α Cell Function and Gene Expression Are Compromised in Type 1 Diabetes

Marcela Brissova, Rachana Haliyur, Diane Saunders, Shristi Shrestha, Chunhua Dai, David M Blodgett, Rita Bottino, Martha Campbell-Thompson, Radhika Aramandla, Gregory Poffenberger, Jill Lindner, Fong Cheng Pan, Matthias G von Herrath, Dale L Greiner, Leonard D Shultz, May Sanyoura, Louis H Philipson, Mark Atkinson, David M Harlan, Shawn E Levy, Nripesh Prasad, Roland Stein, Alvin C Powers, Marcela Brissova, Rachana Haliyur, Diane Saunders, Shristi Shrestha, Chunhua Dai, David M Blodgett, Rita Bottino, Martha Campbell-Thompson, Radhika Aramandla, Gregory Poffenberger, Jill Lindner, Fong Cheng Pan, Matthias G von Herrath, Dale L Greiner, Leonard D Shultz, May Sanyoura, Louis H Philipson, Mark Atkinson, David M Harlan, Shawn E Levy, Nripesh Prasad, Roland Stein, Alvin C Powers

Abstract

Many patients with type 1 diabetes (T1D) have residual β cells producing small amounts of C-peptide long after disease onset but develop an inadequate glucagon response to hypoglycemia following T1D diagnosis. The features of these residual β cells and α cells in the islet endocrine compartment are largely unknown, due to the difficulty of comprehensive investigation. By studying the T1D pancreas and isolated islets, we show that remnant β cells appeared to maintain several aspects of regulated insulin secretion. However, the function of T1D α cells was markedly reduced, and these cells had alterations in transcription factors constituting α and β cell identity. In the native pancreas and after placing the T1D islets into a non-autoimmune, normoglycemic in vivo environment, there was no evidence of α-to-β cell conversion. These results suggest an explanation for the disordered T1D counterregulatory glucagon response to hypoglycemia.

Keywords: alpha cells; glucagon; human; insulin; pancreatic islet; type 1 diabetes.

Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1. T1D β Cells in Recent-Onset…
Figure 1. T1D β Cells in Recent-Onset T1D Retain Secretory Properties and Gene Expression Pattern Similar to Normal β Cells
(A and B) Insulin secretion was assessed in islets isolated from donors with recent-onset T1D (n = 4; ages 12–22 years; donors nos. 1, 3, 4, and 5) and compared to normal controls (n = 7; ages 7–21 years); G 5.6–5.6 mM glucose, G 16.7–16.7 mM glucose, G 16.7 + IBMX 100–16.7 mM glucose + 100 µM isobutylmethylxanthine (IBMX), G 1.7 + Epi 1–1.7 mM glucose + 1 µM epinephrine, KCl 20–20 mM potassium chloride. (A) Insulin secretion normalized to overall islet cell volume (expressed as islet equivalents [IEQs]); ****p INS expression; ***p < 0.0007. (E–G) Expression of β-cell-enriched transcription factors in the native pancreatic tissue from donors with recent-onset T1D (n = 2; ages 12–22 years; donors nos. 1 and 5) was compared to 58-year-old donor with 31 years of T1D duration (donor no. 8) and controls (n = 7; ages 8–55 years). The pancreas of 58-year-old T1D donor did not have any insulin+ islets; only rare β cells were found in exocrine parenchyma. T1D β cells (n = 3 donors; ages 12–58 years; donors nos. 1, 5, and 8) had normal expression of β-cell-enriched transcription factors PDX1 (E) and NKX6.1 (F) but decreased expression of NKX2.2 (G) compared to controls (n = 7 donors; ages 8–55 years); ****p

Figure 2. T1D α Cells in Recent-Onset…

Figure 2. T1D α Cells in Recent-Onset T1D Have Reduced Glucagon Secretion and Dysregulated Gene…

Figure 2. T1D α Cells in Recent-Onset T1D Have Reduced Glucagon Secretion and Dysregulated Gene Expression
The same sets of islets shown in Figures 1A and 1B were simultaneously analyzed for glucagon secretion. The same non-diabetic controls were used as Figure 1. The labeling of islet stimuli is identical to that in Figure 1. (A) Glucagon secretion normalized to overall islet cell volume (expressed as IEQs); p = 0.2470. (B) Glucagon secretion normalized to islet glucagon content; ****p GCG expression; ****p < 0.0001; *p = 0.0184. (E–G) Analysis of native pancreatic tissue for expression of islet-enriched transcription factors. T1D α cells (n = 4 donors; ages 12–58 years; donors nos. 1, 2, 5, and 8) expressed β cell marker NKX6.1 (G) and lost bona fide α cell markers MAFB (E) and ARX (F) in most T1D α cells compared to controls (n = 7 donors; ages 8–55 years); ****p

Figure 3. T1D α Cells Do Not…

Figure 3. T1D α Cells Do Not Show Evidence of α-to-β Cell Reprogramming in Normoglycemic,…

Figure 3. T1D α Cells Do Not Show Evidence of α-to-β Cell Reprogramming in Normoglycemic, Non-autoimmune Environment
(A) Islets from donors with recent-onset and long-standing T1D (n = 3 donors; 12–58 years; donors nos. 1, 5, and 8) depicted in Figures 1 and 2 were transplanted into NSG mice. After 1-month engraftment, mice were treated with either PBS or Ex-4 for an additional 1 month. Representative images of islet grafts are from the 12-year-old individual with 3-year T1D duration (donor no. 1). In control and T1D columns, regions denoted by the dashed line in images on the left (B)–(E) (scale bar in B is 50 µm) are displayed on the right (scale bar is 10 µm). (B) Insulin (INS) and glucagon (GCG) double-positive cells were not detected in either type of T1D islet grafts (PBS or Ex-4). (C–E) As there were no phenotypic differences between PBS and Ex-4 treatment groups, representative images were taken from both cohorts and analyzed for α cell transcription factor expression. Change in number of GCG+ cells expressing MAFB (C), ARX (D), and NKX6.1 (E) in transplanted T1D islets (TX) relative to donor’s native pancreas (Panc) is shown. ****p

Figure 4. Genes Critical to α Cell…

Figure 4. Genes Critical to α Cell Function Are Differentially Expressed in T1D α Cells

Figure 4. Genes Critical to α Cell Function Are Differentially Expressed in T1D α Cells
Transcriptome by RNA-sequencing analysis of purified human α cells from T1D donors (n = 3; ages 14–30 years; donors nos. 3, 6, and 7) and controls (n = 5; ages 26–55 years). (A) Principal-component analysis (PCA) plot shows clustering of α cell samples from control and T1D donors. (B) Heatmap of the pairwise correlation between all samples based on the Spearman correlation coefficient. Perfect correlation is indicated by 1. (C) Genes associated with α cell identity and function are significantly downregulated in the T1D α cells with increased expression of stress response factors and cell-cell contact proteins. Vertical dotted lines represent point of significance for fold change (FC) = 1.53 threshold analysis; p
Similar articles
Cited by
References
    1. Alkorta-Aranburu G, Sukhanova M, Carmody D, Hoffman T, Wysinger L, Keller-Ramey J, Li Z, Johnson AK, Kobiernicki F, Botes S, et al. Improved molecular diagnosis of patients with neonatal diabetes using a combined next-generation sequencing and MS-MLPA approach. J. Pediatr. Endocrinol. Metab. 2016;29:523–531. - PubMed
    1. Balamurugan AN, Chang Y, Fung JJ, Trucco M, Bottino R. Flexible management of enzymatic digestion improves human islet isolation outcome from sub-optimal donor pancreata. Am. J. Transplant. 2003;3:1135–1142. - PubMed
    1. Blodgett DM, Nowosielska A, Afik S, Pechhold S, Cura AJ, Kennedy NJ, Kim S, Kucukural A, Davis RJ, Kent SC, et al. Novel observations from next-generation RNA sequencing of highly purified human adult and fetal islet cell subsets. Diabetes. 2015;64:3172–3181. - PMC - PubMed
    1. Bolli G, de Feo P, Compagnucci P, Cartechini MG, Angeletti G, Santeusanio F, Brunetti P, Gerich JE. Abnormal glucose counterregulation in insulin-dependent diabetes mellitus. Interaction of anti-insulin antibodies and impaired glucagon and epinephrine secretion. Diabetes. 1983;32:134–141. - PubMed
    1. Brissova M, Aamodt K, Brahmachary P, Prasad N, Hong J-Y, Dai C, Mellati M, Shostak A, Poffenberger G, Aramandla R, et al. Islet microenvironment, modulated by vascular endothelial growth factor-A signaling, promotes β cell regeneration. Cell Metab. 2014;19:498–511. - PMC - PubMed
Show all 34 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 2. T1D α Cells in Recent-Onset…
Figure 2. T1D α Cells in Recent-Onset T1D Have Reduced Glucagon Secretion and Dysregulated Gene Expression
The same sets of islets shown in Figures 1A and 1B were simultaneously analyzed for glucagon secretion. The same non-diabetic controls were used as Figure 1. The labeling of islet stimuli is identical to that in Figure 1. (A) Glucagon secretion normalized to overall islet cell volume (expressed as IEQs); p = 0.2470. (B) Glucagon secretion normalized to islet glucagon content; ****p GCG expression; ****p < 0.0001; *p = 0.0184. (E–G) Analysis of native pancreatic tissue for expression of islet-enriched transcription factors. T1D α cells (n = 4 donors; ages 12–58 years; donors nos. 1, 2, 5, and 8) expressed β cell marker NKX6.1 (G) and lost bona fide α cell markers MAFB (E) and ARX (F) in most T1D α cells compared to controls (n = 7 donors; ages 8–55 years); ****p

Figure 3. T1D α Cells Do Not…

Figure 3. T1D α Cells Do Not Show Evidence of α-to-β Cell Reprogramming in Normoglycemic,…

Figure 3. T1D α Cells Do Not Show Evidence of α-to-β Cell Reprogramming in Normoglycemic, Non-autoimmune Environment
(A) Islets from donors with recent-onset and long-standing T1D (n = 3 donors; 12–58 years; donors nos. 1, 5, and 8) depicted in Figures 1 and 2 were transplanted into NSG mice. After 1-month engraftment, mice were treated with either PBS or Ex-4 for an additional 1 month. Representative images of islet grafts are from the 12-year-old individual with 3-year T1D duration (donor no. 1). In control and T1D columns, regions denoted by the dashed line in images on the left (B)–(E) (scale bar in B is 50 µm) are displayed on the right (scale bar is 10 µm). (B) Insulin (INS) and glucagon (GCG) double-positive cells were not detected in either type of T1D islet grafts (PBS or Ex-4). (C–E) As there were no phenotypic differences between PBS and Ex-4 treatment groups, representative images were taken from both cohorts and analyzed for α cell transcription factor expression. Change in number of GCG+ cells expressing MAFB (C), ARX (D), and NKX6.1 (E) in transplanted T1D islets (TX) relative to donor’s native pancreas (Panc) is shown. ****p

Figure 4. Genes Critical to α Cell…

Figure 4. Genes Critical to α Cell Function Are Differentially Expressed in T1D α Cells

Figure 4. Genes Critical to α Cell Function Are Differentially Expressed in T1D α Cells
Transcriptome by RNA-sequencing analysis of purified human α cells from T1D donors (n = 3; ages 14–30 years; donors nos. 3, 6, and 7) and controls (n = 5; ages 26–55 years). (A) Principal-component analysis (PCA) plot shows clustering of α cell samples from control and T1D donors. (B) Heatmap of the pairwise correlation between all samples based on the Spearman correlation coefficient. Perfect correlation is indicated by 1. (C) Genes associated with α cell identity and function are significantly downregulated in the T1D α cells with increased expression of stress response factors and cell-cell contact proteins. Vertical dotted lines represent point of significance for fold change (FC) = 1.53 threshold analysis; p
Similar articles
Cited by
References
    1. Alkorta-Aranburu G, Sukhanova M, Carmody D, Hoffman T, Wysinger L, Keller-Ramey J, Li Z, Johnson AK, Kobiernicki F, Botes S, et al. Improved molecular diagnosis of patients with neonatal diabetes using a combined next-generation sequencing and MS-MLPA approach. J. Pediatr. Endocrinol. Metab. 2016;29:523–531. - PubMed
    1. Balamurugan AN, Chang Y, Fung JJ, Trucco M, Bottino R. Flexible management of enzymatic digestion improves human islet isolation outcome from sub-optimal donor pancreata. Am. J. Transplant. 2003;3:1135–1142. - PubMed
    1. Blodgett DM, Nowosielska A, Afik S, Pechhold S, Cura AJ, Kennedy NJ, Kim S, Kucukural A, Davis RJ, Kent SC, et al. Novel observations from next-generation RNA sequencing of highly purified human adult and fetal islet cell subsets. Diabetes. 2015;64:3172–3181. - PMC - PubMed
    1. Bolli G, de Feo P, Compagnucci P, Cartechini MG, Angeletti G, Santeusanio F, Brunetti P, Gerich JE. Abnormal glucose counterregulation in insulin-dependent diabetes mellitus. Interaction of anti-insulin antibodies and impaired glucagon and epinephrine secretion. Diabetes. 1983;32:134–141. - PubMed
    1. Brissova M, Aamodt K, Brahmachary P, Prasad N, Hong J-Y, Dai C, Mellati M, Shostak A, Poffenberger G, Aramandla R, et al. Islet microenvironment, modulated by vascular endothelial growth factor-A signaling, promotes β cell regeneration. Cell Metab. 2014;19:498–511. - PMC - PubMed
Show all 34 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 3. T1D α Cells Do Not…
Figure 3. T1D α Cells Do Not Show Evidence of α-to-β Cell Reprogramming in Normoglycemic, Non-autoimmune Environment
(A) Islets from donors with recent-onset and long-standing T1D (n = 3 donors; 12–58 years; donors nos. 1, 5, and 8) depicted in Figures 1 and 2 were transplanted into NSG mice. After 1-month engraftment, mice were treated with either PBS or Ex-4 for an additional 1 month. Representative images of islet grafts are from the 12-year-old individual with 3-year T1D duration (donor no. 1). In control and T1D columns, regions denoted by the dashed line in images on the left (B)–(E) (scale bar in B is 50 µm) are displayed on the right (scale bar is 10 µm). (B) Insulin (INS) and glucagon (GCG) double-positive cells were not detected in either type of T1D islet grafts (PBS or Ex-4). (C–E) As there were no phenotypic differences between PBS and Ex-4 treatment groups, representative images were taken from both cohorts and analyzed for α cell transcription factor expression. Change in number of GCG+ cells expressing MAFB (C), ARX (D), and NKX6.1 (E) in transplanted T1D islets (TX) relative to donor’s native pancreas (Panc) is shown. ****p

Figure 4. Genes Critical to α Cell…

Figure 4. Genes Critical to α Cell Function Are Differentially Expressed in T1D α Cells

Figure 4. Genes Critical to α Cell Function Are Differentially Expressed in T1D α Cells
Transcriptome by RNA-sequencing analysis of purified human α cells from T1D donors (n = 3; ages 14–30 years; donors nos. 3, 6, and 7) and controls (n = 5; ages 26–55 years). (A) Principal-component analysis (PCA) plot shows clustering of α cell samples from control and T1D donors. (B) Heatmap of the pairwise correlation between all samples based on the Spearman correlation coefficient. Perfect correlation is indicated by 1. (C) Genes associated with α cell identity and function are significantly downregulated in the T1D α cells with increased expression of stress response factors and cell-cell contact proteins. Vertical dotted lines represent point of significance for fold change (FC) = 1.53 threshold analysis; p
Similar articles
Cited by
References
    1. Alkorta-Aranburu G, Sukhanova M, Carmody D, Hoffman T, Wysinger L, Keller-Ramey J, Li Z, Johnson AK, Kobiernicki F, Botes S, et al. Improved molecular diagnosis of patients with neonatal diabetes using a combined next-generation sequencing and MS-MLPA approach. J. Pediatr. Endocrinol. Metab. 2016;29:523–531. - PubMed
    1. Balamurugan AN, Chang Y, Fung JJ, Trucco M, Bottino R. Flexible management of enzymatic digestion improves human islet isolation outcome from sub-optimal donor pancreata. Am. J. Transplant. 2003;3:1135–1142. - PubMed
    1. Blodgett DM, Nowosielska A, Afik S, Pechhold S, Cura AJ, Kennedy NJ, Kim S, Kucukural A, Davis RJ, Kent SC, et al. Novel observations from next-generation RNA sequencing of highly purified human adult and fetal islet cell subsets. Diabetes. 2015;64:3172–3181. - PMC - PubMed
    1. Bolli G, de Feo P, Compagnucci P, Cartechini MG, Angeletti G, Santeusanio F, Brunetti P, Gerich JE. Abnormal glucose counterregulation in insulin-dependent diabetes mellitus. Interaction of anti-insulin antibodies and impaired glucagon and epinephrine secretion. Diabetes. 1983;32:134–141. - PubMed
    1. Brissova M, Aamodt K, Brahmachary P, Prasad N, Hong J-Y, Dai C, Mellati M, Shostak A, Poffenberger G, Aramandla R, et al. Islet microenvironment, modulated by vascular endothelial growth factor-A signaling, promotes β cell regeneration. Cell Metab. 2014;19:498–511. - PMC - PubMed
Show all 34 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 4. Genes Critical to α Cell…
Figure 4. Genes Critical to α Cell Function Are Differentially Expressed in T1D α Cells
Transcriptome by RNA-sequencing analysis of purified human α cells from T1D donors (n = 3; ages 14–30 years; donors nos. 3, 6, and 7) and controls (n = 5; ages 26–55 years). (A) Principal-component analysis (PCA) plot shows clustering of α cell samples from control and T1D donors. (B) Heatmap of the pairwise correlation between all samples based on the Spearman correlation coefficient. Perfect correlation is indicated by 1. (C) Genes associated with α cell identity and function are significantly downregulated in the T1D α cells with increased expression of stress response factors and cell-cell contact proteins. Vertical dotted lines represent point of significance for fold change (FC) = 1.53 threshold analysis; p

References

    1. Alkorta-Aranburu G, Sukhanova M, Carmody D, Hoffman T, Wysinger L, Keller-Ramey J, Li Z, Johnson AK, Kobiernicki F, Botes S, et al. Improved molecular diagnosis of patients with neonatal diabetes using a combined next-generation sequencing and MS-MLPA approach. J. Pediatr. Endocrinol. Metab. 2016;29:523–531.
    1. Balamurugan AN, Chang Y, Fung JJ, Trucco M, Bottino R. Flexible management of enzymatic digestion improves human islet isolation outcome from sub-optimal donor pancreata. Am. J. Transplant. 2003;3:1135–1142.
    1. Blodgett DM, Nowosielska A, Afik S, Pechhold S, Cura AJ, Kennedy NJ, Kim S, Kucukural A, Davis RJ, Kent SC, et al. Novel observations from next-generation RNA sequencing of highly purified human adult and fetal islet cell subsets. Diabetes. 2015;64:3172–3181.
    1. Bolli G, de Feo P, Compagnucci P, Cartechini MG, Angeletti G, Santeusanio F, Brunetti P, Gerich JE. Abnormal glucose counterregulation in insulin-dependent diabetes mellitus. Interaction of anti-insulin antibodies and impaired glucagon and epinephrine secretion. Diabetes. 1983;32:134–141.
    1. Brissova M, Aamodt K, Brahmachary P, Prasad N, Hong J-Y, Dai C, Mellati M, Shostak A, Poffenberger G, Aramandla R, et al. Islet microenvironment, modulated by vascular endothelial growth factor-A signaling, promotes β cell regeneration. Cell Metab. 2014;19:498–511.
    1. Cano DA, Rulifson IC, Heiser PW, Swigart LB, Pelengaris S, German M, Evan GI, Bluestone JA, Hebrok M. Regulated beta-cell regeneration in the adult mouse pancreas. Diabetes. 2008;57:958–966.
    1. Chakravarthy H, Gu X, Enge M, Dai X, Wang Y, Damond N, Downie C, Liu K, Wang J, Xing Y, et al. Converting adult pancreatic islet α cells into β cells by targeting both Dnmt1 and Arx. Cell Metab. 2017;25:622–634.
    1. Chandra V, Albagli-Curiel O, Hastoy B, Piccand J, Randriamampita C, Vaillant E, Cavé H, Busiah K, Froguel P, Vaxillaire M, et al. RFX6 regulates insulin secretion by modulating Ca2+ homeostasis in human β cells. Cell Rep. 2014;9:2206–2218.
    1. Chera S, Baronnier D, Ghila L, Cigliola V, Jensen JN, Gu G, Furuyama K, Thorel F, Gribble FM, Reimann F, Herrera PL. Diabetes recovery by age-dependent conversion of pancreatic δ-cells into insulin producers. Nature. 2014;514:503–507.
    1. Courtney M, Gjernes E, Druelle N, Ravaud C, Vieira A, Ben-Othman N, Pfeifer A, Avolio F, Leuckx G, Lacas-Gervais S, et al. The inactivation of Arx in pancreatic α-cells triggers their neogenesis and conversion into functional β-like cells. PLoS Genet. 2013;9:e1003934.
    1. Dai C, Kayton NS, Shostak A, Poffenberger G, Cyphert HA, Aramandla R, Thompson C, Papagiannis IG, Emfinger C, Shiota M, et al. Stress-impaired transcription factor expression and insulin secretion in transplanted human islets. J. Clin. Invest. 2016;126:1857–1870.
    1. Dai C, Hang Y, Shostak A, Poffenberger G, Hart N, Prasad N, Phillips N, Levy SE, Greiner DL, Shultz LD, et al. Age-dependent human β cell proliferation induced by glucagon-like peptide 1 and calcineurin signaling. J. Clin. Invest. 2017;127:3835–3844.
    1. Dorrell C, Schug J, Canaday PS, Russ HA, Tarlow BD, Grompe MT, Horton T, Hebrok M, Streeter PR, Kaestner KH, Grompe M. Human islets contain four distinct subtypes of β cells. Nat. Commun. 2016;7:11756.
    1. Gao T, McKenna B, Li C, Reichert M, Nguyen J, Singh T, Yang C, Pannikar A, Doliba N, Zhang T, et al. Pdx1 maintains β cell identity and function by repressing an α cell program. Cell Metab. 2014;19:259–271.
    1. Gerich JE, Langlois M, Noacco C, Karam JH, Forsham PH. Lack of glucagon response to hypoglycemia in diabetes: evidence for an intrinsic pancreatic alpha cell defect. Science. 1973;182:171–173.
    1. Guo S, Dai C, Guo M, Taylor B, Harmon JS, Sander M, Robertson RP, Powers AC, Stein R. Inactivation of specific β cell transcription factors in type 2 diabetes. J. Clin. Invest. 2013;123:3305–3316.
    1. Kayton NS, Poffenberger G, Henske J, Dai C, Thompson C, Aramandla R, Shostak A, Nicholson W, Brissova M, Bush WS, Powers AC. Human islet preparations distributed for research exhibit a variety of insulin-secretory profiles. Am. J. Physiol. Endocrinol. Metab. 2015;308:E592–E602.
    1. Keenan HA, Sun JK, Levine J, Doria A, Aiello LP, Eisenbarth G, Bonner-Weir S, King GL. Residual insulin production and pancreatic β-cell turnover after 50 years of diabetes: Joslin Medalist Study. Diabetes. 2010;59:2846–2853.
    1. Krogvold L, Skog O, Sundström G, Edwin B, Buanes T, Hanssen KF, Ludvigsson J, Grabherr M, Korsgren O, Dahl-Jørgensen K. Function of isolated pancreatic islets from patients at onset of type 1 diabetes: insulin secretion can be restored after some days in a nondiabetogenic environment in vitro: results from the DiViD study. Diabetes. 2015;64:2506–2512.
    1. Marchetti P, Dotta F, Ling Z, Lupi R, Del Guerra S, Santangelo C, Realacci M, Marselli L, Di Mario U, Navalesi R. Function of pancreatic islets isolated from a type 1 diabetic patient. Diabetes Care. 2000;23:701–703.
    1. Mundinger TO, Mei Q, Foulis AK, Fligner CL, Hull RL, Taborsky GJ., Jr Human type 1 diabetes is characterized by an early, marked, sustained, and islet-selective loss of sympathetic nerves. Diabetes. 2016;65:2322–2330.
    1. Muraro MJ, Dharmadhikari G, Grün D, Groen N, Dielen T, Jansen E, van Gurp L, Engelse MA, Carlotti F, de Koning EJP, van Oudenaarden A. A single-cell transcriptome atlas of the human pancreas. Cell Syst. 2016;3:385–394.e3.
    1. Nir T, Melton DA, Dor Y. Recovery from diabetes in mice by beta cell regeneration. J. Clin. Invest. 2007;117:2553–2561.
    1. Oram RA, Jones AG, Besser REJ, Knight BA, Shields BM, Brown RJ, Hattersley AT, McDonald TJ. The majority of patients with long-duration type 1 diabetes are insulin microsecretors and have functioning beta cells. Diabetologia. 2014;57:187–191.
    1. Piccand J, Strasser P, Hodson DJ, Meunier A, Ye T, Keime C, Birling M-C, Rutter GA, Gradwohl G. Rfx6 maintains the functional identity of adult pancreatic β cells. Cell Rep. 2014;9:2219–2232.
    1. Sanyoura M, Jacobsen L, Carmody D, Del Gaudio D, Alkorta-Aranburu G, Arndt K, Hu Y, Kobiernicki F, Kusmartseva I, Atkinson MA, et al. Pancreatic histopathology of human monogenic diabetes due to causal variants in KCNJ11, HNF1A, GATA6, and LMNA. J. Clin. Endocrinol. Metab. 2018;103:35–45.
    1. Segerstolpe Å, Palasantza A, Eliasson P, Andersson E-M, Andréasson A-C, Sun X, Picelli S, Sabirsh A, Clausen M, Bjursell MK, et al. Single-cell transcriptome profiling of human pancreatic islets in health and type 2 diabetes. Cell Metab. 2016;24:593–607.
    1. Sherr J, Tsalikian E, Fox L, Buckingham B, Weinzimer S, Tamborlane WV, White NH, Arbelaez AM, Kollman C, Ruedy KJ, et al. Evolution of abnormal plasma glucagon responses to mixed-meal feedings in youth with type 1 diabetes during the first 2 years after diagnosis. Diabetes Care. 2014;37:1741–1744.
    1. Smith SB, Qu H-Q, Taleb N, Kishimoto NY, Scheel DW, Lu Y, Patch A-M, Grabs R, Wang J, Lynn FC, et al. Rfx6 directs islet formation and insulin production in mice and humans. Nature. 2010;463:775–780.
    1. Taylor BL, Liu F-F, Sander M. Nkx6.1 is essential for maintaining the functional state of pancreatic beta cells. Cell Rep. 2013;4:1262–1275.
    1. Thorel F, Népote V, Avril I, Kohno K, Desgraz R, Chera S, Herrera PL. Conversion of adult pancreatic α-cells to β-cells after extreme β-cell loss. Nature. 2010;464:1149–1154.
    1. Winkler M, Lutz R, Russ U, Quast U, Bryan J. Analysis of two KCNJ11 neonatal diabetes mutations, V59G and V59A, and the analogous KCNJ8 I60G substitution: differences between the channel subtypes formed with SUR1. J. Biol. Chem. 2009;284:6752–6762.
    1. Xu X, D’Hoker J, Stangé G, Bonné S, De Leu N, Xiao X, Van de Casteele M, Mellitzer G, Ling Z, Pipeleers D, et al. Beta cells can be generated from endogenous progenitors in injured adult mouse pancreas. Cell. 2008;132:197–207.
    1. Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature. 2008;455:627–632.

Source: PubMed

3
Se inscrever