Skip to main content
Erschienen in: Endocrine 1/2020

11.04.2020 | Original Article

Clinical features and partial proportional molecular genetics in neonatal diabetes mellitus: a retrospective analysis in southwestern China

verfasst von: Luying Cao, Yi He, Qinrong Huang, Yu Zhang, Pinglan Deng, Weixia Du, Ziyu Hua, Min Zhu, Hong Wei

Erschienen in: Endocrine | Ausgabe 1/2020

Einloggen, um Zugang zu erhalten

Abstract

Purpose

To explore the relationship of phenotype and genotype of neonatal diabetes mellitus (NDM) in southwestern China.

Methods

Sixteen cases of NDM admitted to Children’s Hospital of Chongqing Medical University from May 2009 to May 2019 were included in this study. The clinical features of the included infants were retrospectively analyzed. Peripheral blood samples of the patients and their parents were collected for mutation detection.

Results

Among the 16 cases of NDM, 8 cases were permanent neonatal diabetes mellitus (PNDM) (including 3 clinical syndromes), and 3 cases were transient neonatal diabetes mellitus (TNDM). Mutation detection was performed in six cases. The mutation genes and their loci were FOXP3 p.V408M, KCNJ11 p.C166Y, ABCC8 p.S830P, KCNJ11 p.I182T, KCNJ11 p.G334D, and ZFP57 p.R125X,412. ABCC8 p.S830P was the new found pathogenic site of gene mutation. According to the clinical features and follow-up results, one case was diagnosed as IPEX syndrome, two as DEND syndrome, two as simple PNDM, and one as TNDM. All the TNDM could spontaneously alleviate and then insulin was withdrawn. In PNDM, 75% of those with KATP channel gene mutation could be completely or partially converted to oral sulfonylureas treatment, however, the rest cases needed lifelong insulin replacement therapy.

Conclusion

The clinical manifestations and treatment regimens of patients with NDM vary according to the type of gene mutation. Even the same mutant genotype has differences in phenotype and response to treatment.
Literatur
1.
Zurück zum Zitat K. Nagashima, D. Tanaka, N. Inagaki, Epidemiology, clinical characteristics, and genetic etiology of neonatal diabetes in Japan. Pediatr. Int. 59(2), 129–133 (2017)CrossRef K. Nagashima, D. Tanaka, N. Inagaki, Epidemiology, clinical characteristics, and genetic etiology of neonatal diabetes in Japan. Pediatr. Int. 59(2), 129–133 (2017)CrossRef
2.
Zurück zum Zitat E. Globa, N. Zelinska, D.J. Mackay, K.I. Temple, J.A. Houghton, A.T. Hattersley, S.E. Flanagan, S. Ellard, Neonatal diabetes in Ukraine: incidence, genetics, clinical phenotype and treatment. J. Pediatr. Endocrinol. Metab. 28(11–12), 1279–1286 (2015)PubMedPubMedCentral E. Globa, N. Zelinska, D.J. Mackay, K.I. Temple, J.A. Houghton, A.T. Hattersley, S.E. Flanagan, S. Ellard, Neonatal diabetes in Ukraine: incidence, genetics, clinical phenotype and treatment. J. Pediatr. Endocrinol. Metab. 28(11–12), 1279–1286 (2015)PubMedPubMedCentral
3.
Zurück zum Zitat D. Iafusco, O. Massa, B. Pasquino, C. Colombo, L. Iughetti, C. Bizzarri, C. Mammi, D. Lo Presti, T. Suprani, R. Schiaffini, C.G. Nichols, L. Russo, V. Grasso, F. Meschi, R. Bonfanti, S. Brescianini, F. Barbetti, Minimal incidence of neonatal/infancy onset diabetes in Italy is 1:90,000 live births. Acta Diabetol. 49(5), 405–408 (2012)CrossRef D. Iafusco, O. Massa, B. Pasquino, C. Colombo, L. Iughetti, C. Bizzarri, C. Mammi, D. Lo Presti, T. Suprani, R. Schiaffini, C.G. Nichols, L. Russo, V. Grasso, F. Meschi, R. Bonfanti, S. Brescianini, F. Barbetti, Minimal incidence of neonatal/infancy onset diabetes in Italy is 1:90,000 live births. Acta Diabetol. 49(5), 405–408 (2012)CrossRef
4.
Zurück zum Zitat A.L. Gloyn, E.R. Pearson, J.F. Antcliff, P. Proks, G.J. Bruining, A.S. Slingerland, N. Howard, S. Srinivasan, J.M. Silva, J. Molnes, E.L. Edghill, T.M. Frayling, I.K. Temple, D. Mackay, J.P. Shield, Z. Sumnik, A. van Rhijn, J.K. Wales, P. Clark, S. Gorman, J. Aisenberg, S. Ellard, P.R. Njolstad, F.M. Ashcroft, A.T. Hattersley, Activating mutations in the gene encoding the ATP-sensitive potassium-channel subunit Kir6.2 and permanent neonatal diabetes. N. Engl. J. Med. 350(18), 1838–1849 (2004)CrossRef A.L. Gloyn, E.R. Pearson, J.F. Antcliff, P. Proks, G.J. Bruining, A.S. Slingerland, N. Howard, S. Srinivasan, J.M. Silva, J. Molnes, E.L. Edghill, T.M. Frayling, I.K. Temple, D. Mackay, J.P. Shield, Z. Sumnik, A. van Rhijn, J.K. Wales, P. Clark, S. Gorman, J. Aisenberg, S. Ellard, P.R. Njolstad, F.M. Ashcroft, A.T. Hattersley, Activating mutations in the gene encoding the ATP-sensitive potassium-channel subunit Kir6.2 and permanent neonatal diabetes. N. Engl. J. Med. 350(18), 1838–1849 (2004)CrossRef
5.
Zurück zum Zitat M.B. Lemelman, L. Letourneau, S.A.W. Greeley, Neonatal diabetes mellitus: an update on diagnosis and management. Clin. Perinatol. 45(1), 41–59 (2018)CrossRef M.B. Lemelman, L. Letourneau, S.A.W. Greeley, Neonatal diabetes mellitus: an update on diagnosis and management. Clin. Perinatol. 45(1), 41–59 (2018)CrossRef
6.
Zurück zum Zitat D. Ma, J.P. Shield, W. Dean, I. Leclerc, C. Knauf, R.R. Burcelin, G.A. Rutter, G. Kelsey, Impaired glucose homeostasis in transgenic mice expressing the human transient neonatal diabetes mellitus locus, TNDM. J. Clin. Invest. 114(3), 339–348 (2004)CrossRef D. Ma, J.P. Shield, W. Dean, I. Leclerc, C. Knauf, R.R. Burcelin, G.A. Rutter, G. Kelsey, Impaired glucose homeostasis in transgenic mice expressing the human transient neonatal diabetes mellitus locus, TNDM. J. Clin. Invest. 114(3), 339–348 (2004)CrossRef
7.
Zurück zum Zitat D. Blum, H. Dorchy, T. Mouraux, E. Vamos, Y. Mardens, A. Kumps, C. De Prez, P. Heimann, B. Fowler, R. Baumgartner, Congenital absence of insulin cells in a neonate with diabetes mellitus and mutase-deficient methylmalonic acidaemia. Diabetologia 36(4), 352–357 (1993)CrossRef D. Blum, H. Dorchy, T. Mouraux, E. Vamos, Y. Mardens, A. Kumps, C. De Prez, P. Heimann, B. Fowler, R. Baumgartner, Congenital absence of insulin cells in a neonate with diabetes mellitus and mutase-deficient methylmalonic acidaemia. Diabetologia 36(4), 352–357 (1993)CrossRef
8.
Zurück zum Zitat A. Touati, J. Errea-Dorronsoro, S. Nouri, Y. Halleb, A. Pereda, N. Mahdhaoui, A. Ghith, A. Saad, G. Perez de Nanclares, D. H’Mida Ben Brahim, Transient neonatal diabetes mellitus and hypomethylation at additional imprinted loci: novel ZFP57 mutation and review on the literature. Acta Diabetol. 56(3), 301–307 (2019)CrossRef A. Touati, J. Errea-Dorronsoro, S. Nouri, Y. Halleb, A. Pereda, N. Mahdhaoui, A. Ghith, A. Saad, G. Perez de Nanclares, D. H’Mida Ben Brahim, Transient neonatal diabetes mellitus and hypomethylation at additional imprinted loci: novel ZFP57 mutation and review on the literature. Acta Diabetol. 56(3), 301–307 (2019)CrossRef
9.
Zurück zum Zitat M. Bak, S.E. Boonen, C. Dahl, J.M. Hahnemann, D.J. Mackay, Z. Tumer, K. Gronskov, I.K. Temple, P. Guldberg, N. Tommerup, Genome-wide DNA methylation analysis of transient neonatal diabetes type 1 patients with mutations in ZFP57. BMC Med. Genet. 17, 29 (2016)CrossRef M. Bak, S.E. Boonen, C. Dahl, J.M. Hahnemann, D.J. Mackay, Z. Tumer, K. Gronskov, I.K. Temple, P. Guldberg, N. Tommerup, Genome-wide DNA methylation analysis of transient neonatal diabetes type 1 patients with mutations in ZFP57. BMC Med. Genet. 17, 29 (2016)CrossRef
10.
Zurück zum Zitat S. Quenneville, G. Verde, A. Corsinotti, A. Kapopoulou, J. Jakobsson, S. Offner, I. Baglivo, P.V. Pedone, G. Grimaldi, A. Riccio, D. Trono, In embryonic stem cells, ZFP57/KAP1 recognize a methylated hexanucleotide to affect chromatin and DNA methylation of imprinting control regions. Mol. Cell. 44(3), 361–372 (2011)CrossRef S. Quenneville, G. Verde, A. Corsinotti, A. Kapopoulou, J. Jakobsson, S. Offner, I. Baglivo, P.V. Pedone, G. Grimaldi, A. Riccio, D. Trono, In embryonic stem cells, ZFP57/KAP1 recognize a methylated hexanucleotide to affect chromatin and DNA methylation of imprinting control regions. Mol. Cell. 44(3), 361–372 (2011)CrossRef
11.
Zurück zum Zitat Y. Liu, H. Toh, H. Sasaki, X. Zhang, X. Cheng, An atomic model of Zfp57 recognition of CpG methylation within a specific DNA sequence. Genes Dev. 26(21), 2374–2379 (2012)CrossRef Y. Liu, H. Toh, H. Sasaki, X. Zhang, X. Cheng, An atomic model of Zfp57 recognition of CpG methylation within a specific DNA sequence. Genes Dev. 26(21), 2374–2379 (2012)CrossRef
12.
Zurück zum Zitat I. Baglivo, S. Esposito, L. De Cesare, A. Sparago, Z. Anvar, V. Riso, M. Cammisa, R. Fattorusso, G. Grimaldi, A. Riccio, P.V. Pedone, Genetic and epigenetic mutations affect the DNA binding capability of human ZFP57 in transient neonatal diabetes type 1. FEBS Lett. 587(10), 1474–1481 (2013)CrossRef I. Baglivo, S. Esposito, L. De Cesare, A. Sparago, Z. Anvar, V. Riso, M. Cammisa, R. Fattorusso, G. Grimaldi, A. Riccio, P.V. Pedone, Genetic and epigenetic mutations affect the DNA binding capability of human ZFP57 in transient neonatal diabetes type 1. FEBS Lett. 587(10), 1474–1481 (2013)CrossRef
13.
Zurück zum Zitat E. Gole, S. Oikonomou, S. Ellard, E. De Franco, K. Karavanaki, A Novel KCNJ11 Mutation Associated with Transient Neonatal Diabetes. J. Clin. Res Pediatr. Endocrinol. 10(2), 175–178 (2018)CrossRef E. Gole, S. Oikonomou, S. Ellard, E. De Franco, K. Karavanaki, A Novel KCNJ11 Mutation Associated with Transient Neonatal Diabetes. J. Clin. Res Pediatr. Endocrinol. 10(2), 175–178 (2018)CrossRef
14.
Zurück zum Zitat B. Piccini, C. Coviello, L. Drovandi, A. Rosangela, F. Monzali, E. Casalini, S. Giglio, S. Toni, C. Dani, Transient neonatal diabetes mellitus in a very preterm infant due to ABCC8 mutation. AJP Rep. 8(1), e39–e42 (2018)CrossRef B. Piccini, C. Coviello, L. Drovandi, A. Rosangela, F. Monzali, E. Casalini, S. Giglio, S. Toni, C. Dani, Transient neonatal diabetes mellitus in a very preterm infant due to ABCC8 mutation. AJP Rep. 8(1), e39–e42 (2018)CrossRef
15.
Zurück zum Zitat F.H. Sansbury, S.E. Flanagan, J.A. Houghton, F.L. Shuixian Shen, A.M. Al-Senani, A.M. Habeb, M. Abdullah, A. Kariminejad, S. Ellard, A.T. Hattersley, SLC2A2 mutations can cause neonatal diabetes, suggesting GLUT2 may have a role in human insulin secretion. Diabetologia 55(9), 2381–2385 (2012)CrossRef F.H. Sansbury, S.E. Flanagan, J.A. Houghton, F.L. Shuixian Shen, A.M. Al-Senani, A.M. Habeb, M. Abdullah, A. Kariminejad, S. Ellard, A.T. Hattersley, SLC2A2 mutations can cause neonatal diabetes, suggesting GLUT2 may have a role in human insulin secretion. Diabetologia 55(9), 2381–2385 (2012)CrossRef
16.
Zurück zum Zitat T. Yorifuji, S. Higuchi, Y. Hosokawa, R. Kawakita, Chromosome 6q24-related diabetes mellitus. Clin. Pediatr. Endocrinol. 27(2), 59–65 (2018)CrossRef T. Yorifuji, S. Higuchi, Y. Hosokawa, R. Kawakita, Chromosome 6q24-related diabetes mellitus. Clin. Pediatr. Endocrinol. 27(2), 59–65 (2018)CrossRef
17.
Zurück zum Zitat L.E. Docherty, S. Kabwama, A. Lehmann, E. Hawke, L. Harrison, S.E. Flanagan, S. Ellard, A.T. Hattersley, J.P. Shield, S. Ennis, D.J. Mackay, I.K. Temple, Clinical presentation of 6q24 transient neonatal diabetes mellitus (6q24 TNDM) and genotype-phenotype correlation in an international cohort of patients. Diabetologia 56(4), 758–762 (2013)CrossRef L.E. Docherty, S. Kabwama, A. Lehmann, E. Hawke, L. Harrison, S.E. Flanagan, S. Ellard, A.T. Hattersley, J.P. Shield, S. Ennis, D.J. Mackay, I.K. Temple, Clinical presentation of 6q24 transient neonatal diabetes mellitus (6q24 TNDM) and genotype-phenotype correlation in an international cohort of patients. Diabetologia 56(4), 758–762 (2013)CrossRef
18.
Zurück zum Zitat L. Garcin, D. Kariyawasam, K. Busiah, A.L. Fauret-Amsellem, F. Le Bourgeois, L. Vaivre-Douret, H. Cave, M. Polak, J. Beltrand, Successful off-label sulfonylurea treatment of neonatal diabetes mellitus due to chromosome 6 abnormalities. Pediatr. Diabetes 19(4), 663–669 (2018)CrossRef L. Garcin, D. Kariyawasam, K. Busiah, A.L. Fauret-Amsellem, F. Le Bourgeois, L. Vaivre-Douret, H. Cave, M. Polak, J. Beltrand, Successful off-label sulfonylurea treatment of neonatal diabetes mellitus due to chromosome 6 abnormalities. Pediatr. Diabetes 19(4), 663–669 (2018)CrossRef
19.
Zurück zum Zitat J.L. Fu, T. Wang, X.H. Xiao, Relapsed 6q24-related transient neonatal diabetes mellitus successfully treated with sulfonylurea. Chin. Med J. 132(7), 846–848 (2019)CrossRef J.L. Fu, T. Wang, X.H. Xiao, Relapsed 6q24-related transient neonatal diabetes mellitus successfully treated with sulfonylurea. Chin. Med J. 132(7), 846–848 (2019)CrossRef
20.
Zurück zum Zitat U. Neumann, C. Buhrer, O. Blankenstein, P. Kuhnen, K. Raile, Primary sulphonylurea therapy in a newborn with transient neonatal diabetes attributable to a paternal uniparental disomy 6q24 (UPD6). Diabetes Obes. Metab. 20(2), 474–475 (2018)CrossRef U. Neumann, C. Buhrer, O. Blankenstein, P. Kuhnen, K. Raile, Primary sulphonylurea therapy in a newborn with transient neonatal diabetes attributable to a paternal uniparental disomy 6q24 (UPD6). Diabetes Obes. Metab. 20(2), 474–475 (2018)CrossRef
21.
Zurück zum Zitat C. Reinauer, C. Bergmann, A. Jonasson, V. Soditt, E. Mayatepek, T. Meissner, S. Kummer, ZFP57-related transient neonatal diabetes responsive to oral sulfonylurea treatment. Klin. Padiatr. 231(4), 225–226 (2019)CrossRef C. Reinauer, C. Bergmann, A. Jonasson, V. Soditt, E. Mayatepek, T. Meissner, S. Kummer, ZFP57-related transient neonatal diabetes responsive to oral sulfonylurea treatment. Klin. Padiatr. 231(4), 225–226 (2019)CrossRef
22.
Zurück zum Zitat K. Shimomura, Y. Maejima, KATP channel mutations and neonatal diabetes. Intern Med. 56(18), 2387–2393 (2017)CrossRef K. Shimomura, Y. Maejima, KATP channel mutations and neonatal diabetes. Intern Med. 56(18), 2387–2393 (2017)CrossRef
23.
Zurück zum Zitat P. Proks, H. de Wet, F.M. Ashcroft, Molecular mechanism of sulphonylurea block of K(ATP) channels carrying mutations that impair ATP inhibition and cause neonatal diabetes. Diabetes 62(11), 3909–3919 (2013)CrossRef P. Proks, H. de Wet, F.M. Ashcroft, Molecular mechanism of sulphonylurea block of K(ATP) channels carrying mutations that impair ATP inhibition and cause neonatal diabetes. Diabetes 62(11), 3909–3919 (2013)CrossRef
24.
Zurück zum Zitat C.W. Lin, Y.W. Lin, F.F. Yan, J. Casey, M. Kochhar, E.B. Pratt, S.L. Shyng, Kir6.2 mutations associated with neonatal diabetes reduce expression of ATP-sensitive K+ channels: implications in disease mechanism and sulfonylurea therapy. Diabetes 55(6), 1738–1746 (2006)CrossRef C.W. Lin, Y.W. Lin, F.F. Yan, J. Casey, M. Kochhar, E.B. Pratt, S.L. Shyng, Kir6.2 mutations associated with neonatal diabetes reduce expression of ATP-sensitive K+ channels: implications in disease mechanism and sulfonylurea therapy. Diabetes 55(6), 1738–1746 (2006)CrossRef
25.
Zurück zum Zitat A.P. Babenko, M. Polak, H. Cave, K. Busiah, P. Czernichow, R. Scharfmann, J. Bryan, L. Aguilar-Bryan, M. Vaxillaire, P. Froguel, Activating mutations in the ABCC8 gene in neonatal diabetes mellitus. N. Engl. J. Med. 355(5), 456–466 (2006)CrossRef A.P. Babenko, M. Polak, H. Cave, K. Busiah, P. Czernichow, R. Scharfmann, J. Bryan, L. Aguilar-Bryan, M. Vaxillaire, P. Froguel, Activating mutations in the ABCC8 gene in neonatal diabetes mellitus. N. Engl. J. Med. 355(5), 456–466 (2006)CrossRef
26.
Zurück zum Zitat X. Xiao, T. Wang, W. Li, H. Song, C. Gong, C. Diao, M. Yu, T. Yuan, Y. Zhang, X. Sun, Q. Zhang, K. Lu, H. Wang, O. Schmitz, T. Hansen, Transfer from insulin to sulfonylurea treatment in a chinese patient with permanent neonatal diabetes mellitus due to a KCNJ11 R201H mutation. Horm. Metab. Res. 41(7), 580–582 (2009)CrossRef X. Xiao, T. Wang, W. Li, H. Song, C. Gong, C. Diao, M. Yu, T. Yuan, Y. Zhang, X. Sun, Q. Zhang, K. Lu, H. Wang, O. Schmitz, T. Hansen, Transfer from insulin to sulfonylurea treatment in a chinese patient with permanent neonatal diabetes mellitus due to a KCNJ11 R201H mutation. Horm. Metab. Res. 41(7), 580–582 (2009)CrossRef
27.
Zurück zum Zitat T. Wang, M. Yu, C. Lu, H. Zhang, F. Ping, Q. Zhang, J. Xu, K. Feng, W. Li, J. Zheng, X. Wang, X. Xiao, Molecular and clinical features of 13 cases of ATP-sensitive potassium channel neonatal diabetes mellitus. Chin. J. Diabetes Mellit. 9(6), 350–355 (2017) T. Wang, M. Yu, C. Lu, H. Zhang, F. Ping, Q. Zhang, J. Xu, K. Feng, W. Li, J. Zheng, X. Wang, X. Xiao, Molecular and clinical features of 13 cases of ATP-sensitive potassium channel neonatal diabetes mellitus. Chin. J. Diabetes Mellit. 9(6), 350–355 (2017)
28.
Zurück zum Zitat X. Li, A. Xu, H. Sheng, T.H. Ting, X. Mao, X. Huang, M. Jiang, J. Cheng, L. Liu, Early transition from insulin to sulfonylureas in neonatal diabetes and follow-up: experience from China. Pediatr. Diabetes 19(2), 251–258 (2018)CrossRef X. Li, A. Xu, H. Sheng, T.H. Ting, X. Mao, X. Huang, M. Jiang, J. Cheng, L. Liu, Early transition from insulin to sulfonylureas in neonatal diabetes and follow-up: experience from China. Pediatr. Diabetes 19(2), 251–258 (2018)CrossRef
29.
Zurück zum Zitat S.E. Flanagan, E.L. Edghill, A.L. Gloyn, S. Ellard, A.T. Hattersley, Mutations in KCNJ11, which encodes Kir6.2, are a common cause of diabetes diagnosed in the first 6 months of life, with the phenotype determined by genotype. Diabetologia 49(6), 1190–1197 (2006)CrossRef S.E. Flanagan, E.L. Edghill, A.L. Gloyn, S. Ellard, A.T. Hattersley, Mutations in KCNJ11, which encodes Kir6.2, are a common cause of diabetes diagnosed in the first 6 months of life, with the phenotype determined by genotype. Diabetologia 49(6), 1190–1197 (2006)CrossRef
30.
Zurück zum Zitat K. Balamurugan, B. Kavitha, Z. Yang, V. Mohan, V. Radha, S.L. Shyng, Functional characterization of activating mutations in the sulfonylurea receptor 1 (ABCC8) causing neonatal diabetes mellitus in Asian Indian children. Pediatr. Diabetes 20(4), 397–407 (2019)CrossRef K. Balamurugan, B. Kavitha, Z. Yang, V. Mohan, V. Radha, S.L. Shyng, Functional characterization of activating mutations in the sulfonylurea receptor 1 (ABCC8) causing neonatal diabetes mellitus in Asian Indian children. Pediatr. Diabetes 20(4), 397–407 (2019)CrossRef
31.
Zurück zum Zitat R. Bacchetta, F. Barzaghi, M.G. Roncarolo, From IPEX syndrome to FOXP3 mutation: a lesson on immune dysregulation. Ann. N. Y. Acad. Sci. 1417(1), 5–22 (2018)CrossRef R. Bacchetta, F. Barzaghi, M.G. Roncarolo, From IPEX syndrome to FOXP3 mutation: a lesson on immune dysregulation. Ann. N. Y. Acad. Sci. 1417(1), 5–22 (2018)CrossRef
32.
Zurück zum Zitat O. Rubio-Cabezas, J.A. Minton, R. Caswell, J.P. Shield, D. Deiss, Z. Sumnik, A. Cayssials, M. Herr, A. Loew, V. Lewis, S. Ellard, A.T. Hattersley, Clinical heterogeneity in patients with FOXP3 mutations presenting with permanent neonatal diabetes. Diabetes Care 32(1), 111–116 (2009)CrossRef O. Rubio-Cabezas, J.A. Minton, R. Caswell, J.P. Shield, D. Deiss, Z. Sumnik, A. Cayssials, M. Herr, A. Loew, V. Lewis, S. Ellard, A.T. Hattersley, Clinical heterogeneity in patients with FOXP3 mutations presenting with permanent neonatal diabetes. Diabetes Care 32(1), 111–116 (2009)CrossRef
33.
Zurück zum Zitat T. Wang, M. Li, M. Yu, C. Lu, H. Zhang, F. Ping, Q. Zhang, C. Qi, J. Zheng, X. Wang, J. Fu, X. Xiao, Genetic and clinical features of 3 cases of neonatal diabetes mellitus caused by forkhead box P3 gene mutation. Chin. J. Diabetes Mellit. 9(10), 596–601 (2018) T. Wang, M. Li, M. Yu, C. Lu, H. Zhang, F. Ping, Q. Zhang, C. Qi, J. Zheng, X. Wang, J. Fu, X. Xiao, Genetic and clinical features of 3 cases of neonatal diabetes mellitus caused by forkhead box P3 gene mutation. Chin. J. Diabetes Mellit. 9(10), 596–601 (2018)
34.
Zurück zum Zitat S. Ma, R. Viola, L. Sui, V. Cherubini, F. Barbetti, D. Egli, Beta cell replacement after gene editing of a neonatal diabetes-causing mutation at the insulin locus. Stem Cell Rep. 11(6), 1407–1415 (2018)CrossRef S. Ma, R. Viola, L. Sui, V. Cherubini, F. Barbetti, D. Egli, Beta cell replacement after gene editing of a neonatal diabetes-causing mutation at the insulin locus. Stem Cell Rep. 11(6), 1407–1415 (2018)CrossRef
Metadaten
Titel
Clinical features and partial proportional molecular genetics in neonatal diabetes mellitus: a retrospective analysis in southwestern China
verfasst von
Luying Cao
Yi He
Qinrong Huang
Yu Zhang
Pinglan Deng
Weixia Du
Ziyu Hua
Min Zhu
Hong Wei
Publikationsdatum
11.04.2020
Verlag
Springer US
Erschienen in
Endocrine / Ausgabe 1/2020
Print ISSN: 1355-008X
Elektronische ISSN: 1559-0100
DOI
https://doi.org/10.1007/s12020-020-02279-4

Weitere Artikel der Ausgabe 1/2020

Endocrine 1/2020 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.