Based Clinical Practice Guidelines, Chest 2012. http://stroke.ahajournals.org/content/39/9/2644.full.pdf.
  741. Andrade A, Bigi S, Laughlin S, et al: Association between prolonged seizures and malignant middle cerebral artery infarction in children with acute ischemic stroke, Pediatr Neurol 64:44–51, 2016.
  742. Australian Childhood Stroke Advisory Committee 2017: Guideline for the diagnosis and acute management of childhood stroke.
  743. Baird LC, Smith ER, Ichord R, et al: Moyamoya syndrome associated with alagille syndrome: outcome after surgical revascularization, J Pediatr 166(2):470–473, 2015.
  744. Bernard TJ, Friedman NR, Stence NV, et al: Preparing for a «pediatric stroke Alert», Pediatr Neurol 56:18–24, 2016.
  745. Billinghurst LL, Beslow LA, Abend NS, et al: Incidence and predictors of epilepsy after pediatric arterial ischemic stroke, Neurology 88(7):630–637, 2017.
  746. Buompadre MC, Andres K, Slater LA, et al: Thrombectomy for acute stroke in childhood: a case report, Literature review, and recommendations, Pediatr Neurol 66:21–27, 2017.
  747. deVeber GA, Kirton A, Booth FA, et al: Epidemiology and outcomes of arterial ischemic stroke in children: the Canadian pediatric ischemic stroke registry, Pediatr Neurol 69:58–70, 2017.
  748. De Rosa S, Sievert H, Sabatino J, et al: Percutaneous closure versus medical treatment in stroke patients with patent foramen ovale, Ann Intern Med 168(5):343–350, 2018.
  749. Dunbar M, Kirton A: Perinatal stroke: mechanisms, management, and outcomes of early cerebrovascular brain injury, Lancet 2:666–676, 2018.
  750. Ferriero DM, Fullerton HJ, Bernard TJ, et al: on behalf of the American Heart Association: Management of stroke in neonates and children: A Scientific Statement From the American Heart Association/American Stroke Association, Stroke Council, and Council on Cardiovascular and Stroke Nursing, Stroke 50:2019.
  751. Fullerton HJ, Wintermark M, Hills NK, et al: Risk of recurrent arterial ischemic stroke in childhood: a prospective international study, Stroke 47(1):53–59, 2016.
  752. Greenham M, Gordon A, Anderson V, Mackay MT: Outcome in childhood stroke, Stroke 47:1159–1164, 2016.
  753. Guey S, Tournier-Lasserve E, Herve D, Kossorotoff M: Moyamoya disease and syndromes: from genetics to clinical management, Appl Clin Genet 8:49–68, 2015.
  754. Hankey GJ: Stroke, Lancet 389:641–652, 2017.
  755. Lee PH, Song JK, Kim JS, et al: Cryptogenic stroke and high-risk patent foramen ovale, J Am Coll Cardiol 71(20):2335–2342, 2018.
  756. Mackay MT, Yock-Corrales A, Churilov L, et al: Differentiating childhood stroke from mimics in the emergency department, Stroke 47(10):2476–2481, 2016.
  757. Madaelil TP, Kansagra AP, Cross DT, et al: Mechanical thrombectomy in pediatric acute ischemic stroke: clinical outcomes and literature reviews, Interv Neuroradiol 22(4):426–431, 2016.
  758. Mandalenakis Z, Rosengren A, Lappas G, et al: Ischemic stroke in children and young adults with congenital heart disease, J Am Heart Assoc 5(2):2016.
  759. Meuwissen ME, Halley DJ, Smit LS, et al: The expanding phenotype of COL4a1 and COL4a2 mutations: clinical data on 13 newly identified families and a review of the literature, Genet Med 17(11):843–853, 2015.
  760. Nzwalo H, Santos V, Gradil C, et al: Caucasian familial moyamoya syndrome with rare multisystemic malformations, Pediatr Neurol 48:240–243, 2013.
  761. Powers WJ, Rabinstein AA, Ackerson T, et al: 2018 guidelines for the early management of patients with acute ischemic stroke, Stroke 49:e1–e65, 2018.
  762. Siddiq I, Armstrong D, Surmava AM, et al: Utility of neurovascular imaging in acute neonatal arterial ischemic stroke, J Pediatr 188:110–114, 2017.
  763. Sinclair AJ, Fox CK, Ichord RN, et al: Stroke in children with cardiac disease: report from the international pediatric stroke study group symposium, Pediatr Neurol 52(1):5–15, 2015.
  764. Sondergaard CB, Nielsen JE, Hansen CK: Hereditary cerebral small vessel disease and stroke, Clin Neurol Neurosurg 155:45–57, 2017.
  765. Thomalla G, Simonsen CZ, Boutitie F, et al: MRI-guided thrombolysis for stroke with unknown time of onset, N Engl J Med 379(7):611–622, 2018.
  766. Carducci C, Colafati GS, Figa-Talamanca L, et al: Cerebral sinovenous thrombosis (CSVT) in children: what the pediatric radiologists need to know, Radiol Med 121(5):329–341, 2016.
  767. Kaseka ML, Moharir M, deVeber G, et al: Prognostication value of descending corticospinal tract DWI signal in neonatal cerebral sinovenous thrombosis, Pediatr Neurol 59:90–94, 2016.
  768. Moharir MD, Shroff M, Stephens D, et al: Anticoagulants in pediatric cerebral sinovenous thrombosis: a safety and outcome study, Ann Neurol 67:590–599, 2010.
  769. Monagle P, Chan AK, Goldenberg NA, et al: Antithrombotic therapy in neonates and children: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of chest physicians Evidence-based clinical practice guidelines, Chest 141:e737S–e801S, 2012.
  770. Petrov D, Uohara MY, Ichord R, et al: Pediatric cerebral sinovenous thrombosis following cranial surgery, Childs Nerv Syst 33(3):491–497, 2017.
  771. Suppiej A, Gentilomo C, Saracco P, et al: Paediatric arterial ischaemic stroke and cerebral sinovenous thrombosis. First report from the Italian registry of pediatric thrombosis (R. I. T. I., Registro italiano trombosi infantili), Thromb Haemost 113:1270–1277, 2015.
  772. Bansal S, Brown W, Dayal A, Carpenter JL: Posterior spinal cord infarction due to fibrocartilaginous embolization in a 16-year-old athlete, Pediatrics 134(1):e289–e292, 2014.
  773. Espinosa G, Mendizabal A, Minguez S, et al: Transverse myelitis affecting more than 4 spinal segments associated with systemic lupus erythematosus: clinical, immunological, and radiological characteristics of 22 patients, Semin Arthritis Rheum 39(4):246–256, 2010.
  774. Hsu CY, Cheng CY, Lee JD, et al: Clinical features and outcomes of spinal cord infarction following vertebral artery dissection: a systematic review of the literature, Neurol Res 35(7):676–683, 2013.
  775. Katramados AM, Rabah R, Adams MD, et al: Longitudinal myelitis, aseptic meningitis, and conus medullaris infarction as presenting manifestations of pediatric systemic lupus erythematosus, Lupus 17(4):332–336, 2008.
  776. Li XY, Xiao P, Xiao HB, et al: Myelitis in systemic lupus erythematosus frequently manifests as longitudinal and sometimes occurs at low disease activity, Lupus 23(11):1178–1186, 2014.
  777. Reisner A, Gary MF, Chern JJ, Grattan-Smith JD: Spinal cord infarction following minor trauma in children: fibrocartilaginous embolism as a putative cause, J Neurosurg Pediatr 11(4):445–450, 2013.
  778. Saison J, Costedoat-Chalumeau N, Maucort-Boulch D, et al: Systemic lupus erythematosus-associated acute transverse myelitis: manifestations, treatments, outcomes, and prognostic factors in 20 patients, Lupus 24(1):74–81, 2015.
  779. Beslow LA, Ichord RN, Gindville MC, et al: Frequency of hematoma expansion after spontaneous intracerebral hemorrhage in children, JAMA Neurol 71(2):165–171, 2014.
  780. Beslow LA, Abend NS, Gindville MC, et al: Pediatric intracerebral hemorrhage: acute symptomatic seizures and epilepsy, JAMA Neurol 70:448–454, 2013.
  781. Choquet H, Pawilkowska L, Lawton MT, Kim H: Genetics of cerebral cavernous malformations: current status and future prospects, J Neurosurg Sci 59(3):211–220, 2015.
  782. International multicenter cohort study of pediatric brain arteriovenous malformations. Part 2: Outcomes after stereotactic radiosurgery, Starke RM, Ding D, et al: J Neurosurg Pediatr 19(2):136–148, 2017.
  783. Khan NR, Tsivgoulis G, Lee SL, et al: Fibrinolysis for intraventricular hemorrhage-an updated meta-analysis and systematic review of the literature, Stroke 45:2662–2669, 2014.
  784. Knopman J, Stieg PE: Management of unruptured brain arteriovenous malformations, Lancet 383:581–583, 2014.
  785. Lo WD, Hajek C, Pappa C, et al: Outcomes in children with hemorrhagic stroke, JAMA Neurol 70:66–71, 2013.
  786. Macdonald RL, Schweizer TA: Spontaneous subarachnoid haemorrhage, Lancet 389:655–666, 2017.
  787. Nikolaev SI, Vetiska S, Bonilla X, et al: Somatic activating KRAS mutations in arteriovenous malformations of the brain, N Engl J Med 378(3):250–260, 2018.
  788. Pearl M, Gomez J, Gregg L, Gailloud P: Endovascular management of vein of galen aneurysmal malformations. Influence of the normal venous drainage on the choice of a treatment strategy, Childs Nerv Syst 26:1367–1379, 2010.
  789. Perry JJ, Stiell IG, Sivilotti MLA, et al: Clinical decision rules to rule out subarachnoid hemorrhages for acute headache, JAMA 310:1248–1254, 2013.
  790. Salman RA, White PM, Counsell CE, et al: Outcome after conservative management or intervention for unruptured brain arteriovenous malformations, JAMA 311:1661–1668, 2014.
  791. Singhal NS, Hills NK, Sidney S, Fullerton HJ: Role of trauma and infection in childhood hemorrhagic stroke due to vascular lesions, Neurology 81(6):581–584, 2013.
  792. Thompson BG, Brown RD Jr, Amin-Hanjani S, et al: Guidelines for the management of patients with unruptured intracranial aneurysms, Stroke 46(8):2368–2400, 2015.
  793. Yvon E, Lamotte D, Tiberghien A, et al: Long-term motor, functional, and academic outcome following childhood ischemic and hemorrhagic stroke: a large rehabilitation center-based retrospective study, Dev Neurorehabil 14:1–8, 2016.
  794. Alink J, de Vries TW: Unexplained seizures, confusion or hallucinations: think Hashimoto encephalopathy, Acta Paediatr 97:451–453, 2008.
  795. Aviv RI, Benseler SM, Silverman ED, et al: MR imaging and angiography of primary CNS vasculitis of childhood, AJNR Am J Neuroradiol 27:192–199, 2006.
  796. Benseler SM, deVeber G, Hawkins C, et al: Angiography-negative primary central nervous system vasculitis in children: a newly recognized inflammatory central nervous system disease, Arthritis Rheum 52:2159–2167, 2005.
  797. Benseler SM, Silverman E, Aviv RI, et al: Primary central nervous system vasculitis in children, Arthritis Rheum 54:1291–1297, 2006.
  798. Bernard TJ, Manco-Johnson MJ, Lo W, et al: Towards a consensus-based classification of childhood arterial ischemic stroke, Stroke 43:371–377, 2012.
  799. Calabrese LH, Furlan AJ, Gragg LA, et al: Primary angiitis of the central nervous system: diagnostic criteria and clinical approach, Cleve Clin J Med 59:293–306, 1992.
  800. Calabrese LH, Mallek JA: Primary angiitis of the central nervous system. Report of 8 new cases, review of the literature, and proposal for diagnostic criteria, Medicine (Baltimore) 67:20–39, 1988.
  801. Cellucci T, Tyrrell PN, Pullenayegum E, et al: von Willebrand factor antigen—a possible biomarker of disease activity in childhood central nervous system vasculitis? Rheumatology 51:1838–1845, 2012.
  802. Chabrier S, Rodesch G, Lasjaunias P, et al: Transient cerebral arteriopathy: a disorder recognized by serial angiograms in children with stroke, J Child Neurol 13:27–32, 1998.
  803. Elbers J, Armstrong D, Yau I, Benseler S: Vascular imaging outcomes of childhood primary angiitis of the central nervous system, Pediatr Neurol 63:53–59, 2016.
  804. Elbers J, Halliday W, Hawkins C, et al: Brain biopsy in children with primary small-vessel central nervous system vasculitis, Ann Neurol 68:602–610, 2010.
  805. Eleftheriou D, Cox T, Saunde rs D, et al: Investigation of childhood central nervous system vasculitis: magnetic resonance angiography versus catheter cerebral angiography, Dev Med Child Neurol 52:863–867, 2010.
  806. Gallagher KT, Shaham B, Reiff A, et al: Primary angiitis of the central nervous system in children: 5 cases, J Rheumatol 28:616–623, 2001.
  807. Gowdie P, Twilt M, Benseler SM: Primary and secondary central nervous system vasculitis, J Child Neurol 27:1448–1459, 2012.
  808. Granerod J, Ambrose HE, Davies NW, et al: UK health protection agency (HPA) aetiology of encephalitis study group. Causes of encephalitis and differences in their clinical presentations in England: a multicentre, population-based prospective study, Lancet Infect Dis 10(12):835–844, 2010.
  809. Honda M, Koga M, Kanda T: Treatment for central nervous system vasculitis, Brain Nerve 67:287–293, 2015.
  810. Hutchinson C, Elbers J, Halliday W, et al: Treatment of small vessel primary CNS vasculitis in children: an open-label cohort study, Lancet Neurol 9:1078–1084, 2010.
  811. Katsetos CD, Poletto E, Poletto E, et al: Childhood primary angiitis of the central nervous system with metachronous hemorrhagic infarcts: a postmortem study with clinicopathologic correlation, Semin Pediatr Neurol 21:184–194, 2014.
  812. Koker W, Gaertner S, Nagele T, et al: Vessel wall contrast enhancement: a diagnostic sign of cerebral vasculitis, Cerebrovasc Dis 26:23–29, 2008.
  813. Lanthier S, Armstrong D, Domi T, et al: Post-varicella arteriopathy of childhood: natural history of vascular stenosis, Neurology 64:660–663, 2005.
  814. Lanthier S, Lortie A, Michaud J, et al: Isolated angiitis of the CNS in children, Neurology 56:837–842, 2001.
  815. Mineyko A, Kirton A: Mechanisms of pediatric cerebral arteriopathy: an inflammatory debate, Pediatr Neurol 48:14–23, 2013.
  816. Pagnoux C, Hajj-Ali RA: Pharmacological approaches to CNS vasculitis: where are we at now?, Expert Rev Clin Pharmacol. 9:109–116, 2016.
  817. Salvarani C, Brown RD Jr, Hunder GG: Adult primary central nervous system vasculitis, Lancet 380:767–776, 2012.
  818. Salvarani C, Pipitone N, Hunder GG: Management of primary and secondary central nervous system vasculitis, Curr Opin Rheumatol 28:21–28, 2016.
  819. Torres J, Loomis C, Cucchiara B, et al: Diagnostic yield and safety of brain biopsy for suspected primary central nervous system angiitis, Stroke 47:2127–2129, 2016.
  820. Twilt M, Benseler SM: The spectrum of CNS vasculitis in children and adults, Nat Rev Rheumatol 8:97–107, 2012.
  821. Venkateswaran S, Hawkins C, Wassmer E: Diagnostic yield of brain biopsies in children presenting to neurology, J Child Neurol 23:253–258, 2008.
  822. Aku FY, Lessa FC, Asiedu-Bekoe F, et al: Meningitis outbreak caused by vaccinepreventable bacterial pathogens Northern Ghanam 2016, MMWR 66(30):806–810, 2017.
  823. Avery RA, Shah SS, Licht DJ, et al: Reference range for cerebrospinal fluid opening pressure in children, N Engl J Med 363:891–893, 2010.
  824. Brouwer MC, McIntyre P, Prasad K, van de Beek D: Corticosteroids for acute bacterial meningitis, Cochrane Database Syst Rev (9):CD004405, 2015.
  825. Curtis S, Stobart K, Vandermeer B, et al: Clinical features suggestive of meningitis in children: a systematic review of prospective data, Pediatrics 126:952–960, 2010.
  826. Dando SJ, Mackay-Sim A, Norton R, et al: Pathogens penetrating the central nervous system: infection pathways and the cellular and molecular mechanisms of invasion, Clin Microbiol Rev 27:691–726, 2014.
  827. Egelund GB, Ertner G, Kristensen KL, et al: Cerebrospinal fluid pleocytosis in infectious and noninfectious central nervous system disease—a retrospective cohort study, Medicine (Baltimore) 96:18(e6686), 2017.
  828. Glimaker M, Johansson B, Grindborg O, et al: Adult bacterial meningitis: earlier treatment and improved outcome following guideline revision promoting prompt lumbar puncture, Clin Infect Dis 60(8):1162–1169, 2015.
  829. Guedj R, Chappuy H, Titomanlio L, et al: Risk of bacterial meningitis in children 6 to 11 months of age with a first simple febrile seizure: a retrospective, cross-sectional, observational study, Acad Emerg Med 22(11):1290–1297, 2015.
  830. Kanegaye JT, Soliemanzadeh P, Bradley JS: Lumbar puncture in pediatric bacterial meningitis: defining the time interval for recovery of cerebrospinal fluid pathogens after parenteral antibiotic pretreatment, Pediatrics 108(5):1169–1174, 2001.
  831. Lyons TW, Cruz AT, Freedman SB, et al: Interpretation of cerebrospinal fluid white blood cell counts in young infants with a traumatic lumbar puncture, Ann Emerg Med 69(5):622–631, 2017.
  832. MacNeil JR, Bennett N, Farley MM, et al: Epidemiology of infant meningococcal disease in the United States, 2006–2012, Pediatrics 135:e305–e311, 2015.
  833. Martinez E, Mintegi S, Vilar B, et al: Prevalence and predictors of bacterial meningitis in young infants with fever without a source, Pediatr Infect Dis J 34:494–498, 2015.
  834. McIntyre PB, O’Brien KL, Greenwood B, et al: Effect of vaccines on bacterial meningitis worldwide, Lancet 380:1703–1710, 2012.
  835. Nigrovic LE, Malley R, Macias CG, et al: Effect of antibiotic pretreatment on cerebrospinal fluid profiles of children with bacterial meningitis, Pediatrics 122:726–730, 2008.
  836. Olarte L, Barson WJ, Barson RM, et al: Impact of the 13-valent pneumococcal conjugate vaccine on pneumococcal meningitis in US children, Clin Infect Dis 61(5):767–775, 2015.
  837. Oliveria CR, Morriss MC, Mistrot JG, et al: Brain magnetic resonance imaging of infants with bacterial meningitis, J Pediatr 165:134–139, 2014.
  838. Ouchenir L, Renaud C, Khan S, et al: The epidemiology, management, and outcomes of bacterial meningitis in infants, Pediatrics 140(1):e20170476, 2017.
  839. Thomson J, Sucharew H, Cruz AT, et al: Cerebrospinal fluid reference values for young infants undergoing lumbar puncture, Pediatrics 141(3):e20173405, 2018.
  840. Tunkel AR, Hartman BJ, Kaplan SL, et al: Practice guideline for the management of bacterial meningitis, Clin Infect Dis 39:1267–1284, 2004.
  841. van de Beek D, Brouwer M, Hasbun R, et al: Community-acquired bacterial meningitis, Nat Rev Dis Primers 2:16074, 2016.
  842. van de Beek D, Cabellos C, Dzupova O, et al: ESCMID Study Group for Infections of the Brain (ESGIB). ESCMID guideline: diagnosis and treatment of acute bacterial meningitis, Clin Microbiol Infect 22(Suppl 3):S37–S62, 2016.
  843. Weinberger DM, Malley R, Lipsitch M: Serotype replacement in disease after pneumococcal vaccination, Lancet 378:1962–1973, 2011.
  844. Al-Zaidy SA, MacGregor D, Mahant S, et al: Neurological complications of PCRproven M. pneumoniae infections in children: prodromal illness duration may reflect pathogenetic mechanism, Clin Infect Dis 61:1092–1098, 2015.
  845. Bloch KC, Glaser CA: Encephalitis surveillance through the Emerging Infections Program, 1997-2010, Emerg Infect Dis 21:1562–1567, 2015.
  846. Fowler A, St?dberg T, Eriksson M, et al: Long-term outcomes of acute encephalitis in childhood, Pediatrics 126:e828–e835, 2010.
  847. Gable MS, Sheriff H, Dalmau J, et al: The frequency of autoimmune N-methyl-Daspartate receptor encephalitis surpasses that of individual viral etiologies in young individuals enrolled in the California Encephalitis Project, Clin Infect Dis 54:899–904, 2012.
  848. Geradin P, Couderc T, Bintner M, et al: Chikungunya virus-associated encephalitis, Neurology 86:1–9, 2016.
  849. Glaser CA, Honarmand S, Anderson LJ, et al: Beyond viruses: clinical profiles and etiologies associated with encephalitis, Clin Infect Dis 43:1565–1577, 2006.
  850. Ka A, Britton P, Troedson C, et al: Mild encephalopathy with reversible splenial lesion: an important differential of encephalitis, Eur J Paediatr Neurol 19(3):377–382, 2015.
  851. Kneen R, Michael BD, Menson E, et al: Management of suspected viral encephalitis in children—association, J Infect 64:449–477, 2012.
  852. Krow-Lucal E, Lindsey NP, Lehman J, et al: West Nile Virus and Other Nationally Notifiable Arboviral Diseases—United States, 2015, MMWR Morb Mortal Wkly Rep 66:51–55, 2017.
  853. Pillai SC, Hacohen Y, Tantsis E, et al: Infectious and autoantibody-associated encephalitis: clinical features and long-term outcome, Pediatrics 135:e974–e984, 2015.
  854. Rao S, Elkon B, Flett KB, et al: Long-term outcomes and risk factors associated with acute encephalitis in children, J Pediatr Infect Dis Soc 6(1):20–27, 2017.
  855. Rust RS: Human arboviral encephalitis, Semin Pediatr Neurol 19:130–151, 2012.
  856. Thompson C, Kneen R, Riordan A, et al: Encephalitis in children, Arch Dis Child 2:150–161, 2012.
  857. Tunkel AR, Glaser CA, Bloch KC, et al: The management of encephalitis: clinical practice guidelines by the Infectious Diseases Society of America, Clin Infect Dis 47:303–327, 2008.
  858. Tutolo JW, Staples E, Sosa L, Bennett N: Powassan virus disease in an infant—Connecticut, MMWR 66(15):408–409, 2017.
  859. Tyler KL: Acute viral encephalitis, N Engl J Med 379(6):557–566, 2018.
  860. Venkatesan A, Tunkel AR, Bloch KC, et al: Case definitions, diagnostic algorithms, and priorities in encephalitis: consensus statement of the international encephalitis consortium, Clin Infect Dis 57:1114–1128, 2013.
  861. Vora NM, Holman RC, Mehal JM, et al: Burden of encephalitis-associated hospitalizations in the United States, 1998-2010, Neurology 82:443–451, 2014.
  862. Yokoyama T, Yamada S, Doichi N, Kato E: Rotavirus-infected children with clinically mild encephalopathy with a reversible splenial lesion (MERS), BMJ Case Rep 2013.
  863. Graeff-Teixeira C, Aramburu da Silva AC, Yoshimura K: Update on eosinophilic meningoencephalitis and its clinical relevance, Clin Microbiol Rev 22:322–348, 2009.
  864. Kawakami V, Casto A, Natarajan N, et al: Baylisascaris procyonis encephalomyelitis in a toddler—King County, Washington, 2017, MMWR 67(2):79–80, 2018.
  865. Murray WJ, Kazacos KR: Raccoon roundworm encephalitis, Clin Infect Dis 39: 1484–1492, 2004.
  866. Ramirez-Avila L, Slome S, Schuster FL, et al: Eosinophilic meningitis due to Angiostrongylus and Gnathostoma species, Clin Infect Dis 48:322–327, 2009.
  867. Sircar AD, Abanyie F, Blumberg D, et al: Raccoon roundworm infection associated with central nervous system disease and ocular disease—six states, 2013-2015, MMWR 65(35):930–933, 2016.
  868. Thanaviratananich S, Thanaviratananich S, Ngamjarus C: Corticosteroids for parasitic eosinophilic meningitis, Cochrane Database Syst Rev (2):CD009088, 2015.
  869. Brook I: Microbiology and treatment of brain abscess, J Clin Neurosci 38:8–12, 2017.
  870. Brouwer MC, Coutinho JM, van de Beek D: Clinical characteristics and outcome of brain abscess: systematic review and meta-analysis, Neurology 82(9):806–813, 2014.
  871. Brouwer MC, Tunkel AR, McKhann GM, et al: Brain abscess, N Engl J Med 371:447–456, 2014.
  872. Cole TS, Clark ME, et al: Pediatric focal intracranial suppuration: a UK single-center experience, Childs Nerv Syst 28:2109–2114, 2012.
  873. Felsenstein S, Williams B, Shingadia D, et al: Clinical and microbiologic features guiding treatment recommendations for brain abscesses in children, Pediatr Infect Dis J 32(2):129–135, 2013.
  874. Keller PM, Rampini SK, Bloemberg GV: Detection of mixed infection in a culturenegative brain abscess by broad-spectrum bacterial 16s rRNA gene PCR, J Clin Microbiol 48(6):2250, 2010.
  875. Patel K, Clifford DB: Bacterial brain abscess, Neurohospitalist 4(4):196–204, 2014.
  876. Saez-Llorens X, Nieto Guevara J: Parameningeal infections. In Feigin RD, Cherry JD, Demmler-Harrison GJ, et al, editors: Feigin and Cherry’s textbook of pediatric infectious diseases, ed 7, Ch. 32, Philadelphia, 2013, Saunders Elsevier.
  877. Shachor-Meyouhas Y, Bar-Joseph G, et al: Brain abscess in children–epidemiology, predisposing factors and management in the modern medicine era, Acta Paediatr 99:1163–1167, 2010.
  878. Agid R, Farb RI: Neuroimaging in the diagnosis of idiopathic intracranial hypertension, Minerva Med 97:365–370, 2006.
  879. Avery RA, Shah SS, Licht DJ, et al: Reference range for cerebrospinal fluid opening pressure in children, N Engl J Med 363:891–893, 2010.
  880. Babiker MO: How high is ‘high’ CSF opening pressure in paediatric pseudotumor cerebri?, Eur J Paediatr Neurol 21:248–249, 2017.
  881. Cook G, Tagg T: Idiopathic intracranial hypertension and autonomic neuropathy as the heralding manifestations of neuro-Behcet’s, Neurology 88(16 Suppl):P5–P312, 2017.
  882. Digre KB, Nakamoto BK, Warner JEA, et al: A comparison of idiopathic intracranial hypertension with and without papilledema, Headache 49(2):185–193, 2009.
  883. Friedman DI, Liu GT, Digre KB: Revised diagnostic criteria for the pseudotumor cerebri syndrome in adults and children, Neurology 81:1159–1165, 2013.
  884. Gilbert AL, Heidary G: Update on the evaluation of paediatric idiopathic intracranial hypertension, Curr Opin Ophthalmol 27:493–497, 2016.
  885. Hartmann AJ, Soares BP, Bruce BB, et al: Imaging features of idiopathic intracranial hypertension in children, J Child Neurol 32:120–126, 2017.
  886. Johnson LN, Diehl ML, Hamm CW, et al: Differentiating optic disc edema from optic nerve head drusen on optical coherence tomography, Arch Ophthalmol 127:45–49, 2009.
  887. Kayhanian S, Young AMH, Piper RJ, et al: Radiological correlates of raised intracranial pressure in children: a review, Front Pediatr 6:32, 2018.
  888. Krishnakumar D, Pickard JD, Czosnyka Z, et al: Idiopathic intracranial hypertension in childhood: pitfalls in diagnosis, Dev Med Child Neurol 56(8):749–755, 2014.
  889. Matthews YY, Dean F, Lim MJ, et al: Pseudotumor cerebri syndrome in childhood: incidence, clinical profile and risk factors in national prospective population-based cohort study, Arch Dis Child 102(8):715–721, 2017.
  890. Mishra A, Mordekar SR, Rennie IG, Baxter P: False diagnosis of papilloedema and idiopathic intracranial hypertension, Eur J Paediatr Neurol 11:39–42, 2007.
  891. Mollan SP, Ali F, Hassan-Smith G, et al: Evolving evidence in adult idiopathic intracranial hypertension: pathophysiology and management, J Neurol Neurosurg Psychiatry 87(9):982–992, 2016.
  892. Raffiz M, Abdullah JM: Optic nerve sheath diameter measurement: a means of detecting raised ICP in adult traumatic and non-traumatic neurosurgical patients, Am J Emerg Med 35:150–153, 2017.
  893. Wall M: Update on idiopathic intracranial hypertension, Neurol Clin 35:45–57, 2017.
  894. Albert GW: Spine ultrasounds should not be routinely performed for patients with simple sacral dimples, Acta Paediatr 105(8):890–894, 2016.
  895. Hertzler DA 2nd, DePowell JJ, Stevenson CB, Mangano FT: Tethered cord syndrome: a review of the literature from embryology to adult presentation, Neurosurg Focus 29(1):E1, 2010.
  896. Pang D: Split cord malformation: part II: clinical syndrome, Neurosurgery 31:481–500, 1992.
  897. Proctor MR, Scott RM: Long-term outcome for patients with split cord malformation, Neurosurg Focus 10:e5, 2001.
  898. Magge SN, Smyth MD, Governale LS, et al: Idiopathic syrinx in the pediatric populations: a combined center experience, J Neurosurg Pediatr 7(1):30–36, 2011.
  899. Milhorat TH: Classification of syringomyelia, Neurosurg Focus 8:E1, 2000.
  900. Pindrik J, Johnston JM Jr: Clinical presentation of chiari I malformation and syringomyelia in children, Neurosurg Clin North Am 26(4):509–514, 2015.
  901. Singhal A, Bowen-Roberts T, Steinbok P, et al: Natural history of untreated syringomyelia in pediatric patients, Neurosurg Focus 31(6):E13, 2011.
  902. Hsu W, Jallo GI: Pediatric spinal tumors, Handb Clin Neurol 112:959–965, 2013.
  903. Ravindra VM, Eli IM, Schmidt MH, Brockmeyer DL: Primary osseous tumors of the pediatric spinal column: review of pathology and surgical decision making, Neurosurg Focus 41(2):E3, 2016.
  904. Flores BC, Klinger DR, White JA, Batjer HH: Spinal vascular malformations: treatment strategies and outcome, Neurosurg Rev 40(1):15–28, 2017.

Òðóäû îòå÷åñòâåííûõ àâòîðîâ ïðè ïîäãîòîâêå ñòàòåé ïî ïåäèàòðèè äëÿ ïîëüçîâàòåëåé ñàéòà - íå èñïîëüçîâàëèñü, òàê êàê èõ âû âñåãäà ìîæåòå íàéòè â áèáëèîòåêàõ.

- Âåðíóòüñÿ â ðàçäåë "Ïåäèàòðèÿ"

Ðåäàêòîð: Èñêàíäåð Ìèëåâñêè. Äàòà îáíîâëåíèÿ ïóáëèêàöèè: 8.7.2024

Ìåäóíèâåð Ìû â Telegram Ìû â YouTube Ìû â VK Ôîðóì êîíñóëüòàöèé âðà÷åé Êîíòàêòû, ðåêëàìà
Èíôîðìàöèÿ íà ñàéòå ïîäëåæèò êîíñóëüòàöèè ëå÷àùèì âðà÷îì è íå çàìåíÿåò î÷íîé êîíñóëüòàöèè ñ íèì.
Ñì. ïîäðîáíåå â ïîëüçîâàòåëüñêîì ñîãëàøåíèè.