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 è ïîäðîñòêîâàÿ ñòîìàòîëîãèÿ
Äåòñêàÿ êàðäèîëîãèÿ
Äåòñêàÿ ïóëüìîíîëîãèÿ
Äåòñêàÿ ðåâìàòîëîãèÿ
Äåòñêàÿ ôàðìàêîëîãèÿ
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 âðîæäåííûì íàðóøåíèÿì îáìåíà ó äåòåé

 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
  • 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
  • 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
  • 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
  • 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
  • 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
  • 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
  • ×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 âðîæäåííûì íàðóøåíèÿì îáìåíà ó äåòåé äëÿ ïîëüçîâàòåëåé ñàéòà ÌåäÓíèâåð èñïîëüçîâàíû òðóäû ñëåäóþùèõ àâòîðîâ:

  1. American Academy of Pediatrics: Ethical and policy issues in genetic testing and screening of children, Pediatrics 131:620–622, 2013.
  2. Andersson HC: Newborn screening for spinal muscular atrophy and lysosomal storage disorders takes advantage of novel therapies, J Pediatr 190:9–10, 2017.
  3. Baily MA, Murray TH, editors: Ethics and newborn genetic screening: new technologies, new challenges, Baltimore, 2009, Johns Hopkins University Press.
  4. Bennett MJ: Newborn screening for metabolic diseases: saving children’s lives and improving outcomes, Clin Biochem 47:693–694, 2014.
  5. Landau YE, Lichter-Konecki U, Levy HL: Genomics in newborn screening, J Pediatr 164:14–19, 2014.
  6. Blau N, van Spronsen FJ, Levy HL: Phenylketonuria, Lancet 376:1417–1427, 2010.
  7. Burton BK, Adams DJ, Grange DK, et al: Tetrahydrobiopterin therapy for phenylketonuria in infants and young children, J Pediatr 158:410–415, 2011.
  8. Camp KM, Parisi MA, Acosta PB, et al: Phenylketonuria Scientific Review Conference: state of the science and future research needs, Mol Genet Metab 112:87–122, 2014.
  9. Cederbaum S: Tetrahydrobiopterin and PKU: into the future, J Pediatr 158:351–353, 2011.
  10. Cleary M, Trefz F, Muntau AC, et al: Fluctuations in phenylalanine concentrations in phenylketonuria: a review of possible relationships with outcomes, Mol Genet Metab 110(4):418–423, 2013.
  11. Committee on Genetics: Maternal phenylketonuria, Pediatrics 122:445–449, 2008.
  12. Feillet F, van Spronsen FJ, MacDonald A, et al: Challenges and pitfalls in the management of phenylketonuria, Pediatrics 126:333–341, 2010.
  13. Jahja R, Huijbregts SC, de Sonneville LM, et al: Mental health and social functioning in early treated phenylketonuria: the PKU-COBESO study, Mol Genet Metab 110(Suppl):S57–S61, 2013.
  14. Levy HL: Congenital heart disease in maternal PKU, Mol Genet Metab 107:648–649, 2012.
  15. Mires A, Beveda MD, Leis MR, et al: Risk factors for developing mineral bone disease in phenylketonuric patients, Mol Genet Metab 108:149–154, 2013.
  16. Mitchell JJ: Phenylalanine hydroxylase deficiency. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews [Internet], Seattle, 2000 [updated 2013], University of Washington, Seattle. http://www.ncbi.nlm.nih.gov/books/NBK1504/. 1993–2014.
  17. Singh RH, Rohr F, Frazier D, et al: Recommendations for the nutrition management of phenylalanine hydroxylase deficiency, Genet Med 16:121–131, 2014.
  18. Van Spronsen FJ, de Groot MJ, Hoeksma M, et al: Large neutral amino acids in the treatment of PKU: from theory to practice, J Inherit Metab Dis 33(6):671–676, 2010.
  19. Bendadi F, de Koning TJ, Visser G, et al: Impaired cognitive functioning in patients with tyrosinemia type 1 receiving nitisinone, J Pediatr 164:398–401, 2014.
  20. David CV: Oculocutaneous albinism, Cutis 91:E1–E4, 2013.
  21. De Laet C, Dionisi-Vici C, Leonard JV, et al: Recommendations for the management of tyrosinaemia type 1, Orphanet J Rare Dis 8:8, 2013.
  22. El-Karaksy H, Rashed M, El-Sayed R, et al: Clinical practice. NTBC therapy for tyrosinemia type 1: how much is enough?, Eur J Pediatr 169:689–693, 2010.
  23. Fernendez RM, N??ez-Ramos R, Enguix-Riego MV, et al: Waardenburg syndrome type 4: report of two new cases caused by SOX10 mutations in Spain, Am J Med Genet A 164A:542–547, 2014.
  24. Gahl WA, Huizing M: Hermansky-Pudlak syndrome. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews [Internet], Seattle, 2000 [updated 2013], University of Washington. http://www.ncbi.nlm.nih.gov/books/NBK1287/. 1993–2014.
  25. Gomez-Ospina N, Scott AI, Oh GJ, et al: Expanding the phenotype of hawkinsinuria: new insights from response to N-acetyl-L-cysteine, J Inherit Metab Dis 39:821–829, 2016.
  26. Heylen E, Scherer G, Vincent MF, et al: Tyrosinemia type III detected via neonatal screening: management and outcome, Mol Genet Metab 107(3):605–607, 2012.
  27. Introne WJ, Perry MB, Troendle J, et al: A 3-year randomized therapeutic trial of nitisinone in alkaptonuria, Mol Genet Metab 103(4):307–314, 2011.
  28. Introne WJ, Westbroek W, Golas GA, et al: Chediak-Higashi syndrome. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews [Internet], Seattle, 2009 [updated 2012], University of Washington. http://www.ncbi.nlm.nih.gov/books/NBK5188/. 1993–2014.
  29. Larochelle J, Alvarez F, Bussi?res JF, et al: Effect of nitisinone (NTBC) treatment on the clinical course of hepatorenal tyrosinemia in Qu?bec, Mol Genet Metab107:49–54, 2012
  30. Lewis RA: Oculocutaneous albinism type 1. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews [Internet], Seattle, 2000 [updated 2013], University of Washington. http://www.ncbi.nlm.nih.gov/books/NBK1166/. 1993–2014.
  31. Lewis RA: Oculocutaneous albinism type 2. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews [Internet], Seattle, 2003 [updated 2012], University of Washington, Seattle. http://www.ncbi.nlm.nih.gov/books/NBK1232/. 1993–2014.
  32. Lewis RA: Ocular albinism, X-linked. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2004 [updated 2015], University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK1343/. 1993–2017.
  33. Masurel-Paulet A, Poggi-Bach J, Rolland MO, et al: NTBC treatment in tyrosinaemia type I: long-term outcome in French patients, J Inherit Metab Dis 31:81–87, 2008.
  34. Montoliu L, Gr?nskov K, Wei AH, et al: Increasing the complexity: new genes and new types of albinism, Pigment Cell Melanoma Res 27:11–18, 2014.
  35. Oiso N, Fukai K, Kawada A, et al: Piebaldism, J Dermatol 40:330–335, 2013.
  36. Pingault V, Ente D, Dastot-LeMoal F, et al: Review and update of mutations causing Waardenburg syndrome, Hum Mutat 31:391–406, 2010.
  37. Ranganath LR, Jarvis JC, Gallagher JA: Recent advances in management of alkaptonuria (invited review; best practice article), J Clin Pathol 66:367–373, 2013.
  38. Scott CR: The genetic tyrosinemias, Am J Med Genet C Semin Med Genet 142C:121–126, 2006.
  39. Seward SL Jr, Gahl WA: Hermansky-Pudlak syndrome: health care throughout life, Pediatrics 132:153–160, 2013.
  40. Sniderman King L, Trahms C, Scott CR: Tyrosinemia type I. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2006 [updated 2017], University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK1515/. 1993–2017.
  41. Plass AMC, van El CG, Pieters T, et al: Neonatal screening for treatable and untreatable disorders: prospective parents’ opinions, Pediatrics 125:e99–e106, 2010
  42. Pollitt RJ: Newborn blood spot screening: new opportunities, old problems, J Inherit Metab Dis 32:395–399, 2009.
  43. Shin SY, Fauman EB, Petersen AK, et al: An atlas of genetic influences on human blood metabolites, Nat Genet 46:543–550, 2014.
  44. Sokal EM: Treating inborn errors of liver metabolism with stem cells: current clinical development, J Inherit Metab Dis 37(4):535–539, 2014.
  45. Stevenson T, Millan MT, Wayman K, et al: Long-term outcome following pediatric liver transplant action for metabolic disorders, Pediatr Transplant 14:268–275, 2010.
  46. Taraiol-Graovac M, Shyr C, Ross GA, et al: Exome sequencing and the management of neurometabolic disorders, N Engl J Med 374(23):2246–2255, 2016.
  47. Vernon HJ: Inborn errors of metabolism: advances in diagnosis and therapy, JAMA Pediatr 169(8):778–782, 2015.
  48. Blau N: Defining tetrahydrobiopterin (BH 4 )-responsiveness in PKU, J Inherit Metab Dis 31:2–3, 2008.
  49. Blau N, B?langer-Quintana A, Demirkol M, et al: Management of phenylketonuria in Europe: survey results from 19 countries, Mol Genet Metab 99:109–115, 2010.
  50. Van Spronsen FJ, Derks TGJ: Recombinant phenylalanine ammonia lyase in phenylketonuria, Lancet 384:6–8, 2014.
  51. Vockley J, Andersson HC, Antshel KM, et al: Phenylalanine hydroxylase deficiency: diagnosis and management guideline, Genet Med 16(2):188–200, 2014.
  52. Waisbren SE, White D, van Spronsen F: Phenylketonuria, psychology and the brain, Mol Genet Metab 99(Suppl):S1–S108, 2010.
  53. Zurfluh MR, Zschocke J, Lindner M, et al: Molecular genetics of tetrahydrobiopterinresponsive phenylalanine hydroxylase deficiency, Hum Mutat 29:167–175, 2008.
  54. Bendadi F, de Koning TJ, Visser G, et al: Impaired cognitive functioning in patients with tyrosinemia type 1 receiving nitisinone, J Pediatr 164:398–401, 2014.
  55. David CV: Oculocutaneous albinism, Cutis 91:E1–E4, 2013.
  56. De Laet C, Dionisi-Vici C, Leonard JV, et al: Recommendations for the management of tyrosinaemia type 1, Orphanet J Rare Dis 8:8, 2013.
  57. El-Karaksy H, Rashed M, El-Sayed R, et al: Clinical practice. NTBC therapy for tyrosinemia type 1: how much is enough?, Eur J Pediatr 169:689–693, 2010.
  58. Fernendez RM, N??ez-Ramos R, Enguix-Riego MV, et al: Waardenburg syndrome type 4: report of two new cases caused by SOX10 mutations in Spain, Am J Med Genet A 164A:542–547, 2014.
  59. Gahl WA, Huizing M: Hermansky-Pudlak syndrome. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews [Internet], Seattle, 2000 [updated 2013], University of Washington. http://www.ncbi.nlm.nih.gov/books/NBK1287/. 1993–2014.
  60. Gomez-Ospina N, Scott AI, Oh GJ, et al: Expanding the phenotype of hawkinsinuria: new insights from response to N-acetyl-L-cysteine, J Inherit Metab Dis 39:821–829, 2016.
  61. Heylen E, Scherer G, Vincent MF, et al: Tyrosinemia type III detected via neonatal screening: management and outcome, Mol Genet Metab 107(3):605–607, 2012.
  62. Introne WJ, Perry MB, Troendle J, et al: A 3-year randomized therapeutic trial of nitisinone in alkaptonuria, Mol Genet Metab 103(4):307–314, 2011.
  63. Introne WJ, Westbroek W, Golas GA, et al: Chediak-Higashi syndrome. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews [Internet], Seattle, 2009 [updated 2012], University of Washington. http://www.ncbi.nlm.nih.gov/books/NBK5188/.1993–2014.
  64. Larochelle J, Alvarez F, Bussi?res JF, et al: Effect of nitisinone (NTBC) treatment on the clinical course of hepatorenal tyrosinemia in Qu?bec, Mol Genet Metab 107:49–54, 2012.
  65. Lewis RA: Oculocutaneous albinism type 1. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews [Internet], Seattle, 2000 [updated 2013], University of Washington. http://www.ncbi.nlm.nih.gov/books/NBK1166/. 1993–2014.
  66. Lewis RA: Oculocutaneous albinism type 2. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews [Internet], Seattle, 2003 [updated 2012], University of Washington, Seattle. http://www.ncbi.nlm.nih.gov/books/NBK1232/. 1993–2014.
  67. Lewis RA: Ocular albinism, X-linked. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2004 [updated 2015], University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK1343/. 1993–2017.
  68. Masurel-Paulet A, Poggi-Bach J, Rolland MO, et al: NTBC treatment in tyrosinaemia type I: long-term outcome in French patients, J Inherit Metab Dis 31:81–87, 2008.
  69. Montoliu L, Gr?nskov K, Wei AH, et al: Increasing the complexity: new genes and new types of albinism, Pigment Cell Melanoma Res 27:11–18, 2014.
  70. Oiso N, Fukai K, Kawada A, et al: Piebaldism, J Dermatol 40:330–335, 2013. Pingault V, Ente D, Dastot-LeMoal F, et al: Review and update of mutations causing Waardenburg syndrome, Hum Mutat 31:391–406, 2010. Ranganath LR, Jarvis JC, Gallagher JA: Recent advances in management of alkaptonuria (invited review; best practice article), J Clin Pathol 66:367–373, 2013.
  71. Gunay-Aygun M, Huizing M, Anikster Y: OPA3-related 3-methylglutaconic aciduria. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews [Internet], Seattle, 2006 [updated 2013], University of Washington. http://www.ncbi.nlm.nih.gov/books/NBK1473/. 1993–2014.
  72. Huemer M, Diodato D, Schwahn B, et al: Guidelines for diagnosis and management of the cobalamin-related remethylation disorders cblC, cblD, cblE, cblF, cblG, cblJ and MTHFR deficiency, J Inherit Metab Dis 40(1):21–48, 2017.
  73. Joint WHO/FAO/UNU Expert Consultation: Protein and amino acid requirements in human nutrition, World Health Organ Tech Rep Ser 935:1–265, 2007. back cover.
  74. Knerr I, Weinhold N, Vockley J, et al: Advances and challenges in the treatment of branched-chain amino/keto acid metabolic defects, J Inherit Metab Dis 35:29–40, 2012.
  75. Magner M, Dvorakova V, Tesarova M, et al: TMEM70 deficiency: long-term outcome of 48 patients, J Inherit Metab Dis 38(3):417–426, 2015.
  76. Manoli I, Myles JG, Sloan JL, et al: A critical reappraisal of dietary practices in methylmalonic acidemia raises concerns about the safety of medical foods. Part 1. Isolated methylmalonic acidemias, Genet Med 18(4):386–395, 2016
  77. Manoli I, Myles JG, Sloan JL, et al: A critical reappraisal of dietary practices in methylmalonic acidemia raises concerns about the safety of medical foods. Part 2. Cobalamin C deficiency, Genet Med 18(4):396–404, 2016.
  78. Manoli I, Sloan JL, Venditti CP: Isolated methylmalonic acidemia. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2005 [updated 2016], University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK1231/. 1993–2018.
  79. Mazariegos GV, Morton DH, Sindhi R, et al: Liver transplantation for classical maple syrup urine disease: long-term follow-up in 37 patients and comparative United Network for Organ Sharing experience, J Pediatr 160:116–121, 2012.
  80. Muelly ER, Moore GJ, Bunce SC, et al: Biochemical correlates of neuropsychiatric illness in maple syrup urine disease, J Clin Invest 123:1809–1820, 2013.
  81. Niemi AK, Kim IK, Krueger CE, et al: Treatment of methylmalonic acidemia by liver or combined liver-kidney transplantation, J Pediatr 166:1455–1461, 2015.
  82. Novarino G, El-Fishawy P, Kayserili H, et al: Mutations in BCKD-kinase lead to a potentially treatable form of autism with epilepsy, Science 338(6105):394–397, 2012.
  83. O’Shea CJ, Sloan JL, Wiggs EA, et al: Neurocognitive phenotype of isolated methylmalonicacademia, Pediatrics 129:e1541–e1551, 2012.
  84. Pie J, L?pez-Vi?as E, Puisac B, et al: Molecular genetics of HMG-CoA lyase deficiency, Mol Genet Metab 92:198–209, 2007.
  85. Puckett RL, Lorey F, Rinaldo P, et al: Maple syrup urine disease: further evidence that newborn screening may fail to identify variant forms, Mol Genet Metab100:136–142, 2010.
  86. Rahmandar MH, Bawcom A, Romano ME, Hamid R: Cobalamin C deficiency in an adolescent with altered mental status and anorexia, Pediatrics 134(6):e1709–e1714, 2014.
  87. Ramos M, Menao S, Arnedo M, et al: New case of mitochondrial HMG-CoA synthase deficiency: functional analysis of eight mutations, Eur J Med Genet 56:411–415, 2013.
  88. Rela M, Battula N, Madanur M, et al: Auxiliary liver transplantation for propionic acidemia: a 10-year follow-up, Am J Transplant 7:2200–2203, 2007.
  89. Romano S, Valayannopoulos V, Touati G, et al: Cardiomyopathies in propionic aciduria are reversible after liver transplantation, J Pediatr 156:128–134, 2010.
  90. Scott CR: The genetic tyrosinemias, Am J Med Genet C Semin Med Genet 142C:121–126, 2006.
  91. Seward SL Jr, Gahl WA: Hermansky-Pudlak syndrome: health care throughout life, Pediatrics 132:153–160, 2013.
  92. Sniderman King L, Trahms C, Scott CR: Tyrosinemia type I. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2006 [updated 2017], University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK1515/. 1993–2.
  93. Atwal PS, Scaglia F. Molybdenum cofactor deficiency. Mol Genet Metab.2016;117(1):1–4. Bayram E, Topcu Y, Karakaya P, et al. Molybdenum cofactor deficiency: review of 12 cases (MoCD and review). Eur J Paediatr Neurol .2013;17:1–6
  94. Dhamija R, Gavrilova RH, Wirrell EC. Valproate-induced worsening of seizures: clues to underlying diagnosis. J Child Neurol .2011;26:1319–1321.
  95. Higuchi R, Sugimoto T, Tamura A, et al. Early features in neuroimaging of two siblings with molybdenum cofactordeficiency .Pediatrics.2014;133:e267–e271.
  96. Hitzert MM, Bos AF, Bergman KA, et al. Favorable outcome in a newborn with molybdenum cofactor type A deficiency treated with cPMP. Pediatrics.2012;130:e1005–e1010.
  97. Reiss J, Hahnewald R.Molybdenum cofactor deficiency: mutations in GPHN, MOCS1, and MOCS2.Hum Mutat.2011;32:10–18.
  98. Schwahn BC, Van Spronsen FJ, Belaidi AA, et al. Efficacy and safety of cyclic pyranopterin monophosphate substitution in severe molybdenum cofactor deficiency type A: a prospective cohort study.Lancet.2015;386:1955–1963.
  99. Schwarz G, Mendel RR, Ribbe MW. Molybdenum cofactors, enzymes and pathways. Nature .2009;460:839–84.
  100. Struys EA, Nota B, Bakkali A, et al. Pyridoxine dependent epilepsy with elevated urinary ?-amino adipic semialdehyde in molybdenum cofactor deficiency. Pediatrics. 2012;130:e1716–e1719.
  101. Broer S. Diseases associated with general amino acid transporters of the solute carrier 6 family (SLC6). Curr Mol Pharmacol .2013;6:74–87.
  102. Camargo SM, Singer D, Makrides V, et al. Tissue-specific amino acid transporter partners ACE2 and collectrin differentially interact with Hartnup mutations. Gastroenterology.2009; 136:872–882.
  103. Nozaki J,Dakeishi M, Ohura T,et al. Homozygosity mapping to chromosome 5p15 of a gene responsible for Hartnup disorder. Biochem Biophys Res Commun.2001;284:255–260.
  104. Scriver CR, Mahon B,Levy HL,et al.The Hartnup phenotype: mendelian transport disorder, multifactorial disease.Am J Hum Genet.1987;40:401.
  105. Axler O, Holmquist P: Intermittent maple syrup urine disease: two case reports, Pediatrics 133:e458–e460, 2014.
  106. Bader-Meunier B, Florkin B, Sibilia J, et al: Mevalonate kinase deficiency: a survey of 50 patients, Pediatrics 128:e152–e159, 2011.
  107. Bailey LM, Ivanov RA, Jitrapakdee S, et al: Reduced half-life of holocarboxylase synthetase from patients with severe multiple carboxylase deficiency, Hum Mutat29:e47–e57, 2008.
  108. Baker EH, Sloan JL, Hauser NS, et al: MRI characteristics of globus pallidus infarcts in isolated methylmalonic acidemia, AJNR Am J Neuroradiol 36(1):194–201, 2015.
  109. Baumgartner MR, H?rster F, Dionisi-Vici C, et al: Proposed guidelines for the diagnosis and management of methylmalonic and propionic acidemia, Orphanet J Rare Dis9:130, 2014.
  110. Carmel R, Watkins D, Rosenblatt DS: Megaloblastic anemia. In Orkin SH, Ginsburg D, Nathan DA, et al, editors: Nathan and Oski’s hematology of infancy and childhood, ed 8, Philadelphia, 2014, Elsevier.
  111. Carrillo N, Adams D, Venditti CP: Disorders of intracellular cobalamin metabolism. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2008 [updated 2013], University of Washington. 1993–2018.
  112. Carrillo-Carrasco N, Sloan J, Valle D, et al: Hydroxocobalamin dose escalation improves metabolic control in cblC, J Inherit Metab Dis 32(6):728–731, 2009.
  113. Chapman KA, Gropman A, MacLeod E, et al: Acute management of propionic acidemia, Mol Genet Metab 105:16–25, 2012.
  114. Coelho D, Kim JC, Miouse IR, et al: Mutations in ABCD4 cause a new inborn error of vitamin B12 metabolism, Nat Genet 44:1152–1155, 2012.
  115. Coelho D, Suormala T, Stucki M, et al: Gene identification for the cblD defect of vitamin B12 metabolism, N Engl J Med 358:1454–1564, 2008.
  116. Cosson MA, Benoist JF, Touati G, et al: Long-term outcome in methylmalonic aciduria: a series of 30 French patients, Mol Genet Metab 97:172–178, 2009.
  117. Davey KM, Parboosingh JS, McLeod DR, et al: Mutation of DNAJC19, a human homologue of yeast inner mitochondrial membrane co-chaperones, causes DCMA syndrome, a novel autosomal recessive Barth syndrome-like condition, J Med Genet43:385–393, 2006.
  118. Dercksen M, Duran M, Ijlst L, et al: Clinical variability of isovaleric acidemia in a genetically homogeneous population, J Inherit Metab Dis 35:1021–1029, 2012.
  119. Eminoglu FT, Ozcelik AA, Okur I, et al: 3-Methylcrotonyl-CoA carboxylase deficiency: phenotypic variability in a family, J Child Neurol 24:478–481, 2009.
  120. Forsyth RL, Vockley CW, Edick MJ, et al: Outcomes of cases with 3-methylcrotonyl-CoA carboxylase (3-MCC) deficiency: report from the Inborn Errors of Metabolism Information System, Mol Genet Metab 118(1):15–20, 2016.
  121. Fraser JL, Venditti CP: Methylmalonic and propionic acidemias: clinical management update, Curr Opin Pediatr 28:682–693, 2016.
  122. Fukao T, Mitchell G, Sass JO, et al: Ketone body metabolism and its defects, J Inherit Metab Dis 37(4):541–551, 2014.
  123. Fukao T, Sass JO, Kursula P, et al: Clinical and molecular characterization of five patients with succinyl-CoA:3-ketoacid CoA transferase (SCOT) deficiency, Biochim Biophys Acta 1812:619–624, 2011.
  124. Goldsmith DP: Periodic fever syndromes, Pediatr Rev 30:e34–e41, 2009.
  125. Gomez AR, Couce ML, Garcia-Villoria J, et al: Clinical, genetic and therapeutic diversity in 2 patients with severe mevalonate kinase deficiency, Pediatrics 129:e535–e539, 2012.
  126. Grenert SC, Stucki M, Morscher RJ, et al: 3-methylcrotonyl-CoA carboxylase deficiency: clinical, biochemical, enzymatic and molecular studies in 88 individuals, Orphanet J Rare Dis 7:31, 2012.
  127. Grenert SC, Wendel U, Lindner M, et al: Clinical and neurocognitive outcome in symptomatic isovaleric acidemia, Orphanet J Rare Dis 7:9, 2012.
  128. Rutsch F, Gailus S, Miousse IR, et al: Identification of a putative lysosomal cobalamin exporter mutated in the cblF inborn error of vitamin B12 metabolism, Nat Genet41:234–239, 2009.
  129. Sarafoglou K, Matern D, Redlinger-Grosse K, et al: Siblings with mitochondrial acetoacetyl-CoA thiolase deficiency not identified by newborn screening, Pediatrics128:e246–e250, 2011.
  130. Scholl-B?rgi S, Haberlandt E, Gotwald T, et al: Stroke-like episodes in propionic academia caused by central focal metabolic decompensation, Neuropediatrics40:76–81, 2009.
  131. Scholl-B?rgi S, Sass JO, Zschocke J, et al: Amino acid metabolism in patients with propionic acidaemia, J Inherit Metab Dis 35:65–70, 2012.
  132. Shafqat N, Turnbull A, Zschocke J, et al: Crystal structures of human HMG-CoA synthase isoforms provide insights into inherited ketogenesis disorders and inhibitor design, J Mol Biol 398:497–506, 2010.
  133. Shchelochkov OA, Carrillo N, Venditti C: Propionic acidemia. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2012 [updated 2016], University of Washington. 1993–2018.
  134. Simon E, Flaschker N, Schadewaldt P, et al: Variant maple syrup urine disease (MSUD): the entire spectrum, J Inherit Metab Dis 29:716–724, 2006.
  135. Simon E, Schwarz M, Wendel U: Social outcome in adults with maple syrup urine disease (MSUD), J Inherit Metab Dis 30:264, 2007.
  136. Sloan JL, Johnston JJ, Manoli I, et al: Exome sequencing identifies ACSF3 as a cause of combined malonic and methylmalonic aciduria, Nat Genet 43(9):883–886, 2011.
  137. Strauss KA, Mazariegos GV, Sindhi R, et al: Elective liver transplantation for the treatment of classical maple syrup urine disease, Am J Transplant 6:557–564, 2006.
  138. Strauss KA, Wardley B, Robinson D, et al: Classical maple syrup urine disease and brain development: principles of management and formula design, Mol Genet Metab 99:333–345, 2010.
  139. Sutton VR, Chapman KA, Gropman AL, et al: Chronic management and health supervision ofindividuals with propionic acidemia, Mol Genet Metab 105:26–33, 2012.
  140. Tammachote R, Janklat S, Tongkobpetch S, et al: Holocarboxylase synthetase deficiency: novel clinical and molecular findings, Clin Genet 78:88–93, 2010.
  141. Terlungeanu DC, Deliu E, Dotter CP, et al: Impaired amino acid transport at the blood brain barrier is a cause of autism spectrum disorder, Cell 167(6):1481–1494, e18, 2016.
  142. Ter Haar NM, Jeyaratnam J, Lachmann HJ, et al; Paediatric Rheumatology International Trials Organisation and Eurofever Project: The phenotype and genotype of mevalonate kinase deficiency: a series of 114 cases from the Eurofever Registry, Arthritis Rheumatol 68(11):2795–2805, 2016.
  143. Van der Hilst JC, Bodar EJ, Barron KS, et al: Long-term follow-up, clinical features, and quality of life in a series of 103 patients with hyperimmunoglobulinemia D syndrome, Medicine (Baltimore) 87:301–310, 2008.
  144. Waters PJ, Thuriot F, Clarke JT, et al: Methylmalonyl-coA epimerase deficiency: a new case, with an acute metabolic presentation and an intronic splicing mutation in the MCEE gene, Mol Genet Metab Rep 9:19–24, 2016
  145. Whited K, Baile MG, Currier P, et al: Seven functional classes of Barth syndrome mutation, Hum Mol Genet 22:483–492, 2013.
  146. Wolf B: Clinical issues and frequent questions about biotinidase deficiency, Mol Genet Metab 100:6–13, 2010.
  147. Wolf B: The neurology of biotinidase deficiency, Mol Genet Metab 104:27–34, 2011.
  148. Wortmann SB, de Brouwer APM, Wevers RA, et al: MEGDEL syndrome. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2014, University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK195853/. 1993–2017.
  149. Wortmann SB, Duran M, Anikster Y, et al: Inborn errors of metabolism of 3-methylglutaconicaciduria as discriminative feature: proper classification and nomenclature, J Inherit Metab Dis 36(6):923–928, 2013.
  150. Hart TC, Gorry MC, Hart PS: Mutations of the UMOD gene are responsible for medullary cystic kidney disease 2 and familial juvenile hyperuricaemic nephropathy, J Med Genet 39:882–892, 2002.
  151. Mount DB: The kidney in hyperuricemia and gout, Curr Opin Nephrol Hypertens22(2):216–223, 2013.
  152. Piret SE, Danoy P, Dahan K, et al: Genome-wide study of familial juvenile hyperuricaemic (gouty) nephropathy (FJHN) indicates a new locus, FJHN3, linked to chromosome 2p22.1-p21, Hum Genet 129:51–58, 2011.
  153. Stiburkova B, Majewksi J, Hodanova K: Familial juvenile hyperuricaemic nephropathy (FJHN): linkage analysis in 15 families, physical and transcriptional characterisation of the FJHN critical region on chromosome 16p11.2 and the analysis of seven candidate genes, Eur J Hum Genet 11:145–154, 2003.
  154. Venkat-Raman G, Gast C, Marinaki A, et al: From juvenile hyperuricaemia to dysfunctional uromodulin: an ongoing metamorphosis, Pediatr Nephrol 31(11):2035–2042, 2016.
  155. Augoustides-Savvopoulou P, Papachristou F, Fairbanks LD, et al: Partial hypoxanthineguanine phosphoribosyltransferase deficiency as the unsuspected cause of renal disease spanning three generations: a cautionary tale, Pediatrics 109:E17, 2002.
  156. Ceballos-Picot I, Mockel L, Potier MC, et al: Hypoxanthine-guanine phosphoribosyl transferase regulates early developmental programming of dopamine neurons: implications for Lesch-Nyhan disease pathogenesis, Hum Mol Genet 18:2317–2327, 2009.
  157. Chen BC, Balasubramaniam S, McGown IN, et al: Treatment of Lesch-Nyhan disease with S-adenosylmethionine: experience with five young Malaysians, including a girl, Brain Dev 36(7):593–600, 2014.
  158. Cif L, Biolsi B, Gavarini S, et al: Antero-ventral internal pallidum stimulation improves behavioral disorders in Lesch-Nyhan disease, Mov Disord 22:2126–2129, 2007.
  159. Jinnah HA, Ceballos-Picot I, Torres RJ, et al: Lesch-Nyhan Disease International Study Group: Attenuated variants of Lesch-Nyhan disease, Brain 133(Pt 3):671–689, 2010.
  160. Jinnah HA, Visser JE, Harris JC, et al: Lesch-Nyhan Disease International Study Group: Delineation of the motor disorder of Lesch-Nyhan disease, Brain 129(Pt 5):1201–1217, 2006.
  161. Khasnavis T, Torres RJ, Sommerfeld B, et al: A double-blind, placebo-controlled, crossover trial of the selective dopamine D1 receptor antagonist ecopipam in patients with Lesch-Nyhan disease, Mol Genet Metab 118(3):160–166, 2016.
  162. Nyhan WL, O’Neill JP, Jinnah HA, et al: Lesch-Nyhan syndrome. In Pagon RA, Adam MP, Bird TD, et al, editors: GeneReviews [Internet], Seattle, 2000 [updated 2010]. University of Washington, pp 1993–2013. http://www.ncbi.nlm.nih.gov/books/NBK1149/.
  163. Schretlen DJ, Callon W, Ward RE, et al: Do clinical features of Lesch-Nyhan disease correlate more closely with hypoxanthine or guanine recycling?, J Inherit Metab Dis 39(1):85–91, 2016
  164. Schretlen DJ, Harris JC, Park K, et al: Neurocognitive functioning in Lesch-Nyhan disease and partial hypoxanthine-guanine phosphoribosyltransferase deficiency, J Int Neuropsychol Soc 7:805–812, 2001.
  165. Schretlen DJ, Varvaris M, Ho TE, et al: Regional brain volume abnormalities in Lesch-Nyhan disease and its variants: a cross-sectional study, Lancet Neurol12(12):1151–1158, 2013.
  166. Schretlen DJ, Varvaris M, Vannorsdall T, et al: Brain white matter volume abnormalities in Lesch-Nyhan disease and its variants, Neurology 84(2):190–196, 2015.
  167. Schretlen DJ, Ward J, Meyer SM, et al: Behavioral aspects of Lesch-Nyhan disease and its variants, Dev Med Child Neurol 47:673–677, 2005.
  168. Wong DF, Harris JC, Naidu S, et al: Dopamine transporters are markedly reduced in Lesch-Nyhan disease in vivo, Proc Natl Acad Sci USA 93:5539, 1996
  169. Bollie G, Harambat J, Bensman A, et al: Adenine phosphoribosyltransferase deficiency, Clin J Am Soc Nephrol 7:1521–1527, 2012.
  170. Ceballos-Picot I, Daudon M, Harambat J, et al: 2,8-Dihydroxyadenine urolithiasis: a not so rare inborn error of purine metabolism, Nucleosides Nucleotides Nucleic Acids 33(4–6):241–252, 2014.
  171. Edvardsson VO, Palsson R, Sahota A: Adenine phosphoribosyltransferase deficiency. In Pagon RA, Adam MP, Bird TD, et al, editors: GeneReviews [Internet], Seattle, 2012, University of Washington, pp 1993–2013. http://www.ncbi.nlm.nih.gov/books/NBK100238.
  172. Harambat J, Boll?e G, Daudon M, et al: Adenine phosphoribosyltransferase deficiency in children, Pediatr Nephrol 27:571–579, 2012.
  173. Marra G, Vercelloni PG, Edefonti A, et al: Adenine phosphoribosyltransferase deficiency: an underdiagnosed cause of lithiasis and renal failure, JIMD Rep 5:45–48, 2012.
  174. De Brouwer AP, van Bokhoven H, Nabuurs SB, et al: PRPS1 mutations: four distinct syndromes and potential treatment, Am J Hum Genet 86:506–518, 2010.
  175. Duley JA, Christodoulou J, de Brouwer AP: The PRPP synthetase spectrum: what does it demonstrate about nucleotide syndromes?, Nucleosides Nucleotides Nucleic Acids 30:1129–1139, 2011.
  176. Hove-Jensen B, Andersen KR, Kilstrup M, et al: Phosphoribosyl diphosphate (PRPP): biosynthesis, enzymology, utilization, and metabolic significance, Microbiol Mol Biol Rev 81(1):2016.
  177. Liu XZ, Xie D, Yuan HJ, et al: Hearing loss and PRPS1 mutations: wide spectrum of phenotypes and potential therapy, Int J Audiol 52:23–28, 2013.
  178. Mittal R, Patel K, Mittal J, et al: Association of PRPS1 mutations with disease phenotypes, Dis Markers 2015:1–7, 2015
  179. Jurecka A, Jurkiewicz E, Tylki-Szymanska A: Magnetic resonance imaging of the brain in adenylosuccinate lyase deficiency: a report of seven cases and a review of the literature, Eur J Pediatr 171:131–138, 2012.
  180. Jurecka A, Zikanova M, Jurkiewicz E, et al: Attenuated adenylosuccinate lyase deficiency: a report of one case and a review of the literature, Neuropediatrics 45(1):50–55, 2014.
  181. Jurecka A, Zikanova M, Kmoch S, et al: Adenylosuccinate lyase deficiency, J Inherit Metab Dis 38(2):231–242, 2015.
  182. Lim S, Lowry M, Strongin RM: Progress towards simple and direct detection of adenylosuccinate lyase deficiency in human urine, Aust J Chem 64:1470–1473, 2011.
  183. Race V, Marie S, Vincent MF, et al: Clinical, biochemical and molecular genetic correlations in adenylosuccinate lyase deficiency, Hum Mol Genet 9:2159–2165, 2000.
  184. Ray SP, Deaton MK, Capodagli GC, et al: Structural and biochemical characterization of human adenylosuccinate lyase (ADSL) and the R303C ADSL deficiency-associated mutation, Biochemistry 51:6701–6713, 2012.
  185. Van Werkhoven M, Duley J, McGown I, et al: Early diagnosis of adenylosuccinate lyase deficiency with a high throughput screening method and a trial of oral S-adenosylmethionine, Dev Med Child Neurol 55(11):1060–1064, 2013.
  186. Baggott JE, Tamura T: Evidence for the hypothesis that 10-formyldihydrofolate is the in vivo substrate for aminoimidazolecarboxamide ribotide transformylase, Exp Biol Med (Maywood) 235:271–277, 2010.
  187. Boutchueng-Djidjou M, Collard-Simard G, Fortier S, et al: The last enzyme of the de novo purine synthesis pathway 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/IMP cyclohydrolase (ATIC) plays a central role in insulin signaling and the Golgi/endosomes protein network, Mol Cell Proteomics 14(4):1079–1092, 2015.
  188. Duval N, Luhrs K, Wilkinson TG 2nd, et al: Genetic and metabolomic analysis of AdeD and AdeI mutants of de novo purine biosynthesis: cellular models of de novo purine biosynthesis deficiency disorders, Mol Genet Metab 108:178–189, 2013.
  189. Marie S, Heron B, Bitoun P, et al: AICA-ribosiduria: a novel, neurologically devastating inborn error of purine biosynthesis caused by mutation of ATIC, Am J Hum Genet74:1276–1281, 2004.
  190. Pastore S, Stocco G, Moressa V, et al: 5-Aminoimidazole-4-carboxamideribonucleotidetransformylase and inosine-triphosphate-pyrophosphatase genes variants predict remission rate during methotrexate therapy in patients with juvenile idiopathic arthritis, Rheumatol Int 35(4):619–627, 2015.
  191. Wortmann SB, Kluijtmans LA, Engelke UF, et al: The 3-methylglutaconic acidurias: what’s new?, J Inherit Metab Dis 35:13–22, 2012.
  192. Wortmann SB, Kremer BH, Graham A, et al: 3-Methylglutaconic aciduria type I redefined, Neurology 75(12):1079–1083, 2010.
  193. Wortmann SB, Kluijtmans LA, Rodenburg RJ, et al: 3-Methylglutaconic aciduria—lessons from 50 genes and 977 patients, J Inherit Metab Dis 36:913–921, 2013.
  194. Yu HC, Sloan JL, Scharer G, et al: Mutations in the transcription factor HCFC1 cause a novel X-linked cobalamin disorder (cblX) with a severe neurological phenotype, Am J Hum Genet 93:506–514, 2013.
  195. Zinnanti WJ, Lazovic J, Griffin K, et al: Dual mechanism of brain injury and novel treatment strategy in maple syrup urine disease, Brain 132:903–918, 2009.
  196. Bjoraker KJ, Swanson MA, Coughlin IICR, et al: Neurodevelopmental outcome and treatment efficacy of benzoate and dextromethorphan in siblings with attenuated nonketotic hyperglycemia, J Pediatr 170:234–239, 2016.
  197. Cheillan D, Joncquel-Chevalier Curt M, Briand G, et al: Screening for primary creatine deficiencies in French patients with unexplained neurological symptoms, Orphanet J Rare Dis 7:96, 2012.
  198. Cochat P, Rumsby G: Primary hyperoxaluria, N Engl J Med 369:649–658, 2013.
  199. Coulter-Mackie MB, White CT, Lange D, et al: Primary hyperoxaluria type 1. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2002 [updated 2014], University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK1283/. 1993–2017.
  200. Dhar SU, Scaglia F, Li FY, et al: Expanded clinical and molecular spectrum of guanidinoacetate methyltransferase (GAMT) deficiency, Mol Genet Metab 96:38–43, 2009.
  201. Ellis D, Lieb J: Hyperoxaluria and genitourinary disorders in children ingesting almond milk products, J Pediatr 167:1155–1158, 2015.
  202. Menendez Suso JJ, Del Cerro Mar?n MJ, Dorao Mart?nez-Romillo P, et al: Nonketotic hyperglycinemia presenting as pulmonary hypertensive vascular disease and fatal pulmonary edema in response to pulmonary vasodilator therapy, J Pediatr 161:557–559, 2012.
  203. Mercimek-Mahmutoglu S, Ndika J, Kanhai W, et al: Thirteen new patients with guanidinoacetate methyltransferase deficiency and functional characterization of nineteen novel missense variants in the GAMT gene, Hum Mutat 35:462–469, 2014.
  204. Mercimek-Mahmutoglu S, Salomons GS: Creatine deficiency syndromes. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2009 [updated 2015], University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK3794/. 1993–2017.
  205. Mercimek-Mahmutoglu S, St?ckler-Ipsiroglu S, Salomons GS: Creatine deficiency syndromes. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews [Internet], Seattle, 2009 [updated 2011], University of Washington. http://www.ncbi.nlm.nih.gov/books/NBK3794/. 1993–2014.
  206. Milliner DS, Harris PC, Lieske JC: Primary hyperoxaluria type 3. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2015, University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK316514/. 1993–2017.
  207. Nasrallah F, Feki M, Kaabachi N: Creatine and creatine deficiency syndromes: biochemical and clinical aspects, Pediatr Neurol 42:163–171, 2010. Phillips IR, Shephard EA: Primary trimethylaminuria. https://www.ncbi.nlm.nih.gov/ books/NBK1103/.
  208. Rezvani I, Auerbach VH: Primary hyperoxaluria, N Engl J Med 369:2162–2163, 2013. Rumsby G: Primary hyperoxaluria type 2. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2008 [updated 2011], University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK2692/. 1993–2017.
  209. Sass JO, Fischer K, Wang R, et al: D-Glyceric aciduria is caused by genetic deficiency of D-glycerate kinase (GLYCTK), Hum Mutat 31:1280–1285, 2010.
  210. Stockler-Ipsiroglu S, van Karnebeek C, Longo N, et al: Guanidinoacetate methyltransferase (GAMT) deficiency: outcomes in 48 individuals and recommendations for diagnosis, treatment and monitoring, Mol Genet Metab 111:16–25, 2014.
  211. Valayannopoulos V, Boddaert N, Chabli A, et al: Treatment by oral creatine, L-arginine and L-glycine in six severely affected patients with creatine transporter defect, J Inherit Metab Dis 35:151–157, 2012.
  212. Van de Kamp JM, Betsalel OT, Mercimek-Mahmutoglu S, et al: Phenotype and genotype in 101 males with X-linked creatine transporter deficiency, J Med Genet 50:463–472, 2013.
  213. Van de Kamp JM, Mancini GM, Salomons GS: X-linked creatine transporter deficiency: clinical aspects and pathophysiology, J Inherit Metab Dis 37:715–733, 2014.
  214. Van Hove J, Coughlin C II, Scharer G: Glycine encephalopathy. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews [Internet], Seattle, 2002 [updated 2013], University of Washington. http://www.ncbi.nlm.nih.gov/books/NBK1357/. 1993–2014.
  215. Williams EL, Bockenhauer D, van’t Hoff WG, et al: The enzyme 4-hydroxy-2-oxoglutarate aldolase is deficient in primary hyperoxaluria type 3, Nephrol Dial Transplant 27:3191–3195, 2012
  216. Acuna-Hidalgo R, Schanze D, Kariminejad A, et al: Neu-Laxova syndrome is a heterogeneous metabolic disorder caused by defects in enzymes of the L-serine biosynthesis pathway, Am J Hum Genet 95(3):285–293, 2014.
  217. El-Hattab AW: Serine biosynthesis and transport defects, Mol Genet Metab 118(3):153–159, 2016.
  218. Tabatabaie L, Klomp LW, Berger R, et al: L-serine synthesis in the central nervous system: a review on serine deficiency disorders, Mol Genet Metab 99:256–262, 2010.
  219. Coutelier M, Goizet C, Durr A, et al: Alteration of ornithine metabolism leads to dominant and recessive hereditary spastic paraplegia, Brain 138(Pt 8):2191–2205, 2015.
  220. Dimopoulou A, Fischer B, Gardeitchik T, et al: Genotype-phenotype spectrum of PYCR1-related autosomal recessive cutis laxa, Mol Genet Metab 110(3):352–361, 2013.
  221. Ferreira C, Wang H: Prolidase Deficiency. 2015 Jun 25. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle (WA), 1993-2017, University of Washington, Seattle. Available from: https://www.ncbi.nlm.nih.gov/ books/NBK299584/.
  222. Kurien BT, D’Sousa A, Bruner BF, et al: Prolidase deficiency breaks tolerance to lupus-associated antigens, Int J Rheum Dis 16:674–680, 2013.
  223. Martinelli D, H?berle J, Rubio V, et al: Understanding pyrroline-5-carboxylate synthetase deficiency: clinical, molecular, functional, and expression studies, structure-based analysis, and novel therapy with arginine, J Inherit Metab Dis 35(5):761–776, 2012.
  224. Mitsubuchi H, Nakamura K, Matsumoto S, et al: Inborn errors of proline metabolism, J Nutr 138:2016S–2020S, 2008. Raux G, Bunsel E, Hecketsweiler B, et al: Involvement of hyperprolinemia in cognitive and psychiatric features of the 22q11 deletion syndrome, Hum Mol Genet 16:83–91, 2007.
  225. Raux G, Bunsel E, Hecketsweiler B, et al: Involvement of hyperprolinemia in cognitive and psychiatric features of the 22q11 deletion syndrome, Hum Mol Genet 16:83–91, 2007.
  226. Almusafri F, Elamin HE, Khalaf TE, et al. Clinical and molecular characterization of 6 children with glutamate cysteine ligase deficiency causing hemolytic anemia. Blood Cells Mol Dis.2017; 65:73–77.
  227. Ristoff E, Mayatepek E, Larsson A. Long-term clinical outcome in patients with glutathione synthetasedeficiency. JPediatr.2001;139(1):79–84.
  228. Sass JO, Gemperle-Britschgi C, Tarailo-Graovac M, et al. Unravelling 5-oxoprolinuria (pyroglutamic aciduria) due to bi-allelic OPLAH mutations: 20 new mutations in 14 families. Mol Genet Metab. 2016;119(1–2):44–49.
  229. Acosta MT, Munasinghe J, Pearl PL, et al: Cerebellar atrophy in human and murine succinic semialdehyde dehydrogenase deficiency, J Child Neurol 25:1457–1461, 2010.
  230. Blau N, van Spronsen FJ: Disorders of phenylalanine and tetrahydrobiopterin metabolism. In Blau N, Duran M, Gibson KM, et al, editors: Physician’s guide to the diagnosis, treatment, and follow-up of inherited metabolic diseases, Heidelberg, 2014, Springer, pp 3–21.
  231. Clark JF, Cecil KM: Diagnostic methods and recommendations for the cerebral creatine deficiency syndromes, Pediatr Res 77(3):398–405, 2015.
  232. Friedman J: Sepiapterin reductase deficiency. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2015, University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK304122/. 1993–2017.
  233. Furukawa Y: GTP cyclohydrolase 1-deficient dopa-responsive dystonia. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2002 [updated 2015], University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK1508/. 1993–2017.
  234. Furukawa Y, Kish S: Tyrosine hydroxylase deficiency. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2008 [updated 2017], University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK1437/. 1993–2017.
  235. Hwu WL, Muramatsu S, Tseng SH, et al: Gene therapy for aromatic L-amino acid decarboxylase deficiency, Sci Transl Med 4(134):134ra61, 2012.
  236. Knerr I, Gibson KM, Murdoch G, et al: Neuropathology in succinic semialdehyde dehydrogenase deficiency, Pediatr Neurol 42:255–258, 2010.
  237. Marecos C, Ng J, Kurian MA: What is new for monoamine neurotransmitter disorders?, J Inherit Metab Dis 37(4):619–626, 2014.
  238. Narboux-N?me N, Sagn? C, Doly S, et al: Severe serotonin depletion after conditional deletion of the vesicular monoamine transporter 2 gene in serotonin neurons: neural and behavioral consequences, Neuropsychopharmacology 36:2538–2550, 2011.
  239. Ng J, Zhen J, Meyer E, et al: Dopamine transporter deficiency syndrome: phenotypic spectrum from infancy to adulthood, Brain 137:1107–1119, 2014.
  240. Opladen T, Hoffmann GF, K?hn AA, et al: Pitfalls in phenylalanine loading test inthe diagnosis of dopa-responsive dystonia, Mol Genet Metab 108:195–197, 2013.
  241. Pearl PL: Monoamine neurotransmitter deficiencies, Handb Clin Neurol 113:1819–1825, 2013.
  242. Pearl PL, Wiwattanadittakul N, Roullet JB, et al: Succinic semialdehyde dehydrogenase deficiency. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2004 [updated 2016], University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK1195/. 1993–2017.
  243. Rilstone JJ, Alkhater RA, Minassian BA: Brain dopamine-serotonin vesicular transport disease and its treatment, N Engl J Med 368:543–550, 2013.
  244. Robertson D, Garland EM: Dopamine beta-hydroxylase deficiency. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2003 [updated 2015], University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK1474/. 1993–2017.
  245. Segawa M: Hereditary progressive dystonia with marked diurnal fluctuation, Brain Dev 33:195–201, 2011.
  246. Van Hove JLK, Thomas JA: Disorders of glycine, serine, GABA and proline metabolism.In Blau N, Duran M, Gibson KM, et al, editors: Physician’s guide to the diagnosis, treatment, and follow-up of inherited metabolic diseases, Heidelberg, 2014, Springer, pp 63–84.
  247. Vogel KR, Pearl PL, Theodore WH, et al: Thirty years beyond discovery—clinical trials in succinic semialdehyde dehydrogenase deficiency, a disorder of GABA metabolism, J Inherit Metab Dis 36:401–410, 2013.
  248. Adam S, Almeida MF, Assoun M, et al: Dietary management of urea cycle disorders: European practice, Mol Genet Metab 110:439–445, 2013.
  249. Bergmann KR, McCabe J, Smith RT, et al: Late-onset ornithine transcarbamylase deficiency: treatment and outcome of hyperammonemic crisis, Pediatrics133:e1072–e1076, 2014.
  250. Camacho J, Rioseco-Camacho N: Hyperornithinemia-hyperammonemia-homocitrullinuriasyndrome. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews [Internet], Seattle, 2012, University of Washington. http://www.ncbi.nlm.nih.gov/books/NBK97260/. 1993–2014.
  251. Cederbaum S, LeMons C, Lee B: New developments and future directions for urea cycle disorders, Mol Genet Metab 100(Suppl):1–106, 2010.
  252. Diaz GA, Krivitzky LS, Mokhtarani M, et al: Ammonia control and neurocognitive outcome among urea cycle disorder patients treated with glycerol phenylbutyrate, Hepatology 57:2171, 2013.
  253. Enns GM, Berry SA, Berry GT, et al: Survival after treatment with phenylacetate and benzoate for urea-cycle disorders, N Engl J Med 356:2282–2292, 2007.
  254. Ficicioglu C, Mandell R, Shih VE: Argininosuccinate lyase deficiency: long-term outcome of 13 patients detected by newborn screening, Mol Genet Metab 98:273–277, 2009.
  255. Gallagher RC, Lam C, Wong D, et al: Significant hepatic involvement in patients with ornithine transcarbamylase deficiency, J Pediatr 164(4):720–725, 2014.
  256. Gardeitichik T, Humphrey M, Nation J, et al: Early clinical manifestations and eating patterns in patients with urea cycle disorders, J Pediatr 161:328–332, 2012.
  257. Gropman AL, Seltzer RR, Yudkoff M, et al: 1H MRS allows brain phenotype differentiation in sisters with late onset ornithine transcarbamylase deficiency (OTCD) and discordant clinical presentations, Mol Genet Metab 94:52–56, 2008.
  258. Heberle J, Boddaert N, Burlina A, et al: Suggested guidelines for the diagnosis and management of urea cycle disorders, Orphanet J Rare Dis 7:32, 2012.
  259. Heberle J, G?rg B, Toutain A, et al: Inborn error of amino acid synthesis; human glutamine synthetase deficiency, J Inherit Metab Dis 29:352–358, 2006.
  260. Heberle J, Shahbeck N, Ibrahim K, et al: Natural course of glutamine synthetase deficiency in a 3 year old patient, Mol Genet Metab 103:89–91, 2011.
  261. Heberle J, Shahbeck N, Ibrahim K, et al: Glutamine supplementation in a child with inherited GS deficiency improves the clinical status and partially corrects the peripheral and central amino acid imbalance, Orphanet J Rare Dis 7:48, 2012
  262. Kim SZ, Song WJ, Nyhan WL, et al: Long-term follow-up of four patients affected by HHH syndrome, Clin Chim Acta 413:1151–1155, 2012.
  263. Kobayashi K, Saheki T, Song YZ: Citrin deficiency. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews [Internet], Seattle, 2005 [updated 2012], University of Washington. http://www.ncbi.nlm.nih.gov/books/NBK1181/. 1993–2014.
  264. Lichter-Konecki U, Caldovic L, Morizono H, et al: Ornithine transcarbamylase deficiency. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews [Internet], Seattle, 2013, University of Washington. http://www.ncbi.nlm.nih.gov/books/NBK154378/. 1993–2014.
  265. Martinelli D, Diodato D, Ponzi E, et al: The hyperprnithinemia-hyperammonemiahomocitrullinuria syndrome, Orph J Rare Dis 10:29, 2015.
  266. The Medical Letter: Glycerol phenylbutyrate (Ravicti) for urea cycle disorders, Med Lett 56:77–78, 2014.
  267. Mew NA, Krivitzky L, McCarter R, et al: Clinical outcomes of neonatal onset proximal versus distal urea cycle disorders do not differ, J Pediatr 162:324–329, 2013.
  268. Nagamani SC, Lee B, Erez A: Optimizing therapy for argininosuccinic aciduria, Mol Genet Metab 107:10–14, 2012.
  269. Smith W, Diaz GA, Lichter-Konecki U, et al: Ammonia control in children ages 2 months through 5 years with urea cycle disorders comparison of sodium phenylbutyrate and glycerol phenylbutyrate, J Pediatr 162:1228–1234, 2013.
  270. Summar ML, Koelker S, Freedenberg D, et al: The incidence of urea cycle disorders, Mol Genet Metab 110:179–180, 2013.
  271. Tuchman M, Lee B, Lichter-Konecki U, et al; Urea Cycle Disorders Consortium of the Rare Diseases Clinical Research Network: Cross-sectional multicenter study of patients with urea cycle disorders in the United States, Mol Genet Metab 94:397–402, 2008.
  272. Van Karnebeek C, H?berle J: Carbonic anhydrase VA deficiency. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2015, University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK284774. 1993–2017.
  273. Yudkoff M, Mew NA, Daikhin Y, et al: Measuring in vivo ureagenesis with stable isotopes, Mol Genet Metab 100(Suppl 1):S37–S41, 2010
  274. Castellan Baldan L, Williams KA, et al: Histidine decarboxylase deficiency causes Tourette syndrome: parallel findings in humans and mice, Neuron 81:77–90, 2014.
  275. Ercan-Sencicek AG, Stillman AA, Ghosh AK, et al: L-histidine decarboxylase and Tourette’s syndrome, N Engl J Med 362:1901–1908, 2010.
  276. Basura GJ, Hagland SP, Wiltse AM, et al: Clinical features and the management of pyridoxine-dependent and pyridoxine-responsive seizures: review of 63 North American cases submitted to a patient registry, Eur J Pediatr 168:697–704, 2009.
  277. Boy N, Haege G, Heringer J, et al: Low lysine diet in glutaric aciduria type I—effect on anthropometric and biochemical follow-up parameters, J Inherit Metab Dis 36:525–533, 2013.
  278. Gospe SM Jr: Pyridoxine-dependent epilepsy. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2001 [updated 2017], University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK1486/. 1993–2017.
  279. Jafari P, Braissant O, Bonaf? L, et al: The unsolved puzzle of neuropathogenesis in glutaric aciduria type I, Mol Genet Metab 104:425–437, 2011. K?lker S, Boy SP, Heringer J, et al: Complementary dietary treatment using lysine-free, arginine-fortified amino acid supplements in glutaric aciduria type I—a decade of experience, Mol Genet Metab 107:72–80, 2012.
  280. Kelker S, Christensen E, Leonard JV, et al: Diagnosis and management of glutaric aciduria type I — revised recommendations, J Inherit Metab Dis 34:677–694, 2011.
  281. Nunes J, Loureiro S, Carvalho S, et al: Brain MRI findings as an important diagnostic clue in glutaric aciduria type 1, Neuroradiol J 26:155–161, 2013.
  282. Ogier de Baulny H, Schiff M, Dionisi-Vici C: Lysinuric protein intolerance (LPI): a multi organ disease by far more complex than a classic urea cycle disorder, Mol Genet Metab 106:12–17, 2012
  283. Sebastio G, Nunes V: Lysinuric protein intolerance. In Adam MP, Ardinger HH, Pagon RA, et al, editors: GeneReviews [Internet], Seattle, 2006 [updated 2011], University of Washington. https://www.ncbi.nlm.nih.gov/books/NBK1361/. 1993–2017.
  284. Stockler S, Plecko B, Gospe SM Jr, et al: Pyridoxine dependent epilepsy and antiquitin deficiency: clinical and molecular characteristics and recommendations for diagnosis, treatment and follow-up, Mol Genet Metab 104:48–60, 2011.
  285. Strauss KA, Donnelly P, Wintermark M: Cerebral haemodynamics in patients with glutaryl-coenzyme A dehydrogenase deficiency, Brain 133(Pt 1):76–92, 2010.
  286. Struys EA, Nota B, Bakkali A, et al: Pyridoxine-dependent epilepsy with elevated urinary ?-amino adipic semialdehyde in molybdenum cofactor deficiency, Pediatrics 130:e1716–e1719, 2012.
  287. Tort F, Ugarteburu O, Torres MA, et al: Lysine restriction and pyridoxal phosphate administration in a NADK2 patient, Pediatrics 138(5):e20154534, 2016.
  288. Tortorelli S, Hahn SH, Cowan TM, et al: The urinary excretion of glutarylcarnitine is an informative tool in the biochemical diagnosis of glutaric acidemia type I, Mol Genet Metab 84(2):137–143, 2005.
  289. Van Karnebeek CD, Stockler-Ipsiroglu S, Jaggumantri S, et al: Lysine-restricted diet as adjunct therapy for pyridoxine-dependent epilepsy: the PDE Consortium Consensus Recommendations, JIMD Rep 15:1–11, 2014.
  290. Zinnanti WJ, Lazovic J: Mouse model of encephalopathy and novel treatment strategies with substrate competition in glutaric aciduria type I, Mol Genet Metab 100(Suppl 1):S88–S91, 2010.
  291. Baslow MH, Guilfoyle DN: Canavan disease, a rare early-onset human spongiform leukodystrophy: insights into its genesis and possible clinical interventions, Biochimie 95(4):946–956, 2013.
  292. Gao G, Su Q, Michals-Matalon K, et al: Efficacious and safe gene therapy for Canavan disease: a novel approach, J Inherit Metab Dis 34(Suppl 3):234, 2011.
  293. Gessler DJ, Li D, Xu H, et al: Redirecting N-acetylaspartate metabolism in the central nervous system normalizes myelination and rescues Canavan disease, JCI Insight 2017. 2.3.
  294. Leone P, Shera D, McPhee SW, et al: Long-term follow-up after gene therapy for Canavan disease, Sci Transl Med 4:165ra163, 2012.
  295. Matalon R, Michals K, Sebasta D, et al: Aspartoacylase deficiency and N-acetylaspartic aciduria in patients with Canavan disease, Am J Med Genet 29:463–471, 1988.
  296. Matalon R, Michals-Matalon K, Surendran S, et al: Canavan disease: studies on the knockout mouse, Adv Exp Med Biol 576:77–93, 2006.
  297. Matalon R, Michals-Matalon K: Canavan disease. In Pagon RA, Bird TD, Colan CR, et al, editors: GeneReviews [Internet], Seattle, 1999 [updated 2011], University of Washington. http://www.ncbi.nlm.nih.gov/books/NBK1234/. 1993–2014.
  298. Michals K, Matalon R: Canavan disease. In Raymond GV, Eichler F, Fatemi A, et al, editors: Leukodystrophies, London, 2011, Mac Keith Press, pp 156–169.
  299. Sommer A, Sass JO: Expression of aspartoacylase (ASPA) and Canavan disease, Gene 505(2):206–210, 2012.
  300. Taylor DL, Davies SE, Obrenovitch TP, et al: Investigation into the role of N-acetylaspartate in cerebral osmoregulation, J Neurochem 65:275–281, 1995.
  301. Zano S, Malik R, Szucs S, et al: Modification of aspartoacylase for potential use in enzyme replacement therapy for the treatment of Canavan disease, Mol Genet Metab 102:176–180, 2011.
  302. Zano S, Wijayasinghe YS, Malik R, et al: Relationship between enzyme properties and disease progression in Canavan disease, J Inherit Metab Dis 36(1):159–160, 2013.
  303. Ahrens-Nicklas RC, Pyle LC, Ficicioglu C: Morbidity and mortality among exclusively breastfed neonates with medium-chain acyl-CoA dehydrogenase deficiency, Genet Med 18:1315–1319,2016.
  304. Dykema DM: Carnitine palmitoyltransferase-1A deficiency: a look at classic and arctic variants, Adv Neonatal Care 12:23–27, 2012.
  305. Ferdinandusse S, Friederich MW, Burlina A, et al: Clinical and biochemical characterization of four patients with mutations in ECHS1, Orphanet J Rare Dis 10:79, 2015.
  306. Fukao T, Sass JO, Kursula P, et al: Clinical and molecular characterization of five patients with succinyl-CoA:3-ketoacid CoA transferase (SCOT) deficiency, Biochim Biophys Acta 1812:619–624, 2011.
  307. Gillingham MB, Weleber RG, Neuringer M, et al: Effect of optimal dietary therapy upon visual function in children with long-chain 3-hydroxyacyl CoA dehydrogenase and trifunctional protein deficiency, Mol Genet Metab 86:124–133, 2005.
  308. Greenberg CR, Dilling LA, Thompson GR, et al: The paradox of the carnitine palmitoyltransferase type 1a P479L variant in Canadian Aboriginal populations, Mol Genet Metab 96:201–207, 2009.
  309. Hsu HW, Zytkovicz TH, Comeau AM, et al: Spectrum of medium-chain acyl-CoA dehydrogenase deficiency detected by newborn screening, Pediatrics 121:e1108–e1114, 2008.
  310. Jethva R, Bennett MJ, Vockley J: Short-chain acyl-coenzyme A dehydrogenase deficiency, Mol Genet Metab 95:195–200, 2008.
  311. Li C, Chen P, Palladino A, et al: Mechanism of hyperinsulinism in short-chain 3-hydroxyacyl-CoA dehydrogenase deficiency involves activation of glutamate dehydrogenase, J Biol Chem 285:31806–31818, 2010.
  312. Longo N, di San Filippo CA, Pasquali M: Disorders of carnitine transport and the carnitine cycle, Am J Med Genet C Semin Med Genet 142C:77–85, 2006.
  313. Loughrey C, Bennett MJ: Screening for MCAD deficiency in newborns, BMJ338:843–846, 2009.
  314. Molven A, Matre GE, Duran M, et al: Familial hyperinsulinemic hypoglycemia caused by a defect in the SCHAD enzyme of mitochondrial fatty acid oxidation, Diabetes53:221–227, 2004.
  315. Peters H, Buck N, Wanders R, et al: ECHS1 mutations in Leigh disease: a new inborn error of metabolism affecting valine metabolism, Brain 137:2903–2908, 2014.
  316. Sass JO: Inborn errors of ketogenesis and ketone body utilization, J Inherit Metab Dis 35:23–28, 2012.
  317. Schulze A, Matern D, Hoffmann GF: Newborn screening. In Sarafoglou K, Hoffmann GF, Roth KS, editors: Pediatric endocrinology and inborn errors of metabolism, New York, 2009, McGraw-Hill, pp 17–32.
  318. Shekhawat PS, Matern D, Strauss AW: Fetal fatty acid oxidation disorders, their effect on maternal health and neonatal outcome: impact of expanded newborn screening on their diagnosis and management, Pediatr Res 57:78R–86R, 2005.
  319. Sinclair GB, Collinc S, Popescu O, et al: Carnitine palmitoyltransferase I and sudden unexpected infant death in British Columbia First Nations, Pediatrics130:e1162–e1169, 2012.
  320. Stanley CA: Perspective on the genetics and diagnosis of congenital hyperinsulinism disorders, J Clin Endocrinol Metab 101:815–826, 2016.
  321. Strauss AW, Andresen BS, Bennett MJ: Mitochondrial fatty acid oxidation defects. In Sarafoglou K, Hoffmann GF, Roth KS, editors: Pediatric endocrinology and inborn errors of metabolism, New York, 2009, McGraw-Hill, pp 51–70.
  322. Van Hasselt PM, Ferdinandusse S, Monroe GR, et al: Monocarboxylate transporter 1 deficiency and ketone utilization, N Engl J Med 371(20):1900–1907, 2014.
  323. Van Maldegem BT, Duran M, Wanders RJA, et al: Fasting and fat-loading tests providepathophysiological insight into short-chain acyl-coenzyme A dehydrogenase deficiency, J Pediatr 156:121–127, 2010.
  324. Wilcken B, Haas M, Joy P, et al: Outcome of neonatal screening for medium-chain acyl-CoA dehydrogenase deficiency in Australia: a cohort study, Lancet 369:37–42, 2007.
  325. Wiles JR, Leslie N, Knilans TK, Akinbi H: Prolonged QTc interval in association with medium-chain acyl-coenzyme A dehydrogenase deficiency, Pediatrics133(6):e1781–e1786, 2014.
  326. Wolfe L, Jethva R, Oglesbee D, et al: Short-chain acyl-CoA dehydrogenase deficiency. In Pagon RA, Bird TD, Dolan CR, et al, editors: GeneReviews [Internet], Seattle, 1993–2011, University of Washington. http://www.ncbi.nlm.nih.gov/books/NBK63582/.
  327. Braverman NE, Raymond GV, Rizzo WB, et al: Peroxisome biogenesis disorders in the Zellweger spectrum: an overview of current diagnosis, clinical manifestations, and treatment guidelines, Mol Genet Metab 117(3):313–321, 2016.
  328. Berger J, Dorninger F, Forss-Petter S, Kunze M: Peroxisomes in brain development and function, Biochim Biophys Acta 1863(5):934–955, 2016.
  329. Klouwer FC, Berendse K, Ferdinandusse S, et al: Zellweger spectrum disorders: clinical overview and management approach, Orphanet J Rare Dis 10:151, 2015.
  330. Aubourg P: Gene therapy for rare central nervous system diseases comes to age, Endocr Dev 30:141–146, 2016.
  331. Berger J, Forss-Petter S, Eichler FS: Pathophysiology of X-linked adrenoleukodystrophy, Biochimie 98:135–142, 2014.
  332. Deon M, Marchetti DP, Donida B, et al: Oxidative stress in patients with X-Linked adrenoleukodystrophy, Cell Mol Neurobiol 36(4):497–512, 2016.
  333. Engelen M, Kemp S, Poll-The BT: X-linked adrenoleukodystrophy: pathogenesis and treatment, Curr Neurol Neurosci Rep 14(10):486, 2014.
  334. Kemp S, Huffnagel IC, Linthorst GE, et al: Adrenoleukodystrophy: neuroendocrine pathogenesis and redefinition of natural history, Nat Rev Endocrinol 12(10):606–615, 2016.
  335. Steinberg SJ, Moser AB, Raymond GV: X-linked adrenoleukodystrophy, 1999 [updated 2015]. In Pagon RA, Adam MP, Ardinger HH, et al, editors: GeneReviews [Internet], Seattle, 1993–2017, University of Washington. http://www-ncbi-nlm-nih-gov.ezp1.lib.umn.edu/books/NBK1315/.
  336. Wiesinger C, Eichler FS, Berger J: The genetic landscape of X-linked adrenoleukodystrophy: inheritance, mutations, modifier genes, and diagnosis, Appl Clin Genet8:109–121, 2015
  337. Austin MA, Hutter CH, Zimmern RL, et al: Familial hypercholesterolemia and coronary heart disease: a huge association review, Am J Epidemiol 160:421–429, 2004.
  338. Bhatnagar D, Soran H, Durrington PN: Hypercholesterolaemia and its management, BMJ 337:503–508, 2008.
  339. Bremer AA, Mietus-Snyder M, Lustig RH: Toward a unifying hypothesis of metabolic syndrome, Pediatrics 129:557–570, 2012.
  340. Brunzell JD: Hypertriglyceridemia, N Engl J Med 357:1009–1016, 2007.
  341. Buonuomo PS, Malamisura M, Macchiaiolo M, et al: Eruptive xanthomas in pipoprotein lipase deficiency, J Pediatr 187:330, 2017.
  342. Centers for Disease Control and Prevention: Prevalence of abnormal lipid levels among youths — United States, 1999–2006, MMWR Morb Mortal Wkly Rep 59:29–33, 2010.
  343. Chan YM, Merkens LS, Connor WB, et al: Effects of dietary cholesterol and simvastatin on cholesterol synthesis in Smith-Lemli-Opitz syndrome, Pediatr Res 65:681–685, 2009.
  344. Cuchel M, Meagher EA, du Toit Theron H, et al: Efficacy and safety of a microsomal triglyceride transfer protein inhibitor in patients with homozygous familial hypercholesterolaemia: a single-arm, open-label, phase 3 study, Lancet 381:40–46, 2013.
  345. Daniels SR, Greer FR, et al: Lipid screening and cardiovascular health in childhood, Pediatrics 122:198–208, 2008.
  346. Expert Panel on Integrated Guidelines for cardiovascular health and risk reduction in children and adolescents: summary report, Pediatrics 128:S1–S44, 2011.
  347. Ford ES, Li C, Zhao G, et al: Concentrations of low-density lipoprotein cholesterol and total cholesterol among children and adolescents in the United States, Circulation 119:1108–1115, 2009.
  348. Gillman MW, Daniels SR: Is universal pediatric lipid screening justified?, JAMA 307:259–260, 2012. Grundy SM, Hansen B, Smith SC, et al: Clinical management of metabolic syndrome: report of the American Heart Association/National Heart, Lung, Blood Institute/ American Diabetes Association Conference on Scientific Issues Related to Management, Circulation 109:551–556, 2004
  349. Jergensen AB, Frikke-Schmidt R, Nordestgaard BG, et al: Loss-of-function mutations in APOC3 and risk of ischemic vascular disease, N Engl J Med 371:32–40, 2014.
  350. Kastelein JJP, Besseling J, Shah S, et al: Anacetrapib as lipid-modifying therapy in patients with heterozygous familial hypercholesterolaemia (REALIZE): a randomized, double-blind, placebo-controlled, phase 3 study, Lancet 385:2153–2160, 2015.
  351. Kavey REW, Mietus-Snyder M: Beyond cholesterol: the atherogenic consequences of combined dyslipidemia, J Pediatr 161:977–979, 2012.
  352. Khera AV, et al: Diagnostic yield and clinical utility of sequencing familial hypercholesterolemia genes in patients with severe hypercholesterolemia, J Am Coll Cardiol 67(22):2578–2589, 2016.
  353. Klancar G, et al: Universal screening for familial hypercholesterolemia in children, J Am Coll Cardiol 66(11):1250–1257, 2015.
  354. Kumanyika SK, Obarzanek E: Population-based prevention of obesity: the need for comprehensive practices of healthful eating, physical activity and energy balance. A scientific statement from American Heart Association Council on Epidemiology and Prevention, Circulation 118:428–464, 2008.
  355. Kusters DM, Caceres M, Coll M, et al: Efficacy and safety of ezetimibe monotherapy in children with heterozygous familial or nonfamilial hypercholesterolemia, J Pediatr 166:1377–1384, 2015.
  356. Lebenthal Y, Horvath A, Dziechciarz P, et al: Are treatment targets for hypercholesterolemia evidence-based? Systematic review and meta-analysis of randomized controlled trials, Arch Dis Child 95:673–680, 2010.
  357. Lozano P, Henrikson NB, Dunn J, et al: Lipid screening in childhood and adolescence for detection of familial hypercholesterolemia: evidence report and systematic review for the US Preventive Services Task Force, JAMA 316(6):645–655, 2016.
  358. Lozano P, Henrikson NB, Morrison CC, et al: Lipid screening in childhood and adolescence for detection of multifactorial dyslipidemia: evidence report and systematic review for the US Preventive Services Task Force, JAMA 316(6):634–644, 2016.
  359. Lufti R, Huang J, Wong HR: Plasmapheresis to treat hypertriglyceridemia in a child with diabetic ketoacidosis and pancreatitis, Pediatrics 129:e195–e198, 2012.
  360. Macchiaiolo M, Gagliardi MG, Toscano A, et al: Homozygous familial hypercholesterolaemia, Lancet 379:1330, 2012.
  361. Magnussen CG, Raitakari OT, Thomson R, et al: Utility of currently recommended pediatric dyslipidemia classifications in predicting dyslipidemia in adulthood, Circulation 117:32–42, 2008.
  362. Manlhiot C, Larsson P, Gurofsky R, et al: Spectrum and management of hypertriglyceridemia among children in clinical practice, Pediatrics 123:458–465, 2009.
  363. Marks D, et al: A review on the diagnosis, natural history, and treatment of familial hypercholesterolaemia, Atherosclerosis 168(1):1–14, 2003.
  364. McCrindle BW, Gidding SS: What should be the screening strategy for familial hypercholesterolemia?, N Engl J Med 375(17):1685–1686, 2016.
  365. The Medical Letter: Lipid-lowering drugs, Med Lett Drugs Ther 58:133–140, 2016.
  366. The Medical Letter: Fenofibric acid (Trilipix), Med Lett Drugs Ther 51:33–34, 2009.
  367. The Medical Letter: Icosapent ethyl (Vascepa) for severe hypertriglyceridemia, Med Lett Drugs Ther 55:33–34, 2013.
  368. The Medical Letter: Drugs for hypertriglyceridemia, Med Lett Drugs Ther 55:17–20, 2013.
  369. The Medical Letter: Two new drugs for homozygous familial hypercholesterolemia, Med Lett Drugs Ther 55:25–26, 2013.
  370. Merkens LS, Connor WE, Linck LM, et al: Effects of dietary cholesterol on plasma lipoproteins in Smith-Lemli-Opitz syndrome, Pediatr Res 56:726–732, 2004.
  371. Othman RA, Myrie SB, Mymin D, et al: Ezetimibe reduces plant sterol accumulation and favorably increases platelet count in sitosterolemia, J Pediatr 166:125–131, 2015.
  372. Psaty BM, Rivara FP: Universal screening and drug treatment of dyslipidemia in children and adolescents, JAMA 307:257–258, 2012.
  373. Raal FJ: Lomitapide for homozygous familial hypercholesterolaemia, Lancet 381:7–8, 2013.
  374. Raitakari OT: Arterial abnormalities in children with familial hypercholesteremia, Lancet 363:342–343, 2004.
  375. Roth EM, McKenney JM, Hanotin C, et al: Atorvastatin with or without an antibody to PCSK9 in primary hypercholesterolemia, N Engl J Med 367:1891–1900, 2012.
  376. Silverstein J, Haller M: Coronary artery disease in youth: present markers, future hope?, J Pediatr 157(4):523–524, 2010.
  377. Stein EA, Raal FJ: Polygenic familial hypercholesterolaemia: does it matter?, Lancet 381:1255–1258, 2013.
  378. Steiner MJ, Skinner AC, Perrin EM: Fasting might not be necessary before lipid screening: a nationally representative cross-sectional study, Pediatrics 128(3):463–470, 2011.
  379. The SEARCH Collaborative Group: SLCO1B1 variants and statin-induced myopathy — a genomewide study, N Engl J Med 359:789–799, 2008.
  380. The TG and HDL Working Group of the Exome Sequencing Project, National Heart, Lung, and Blood Institute: Loss-of-function mutations in APOC3 triglycerides, and coronary disease, N Engl J Med 371:22–30, 2014.
  381. Urbina EM, de Ferranti SD: Lipid screening in children and adolescents, JAMA 316(6):589–591, 2016
  382. US Preventive Services Task Force: Screening for lipid disorders in children and adolescents, JAMA 316(6):625–632, 2016.
  383. Vinci S, et al: Cholesterol testing among children and adolescents during health visits, JAMA 311(17):1804–1806, 2014.
  384. Wald DS, Bestwick JP, Morris JK, et al: Child-parent familial hypercholesterolemia screening in primary care, N Engl J Med 375(17):1628–1636, 2016.
  385. Wald DS, Kasturiratne A, Godoy A, et al: Child-parent screening for familial hypercholesterolaemia, J Pediatr 159:865–867, 2011.
  386. Webber BJ, Seguin PG, Burnett DG, et al: Prevalence of and risk factors for autopsydetermined atherosclerosis among US service members, JAMA 308:2577–2582, 2012.
  387. Andersson HC: Newborn screening + enzyme replacement therapy = improved lysosomal storage disorder: outcomes in infantile-onset Pompe disease, J Pediatr 166(4):800–801, 2015.
  388. Andersson H, Kaplan P, Kacena K, et al: Eight-year clinical outcomes of long-term enzyme replacement therapy for 884 children with Gaucher disease type 1, Pediatrics 122:1182–1190, 2008.
  389. Balwani M, Burrow TA, Charrow J, et al: Recommendations for the use of eliglustat in the treatment of adults with Gaucher disease type 1 in the United States, Mol Genet Metab 117:95–103, 2015.
  390. Burton BK, Balwani M, Feillet F, et al: A phase 3 trial of sebelipase alfa in lysosomal acid lipase deficiency, N Engl J Med 373:1010–1020, 2015.
  391. Burton BK, Charrow J, Hoganson GE, et al: Newborn screening for lysosomal storage disorders in Illinois: the initial 15-month experience, J Pediatr 190:130–135, 2017.
  392. Cox TM, Drelichman G, Cravo R, et al: Eliglustat compared with imiglucerase in patients with Gaucher’s disease type 1 stabilized on enzyme replacement therapy: a phase 3, randomized, open-label, non-inferiority trial, Lancet 385:1355–2362, 2015.
  393. Desnick RJ, Schuchman EH: Enzyme replacement therapy for lysosomal diseases: lessons from 20 years of experience and remaining challenges, Annu Rev Genomics Hum Genet 13:307–335, 2012.
  394. Erickson RP, Fiorenza MT: A hopeful therapy for Niemann-Pick C diseases, Lancet 390:1720–1721, 2017.
  395. Escolar ML, Poe MD, Provenzale JM, et al: Transplantation of umbilical-cord blood in babies with infantile Krabbe’s disease, N Engl J Med 20:2069–2080, 2005.
  396. Germani DP, Hughes DA, Nicholls K, et al: Treatment of Fabry’s disease with the pharmacologic chaperone migalastat, N Engl J Med 375(6):545–554, 2016.
  397. Giugliani R, Vairo F, Kubaski F, et al: Neurological manifestations of lysosomal disorders and emerging therapies targeting the CNS, Lancet 2:56–68, 2018.
  398. Grabowski GA: Phenotype, diagnosis, and treatment of Gaucher’s disease, Lancet 372:1263–1271, 2008.
  399. Guggenbuhl P, Grosbois B, Chal?s G: Gaucher disease, Joint Bone Spine 75:116–124, 2008.
  400. Hughes DA, Pastores GM: Eliglustat for Gaucher’s disease: trippingly on the tongue, Lancet 385:2328–2330, 2015.
  401. Kaplan P, Andersson HC, Kacena KA, Yee JD: The clinical and demographic characteristics of nonneuronopathic Gaucher disease in 887 children at diagnosis, Arch Pediatr Adolesc Med 160:603–608, 2006.
  402. Lachmann RH: Enzyme replacement therapy for lysosomal storage diseases, Curr Opin Pediatr 23:588–593, 2011.
  403. Marchesoni CL, Roa N, Pardal AM, et al: Misdiagnosis in Fabry disease, J Pediatr 156:828–831, 2010.
  404. Martin HR, Poe MD, Provenzale JM: Neurodevelopmental outcomes of umbilical cord blood transplantation in metachromatic leukodystrophy, Biol Blood Marrow Transplant 19:616–624, 2013.
  405. McGovern MM, Avetisyan R, Sanson BJ, Lidove O: Disease manifestations and burden of illness in patients with acid sphingomyelinase deficiency (ASMD), Orphanet J Rare Dis 12:41–53, 2017.
  406. Mechtler TP, Stary S, Metz TF, et al: Neonatal screening for lysosomal storage disorders: feasibility and incidence from a nationwide study in Austria, Lancet 379:335–340, 2012.
  407. Mistry PK, Belmatoug N, vom Dahl S, Giugliani R: Understanding the natural history of Gaucher disease, Am J Hematol 90(51):S6–S11, 2015.
  408. Mistry PK, Lukina E, Turkia HB, et al: Effect of oral eliglustat on splenomegaly in patients with Gaucher disease type 1: the ENGAGE randomized clinical trial, JAMA 313(7):695–706, 2015.
  409. Ory DS, Ottinger EA, Farhat NY, et al: Intrathecal 2-hydroxypropropyl-?-cyclodextrin decreases neurological disease progression in Niemann-Pick disease type C1: a non-randomised, open-label, phase 1-2 trial, Lancet 390:1758–1768, 2017.
  410. Park NJ, Morgan C, Sharma R, et al: Improving accuracy of Tay Sachs carrier screening of the non-Jewish population: analysis of 34 carriers and six late-onset patients with HEXA enzyme and DNA sequence analysis, Pediatr Res 67:217–220, 2010.
  411. Ranieri M, Bedini G, Parati EA, Bersano A: Fabry disease: recognition, diagnosis, and treatment of neurological features, Curr Treat Options Neurol 18:33, 2016.
  412. Rossi L, Zulian F, Stirnemann J: Bone involvement as presenting sign of pediatric-onset Gaucher disease, Joint Bone Spine 78:70–74, 2011.
  413. Sethuraman G, Chouhan K, Kaushi S, et al: Fabry’s disease, Lancet 378:1254, 2011. Sivley MD: Fabry disease: a review of ophthalmic and systemic manifestations, Optom Vis Sci 90(2):e63–e78, 2013.
  414. Staretz-Chachem O, Lang TC, LaMarca ME, et al: Lysosomal storage disorders in the newborn, Pediatrics 123:1191–1207, 2009.
  415. Stirnemann J, Belmatoug N, Canou F, et al: A review of Gaucher disease pathophysiology, clinical presentation and treatments, Int J Mol Sci 18:441, 2017.
  416. Stirnemann J, Vigan M, Hamroun D, et al: The French Gaucher’s Disease Registry: clinical characteristics, complications and treatment of 562 patients, Orphanet J Rare Dis 7:77, 2012.
  417. Wraith JE, Tylki-Szymanska A, Guffon N, et al: Safety and efficacy of enzyme replacement therapy with agalsidase beta: an international, open-label study in pediatric patients with Fabry disease, J Pediatr 152:563–570, 2008.
  418. Yang AC, Bier L, Overbey JR, et al: Early manifestations of type 1 Gaucher disease in presymptomatic children diagnosed after parental carrier screening, Genet Med 19:652–658, 2016.
  419. Zhou H, Fernhoff P, Vogt RF: Newborn bloodspot screening for lysosomal storage disorders, J Pediatr 159:7–13, 2011.
  420. Kudo M, Brem MS, Canfield WM. Mucolipidosis II (I-cell disease) andmucolipidosis IIIA (classical pseudo-Hurler polydystrophy) arecausedby mutationsin the GlcNAcphosphotransferase alpha/beta-subunits precursor gene. Am J Hum Genet.2006;78:451–463.
  421. Akman HO, Aykit Y, Amuk OC, et al: Late-onset polyglucosan body myopathy in five patients with a homozygous mutation in GYG1, Neuromuscul Disord 26(1):16–20, 2016.
  422. Byrne BJ, Falk DJ, Pacel CA, et al: Pompe disease gene therapy, Hum Mol Genet 20:R61–R68, 2011.
  423. Case LE, Beckemeyer AA, Kishnani PS: Infantile Pompe disease on ERT—update on clinical presentation, musculoskeletal management, and exercise considerations, Am J Med Genet C Semin Med Genet 160C:69–79, 2012.
  424. Chiang SC, Hwu WL, Lee NC, et al: Algorithm for Pompe disease newborn screening: results from the Taiwan screening program, Mol Genet Metab 106(3):281–286, 2012.
  425. Chien YH, Lee NC, Huang HJ, et al: Later-onset Pompe disease: early detection and early treatment initiation enabled by newborn screening, J Pediatr 158:1023–1027, 2011.
  426. D’Souza R, Levandowski C, Slavov D, et al: Danon disease: clinical features, evaluation, and management, Circ Heart Fail 7(5):843–849, 2014.
  427. DiMauro S, Spiegel R: Progress and problems in muscle glycogenoses, Acta Myol30(2):96–102, 2011.
  428. Elder ME, Nayak S, Collins SW, et al: B-cell depletion and immunomodulation before initiation of enzyme replacement therapy blocks the immune response to acid alpha-glucosidase in infantile-onset Pompe disease, J Pediatr 163:847–854, 2013.
  429. Hopkins PV, Campbell C1, Klug T, et al: Lysosomal storage disorder screening implementation: findings from the first six months of full population pilot testing in Missouri, J Pediatr 166(1):172–177, 2015.
  430. Kishnani PS, Austin SL, Abdenur JE, et al: Diagnosis and management of glycogen storage disease type I: a practice guideline of the American College of Medical Genetics and Genomics, Genet Med 16(11):e1, 2014.
  431. Kishnani PS, Austin SL, Arn P, et al: Glycogen storage disease type III diagnosis and management guidelines, Genet Med 12(7):446–463, 2010
  432. Kronn DF, Day-Salvatore D, Hwu WL, et al: Management of confirmed newbornscreened patients with Pompe disease across the disease spectrum, Pediatrics140(1):e20160280, 2016.
  433. Maga JA, Zhou J, Kambampati R, et al: Glycosylation-independent lysosomal targeting of acid ?-glucosidase enhances muscle glycogen clearance in Pompe mice, J Biol Chem 288(3):1428–1438, 2013.
  434. Miteff F, Potter HC, Allen J, et al: Clinical and laboratory features of patients with myophosphorylase deficiency (McArdle disease), J Clin Neurosci 18(8):1055–1058, 2011.
  435. Poelman E, Hoogeveen-Westerveld M, Kross-de Haan MA, et al: High sustained antibody titers in patients with classic infantile Pompe disease following immunomodulation at start of enzyme replacement therapy, J Pediatr 195:236–243, 2018.
  436. Porto A, Brun F, Severini G, et al: Clinical spectrum of PRKAG2 syndrome, Circ Arrhythm Electrophysiol 9(1):e003121, 2016.
  437. Rimoin DL, Connor JM, Pyeritz RE, et al: Emery and Rimoin’s principles and practice of medical genetics, New York, 2011, Churchill Livingstone Elsevier.
  438. Roscher A, Patel J, Hewson S, et al: The natural history of glycogen storage disease types VI and IX: long-term outcome from the largest metabolic center in Canada, Mol Genet Metab 113(3):171–176, 2014.
  439. Valayannopoulos V, Bajolle F, Arnoux JB, et al: Successful treatment of severe cardiomyopathy in glycogen storage disease type III with D,L-3-hydroxybutyrate, ketogenic and high-protein diet, Pediatr Res 70:638–641, 2011.
  440. Valle D, Beaudet AL, Vogelstein B, et al: Online metabolic and molecular bases of inherited disease, 2006 [updated 2011]. http://www.ommbid.com.
  441. Wang DQ, Fiske LM, Carreras CT, et al: Natural history of hepatocellular adenoma formation in glycogen storage disease type 1, J Pediatr 159:442–446, 2011.
  442. Yang CF, Yang CC, Liao HC, et al: Very early treatment for infantile-onset Pompe disease contributes to better outcomes, J Pediatr 169:174–180, 2016
  443. Fridovich-Keil JL,Walter JH.Galactosemia.The online metabolic & molecular bases of inherited disease. McGraw-Hill:NewYork;2009 http://www.ommbid.com
  444. Potter NL, Nievergelt Y, Shriberg LD. Motor and speech disorders in classic galactosemia. JIMDRep. 2013;11:31–41.
  445. Rimoin DL, Connor JM, Pyeritz RE,et al. Emery and Rimoin's principles and practice of medical genetics.Churchill Livingstone Elsevier: New York; 2011.
  446. Waisbren SE, Potter NL, Gordon CM, et al.The adult galactosemic phenotype. JInherit Metab Dis. 2012;35(2):279–286.
  447. Bouteldja N, TimsonDJ. The biochemical basis ofhereditary fructose intolerance. J Inherit MetabDis.2010;33(2):105– 112.
  448. Rimoin DL, Connor JM, Pyeritz RE,et al. Emery and Rimoin's principlesand practice of medical genetics.Churchill Livingstone Elsevier: New York;2011.
  449. Steinmann B,Gitzemann R,van den Berghe G. Disordersof fructose metabolism. Theonline metabolic andmolecular bases of inheriteddisease–OMMBID. McGraw-Hill: NewYork; 2009
  450. Barnerias C, Saudubray JM, Touti G, et al: Pyruvate dehydrogenase complex deficiency: four neurological phenotypes with differing pathogenesis, Dev Med Child Neurol 52(2):1–9, 2010.
  451. Davis RL, Liang C, Sue CM: A comparison of current serum biomarkers as diagnostic indicators of mitochondrial diseases, Neurology 86:2010–2015, 2016.
  452. Fassone E, Rahman S: Complex I deficiency: clinical features, biochemistry, and molecular genetics, J Med Genet 49(9):578–590, 2012.
  453. Ghaddhab C, Morin C, Brunel-Guitton C, et al: Premature ovarian failure in French Canadian Leigh syndrome, J Pediatr 184:227–229, 2017.
  454. Koopman WJH, Distelmaier F, Smeitink JA, et al: OXPHOS mutations and neurodegeneration, EMBO J 32(1):9–29, 2013.
  455. Koopman WJH, Willema PHGM, Smeitink JAM: Monogenic mitochondrial disorders, N Engl J Med 366:1132–1140, 2012.
  456. Mercimek-Mahmutoglu S, Horvath GA, Coulter-Mackie M, et al: Profound neonatal hypoglycemia and lactic acidosis caused by pyridoxine-dependent epilepsy, Pediatrics 129:e1368–e1372, 2012
  457. Montero R, Yubero D, Villarroya J, et al: GDF-15 is elevated in children with mitochondrial diseases and is induced by mitochondrial dysfunction, PLoS ONE 11(2):e0148709, 2016.
  458. Perez-Dueoas B, Serrano M, Rebollo M, et al: Reversible lactic acidosis in a newborn with thiamine transporter-2 deficiency, Pediatrics 131:e1670–e1675, 2013.
  459. Rimoin DL, Connor JM, Pyeritz RE, et al: Emery and Rimoin’s principles and practice of medical genetics, New York, 2011, Churchill Livingstone Elsevier. Steinmann B, Gitzemann R, van den Berghe G: Disorders of fructose metabolism. In The online metabolic and molecular bases of inherited disease–OMMBID, New York, 2009, McGraw-Hill. http://www.ommbid.com.
  460. Vernon C, LeTourneau JL: Lactic acidosis: recognition, kinetics, and associated prognosis, Crit Care Clin 26:255–283, 2010. Vernon HJ: Inborn errors of metabolism: advances in diagnosis and therapy, JAMA Pediatr 169(8):778–782, 2015.
  461. Wallace DC: Mitochondrial DNA mutations in disease and aging, Environ Mol Mutagen 51(5):440–450, 2010
  462. Copeland WC. Defects of mitochondrial DNA replication.J Child Neuro2014;29(9):1216–1224.
  463. Pierce SB, Spurrell CH, Mandell JB, et al. Garrod's fourth inbornerror of metabolism solved by the identification of mutations causing pentosuria. Proc Natl AcadSci USA. 2011; 108(45): 18313–18317.
  464. Rimoin DL, Connor JM, Pyeritz RE,et al. Emery and Rimoin's principlesand practiceofmedical genetics.Churchill Livingstone Elsevier: New York;2011.
  465. Valle D, Beaudet AL,Vogelstein B,et al. Online metabolic and molecular baseof inherited disease2006.[updated2011] http://www.ommbid.com;.
  466. Wamelink MMC, Struys EA,Jakobs C. The biochemistry, metabolism and inherited defects of the pentose phosphate pathway: a review. J InheritMetabDis.2008;31:703–717.
  467. Arvio M, Mononen I: Aspartylglycosaminuria: a review, Orphanet J Rare Dis 11:162–172, 2016.
  468. Borgwardt L, Stensland HM, Olsen KJ, et al: Alpha-mannosidosis: correlation between phenotype, genotype and mutant MAN2B1 subcellular localization, Orphanet J Rare Dis 10:70–86, 2015.
  469. Freeze HH, Eklund EA, Ng BG, et al: Neurological aspects of human glycosylation disorders, Annu Rev Neurosci 38:105–125, 2015
  470. Stroobants S, Damme M, Van der Jeugd A, et al: Long-term enzyme replacement therapy improves neurocognitive functioning and hippocampal synaptic plasticity in immune-tolerant alpha-mannosidosis mice, Neurobiol Dis 106:255–268, 2017.
  471. Tegtmeyer LC, Rust S, van Scherpenzeel M, et al: Multiple phenotypes in phosphoglucomutase 1 deficiency, N Engl J Med 370:533–542, 2014.
  472. De Lonlay P, Seta N: The clinical spectrum of phosphomannose isomerase deficiency, with an evaluation of mannose treatment for CDG-Ib, Biochim Biophys Acta1792(9):841–843, 2009.
  473. Freeze HH, Chong JX, Bamshad MJ, Ng BG: Solving glycosylation disorders: fundamental approaches reveal complicated pathways, Am J Hum Genet 94:161–175, 2014.
  474. Freeze HH, Eklund EA, Ng BG, Patterson MC: Neurology of inherited glycosylation disorders, Lancet Neurol 11:453–466, 2012.
  475. Funke S, Gardeitchik T, Kouwenberg D, et al: Perinatal and early infantile symptoms in congenital disorders of glycosylation, Am J Med Genet 161A:578–584, 2013.
  476. Jaeken J, Hennet T, Matthijs G, Freeze HH: CDG nomenclature: time for a change!, Biochim Biophys Acta 1792:825–826, 2009.
  477. Jaeken J, et al: Sialic acid-deficient serum and cerebrospinal fluid transferrin in a newly recognized genetic syndrome, Clin Chim Acta 144:245–247, 1984.
  478. Lam C, Ferreira C, Krasnewich D, et al: Prospective phenotyping of NGLY1-CDDG, the first congenital disorder of deglycosylation, Genet Med 19(2):160–168, 2017.
  479. Lefeber DJ, Morava E, Jaeken J: How to find and diagnose a CDG due to defective N-glycosylation, J Inherit Metab Dis 34(4):849–852, 2011.
  480. Mohamed M, Kouwenberg D, Gardeitchik T, et al: Metabolic cutis laxa syndromes, J Inherit Metab Dis 34:907–916, 2011
  481. Morava E: Galactose supplementation in 1 phosphoglucomutase-1 deficiency: review and outlook for a novel treatable CDG, Mol Genet Metab 112:275–279, 2014.
  482. Morava E, Tiemes V, Thiel C, et al: ALG6-CDG: a recognizable phenotype with epilepsy, proximal muscle weakness, ataxia and behavioral and limb anomalies, J Inherit Metab Dis 39(5):713–723, 2016.
  483. Morava E, Wevers RA, Cantagrel V, et al: A novel cerebello-ocular syndrome with abnormal glycosylation due to abnormalities in dolichol metabolism, Brain 133:3210–3220, 2010.
  484. Perez-Cerde C, Gir?s ML, Serrano M, et al: A population-based study on congenital disorders of protein N- and combined with O-glycosylation: experience in clinical and genetic diagnosis, J Pediatr 183:170–177, 2017.
  485. Rymen D, Winter J, van Hasselt PM, et al: Key features and clinical variability of COG6-CDG, Mol Genet Metab 116(3):163–170, 2015.
  486. Scott K, Gadomski T, Kozicz T, Morava E: Congenital disorders of glycosylation: new defects and still counting, J Inherit Metab Dis 37:609–617, 2014.
  487. Wong SY, Beamer LJ, Gadomski T, et al: Defining the phenotype and assessing severity in phosphoglucomutase-1 deficiency, J Pediatr 175:130–136.e8, 2016.
  488. Alston CL, Rocha MC, Lax NZ, et al: The genetics and pathology of mitochondrial disease, J Pathol 241(2):236–250, 2017.
  489. Avula S, et al: Treatment of mitochondrial disorders, Curr Treat Options Neurol16(6):292, 2014.
  490. Calvo SE, Clauser KR, Mootha VK: MitoCarta2.0: an updated inventory of mammalian mitochondrial proteins, Nucleic Acids Res 44(D1):D1251–D1257, 2016.
  491. Camp KM, et al: Nutritional interventions in primary mitochondrial disorders: Developing an evidence base, Mol Genet Metab 119(3):187–206, 2016.
  492. Chinnery P, et al: Treatment for mitochondrial disorders, Cochrane Database Syst Rev (1):CD004426, 2006.
  493. Copeland WC: Defects of mitochondrial DNA replication, J Child Neurol 29(9):1216–1224, 2014.
  494. Davis RL, Liang C, Sue CM: A comparison of current serum biomarkers as diagnostic indicators of mitochondrial diseases, Neurology 86:2010–2015, 2016.
  495. Di Meo I, Lamperti C, Tiranti V: Mitochondrial diseases caused by toxic compound accumulation: from etiopathology to therapeutic approaches, EMBO Mol Med7:1257–1266, 2015.
  496. Distelmaier F, Haack TB, Wortmann SB, et al: Treatable mitochondrial diseases: cofactor metabolism and beyond, Brain 140(2):e11, 2017.
  497. Falk MJ, Decherney A, Kahn JP: Mitochondrial Replacement Techniques–Implications for the Clinical Community, N Engl J Med 374(12):1103–1106, 2016.
  498. Falk MJ, et al: Mitochondrial Disease Sequence Data Resource (MSeqDR): a global grass-roots consortium to facilitate deposition, curation, annotation, and integrated analysis of genomic data for the mitochondrial disease clinical and research communities, MolGenetMetab 114(3):388–396, 2015.
  499. Finsterer J, Ahting U: Mitochondrial depletion syndromes in children and adults, Can J NeurolSci 40:635–644, 2013.
  500. Gorman GS, et al: Mitochondrial diseases, Nat Rev Dis Primers 2:16080, 2016.
  501. Gorman GS, et al: Prevalence of nuclear and mitochondrial DNA mutations related to adult mitochondrial disease, Ann Neurol 77(5):753–759, 2015.
  502. Haas RH, Parikh S, Falk MJ, et al: The in-depth evaulation of suspected mitochondrial disease, Mol Genet Metab 94(1):16–37, 2008.
  503. Hopkins SE, Somoza A, Gilbert DL: Rare autosomal dominant POLGI mutation in a family with metabolic strokes, posterior column spinal degeneration and multi-endocrine disease, J Child Neurol 25(6):752–756, 2010.
  504. Khan NA, Govindaraj P, Meena AK: Mitochondrial disorders: challenges in diagnosis and treatment, Indian J Med Res 141(1):13–26, 2015.
  505. Paschke E, Milos I, Kreimer-Erlacher H, et al: Mutation analyses in 17 patients with deficiency in acid beta-galactosidase: three novel point mutations and high correlation of mutation W273L with Morquio disease type B, Hum Genet 109:159–166, 2001.
  506. Poe MD, Chagnon SL, Escolar L: Early treatment is associated with improved cognition in Hurler syndrome, Ann Neurol 76:747–753, 2014.
  507. Rodgers NJ, Kaizer AM, Miller WP: Mortality after hematopoietic stem cell transplantation for severe mucopolysaccharidosis type I: the 30-year University of Minnesota experience, J Inherit Metab Dis 40:271–280, 2017.
  508. Scarpa M: Mucopolysaccharidosis type II. In Pagon RA, Adam MP, Ardinger HH, et al, editors: Gene Reviews (Internet). Seattle, 2015.
  509. Shapiro EG, Nestrasil I, Delaney KA, et al: A prospective natural history study of mucopolysaccharidosis type IIIA, J Pediatr 170:278–287, 2016.
  510. Solanki GA, Martin KW, Theroux MC, et al: Spinal involvement in mucopolysaccharidosis IVA (Morquio-Brailsford or Morquio A syndrome): presentation, diagnosis and management, J Inherit Metab Dis 36:339–355, 2013.
  511. Tomatsu S, Alm?ciga-Diaz CJ, Montano AM, et al: Therapies for the bone in mucopolysaccharidoses, Mol Genet Metab 114:94–108, 2015.
  512. Triggs-Raine B, Salo TJ, Zhang H, et al: Mutations in HYAL1, a member of a tandemly distributed multigene family encoding disparate hyaluronidase activities, cause a newly described lysosomal disorder, mucopolysaccharidosis IX, Proc Natl Acad Sci USA 96:6296–6300, 1999.
  513. Tylki-Szymanska A, De Meirleir L, Di Rocco M, et al: Easy-to use algorithm would provide faster diagnoses of mucopolysaccharidosis type I and enable patients to receive earlier treatment, Acta Paediatr 107:1402–1408, 2018.
  514. Valayannopoulos V, Nicely H, Harmatz P, Turbeville S: Mucopolysaccharidosis VI, Orphanet J Rare Dis 5:5, 2010.
  515. Valstar MJ, Ruijter GJ, Van Diggelen OP, et al: Sanfilippo syndrome: a mini-review, J Inherit Metab Dis 31:240–252, 2008.
  516. Whitley CB, Cleary M, Mengel KE, et al: Observational prospective natural history of patients with Sanfilippo syndrome type B, J Pediatr 197:198–206, 2018.
  517. Yasuda E, Suzuki Y, Shimada T, et al: Activity of daily living for Morquio A syndrome, Mol Genet Metab 118:111122, 2016
  518. Balasubramaniam S, Duley JA, Christodoulou J: Inborn errors of purine metabolism: clinical update and therapies, J Inherit Metab Dis 37(5):669–686, 2014.
  519. Balasubramaniam S, Duley JA, Christodoulou J: Inborn errors of pyrimidine metabolism: clinical update and therapy, J Inherit Metab Dis 37(5):687–698, 2014.
  520. Bleyer AJ, Kidd K, ?ivn? M, et al: Autosomal dominant tubulointerstitial kidney disease, Adv Chronic Kidney Dis 24(2):86–93, 2017.
  521. Camici M, Micheli V, Ipata PL, et al: Pediatric neurological syndromes and inborn errors of purine metabolism, Neurochem Int 56:367–378, 2010.
  522. Castro M, Carrillo R, Garc?a F, et al: Thirteen years experience with selective screening for disorders in purine and pyrimidine metabolism, Nucleosides Nucleotides Nucleic Acids 33(4–6):233–240, 2014.
  523. Gettle M, Burhenne H, Sutcliffe D, et al: Purine metabolism during neuronal differentiation: the relevance of purine synthesis and recycling, J Neurochem 127(6):805–818, 2013.
  524. Jinnah HA, Sabina RL, van den Berghe G: Metabolic disorders of purine metabolism affecting the nervous system, Handb Clin Neurol 113:1827–1836, 2013.
  525. Loffler M, Fairbanks LD, Zameitat E, et al: Pyrimidine pathways in health and disease, Mol Med 11:430–437, 2005.
  526. Koopman WJ, Willems PH, Smeitink JA: Monogenic mitochondrial disorders, N Engl J Med 366(12):1132–1141, 2012.
  527. Lake NJ, et al: Leigh syndrome: One disorder, more than 75 monogenic causes, Ann Neurol 79(2):190–203, 2016.
  528. McCormick EM, Muraresku CC, Falk MJ: Mitochondrial genomics: a complex filed now coming of age, Curr Genet Med Rep 6:52–61, 2018.
  529. McCormick E, Place E, Falk MJ: Molecular genetic testing for mitochondrial disease: from one generation to the next, Neurother 10(2):251–261, 2013.
  530. Mitochondrial Medicine Society’s Committee on, D., et al: The in-depth evaluation of suspected mitochondrial disease, Mol Genet Metab 94(1):16–37, 2008.
  531. Montero R, Yubero D, Villarroya J, et al: GDF-15 elevated in children with mitochondrial diseases and is induced by mitochondrial dysfunction, PLoS ONE 11(2):e148709, 2016.
  532. Muraresku CC, McCormick EM, Falk MJ: Mitochondrial disease: advances in clinical diagnosis, management, therapeutic development, and preventative strategies, Curr Genet Med Rep 6:62–72, 2018.
  533. Niezgoda J, Morgan PG: Anesthetic considerations in patients with mitochondrial defects, PaediatrAnaesth 23:785–793, 2013.
  534. Ohno K, Yamamoto M, Engel AG, et al: MELAS- and Kearns-Sayre-type commutation with myopathy and autoimmune polyendocrinopathy, Ann Neurol 39(6):761–766, 1996.
  535. Parikh S, et al: A modern approach to the treatment of mitochondrial disease, Curr Treat Options Neurol 11(6):414–430, 2009.
  536. Parikh S, et al: Diagnosis and management of mitochondrial disease: a consensus statement from the Mitochondrial Medicine Society, Genet Med 17(9):689–701, 2015.
  537. Parikh S, et al: Practice patterns of mitochondrial disease physicians in North America. Part 1: diagnostic and clinical challenges, Mitochondrion 14(1):26–33, 2014.
  538. Pfeffer G, et al: New treatments for mitochondrial disease-no time to drop our standards, Nat Rev Neurol 9(8):474–481, 2013.
  539. Quinzil CM, Hirano M: Primary and secondary CoQ10 deficiencies in humans, Intern Union BiochemMolBiol 37(5):361–365, 2011.
  540. Suomalainen A, Isohanni P: Mitochondrial DNA depletion syndromes - many genes, common mechanisms, NeuromusculDisord 20:429–437, 2010.
  541. Taylor RW, Pyle A, Griffin H, et al: Use of whole-exome sequencing to determine the genetic basis of multiple mitochondrial respiratory chain complex deficiencies, JAMA 312(1):68–77, 2014.
  542. Tulinius MH, Oldfors A, Holme E, et al: Atypical presentation of multisystem disorders in two girls with mitochondrial DNA deletions, Eur J Pediatr 154:35–42, 1995.
  543. Tzoufi M, Makis A, Chaliasos N, et al: A rare case of simultaneous presentation of myopathy, Addison’s disease primary hypoparathyroidism and Fanconi syndrome in a child diagnosed with Kearns-Sayre syndrome, Eur J Pediatr 172:557–561, 2013.
  544. Uusimaa J, Evans J, Smith C, et al: Clinical, biochemical, cellular and molecular characterization of mitochondrial DNA depletion syndrome due to novel mutations in the MPV17 gene, Eur J Hum Genet 22:184–191, 2014.
  545. Vernon HJ: Inborn errors of metabolism - advances in diagnosis and therapy, JAMA Pediatr 169(8):778–782, 2015.
  546. Zhang Z, et al: Primary respiratory chain disease causes tissue-specific dysregulation of the global transcriptome and nutrient-sensing signaling network, PLoS ONE8(7):e69282, 2013.
  547. Zolkipli-Cunningham Z, Xiao R, Stoddart A, et al: Mitochondrial disease patient motivations and barriers to participate in clinical trials, PLoS ONE 13(5):e01975132018, 2018. doi:10.1371/journal.pone.0197513. eCollection 2018.
  548. Burton BK, Jego V, Mikl J, et al: Survival in idursulfase-treated and untreated patients with mucopolysaccharidosis type II: data from the Hunter Outcome survey (HOS), J Inherit Metab Dis 40:867–874, 2017.
  549. Clarke LA: Mucopolysaccharidosis type I. In Pagon RA, Adam MP, Ardinger HH, et al, editors: Gene reviews (internet), Seattle, 2016.
  550. Clarke LA, Atherton AM, Burton BK, et al: Mucopolysaccharidosis type I newborn screening: best practices for diagnosis and management, J Pediatr 182:363–370, 2017.
  551. Dvorakova L, Vlaskova H, Sarjlija A, et al: Genotype-phenotype correlation in 44 Czech, Slovak, Croatian and Serbian patients with mucopolysaccharidosis type II, Clin Genet 91:787–796, 2017.
  552. Eisengart JV, Rudser KD, Xue Y, et al: Long-term outcomes of systemic therapies for Hurler syndrome: an international multicenter comparison, Genet Med 2018. doi:10.1038/gim.2018.29.
  553. Escolar ML, Jones SA, Shapiro EG, et al: Practical management of behavioral problems in mucopolysaccharidoses disorders, Mol Genet Metab 122:35–40, 2017.
  554. Ghosh A, Miller W, Orachard PJ, et al: Enzyme replacement therapy prior to haematopoietic stem cell transplantation in mucopolysaccharidosis type I: 10 year combined experience of 2 centers, Mol Genet Metab 117:373–377, 2016.
  555. Giugliani R, Lampe C, Guffon N, et al: Natural history and galsulfase treatment in mucopolysaccharidosis VI (MPS VI, Maroteaux-Lamy syndrome): 10-year follow-up of patients who previously participated in an MPS VI survey study, Am J Med Genet 164:1953–1964, 2014.
  556. Harmatz P, Hendriksz CJ, Lampe C, et al: The effect of galsulfase enzyme replacement therapy on the growth of patients with mucopolysaccharidosis VI (Maroteaux-Lamy syndrome), Mol Genet Metab 122:107–112, 2017.
  557. Hendriksz CJ, Berger KI, Giugliani R, et al: International guidelines for the management and treatment of Morquio A syndrome, Am J Med Genet 167:1125, 2015.
  558. Hendricksz C, Parini R, Moeenaldeen D, et al: Long-term endurance and safety of elosulfase alfa enzyme replacement therapy in patients with Morquio A syndrome, Mol Genet Metab 119:131–143, 2016.
  559. Hughes D, Giogliani R, Guffon N, et al: Clinical outcomes in a subpopulation of adults with Morquio A syndrome: results from a long-term extension study of elosulfase alfa, Orphanet J Rare Dis 12:98–106, 2017.
  560. Jameson E, Jones S, Remmington T: Enzyme replacement therapy with laronidase (Aldurozyme) for treating mucopolysaccharidosis type I, Cochrane Database Syst Rev (4):CD009354, 2016.
  561. Jones A, Breen C, Heap F, et al: A phase 1/2 study of intrathecal heparan-N-sulfatase in patients with mucopolysaccharidosis IIIA, Mol Genet Metab 118:198–205, 2016.
  562. Khan SA, Peracha H, Ballhausen D, et al: Epidemiology of mucopolysaccharidoses, Mol Genet Metab 121(3):227–240, 2017.
  563. Kondo H, Maksimova N, Otomo T, et al: Mutations in VPS33A affects metabolism of glycosaminoglycans: a new type of mucopolysaccharidosis with severe systemic symptoms, Hum Mol Genet 26:173–186, 2017.
  564. Kosuga M, Mashima R, Hirkiyama A, et al: Molecular diagnosis of 6 families with mucopolysaccharidosis type II (Hunter syndrome) characterized by 16 novel mutations in the IDS gene: genetic, pathological, and structural studies on iduronate-2-sulfatase, Mol Genet Metab 118:190–197, 2016.
  565. Kubaski F, Suzuki Y, Orii K, et al: Glycosaminoglycan levels in dried blood spots of patients with mucopolysaccharidoses and mucolipidoses, Mol Genet Metab120:247–254, 2017.
  566. Lin HY, Chuang CK, Lee L, et al: Mucopolysaccharidosis III in Taiwan: Natural history, clinical and molecular characteristic of 28 patients diagnosed during a 21-year period, Am J Med Genet 176:1799–1809, 2018.
  567. Lonardo F, DiNatale P, Lualdi S: Mucopolysaccharidosis type II in a female patient with a reciprocal X;0 translocation and skewed X chromosome inactivation, Am J Med Genet 164:2627–2632, 2014.
  568. Lum SH, Stepien M, Ghos A, et al: Long term survival and cardiopulmonary outcome in childen with Hurler syndrome after haematopoietic stem cell transplantation, J Inherit Metab Dis 40:455–460, 2017.
  569. Montano AM, Lock-Hock N, Steiner RD, et al: Clinical course of Sly syndrome (mucopolysaccharidosis type VII), J Med Genet 53:403–418, 2016.
  570. Natowicz MR, Short MP, Wang Y, et al: Clinical and biochemical manifestations of hyaluronidase deficiency, N Engl J Med 335:1029–1033, 1996.
  571. Micheli V, Camici M, Tozzi MG, et al: Neurological disorders of purine and pyrimidine metabolism, Curr Top Med Chem 11:923–947, 2011.
  572. Nyhan WL: Disorders of purine and pyrimidine metabolism, Mol Genet Metab86:25–33, 2005.
  573. Simmonds HA, Duley JA, Fairbanks LD, et al: When to investigate for purine and pyrimidine disorders: introduction and review of clinical and laboratory indications, J Inherit Metab Dis 20:214, 1997.
  574. Bingham C, Ellard S, van’t Hoff WG, et al: Atypical familial juvenile hyperuricemic nephropathy associated with a hepatocyte nuclear factor-1beta gene mutation, Kidney Int 63:1645–1651, 2003.
  575. Cameron JS, Simmonds HA: Hereditary hyperuricemia and renal disease, Semin Nephrol 25:9–18, 2005.
  576. Landau ME, Kenney K, Deuster P, et al: Exertional rhabdomyolysis: a clinical review with a focus on genetic influences, J Clin Neuromuscul Dis 13:122–136, 2012.
  577. Tarnopolsky MA: What can metabolic myopathies teach us about exercise physiology?, Appl Physiol Nutr Metab 31:21–30, 2006.
  578. Van Adel BA, Tarnopolsky MA: Metabolic myopathies: update 2009, J Clin Neuromuscul Dis 10:97–121, 2009.
  579. Agarwal A, Banerjee A, Banerjee UC: Xanthine oxidoreductase: a journey from purine metabolism to cardiovascular excitation-contraction coupling, Crit Rev Biotechnol31:264–280, 2011.
  580. Atwal PS, Scaglia F: Molybdenum cofactor deficiency, Mol Genet Metab 117(1):1–4, 2016.
  581. Bayram E, Topcu Y, Karakaya P, et al: Molybdenum cofactor deficiency: review of 12 cases (MoCD and review), Eur J Paediatr Neurol 17:1–6, 2013.
  582. Hitzert MM, Bos AF, Bergman KA, et al: Favorable outcome in a newborn with molybdenum cofactor type A deficiency treated with cPMP, Pediatrics 130:e1005–e1010, 2012.
  583. Ichida K, Amaya Y, Okamoto K, et al: Mutations associated with functional disorder of xanthine oxidoreductase and hereditary xanthinuria in humans, Int J Mol Sci13:15475–15495, 2012
  584. Bailey CJ: Orotic aciduria and uridine monophosphate synthase: a reappraisal, J Inherit Metab Dis 32(Suppl 1):S227–S233, 2009.
  585. Leffler M, Carrey EA, Zameitat E: Orotate (orotic acid): an essential and versatile molecule, Nucleosides Nucleotides Nucleic Acids 35(10–12):566–577, 2016.
  586. Leffler M, Carrey EA, Zameitat E: Orotic Acid, More Than Just an Intermediate of Pyrimidine de novo Synthesis, J Genet Genomics. 42(5):207–219, 2015.
  587. Ferrara C, Patel P, Becker S, et al: Biomarkers of insulin for the diagnosis of hyperinsulinemic hypoglycemia in infants and children, J Pediatr 168:212–219, 2016.
  588. Filan PM, Inder TE, Cameron FJ, et al: Neonatal hypoglycemia and occipital cerebral injury, J Pediatr 148:552–555, 2006.
  589. Flanagan SE, Vairo F, Johnson MB, et al: A CACNA1D mutation in a patient with persistent hyperinsulinemic hypoglycemia, congenital heart defects and severe hypotonia, Pediatr Diabetes 18(4):320–323, 2017.
  590. Ghosh A, Banerjee I, Morris AAM: Recognition, assessment and management of hypoglycaemia in childhood, BMJ 101:575–580, 2016.
  591. Giurgea I, Ulinski T, Touati G, et al: Factitious hyperinsulinism leading to pancreatectomy: severe forms of Munchausen syndrome by proxy, Pediatrics 116:e145–e148, 2005.
  592. Goode RH, Rettiganti M, Li J, et al: Developmental outcomes of preterm infants with neonatal hypoglycemia, Pediatrics 138(6):e20161424, 2016.
  593. Guemes M, Hussain K: Hyperinsulinemic hypoglycemia, Pediatr Clin North Am62(4):1017–1036, 2015.
  594. Guemes M, Rahman SA, Hussain K: What is a normal blood glucose?, Arch Dis Child 101:569–574, 2016.
  595. Harris DL, Weston PJ, Signal M, et al: Dextrose gel for neonatal hypoglycaemia (the sugar babies study): a randomized, double-blind, placebo-controlled trial, Lancet382:2077–2083, 2013.
  596. Hegarty JE, Harding JE, Gamble GD: Prophylactic oral dextrose gel for newborn babies at risk of neonatal hypoglycaemia: a randomized controlled dose-finding trial (the Pre-hPOD study), PLoS Med 13(10):e1002155, 2016.
  597. Herrera A, Vajravelu ME, Givler S, et al: Prevalence of adverse events in children with congenital hyperinsulinism treated with diazoxide, J Clin Endocrinol Metab103(12):4365–4372, 2018.
  598. Inder T: How low can I go? The impact of hypoglycemia on the immature brain, Pediatrics 122:440–441, 2008.
  599. Kapoor RR, James C, Hussain K: Hyperinsulinism in developmental syndromes, Endocr Dev 14:95–113, 2009.
  600. Lord K, Radcliffe J, Gallagher PR, et al: High risk of diabetes and neurobehavioral deficits in individuals with surgically treated hyperinsulinism, J Clin Endocrinol Metab 100(11):4133–4139, 2015.
  601. Meder U, Bokodi G, Balogh L, et al: Severe hyperinsulinemic hypoglycemia in a neonate: response to sirolimus therapy, Pediatrics 136(5):e1369–e1372, 2015.
  602. Minute M, Patti G, Tornese G, et al: Sirolimus therapy in congenital hyperinsulinism: a successful experience beyond infancy, Pediatrics 136(5):e1373–e1376, 2015.
  603. Neal JM, Han W: Insulin immunoassays in the detection of insulin analogues in factitious hypoglycemia, Endocr Pract 14:1006–1010, 2008.
  604. Palladino AA, Stanley CA: A specialized team approach to diagnosis and medical versus surgical treatment of infants with congenital hyperinsulinism, Semin Pediatr Surg 20:32–37, 2011.
  605. Regellman MO, Sperling MA: Neurodevelopmental consequences of neonatal hypoglycemia. In Stevenson DK, Benitz WE, Sunshine P, et al, editors: Fetal and neonatal brain injury, ed 5, Cambridge UK, 2018, Cambridge University Press, pp 412–422.
  606. Rekate HL, Nadkarni TD, Teaford PA, Wallace D: Brainstem dysfunction in Chiari malformation presenting as profound hypoglycemia: presentation of four cases, review of the literature, and conjecture as to mechanism, Neurosurgery 45(2):386, 1999.
  607. Senniappan S, Alexandrescu S, Tatevian N, et al: Sirolimus therapy in infants with severe hyperinsulinemic hypoglycemia, N Engl J Med 370:1131–1136, 2014.
  608. Senniappan S, Shanti B, James C, et al: Hyperinsulinaemic hypoglycaemia: genetic mechanisms, diagnosis and management, J Inherit Metab Dis 35:589–601, 2012.
  609. Shrivastava A, Kumar A, Thomas JD, et al: Association of acute toxic encephalopathy with litchi consumption in an outbreak in Muzaffarpur, India, 2014: a case control study, Lancet Glob Health 5(4):e458–e466, 2017.
  610. Shrivastava A, Srikantiah P, Kumar A, et al: Outbreaks of unexplained neurologic illness: Muzaffarpur, India, 2013–2014, MMWR Morb Mortal Wkly Rep 64(3):49–53, 2015.
  611. Salerno C, Crif? C: Diagnostic value of urinary orotic acid levels: applicable separation methods, J Chromatogr B Analyt Technol Biomed Life Sci 781(1/2):57–71, 2002.
  612. Alves CN, Silva JR, Roitberg AE: Insights into the mechanism of oxidation of dihydroorotate to orotate catalysed by human class 2 dihydroorotate dehydrogenase: a QM/MM free energy study, Phys Chem Chem Phys 17(27):17790–17796, 2015.
  613. Duley JA, Henman MG, Carpenter KH, et al: Elevated plasma dihydroorotate in Miller syndrome: biochemical, diagnostic and clinical implications, and treatment with uridine, Mol Genet Metab 119(1/2):83–90, 2016.
  614. Fang J, Uchiumi T, Yagi M, et al: Protein instability and functional defects caused by mutations of dihydro-orotate dehydrogenase in Miller syndrome patients, Biosci Rep 32(6):631–639, 2012.
  615. Fang J, Yamaza H, Uchiumi T, et al: Dihydroorotate dehydrogenase depletion hampers mitochondrial function and osteogenic differentiation in osteoblasts, Eur J Oral Sci 124(3):241–245, 2016.
  616. Rainger J, Bengani H, Campbell L, et al: Miller (Genee-Wiedemann) syndrome represents a clinically and biochemically distinct subgroup of postaxial acrofacial dysostosis associated with partial deficiency of DHODH, Hum Mol Genet21(18):3969–3983, 2012.
  617. Del Re M, Restante G, Di Paolo A, et al: Pharmacogenetics and metabolism from science to implementation in clinical practice: the example of dihydropyrimidine dehydrogenase, Curr Pharm Des 2017.
  618. Lunenburg CA, Henricks LM, Guchelaar HJ, et al: Prospective DPYD genotyping to reduce the risk of fluoropyrimidine-induced severe toxicity: ready for prime time?, Eur J Cancer 54:40–48, 2016.
  619. Meulendijks D, Cats A, Beijnen JH, et al: Improving safety of fluoropyrimidine chemotherapy by individualizing treatment based on dihydropyrimidine dehydrogenase activity: ready for clinical practice?, Cancer Treat Rev 50:23–34, 2016.
  620. Meulendijks D, Henricks LM, Sonke GS, et al: Clinical relevance of DPYD variants c.1679T>G, c.1236G>A/HapB3, and c.1601G>A as predictors of severe fluoropyrimidine-associated toxicity: a systematic review and meta-analysis of individual patient data, Lancet Oncol 16(16):1639–1650, 2015.
  621. Teh LK, Hamzah S, Hashim H, et al: Potential of dihydropyrimidine dehydrogenase genotypes in personalizing 5-fluorouracil therapy among colorectal cancer patients, Ther Drug Monit 35(5):624–630, 2013.
  622. Van Staveren MC, Guchelaar HJ, van Kuilenburg AB, et al: Evaluation of predictive tests for screening for dihydropyrimidine dehydrogenase deficiency, Pharmacogenomics J 13(5):389–395, 2013.
  623. Hsu CC, Lu LY, Yang YS: From sequence and structure of sulfotransferases and dihydropyrimidinases to an understanding of their mechanisms of action and function, Expert Opin Drug Metab Toxicol 6:591–601, 2010.
  624. Ito T: Children’s toxicology from bench to bed—Liver injury (1): Drug-induced metabolic disturbance—toxicity of 5-FU for pyrimidine metabolic disorders and pivalic acid for carnitine metabolism, J Toxicol Sci 34(Suppl 2):SP217–SP222, 2009.
  625. Thomas HR, Ezzeldin HH, Guarcello V, et al: Genetic regulation of dihydropyrimidinase and its possible implication in altered uracil catabolism, Pharmacogenet Genomics17(11):973–987, 2007.
  626. Van Kuilenburg AB, Dobritzsch D, Meijer J: Dihydropyrimidinase deficiency: phenotype, genotype and structural consequences in 17 patients, Biochim Biophys Acta 1802(7/8):639–648, 2010
  627. Fidlerova J, Kleiblova P, Kormunda S, et al: Contribution of the ?-ureidopropionase (UPB1) gene alterations to the development of fluoropyrimidine-related toxicity, Pharmacol Rep 64(5):1234–1242, 2012.
  628. Kummer D, Froehlich TK, Joerger M, et al: Dihydropyrimidinase and ?-ureidopropionase gene variation and severe fluoropyrimidine-related toxicity, Pharmacogenomics16(12):1367–1377, 2015.
  629. Nakajima Y, Meijer J, Dobritzsch D, et al: Clinical, biochemical and molecular analysis of 13 Japanese patients with ?-ureidopropionase deficiency demonstrates high prevalence of the c.977G > A (p.R326Q) mutation [corrected], J Inherit Metab Dis 37(5):801–812, 2014.
  630. Thomas HR, Ezzeldin HH, Guarcello V: Genetic regulation of beta-ureidopropionase and its possible implication in altered uracil catabolism, Pharmacogenet Genomics18:25–35, 2008.
  631. Van Kuilenburg AB, Dobritzsch D, Meijer J, et al: ?-Ureidopropionase deficiency: phenotype, genotype and protein structural consequences in 16 patients, Biochim Biophys Acta 1822(7):1096–1108, 2012.
  632. Van Kuilenburg AB, Meinsma R, Beke E, et al: Beta-ureidopropionase deficiency: an inborn error of pyrimidine degradation associated with neurological abnormalities, Hum Mol Genet 13:2793–2801, 2004.
  633. Yaplito-Lee J, Pitt J, Meijer J, et al: Beta-ureidopropionase deficiency presenting with congenital anomalies of the urogenital and colorectal systems, Mol Genet Metab93:190–194, 2008.
  634. Altounian L, Perrin J, Fouyssac F, et al: Pyrimidine 5?-nucleotidase deficiency associated to a polymalformative syndrome, Ann Biol Clin (Paris) 73(5):587, 2015.
  635. Barasa BA, van Oirschot BA, Bianchi P, et al: Proteomics reveals reduced expression of transketolase in pyrimidine 5?-nucleotidase deficient patients, Proteomics Clin Appl 10(8):859–869, 2016.
  636. Chiarelli LR, Fermo E, Zanella A, et al: Hereditary erythrocyte pyrimidine 5?-nucleotidase deficiency: a biochemical, genetic and clinical overview, Hematology 11:67–72, 2006.
  637. Marinaki AM, Escuredo E, Duley JA, et al: Genetic basis of hemolytic anemia caused by pyrimidine 5? nucleotidase deficiency, Blood 97:3327–3332, 2001.
  638. Rees DC, Duley JA, Marinaki AM: Pyrimidine 5? nucleotidase deficiency, Br J Haematol120:375–383, 2003.
  639. Tozzi MG, Pesi R, Allegrini S: On the physiological role of cytosolic 5?-nucleotidase II (cN-II): pathological and therapeutical implications, Curr Med Chem20(34):4285–4291, 2013.
  640. Zanella A, Bianchi P, Fermo E, et al: Hereditary pyrimidine 5?-nucleotidase deficiency: from genetics to clinical manifestations, Br J Haematol 133:113–123, 2006.
  641. Eriksson S, Wang L: Molecular mechanisms of mitochondrial DNA depletion diseases caused by deficiencies in enzymes in purine and pyrimidine metabolism, Nucleosides Nucleotides Nucleic Acids 27:800–808, 2008.
  642. Galmarini CM, Jordheim L, Dumontet C: Role of IMP-selective 5?-nucleotidase (cN-II) in hematological malignancies, Leuk Lymphoma 44:1105–1111, 2003
  643. Grazia TM: Inborn errors in purine metabolism: role of 5?-nucleotidases and their involvement in the etiology of neurological impairments, Nucleosides Nucleotides Nucleic Acids 30:1276–1283, 2011.
  644. Ipata PL, Balestri F: The functional logic of cytosolic 5?-nucleotidases, Curr Med Chem 20(34):4205–4216, 2013.
  645. Magni G, Amici A, Orsomando G: The enzymology of cytosolic pyrimidine 5?-nucleotidases: functional analysis and physiopathological implications, Curr Med Chem 20(34):4304–4316, 2013.
  646. Tozzi MG, Pesi R, Allegrini S: On the physiological role of cytosolic 5?-nucleotidase II (cN-II): pathological and therapeutical implications, Curr Med Chem 20(34):4285–4291, 2013.
  647. Fairbanks LD, Marinaki AM, Carrey EA, et al: Deoxyuridine accumulation in urine in thymidine phosphorylase deficiency (MNGIE), J Inherit Metab Dis 25:603–604, 2003.
  648. Hirano M, Garone C, Quinzii CM: CoQ(10) deficiencies and MNGIE: two treatable mitochondrial disorders, Biochim Biophys Acta 1820:625–631, 2012.
  649. Karyampudi A, Srivastava P, Mandal K, et al: Novel sequence variations in the thymidine phosphorylase gene causing mitochondrial neurogastrointestinal encephalopathy, Clin Dysmorphol 25(4):156–162, 2016.
  650. Mohamed S, Caporali L, De Giorgio R, et al: HPLC-UV analysis of thymidine and deoxyuridine in plasma of patients with thymidine phosphorylase deficiency, J Chromatogr B Analyt Technol Biomed Life Sci 949(950):58–62, 2014.
  651. Nishino I, Spinazzola A, Hirano M: MNGIE: from nuclear DNA to mitochondrial DNA, Neuromuscul Disord 11:7–10, 2001.
  652. Peedikayil MC, Kagevi EI, Abufarhaneh E, et al: Mitochondrial neurogastrointestinal encephalomyopathy treated with stem cell transplantation: a case report and review of literature, Hematol Oncol Stem Cell Ther 8(2):85–90, 2015
  653. Sivadasan A, Muthusamy K, Patil A, et al: Pearls & Oysters. Mitochondrial neurogastrointestinal encephalomyopathy: diagnosis and response to peritoneal dialysis, Neurology 86(14):e147–e150, 2016.
  654. Wong LJ: Mitochondrial syndromes with leukoencephalopathies, Semin Neurol32:55–61, 2012.
  655. Yadak R, Sillevis Smitt P, van Gisbergen MW, et al: Mitochondrial neurogastrointestinal encephalomyopathy caused by thymidine phosphorylase enzyme deficiency: from pathogenesis to emerging therapeutic options, Front Cell Neurosci 11:31, 2017.
  656. Bartesaghi S, Betts-Henderson J, Cain K, et al: Loss of thymidine kinase 2 alters neuronal bioenergetics and leads to neurodegeneration, Hum Mol Genet 19:1669–1677, 2010.
  657. Behin A, Jardel C, Claeys KG, et al: Adult cases of mitochondrial DNA depletion due to TK2 defect: an expanding spectrum, Neurology 78:644–648, 2012.
  658. Cemara Y, Carre?o-Gago L, Mart?n MA, et al: Severe TK2 enzyme activity deficiency in patients with mild forms of myopathy, Neurology 84(22):2286–2288, 2015.
  659. Chanprasert S, Wang J, Weng SW, et al: Molecular and clinical characterization of the myopathic form of mitochondrial DNA depletion syndrome caused by mutations in the thymidine kinase (TK2) gene, Mol Genet Metab 110(1/2):153–161, 2013.
  660. Lopez-Gomez C, Levy RJ, Sanchez-Quintero MJ, et al: Deoxycytidine and deoxythymidine treatment for thymidine kinase 2 deficiency, Ann Neurol 2017.
  661. Mazurova S, Magner M, Kucerova-Vidrova V, et al: Thymidine kinase 2 and alanyl-tRNA synthetase 2 deficiencies cause lethal mitochondrial cardiomyopathy: case reports and review of the literature, Cardiol Young 14:1–9, 2016.
  662. Saada-Reisch A: Deoxyribonucleoside kinases in mitochondrial DNA depletion, Nucleosides Nucleotides Nucleic Acids 23:1205–1215, 2004.
  663. Cleveland RH, Gordon LB, Kleinman ME, et al: A prospective study of radiographic manifestations in Hutchinson-Gilford progeria syndrome, PediatrRadiol 42(9):1089–1098, 2012.
  664. Gordon LB, Brown WT, Collis FS: Hutchinson-Gilford progeria syndrome. In GeneReviews at GeneTests, editor: Medical Genetics Information Resource, 2017. http://www.ncbi.nlm.nih.gov/sites/GeneTests/review/disease/progeria?db=genetests&search_param=contains.
  665. Gordon AS, Gordon LB: The Progeria Research Foundation: its remarkable journey from obscurity to treatment, Expert Opin Orphan Drugs 2(11):1–9, 2014.
  666. Gordon CM, Gordon LB, Snyder BD, et al: Hutchinson-Gilford progeria is a skeletal dysplasia, J Bone Miner Res 26(7):1670–1679, 2011.
  667. Gordon LB, Kleinman ME, Massaro J, et al: Clinical trial of the protein farnesylation inhibitors lonafarnib, pravastatin, and zoledronic acid in children with Hutchinson Gilford progeria syndrome, Circulation 134(2):114–125, 2016.
  668. Gordon LB, Kleinman ME, Miller DT, et al: Clinical trial of a farnesyltransferase inhibitor in children with Hutchinson-Gilford progeria syndrome, Proc Natl AcadSci USA 109(41):16666–16671, 2012.
  669. Gordon LB, Massaro J, D’Agostino RB, et al: Impact of farnesylation inhibitors on survival in Hutchinson-Gilford progeria syndrome, Circulation 130(1):27–34, 2014.
  670. Gordon LB, McCarten KM, Giobbie-Hurder A, et al: Disease progression in Hutchinson-Gilford progeria syndrome: impact on growth and development, Pediatrics 120(4):824–833, 2007
  671. Guardiani E, Zalewski C, Brewer C, et al: Otologic and audiologic manifestations of Hutchinson-Gilford progeria syndrome, Laryngoscope 121(10):2250–2255, 2011.
  672. Jung HJ, Coffinier C, Choe Y, et al: Regulation of prelamin A but not lamin C by miR-9, a brain-specific microRNA, Proc Natl AcadSci USA 109(7):E423–E431, 2012.
  673. Merideth MA, Gordon L, Clauss S, et al: Phenotype and course of Hutchinson-Gilford progeria syndrome, N Engl J Med 358(6):592–604, 2008.
  674. Olive M, Harten I, Mitchell R, et al: Cardiovascular pathology in Hutchinson-Gilford progeria: correlation with the vascular pathology of aging, ArteriosclerThrombVascBiol 30(11):2301–2309, 2010.
  675. Rork JF, Huang JT, Gordon LB, et al: Initial cutaneous manifestations of Hutchinson-Gilford progeria syndrome, PediatrDermatol 31(2):196–202, 2014.
  676. Silvera VM, Gordon LB, Orbach DB, et al: Imaging characteristics of cerebrovascular arteriopathy and stroke in Hutchinson-Gilford progeria syndrome, AJNR Am J Neuroradiol 34(5):1091–1097, 2013.
  677. Sinensky M, Fantle K, Trujillo M, et al: The processing pathway of prelamin A, J Cell Sci 107(Pt 1):61–67, 1994.
  678. Ullrich NJ, Kieran MW, Miller DT, et al: Neurologic features of Hutchinson-Gilford progeria syndrome after lonafarnib treatment, Neurology 81:427–430, 2013.
  679. Bonkovsky HL, Guo JT, Hou W, et al: Porphyrin and heme metabolism and the porphyrias, Compr Physiol 3(1):365–401, 2013.
  680. Phillips JD, Anderson KE: The porphyrias. In Kaushansky K, Lichtman MA, Beutler E, et al, editors: Williams hematology, ed 9, New York, 2016, McGraw-Hill, pp 889–914.
  681. Acute Porphyrias
  682. Anderson KE, Bloomer JR, Bonkovsky HL, et al: Recommendations for the diagnosis and treatment of the acute porphyrias, Ann Intern Med 142(6):439–450, 2005.
  683. Besur S, Schmeltzer P, Bonkovsky HL: Acute porphyrias, J Emerg Med 49(3):305–312, 2015.
  684. Bissell DM, Anderson KE, Bonkovsky HL: Porphyria, N Engl J Med 377(9):862–872, 2017.
  685. Horner ME, Alikhan A, Tintle S, et al: Cutaneous porphyrias. Part 1. Epidemiology, pathogenesis, presentation, diagnosis, and histopathology, Int J Dermatol 52:1464–1480, 2013.
  686. Singal AK, Parker C, Bowden C, et al: Liver transplantation in the management of porphyria, Hepatology 60(3):1082–1089, 2014.
  687. Stein P, Badminton M, Barth J, et al: Best practice guidelines on clinical management of acute attacks of porphyria and their complications, Ann Clin Biochem 50(Pt 3):217–223, 2013.
  688. Congenital Erythropoietic Porphyria
  689. Katugampola RP, Anstey AV, Finlay AY, et al: A management algorithm for congenital erythropoietic porphyria derived from a study of 29 cases, Br J Dermatol 167(4):888–900, 2012.
  690. Porphyria Cutanea Tarda
  691. Singal AK, Kormos-Hallberg C, Lee C, et al: Low-dose hydroxychloroquine is as effective as phlebotomy in treatment of patients with porphyria cutanea tarda, Clin Gastroenterol Hepatol 10(12):1402–1409, 2012.
  692. Thawani RJ, Moghe A, Idhate T, et al: Porphyria cutanea tarda in a child with acute lymphoblastic leukemia, Q J Med 109(3):191–192, 2016.
  693. Erythropoietic Protoporphyria and X-Linked Protoporphyria
  694. Balwani M, Doheny D, Bishop DF, et al: Loss-of-function ferrochelatase and gainof-function erythroid-specific 5-aminolevulinate synthase mutations causing erythropoietic protoporphyria and X-linked protoporphyria in North American patients reveal novel mutations and a high prevalence of X-linked protoporphyria, Mol Med 19:26–35, 2013.
  695. Gou E, Balwani M, Bissell DM, et al: Pitfalls in erythrocyte protoporphyrin measurement for diagnosis and monitoring of protoporphyrias, Clin Chem 61(12):1453–1456, 2015.
  696. Langendonk JG, Balwani M, Anderson KE, et al: Afamelanotide for erythropoietic protoporphyria, N Engl J Med 373(1):48–59, 2015.
  697. Minder EI, Schneider-Yin X, Steurer J, et al: A systematic review of treatment options for dermal photosensitivity in erythropoietic protoporphyria, Cell Mol Biol (NoisyLe-Grand) 55:84–97, 2009.
  698. Whatley SD, Mason NG, Holme SA, et al: Molecular epidemiology of erythropoietic protoporphyria in the UK, Br J Dermatol 162(3):642–646, 2010.
  699. Adamkin DH: Clinical report: postnatal glucose homeostasis in late-preterm and term infants, Pediatrics 127(3):575–579, 2011.
  700. Adamkin DH, Polin RA: Imperfect advice: neonatal hypoglycemia, J Pediatr 176:195–196, 2016.
  701. Alkalay AL, Flores-Sarnat L, Sarnat HB, et al: Plasma glucose concentrations in profound neonatal hypoglycemia, Clin Pediatr (Phila) 45:550–558, 2006.
  702. Alves C, Constanca J, Leon DD, et al: A novel atypical presentation of insulin autoimmune syndrome (Hirata’s disease) in a child, J Pediatr Endocrinol Metab11:1–4, 2013.
  703. Bateman BT, Patorno E, Desai RJ, et al: Late pregnancy ? blocker exposure and risks of neonatal hypoglycemia and bradycardia, Pediatrics 138(3):e20160731, 2016.
  704. Beltrand J, Caquard M, Arnoux JB, et al: Glucose metabolism in 105 children and adolescents after pancreatectomy for congenital hyperinsulinism, Diabetes Care35:198–203, 2012.
  705. Boluyt N, van Kempen A, Offringa M: Neurodevelopment after neonatal hypoglycemia: a systematic review and design of an optimal future study, Pediatrics 117:2231–2243, 2006.
  706. Brown LM, Corrado MM, van der Ende RM, et al: Evaluation of glycogen storage disease as a cause of ketotic hypoglycemia in children, J Inherit Metab Dis 38(3):489–493, 2015.
  707. Calabria AC, Galagher PR, Simmons R, et al: Postoperative surveillance and detection of postprandial hypoglycemia after fundoplasty in children, J Pediatr 159:597–601, 2011.
  708. Cryer PE: Mechanisms of hypoglycemia-associated autonomic failure in diabetes, N Engl J Med 369:362–370, 2013.
  709. De Leon DD, Stanley CA: Congenital hypoglycemia disorders: new aspects of etiology, diagnosis, treatments and outcomes, Pediatr Diabetes 18(1):3–9, 2017.
  710. Dekelbab BH, Sperling MA: Hypoglycemia in newborns and infants, Adv Pediatr53:5–22, 2006.
  711. Sperling MA: New insights and new conundrums in neonatal hypoglycemia: enigmas wrapped in mystery, Diabetes 62:1373–1375, 2013.
  712. Stanley CA, Rozance PJ, Thornton PS, et al: Re-evaluating “transitional hypoglycemia”: mechanism and implication for management, J Pediatr 166(6):1520–1525, 2015.
  713. Tam EWY, Haeusslein LA, Bonifacio SL, et al: Hypoglycemia is associated with increased risk for brain injury and adverse neurodevelopmental outcome in neonates at risk for encephalopathy, J Pediatr 161:88–93, 2012.
  714. Thornton PS, Stanley CA, De Leon DD, et al: Recommendations from the Pediatric Endocrine Society for evaluation and management of persistent hypoglycemia in neonates, infants and children, J Pediatr 167(2):238–245, 2015.
  715. Treglia G, Mirk P, Giordano A, et al: Diagnostic performance of fluorine18dihydroxyphenylalanine positron emission tomography in diagnosing and localizing the focal form of congenital hyperinsulinism: a meta-analysis, PediatrRadiol 42:1372–1379, 2012.
  716. Tsilianidis LA, Fiske LM, Siegel S, et al: Aggressive therapy improves cirrhosis in glycogen storage disease type IX, Mol Genet Metab 109(2):179–182, 2013.
  717. Weston PJ, Harris DL, Battin M, et al: Oral dextrose gel for the treatment of hypoglycaemia in newborn infants (review), Cochrane Database Syst Rev (5):CD011027, 2016.
  718. Wilcken B: Fatty acid oxidation disorders: outcome and long-term prognosis, J Inherit Metab Dis 33:501–506, 2010.

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 "òðàâìàòîëîãèÿ"

Ðåäàêòîð: Èñêàíäåð Ìèëåâñêè. Äàòà ïóáëèêàöèè: 25.1.2024

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.