Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Diabetologia ; 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38871836

RESUMO

AIMS/HYPOTHESIS: Stem cell-derived islets (SC-islets) are being used as cell replacement therapy for insulin-dependent diabetes. Non-invasive long-term monitoring methods for SC-islet grafts, which are needed to detect misguided differentiation in vivo and to optimise their therapeutic effectiveness, are lacking. Positron emission tomography (PET) has been used to monitor transplanted primary islets. We therefore aimed to apply PET as a non-invasive monitoring method for SC-islet grafts. METHODS: We implanted different doses of human SC-islets, SC-islets derived using an older protocol or a state-of-the-art protocol and SC-islets genetically rendered hyper- or hypoactive into mouse calf muscle to yield different kinds of grafts. We followed the grafts with PET using two tracers, glucagon-like peptide 1 receptor-binding [18F]F-dibenzocyclooctyne-exendin-4 ([18F]exendin) and the dopamine precursor 6-[18F]fluoro-L-3,4-dihydroxyphenylalanine ([18F]FDOPA), for 5 months, followed by histological assessment of graft size and composition. Additionally, we implanted a kidney subcapsular cohort with different SC-islet doses to assess the connection between C-peptide and stem cell-derived beta cell (SC-beta cell) mass. RESULTS: Small but pure and large but impure grafts were derived from SC-islets. PET imaging allowed detection of SC-islet grafts even <1 mm3 in size, [18F]exendin having a better detection rate than [18F]FDOPA (69% vs 44%, <1 mm3; 96% vs 85%, >1 mm3). Graft volume quantified with [18F]exendin (r2=0.91) and [18F]FDOPA (r2=0.86) strongly correlated with actual graft volume. [18F]exendin PET delineated large cystic structures and its uptake correlated with graft SC-beta cell proportion (r2=0.68). The performance of neither tracer was affected by SC-islet graft hyper- or hypoactivity. C-peptide measurements under fasted or glucose-stimulated conditions did not correlate with SC-islet graft volume or SC-beta cell mass, with C-peptide under hypoglycaemia having a weak correlation with SC-beta cell mass (r2=0.52). CONCLUSIONS/INTERPRETATION: [18F]exendin and [18F]FDOPA PET enable non-invasive assessment of SC-islet graft size and aspects of graft composition. These methods could be leveraged for optimising SC-islet cell replacement therapy in diabetes.

2.
J Theor Biol ; 587: 111822, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38589006

RESUMO

Obesity and diabetes are a progressively more and more deleterious hallmark of modern, well fed societies. In order to study the potential impact of strategies designed to obviate the pathological consequences of detrimental lifestyles, a model for the development of Type 2 diabetes geared towards large population simulations would be useful. The present work introduces such a model, representing in simplified fashion the interplay between average glycemia, average insulinemia and functional beta-cell mass, and incorporating the effects of excess food intake or, conversely, of physical activity levels. Qualitative properties of the model are formally established and simulations are shown as examples of its use.


Assuntos
Glicemia , Diabetes Mellitus Tipo 2 , Insulina , Modelos Biológicos , Humanos , Insulina/metabolismo , Glicemia/metabolismo , Células Secretoras de Insulina/patologia , Obesidade , Simulação por Computador , Estudos Longitudinais , Exercício Físico/fisiologia
3.
Diabetologia ; 66(2): 367-375, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36394644

RESUMO

AIMS/HYPOTHESIS: The role of beta cell mass in the balance of glucose control and hypoglycaemic burden in people with type 1 diabetes is unclear. We applied positron emission tomography (PET) imaging with radiolabelled exendin to compare beta cell mass among people with type 1 diabetes and either low glucose variability (LGV) or high glucose variability (HGV). METHODS: All participants with either LGV (n=9) or HGV (n=7) underwent a mixed-meal tolerance test to determine beta cell function and wore a blinded continuous glucose monitor for a week. After an i.v. injection with [68Ga]Ga-NODAGA-exendin-4, PET images were acquired for the quantification of pancreatic uptake of radiolabelled exendin. The mean standardised uptake value (SUVmean) of the pancreas was used to determine the amount of beta cell mass. RESULTS: Participants with LGV had lower HbA1c (46.0 mmol/mol [44.5-52.5] [6.4% (6.3-7)] vs 80 mmol/mol [69.0-110] [9.5% (8.5-12.2)], p=0.001) and higher time in range (TIR) (75.6% [73.5-90.3] vs 38.7% [25.1-48.5], p=0.002) than those with HGV. The SUVmean of the pancreas was higher for the LGV than for the HGV group (5.1 [3.6-5.6] vs 2.9 [2.1-3.4], p=0.008). The AUCC-peptide:AUCglucose ratio was numerically, but not statistically, higher in the LGV compared with the HGV group (2.7×10-2 [6.2×10-4-5.3×10-2] vs 9.3×10-4 [4.7×10-4-5.2×10-3], p=0.21). SUVmean correlated with the AUCC-peptide:AUCglucose ratio (Pearson r=0.64, p=0.01), as well as with the TIR (r=0.64, p=0.01) and the SD of interstitial glucose levels (r=-0.66, p=0.007). CONCLUSION/INTERPRETATION: Our data show higher beta cell mass in people with type 1 diabetes and LGV than in those with HGV, independent of beta cell function.


Assuntos
Diabetes Mellitus Tipo 1 , Humanos , Diabetes Mellitus Tipo 1/metabolismo , Peptídeo C/metabolismo , Controle Glicêmico , Pâncreas/metabolismo , Glicemia/metabolismo , Glucose/metabolismo
4.
Physiol Genomics ; 55(9): 381-391, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37458461

RESUMO

This study investigated the effects of different multiple low doses of streptozotocin (STZ), namely 35 and 55 mg/kg, on the onset and progression of diabetes in mice. Both doses are commonly used in research, and although both induced a loss of beta cell mass, they had distinct effects on whole glucose tolerance, beta cell function, and gene transcription. Mice treated with 55 mg/kg became rapidly glucose intolerant, whereas those treated with 35 mg/kg had a slower onset and remained glucose tolerant for up to a week before becoming equally glucose intolerant as the 55 mg/kg group. Beta cell mass loss was similar between the two groups, but the 35 mg/kg-treated mice had improved glucose-stimulated insulin secretion in gold-standard hyperglycemic clamp studies. Transcriptomic analysis revealed that the 55 mg/kg dose caused disruptions in nearly five times as many genes as the 35 mg/kg dose in isolated pancreatic islets. Pathways that were downregulated in both doses were more downregulated in the 55 mg/kg-treated mice, whereas pathways that were upregulated in both doses were more upregulated in the 35 mg/kg-treated mice. Moreover, we observed a differential downregulation in the 55 mg/kg-treated islets of beta cell characteristic pathways, such as exocytosis or hormone secretion. On the other hand, apoptosis was differentially upregulated in 35 mg/kg-treated islets, suggesting different transcriptional mechanisms in the onset of STZ-induced damage in the islets. This study demonstrates that the two STZ doses induce distinctly mechanistic progressions for the loss of functional beta cell mass.


Assuntos
Diabetes Mellitus Experimental , Células Secretoras de Insulina , Ilhotas Pancreáticas , Camundongos , Animais , Estreptozocina/efeitos adversos , Estreptozocina/metabolismo , Ilhotas Pancreáticas/metabolismo , Células Secretoras de Insulina/metabolismo , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Glicemia/metabolismo
5.
Semin Cell Dev Biol ; 103: 83-93, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32417220

RESUMO

ß cells uniquely produce and secrete insulin under the control of several, integrated signals, to maintain blood glucose concentrations within a narrow physiological interval. ß cell failure is key to the onset and progression of type 2 diabetes, due to impaired function and reduced mass. In this review we focus on several features of human ß cell dysfunction and pathology in type 2 diabetes, as revealed by direct assessment of isolated islet traits and examination of pancreatic tissue from organ donors, surgical samples or autoptic specimens. Insulin secretion defects and pathology findings are discussed in relation to some of the major underlying mechanisms, to also provide clues for conceiving better prevention and treatment of type 2 diabetes by targeting the pancreatic ß cells.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Humanos
6.
Diabetologia ; 65(3): 490-505, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34932133

RESUMO

AIMS/HYPOTHESIS: Second-generation antipsychotic (SGA) drugs have been associated with the development of type 2 diabetes and the metabolic syndrome in patients with schizophrenia. In this study, we aimed to investigate the effects of two different SGA drugs, olanzapine and aripiprazole, on metabolic state and islet function and plasticity. METHODS: We analysed the functional adaptation of beta cells in 12-week-old B6;129 female mice fed an olanzapine- or aripiprazole-supplemented diet (5.5-6.0 mg kg-1 day-1) for 6 months. Glucose and insulin tolerance tests, in vivo glucose-stimulated insulin secretion and indirect calorimetry were performed at the end of the study. The effects of SGAs on beta cell plasticity and islet serotonin levels were assessed by transcriptomic analysis and immunofluorescence. Insulin secretion was assessed by static incubations and Ca2+ fluxes by imaging techniques. RESULTS: Treatment of female mice with olanzapine or aripiprazole for 6 months induced weight gain (p<0.01 and p<0.05, respectively), glucose intolerance (p<0.01) and impaired insulin secretion (p<0.05) vs mice fed a control chow diet. Aripiprazole, but not olanzapine, induced serotonin production in beta cells vs controls, likely by increasing tryptophan hydroxylase 1 (TPH1) expression, and inhibited Ca2+ flux. Of note, aripiprazole increased beta cell size (p<0.05) and mass (p<0.01) vs mice fed a control chow diet, along with activation of mechanistic target of rapamycin complex 1 (mTORC1)/S6 signalling, without preventing beta cell dysfunction. CONCLUSIONS/INTERPRETATION: Both SGAs induced weight gain and beta cell dysfunction, leading to glucose intolerance; however, aripiprazole had a more potent effect in terms of metabolic alterations, which was likely a result of its ability to modulate the serotonergic system. The deleterious metabolic effects of SGAs on islet function should be considered while treating patients as these drugs may increase the risk for development of the metabolic syndrome and diabetes.


Assuntos
Antipsicóticos , Diabetes Mellitus Tipo 2 , Ilhotas Pancreáticas , Animais , Antipsicóticos/efeitos adversos , Aripiprazol/metabolismo , Aripiprazol/farmacologia , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Humanos , Ilhotas Pancreáticas/metabolismo , Camundongos , Olanzapina/efeitos adversos , Olanzapina/metabolismo
7.
Diabet Med ; 39(12): e14962, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36151994

RESUMO

AIMS: Beta cell endoplasmic reticulum (ER) stress can cause cellular death and dysfunction and has been implicated in the pathogenesis of diabetes. Animal models of beta cell ER stress are critical in further understanding this and for testing novel diabetes therapeutics. The KINGS mouse is a model of beta cell ER stress driven by a heterozygous mutation in Ins2. In this study, we investigated how beta cell ER stress in the KINGS mouse drives diabetes. METHODS: We investigated whether the unfolded protein response (UPR) was activated in islets isolated from male and female KINGS mice and whether this impacted beta cell mass and turnover. RESULTS: Whilst the UPR was up-regulated in KINGS islets, with increased protein expression of markers of all three UPR arms, this was not associated with a mass loss of beta cells; beta cell apoptosis rates did not increase until after the development of overt diabetes, and did not lead to substantial changes in beta cell mass. CONCLUSION: We propose that the KINGS mouse represents a model where beta cell maladaptive UPR signalling drives diabetes development without causing mass beta cell loss.


Assuntos
Diabetes Mellitus , Células Secretoras de Insulina , Feminino , Masculino , Camundongos , Humanos , Animais , Estresse do Retículo Endoplasmático/fisiologia , Células Secretoras de Insulina/metabolismo , Resposta a Proteínas não Dobradas , Diabetes Mellitus/metabolismo , Apoptose
8.
Cell Mol Life Sci ; 78(1): 287-298, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32189007

RESUMO

Unveiling the key pathways underlying postnatal beta-cell proliferation can be instrumental to decipher the mechanisms of beta-cell mass plasticity to increased physiological demand of insulin during weight gain and pregnancy. Using transcriptome and global Serine Threonine Kinase activity (STK) analyses of islets from newborn (10 days old) and adult rats, we found that highly proliferative neonatal rat islet cells display a substantially elevated activity of the mitogen activated protein 3 kinase 12, also called dual leucine zipper-bearing kinase (Dlk). As a key upstream component of the c-Jun amino terminal kinase (Jnk) pathway, Dlk overexpression was associated with increased Jnk3 activity and was mainly localized in the beta-cell cytoplasm. We provide the evidence that Dlk associates with and activates Jnk3, and that this cascade stimulates the expression of Ccnd1 and Ccnd2, two essential cyclins controlling postnatal beta-cell replication. Silencing of Dlk or of Jnk3 in neonatal islet cells dramatically hampered primary beta-cell replication and the expression of the two cyclins. Moreover, the expression of Dlk, Jnk3, Ccnd1 and Ccnd2 was induced in high replicative islet beta cells from ob/ob mice during weight gain, and from pregnant female rats. In human islets from non-diabetic obese individuals, DLK expression was also cytoplasmic and the rise of the mRNA level was associated with an increase of JNK3, CCND1 and CCND2 mRNA levels, when compared to islets from lean and obese patients with diabetes. In conclusion, we find that activation of Jnk3 signalling by Dlk could be a key mechanism for adapting islet beta-cell mass during postnatal development and weight gain.


Assuntos
Células Secretoras de Insulina/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Transdução de Sinais , Animais , Proliferação de Células/efeitos dos fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Ciclina D2/genética , Ciclina D2/metabolismo , Feminino , Glucose/farmacologia , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/citologia , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 10 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 10 Ativada por Mitógeno/genética , Obesidade/metabolismo , Obesidade/patologia , Pâncreas/crescimento & desenvolvimento , Pâncreas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
9.
Endocr J ; 69(12): 1407-1414, 2022 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-35934795

RESUMO

The development of pancreatic cancer (PC) is associated with worsening of glucose tolerance. However, there is limited information about the effects of PC on islet morphology. The aim of this study was to elucidate changes in alpha and beta cell mass in patients with PC. We enrolled 30 autopsy cases with death due to PC (9 with diabetes; DM) and 31 age- and BMI-matched autopsy cases without PC (controls, 12 with DM). Tumor-free pancreatic sections were stained for insulin and glucagon, and fractional beta cell (BCA) and alpha cell area (ACA) were quantified. In addition, expression of de-differentiation markers, i.e., ALDH1A3 and UCN3, was qualitatively evaluated. The pancreas of subjects with PC showed atrophic and fibrotic changes. There was no significant difference in BCA in subjects with PC compared to controls (1.53 ± 1.26% vs. 0.95 ± 0.42%, p = 0.07). However, ACA and ACA to BCA ratio were significantly higher in subjects with PC compared to controls (2.48 ± 2.39% vs. 0.53 ± 0.26% and 1.94 ± 1.93 vs. 0.59 ± 0.26, respectively, both p < 0.001). Increased ACA to BCA ratio was observed in subjects with PC irrespective of the presence of DM. Qualitative evaluation of ALDH1A3 and UCN3 expression showed no significant difference between the groups. In conclusion, in subjects with PC, alpha to beta cell mass ratio is increased, which may contribute to the increased risk of worsening glucose metabolism. Further studies are warranted to elucidate the mechanisms of increased alpha to beta cell mass in patients with PC.


Assuntos
Diabetes Mellitus , Células Secretoras de Glucagon , Células Secretoras de Insulina , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/complicações , Insulina , Neoplasias Pancreáticas
10.
Diabetologia ; 64(8): 1816-1821, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33938968

RESUMO

AIMS/HYPOTHESIS: Type 2 diabetes is characterised by reduced beta cell mass (BCM). However, it remains uncertain whether the reduction in BCM in type 2 diabetes is due to a decrease in size or number of beta cells. Our aim was to examine the impact of beta cell size and number on islet morphology in humans with and without type 2 diabetes. METHODS: Pancreas samples were obtained from 64 Japanese adults with (n = 26) and without (n = 38) type 2 diabetes who underwent pancreatectomy. Using pancreatic tissues stained for insulin, we estimated beta cell size based on beta cell diameter. Beta cell number was estimated from the product of fractional beta cell area and pancreas volume divided by beta cell size. The associations of beta cell size and number with islet morphology and metabolic status were examined. RESULTS: Both beta cell size (548.7 ± 58.5 vs 606.7 ± 65.0 µm3, p < 0.01) and number (5.10 × 108 ± 2.35 × 108 vs 8.16 × 108 ± 4.27 × 108, p < 0.01) were decreased in participants with type 2 diabetes compared with those without diabetes, with the relative reduction in beta cell number (37%) being greater than for beta cell size (10%). Beta cell number but not size was positively correlated with BCM in participants with and without type 2 diabetes (r = 0.97 and r = 0.98, both p < 0.01) and negatively correlated with HbA1c (r = -0.45, p < 0.01). CONCLUSIONS/INTERPRETATION: Both beta cell size and number were reduced in participants with type 2 diabetes, with the relative reduction in beta cell number being greater. Decrease in beta cell number appears to be a major contributor to reduced BCM in type 2 diabetes.


Assuntos
Contagem de Células , Tamanho Celular , Diabetes Mellitus Tipo 2/patologia , Células Secretoras de Insulina/patologia , Idoso , Cromatografia Líquida de Alta Pressão , Diabetes Mellitus Tipo 2/sangue , Feminino , Hemoglobinas Glicadas/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Pancreatectomia
11.
Molecules ; 26(5)2021 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-33800801

RESUMO

Diabetes remains one of the fastest growing chronic diseases and is a leading source of morbidity and accelerated mortality in the world. Loss of beta cell mass (BCM) and decreased sensitivity to insulin underlie diabetes pathogenesis. Yet, the ability to safely and directly assess BCM in individuals with diabetes does not exist. Measures such as blood glucose provide only a crude indirect picture of beta cell health. PET imaging could, in theory, allow for safe, direct, and precise characterization of BCM. However, identification of beta cell-specific radiolabeled tracers remains elusive. G-protein coupled receptor 44 (GPR44) is a transmembrane protein that was characterized in 2012 as highly beta cell-specific within the insulin-positive islets of Langerhans. Accordingly, radiolabeling of existing GPR44 antagonists could be a viable method to accelerate PET tracer development. The present study aims to evaluate and summarize published analogues of the GPR44 antagonist ramatroban to develop 18F-labeled PET tracers for BCM analysis. The 77 corresponding ramatroban analogues containing a fluorine nuclide were characterized for properties including binding affinity, selectivity, and pharmacokinetic and metabolic profile, and 32 compounds with favorable properties were identified. This review illustrates the potential of GPR44 analogues for the development of PET tracers.


Assuntos
Carbazóis/química , Radioisótopos de Flúor/metabolismo , Células Secretoras de Insulina/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Traçadores Radioativos , Compostos Radiofarmacêuticos/metabolismo , Receptores Imunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Sulfonamidas/química , Humanos , Células Secretoras de Insulina/química , Células Secretoras de Insulina/citologia , Inibidores da Agregação Plaquetária/química
12.
Diabetologia ; 63(6): 1199-1210, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32239263

RESUMO

AIMS/HYPOTHESIS: Low birthweight is associated with a high risk of diabetes, but there are no reports discussing birthweight and pancreatic tissues in humans. The purpose of this study was to examine the correlation between birthweight and beta and alpha cell mass in humans. METHODS: Sixty-four Japanese adults with and without diabetes who underwent pancreatectomy and were able to recall their weight history including birthweight were included. Pancreatic tissues were stained for insulin and glucagon, and fractional beta cell area (BCA) and alpha cell area (ACA) were quantified. Islet size and density and beta cell replication were also quantified and their associations with birthweight were evaluated. RESULTS: In participants without diabetes, there was a weak positive correlation between birthweight and BCA (R = 0.34, p = 0.03). The group with a history of childhood obesity, but not the group with a history of obesity in adulthood only, showed higher BCA compared with those without a history of obesity (1.78 ± 0.74% vs 0.99 ± 0.53%, p = 0.01), and the correlation coefficient between birthweight and BCA increased after excluding those with a history of childhood obesity (R = 0.51, p < 0.01). In those with diabetes, there was no correlation between birthweight and BCA. No correlation was found between birthweight and ACA in either those with or without diabetes. CONCLUSIONS/INTERPRETATION: Birthweight and beta, but not alpha, cell mass are positively correlated in non-diabetic adults, and a history of childhood obesity may affect beta cell mass. Graphical abstract.


Assuntos
Peso ao Nascer/fisiologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Obesidade Infantil/metabolismo , Obesidade Infantil/fisiopatologia , Humanos , Japão/epidemiologia , Pâncreas/metabolismo , Obesidade Infantil/epidemiologia , Inquéritos e Questionários
13.
Diabetologia ; 63(5): 987-1001, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32072192

RESUMO

AIMS/HYPOTHESIS: High-mobility group box 1 (HMGB1), an evolutionarily conserved chromosomal protein, was rediscovered to be a 'danger signal' (alarmin) that alerts the immune system once released extracellularly. Therefore, it has been recognised contributing to the pathogenesis of autoimmune diabetes, but its exact impact on the initiation and progression of type 1 diabetes, as well as the related molecular mechanisms, are yet to be fully characterised. METHODS: In the current report, we employed NOD mice as a model to dissect the impact of blocking HMGB1 on the prevention, treatment and reversal of type 1 diabetes. To study the mechanism involved, we extensively examined the characteristics of regulatory T cells (Tregs) and their related signalling pathways upon HMGB1 stimulation. Furthermore, we investigated the relevance of our data to human autoimmune diabetes. RESULTS: Neutralising HMGB1 both delayed diabetes onset and, of particular relevance, reversed diabetes in 13 out of 20 new-onset diabetic NOD mice. Consistently, blockade of HMGB1 prevented islet isografts from autoimmune attack in diabetic NOD mice. Using transgenic reporter mice that carry a Foxp3 lineage reporter construct, we found that administration of HMGB1 impairs Treg stability and function. Mechanistic studies revealed that HMGB1 activates receptor for AGE (RAGE) and toll-like receptor (TLR)4 to enhance phosphatidylinositol 3-kinase (PI3K)-Akt-mechanistic target of rapamycin (mTOR) signalling, thereby impairing Treg stability and functionality. Indeed, high circulating levels of HMGB1 in human participants with type 1 diabetes contribute to Treg instability, suggesting that blockade of HMGB1 could be an effective therapy against type 1 diabetes in clinical settings. CONCLUSIONS/INTERPRETATION: The present data support the possibility that HMGB1 could be a viable therapeutic target to prevent the initiation, progression and recurrence of autoimmunity in the setting of type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Proteína HMGB1/metabolismo , Linfócitos T Reguladores/metabolismo , Animais , Anticorpos Neutralizantes/farmacologia , Western Blotting , Células Cultivadas , Colite/imunologia , Colite/metabolismo , Colite/patologia , Diabetes Mellitus Tipo 1/patologia , Feminino , Proteína HMGB1/antagonistas & inibidores , Humanos , Transplante das Ilhotas Pancreáticas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Fosfatidilinositol 3-Quinases/metabolismo
14.
Cell Tissue Res ; 381(1): 163-175, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32060653

RESUMO

The ß1 integrin subunit contributes to pancreatic beta cell growth and function through communication with the extracellular matrix (ECM). The effects of in vitro and in vivo ß1 integrin knockout have been extensively studied in mature islets, yet no study to date has examined how the loss of ß1 integrin during specific stages of pancreatic development impacts beta cell maturation. Beta-cell-specific tamoxifen-inducible Cre recombinase (MIP-CreERT) mice were crossed with mice containing floxed Itgb1 (ß1 integrin) to create an inducible mouse model (MIPß1KO) at the second transition stage (e13.5) of pancreas development. By e19.5-20.5, the expression of beta-cell ß1 integrin in fetal MIPß1KO mice was significantly reduced and these mice displayed decreased beta cell mass, density and proliferation. Morphologically, fetal MIPß1KO pancreata exhibited reduced islet vascularization and nascent endocrine cells in the ductal region. In addition, decreased ERK phosphorylation was observed in fetal MIPß1KO pancreata. The expression of transcription factors needed for beta-cell development was unchanged in fetal MIPß1KO pancreata. The findings from this study demonstrate that ß1 integrin signaling is required during a transition-specific window in the developing beta-cell to maintain islet mass and vascularization.


Assuntos
Células Secretoras de Insulina/citologia , Integrina beta1/fisiologia , Pâncreas/embriologia , Animais , Diferenciação Celular , Feminino , Desenvolvimento Fetal , Integrinas/deficiência , Masculino , Camundongos , Camundongos Knockout , Pâncreas/irrigação sanguínea
15.
Diabetologia ; 62(6): 1000-1010, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30852627

RESUMO

AIMS/HYPOTHESIS: Islet amyloid polypeptide (IAPP) misfolding and toxic oligomers contribute to beta cell loss and stress in type 2 diabetes. Pregnancy-related diabetes predicts subsequent risk for type 2 diabetes but little is known about the impact of pregnancy on beta cell mass, turnover and stress. Availability of human pancreas tissue in pregnancy is limited and most widely used mouse models of type 2 diabetes do not develop pregnancy-related diabetes, possibly because rodent IAPP is not prone to form toxic oligomers. We hypothesised that mice transgenic for human IAPP (hIAPP) are prone to pregnancy-related diabetes with beta cell responses reflective of those in type 2 diabetes. METHODS: We evaluated the impact of a first and second pregnancy on glucose homeostasis, beta cell mass and turnover and markers of beta cell stress in hIAPP transgenic (hTG) mice. RESULTS: Pregnancy induced both endoplasmic reticulum stress and oxidative stress and compromised autophagy in beta cells in hTG mice, which are characteristic of beta cells in type 2 diabetes. Beta cell stress persisted after pregnancy, resulting in subsequent diabetes before or during a second pregnancy. CONCLUSIONS/INTERPRETATION: High expression of hIAPP in response to pregnancy recapitulates mechanisms contributing to beta cell stress in type 2 diabetes. We hypothesise that, in individuals prone to type 2 diabetes, pregnancy-induced increased expression of IAPP inflicts beta cell damage that persists and is compounded by subsequent additive stress such as further pregnancy. The hTG mouse model is a novel model for pregnancy-related diabetes.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Animais , Autofagia/fisiologia , Dano ao DNA/genética , Dano ao DNA/fisiologia , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/fisiologia , Feminino , Humanos , Insulina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Camundongos , Camundongos Transgênicos , Estresse Oxidativo/fisiologia , Gravidez
16.
Diabetologia ; 62(4): 567-577, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30767048

RESUMO

Histological analysis of donor pancreases coupled with measurement of serum C-peptide in clinical cohorts has challenged the idea that all beta cells are eventually destroyed in type 1 diabetes. These findings have raised a number of questions regarding how the remaining beta cells have escaped immune destruction, whether pools of 'sleeping' or dysfunctional beta cells could be rejuvenated and whether there is potential for new growth of beta cells. In this Review, we describe histological and in vivo evidence of persistent beta cells in type 1 diabetes and discuss the limitations of current methods to distinguish underlying beta cell mass in comparison with beta cell function. We highlight that evidence for new beta cell growth in humans many years from diagnosis is limited, and that this growth may be very minimal if at all present. We review recent contributions to the debate around beta cell abnormalities contributing to the pathogenesis of type 1 diabetes. We also discuss evidence for restoration of beta cell function, as opposed to mass, in recent-onset type 1 diabetes, but highlight the absence of data supporting functional recovery in the setting of long-duration diabetes. Finally, future areas of research are suggested to help resolve the source and phenotype of residual beta cells that persist in some, but not all, people with type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/fisiopatologia , Células Secretoras de Insulina/fisiologia , Biomarcadores/sangue , Peptídeo C/sangue , Diabetes Mellitus Tipo 1/sangue , Humanos , Insulina/metabolismo , Pâncreas/metabolismo , Fenótipo , Projetos de Pesquisa
17.
Curr Diab Rep ; 19(8): 49, 2019 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-31250117

RESUMO

PURPOSE OF REVIEW: Quantitative markers for beta-cell mass (BCM) in human pancreas are currently lacking. Medical imaging using positron emission tomography (PET) markers for beta-cell restricted targets may provide an accurate and non-invasive measurement of BCM, to assist diagnosis and treatment of metabolic disease. GPR44 was recently discovered as a putative marker for beta cells and this review summarizes the developments so far. RECENT FINDINGS: Several small molecule binders targeting GPR44 have been radiolabeled for PET imaging and evaluated in vitro and in small and large animal models. 11C-AZ12204657 and 11C-MK-7246 displayed a dose-dependent and GPR44-mediated binding to beta cells both in vitro and in vivo, with negligible uptake in exocrine pancreas. GPR44 represents an attractive target for visualization of BCM. Further progress in radioligand development including clinical testing is expected to clarify the role of GPR44 as a surrogate marker for BCM in humans.


Assuntos
Células Secretoras de Insulina , Animais , Biomarcadores , Humanos , Pâncreas , Tomografia por Emissão de Pósitrons
18.
Adv Exp Med Biol ; 1122: 27-40, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30937861

RESUMO

Glucose homeostasis relies on tightly regulated insulin secretion from pancreatic beta-cells, and its loss in diabetes is associated with the dysfunction of these cells. Beta-cells reside in the islets of Langerhans, which are highly vascularized by a dense capillary network comprised of endothelial cells and pericytes. While the requirement of the endothelium for the proper pancreatic function is well established, the role of pancreatic pericytes has only recently begun to unveil. Recent studies described multiple roles for pancreatic pericytes in glucose homeostasis, highlighting their function as both regulators of islet blood flow and as a source of critical signals that support proper beta-cell function and mass. Furthermore, recent findings point to the contribution of pericytic abnormalities to beta-cell dysfunction in type 2 diabetes, implicating the involvement of pancreatic pericytes in both the initiation and the progression of this disease. This newly gained data implicate pancreatic pericytes as critical components of the cellular network required for glucose regulation.


Assuntos
Diabetes Mellitus Tipo 2 , Glucose/metabolismo , Homeostase , Células Secretoras de Insulina/citologia , Pericitos/citologia , Humanos , Insulina , Ilhotas Pancreáticas
19.
Diabetologia ; 61(12): 2598-2607, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29721633

RESUMO

AIMS/HYPOTHESIS: The progressive loss of beta cell function is part of the natural history of type 2 diabetes. Autopsy studies suggest that this is, in part, due to loss of beta cell mass (BCM), but this has not been confirmed in vivo. Non-invasive methods to quantify BCM may contribute to a better understanding of type 2 diabetes pathophysiology and the development of therapeutic strategies. In humans, the localisation of vesicular monoamine transporter type 2 (VMAT2) in beta cells and pancreatic polypeptide cells, with minimal expression in other exocrine or endocrine pancreatic cells, has led to its development as a measure of BCM. We used the VMAT2 tracer [18F]fluoropropyl-(+)-dihydrotetrabenazine to quantify BCM in humans with impaired glucose tolerance (prediabetes) or type 2 diabetes, and in healthy obese volunteers (HOV). METHODS: Dynamic positron emission tomography (PET) data were obtained for 4 h with metabolite-corrected arterial blood measurement in 16 HOV, five prediabetic and 17 type 2 diabetic participants. Eleven participants (six HOV and five with type 2 diabetes) underwent two abdominal PET/computed tomography (CT) scans for the assessment of test-retest variability. Standardised uptake value ratio (SUVR) was calculated in pancreatic subregions (head, body and tail), with the spleen as a reference region to determine non-specific tracer uptake at 3-4 h. The outcome measure SUVR minus 1 (SUVR-1) accounts for non-specific tracer uptake. Functional beta cell capacity was assessed by C-peptide release following standard (arginine stimulus test [AST]) and acute insulin response to the glucose-enhanced AST (AIRargMAX). Pearson correlation analysis was performed between the binding variables and the C-peptide AUC post-AST and post-AIRargMAX. RESULTS: Absolute test-retest variability (aTRV) was ≤15% for all regions. Variability and overlap of SUVR-1 was measured in all groups; HOV and participants with prediabetes and with type 2 diabetes. SUVR-1 showed significant positive correlations with AIRargMAX (all groups) in all pancreas subregions (whole pancreas p = 0.009 and pancreas head p = 0.009; body p = 0.019 and tail p = 0.023). SUVR-1 inversely correlated with HbA1c (all groups) in the whole pancreas (p = 0.033) and pancreas head (p = 0.008). SUVR-1 also inversely correlated with years since diagnosis of type 2 diabetes in the pancreas head (p = 0.049) and pancreas tail (p = 0.035). CONCLUSIONS/INTERPRETATION: The observed correlations of VMAT2 density in the pancreas and pancreas regions with years since diagnosis of type 2 diabetes, glycaemic control and beta cell function suggest that loss of BCM contributes to deficient insulin secretion in humans with type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Pâncreas/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Proteínas Vesiculares de Transporte de Monoamina/metabolismo , Feminino , Humanos , Células Secretoras de Insulina/metabolismo , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade
20.
Diabetologia ; 61(12): 2512-2515, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29955934

RESUMO

In this commentary, we describe the limitations of positron emission tomography (PET) in visualising and characterising beta cell mass in the native pancreas in healthy individuals and those diagnosed with diabetes. Imaging with PET requires a large mass of targeted cells or other structures in the range of approximately 8-10 cm3. Since islets occupy only 1% of the pancreatic volume and are dispersed throughout the organ, it is our view that uptake of PET tracers, including [18F]fluoropropyl-(+)-dihydrotetrabenazine, in islets cannot be successfully detected by current imaging modalities. Therefore, we dispute the feasibility of PET imaging for the detection of loss of beta cells in the native pancreas in individuals with diabetes. However, we believe this novel approach can be successfully employed to visualise beta cell mass in individuals with hyperinsulinism and transplanted islets.


Assuntos
Células Secretoras de Insulina/patologia , Futilidade Médica , Pâncreas/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA