Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.484
Filter
1.
Int J Mol Sci ; 25(10)2024 May 07.
Article in English | MEDLINE | ID: mdl-38791099

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by progressive loss of motor neurons. Emerging evidence suggests a potential link between metabolic dysregulation and ALS pathogenesis. This study aimed to investigate the relationship between metabolic hormones and disease progression in ALS patients. A cross-sectional study was conducted involving 44 ALS patients recruited from a tertiary care center. Serum levels of insulin, total amylin, C-peptide, active ghrelin, GIP (gastric inhibitory peptide), GLP-1 active (glucagon-like peptide-1), glucagon, PYY (peptide YY), PP (pancreatic polypeptide), leptin, interleukin-6, MCP-1 (monocyte chemoattractant protein-1), and TNFα (tumor necrosis factor alpha) were measured, and correlations with ALSFRS-R, evolution scores, and biomarkers were analyzed using Spearman correlation coefficients. Subgroup analyses based on ALS subtypes, progression pattern of disease, and disease progression rate patterns were performed. Significant correlations were observed between metabolic hormones and ALS evolution scores. Insulin and amylin exhibited strong correlations with disease progression and clinical functional outcomes, with insulin showing particularly robust associations. Other hormones such as C-peptide, leptin, and GLP-1 also showed correlations with ALS progression and functional status. Subgroup analyses revealed differences in hormone levels based on sex and disease evolution patterns, with male patients showing higher amylin and glucagon levels. ALS patients with slower disease progression exhibited elevated levels of amylin and insulin. Our findings suggest a potential role for metabolic hormones in modulating ALS progression and functional outcomes. Further research is needed to elucidate the underlying mechanisms and explore the therapeutic implications of targeting metabolic pathways in ALS management.


Subject(s)
Amyotrophic Lateral Sclerosis , Biomarkers , Insulin , Islet Amyloid Polypeptide , Humans , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/blood , Male , Female , Middle Aged , Aged , Islet Amyloid Polypeptide/metabolism , Islet Amyloid Polypeptide/blood , Cross-Sectional Studies , Biomarkers/blood , Insulin/metabolism , Insulin/blood , Disease Progression , Leptin/blood , Leptin/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/blood , C-Peptide/blood , C-Peptide/metabolism , Ghrelin/metabolism , Ghrelin/blood , Glucagon/blood , Glucagon/metabolism , Adult , Hormones/metabolism , Hormones/blood
2.
Sci Rep ; 14(1): 11640, 2024 05 21.
Article in English | MEDLINE | ID: mdl-38773268

ABSTRACT

Porcine islet xenotransplantation is a promising therapy for severe diabetes mellitus. Maintenance of the quality and quantity of porcine islets is important for the success of this treatment. Here, we aimed to elucidate the influence of relatively short-term (14 days) culture on adult porcine islets isolated from three micro-minipigs (P111, P112 and P121). Morphological characteristics of islets changed little after 14 days of culture. The viability of cultured islets was also maintained at a high level (> 80%). Furthermore, cultured islets exhibited similar glucose-stimulated insulin secretion and insulin content at Day 14 were preserved comparing with Day 1, while the expressions of Ins, Gcg and Sst were attenuated at Day 14. Xenotransplantation using diabetic nude mice showed no normalization of blood glucose but increased levels of plasma porcine C-peptide after the transplantation of 14 day cultured porcine islets. Histological assessment revealed that relatively short-term cultured porcine islets were successfully engrafted 56 days following transplantation. These data show that relatively short-term culture did not impair the quality of adult porcine islets in regard to function, morphology, and viability. Prevention of impairment of gene correlated with endocrine hormone is warranted for further improvement.


Subject(s)
Insulin , Islets of Langerhans Transplantation , Islets of Langerhans , Transplantation, Heterologous , Animals , Islets of Langerhans/metabolism , Islets of Langerhans/cytology , Swine , Islets of Langerhans Transplantation/methods , Insulin/metabolism , Mice , Mice, Nude , Insulin Secretion , Diabetes Mellitus, Experimental/therapy , Blood Glucose/metabolism , Swine, Miniature , Cell Survival , C-Peptide/metabolism , C-Peptide/blood
3.
Rev Med Suisse ; 20(876): 1069-1073, 2024 May 29.
Article in French | MEDLINE | ID: mdl-38812338

ABSTRACT

C-peptide measurement allows an estimation of the residual endogenous insulin secretion in diabetic patients. Nowadays plasmatic testing is convenient and unexpensive, but we lack standardized tests. Therefore, there are no official recommendation regarding its use. As an indication, in some circumstances, C-peptide measurement could be used to specify the type of diabetes, help guide the treatment strategy and potentially assess the risk for complications. Its use is still limited and not recommended on a routine base for all patients living with diabetes, but in the future, tests standardization and establishment of reference ranges could give more insight on the clinical relevance of C-peptide measurement.


Le dosage du peptide-C est une mesure permettant d'évaluer la sécrétion endogène résiduelle d'insuline chez les patients diabétiques. Le dosage plasmatique est facilement réalisable actuellement, pour un coût modeste, mais l'absence de standardisation des tests ne permet pas d'émettre des recommandations officielles par rapport à son utilisation. À titre indicatif, dans certaines situations, le dosage du peptide-C peut être utilisé pour préciser le type de diabète, guider les traitements médicamenteux et potentiellement évaluer les risques de complications. Son utilisation est pour le moment limitée et n'est pas recommandée en routine pour tous les patients atteints de diabète, mais à l'avenir, la formalisation du dosage et l'établissement de valeurs de référence pourraient permettre de définir son utilisation clinique.


Subject(s)
C-Peptide , Insulin Secretion , Insulin , Humans , C-Peptide/blood , C-Peptide/metabolism , Insulin/metabolism , Insulin Secretion/physiology , Diabetes Mellitus/blood , Diabetes Mellitus/metabolism , Diabetes Mellitus/diagnosis
4.
Cell Transplant ; 33: 9636897241243014, 2024.
Article in English | MEDLINE | ID: mdl-38659255

ABSTRACT

Stress-induced islet graft loss during the peri-transplantation period reduces the efficacy of islet transplantation. In this prospective, randomized, double-blind clinical trial, we evaluated the safety and efficacy of 60 mg/kg human alpha-1 antitrypsin (AAT) or placebo infusion weekly for four doses beginning before surgery in chronic pancreatitis (CP) patients undergoing total pancreatectomy and islet autotransplantation (TP-IAT). Subjects were followed for 12 months post-TP-IAT. The dose of AAT was safe, as there was no difference in the types and severity of adverse events in participants from both groups. There were some biochemical signals of treatment effect with a higher oxygen consumption rate in AAT islets before transplantation and a lower serum C-peptide (an indicator of islet death) in the AAT group at 15 min after islet infusion. Findings per the statistical analysis plan using a modified intention to treat analysis showed no difference in the C-peptide area under the curve (AUC) following a mixed meal tolerance test at 12 months post-TP-IAT. There was no difference in the secondary and exploratory outcomes. Although AAT therapy did not show improvement in C-peptide AUC in this study, AAT therapy is safe in CP patients and there are experiences gained on optimal clinical trial design in this challenging disease.


Subject(s)
Islets of Langerhans Transplantation , Pancreatectomy , Pancreatitis, Chronic , Transplantation, Autologous , alpha 1-Antitrypsin , Humans , Islets of Langerhans Transplantation/methods , Pancreatitis, Chronic/surgery , Pancreatitis, Chronic/therapy , alpha 1-Antitrypsin/therapeutic use , Male , Female , Pancreatectomy/methods , Middle Aged , Transplantation, Autologous/methods , Adult , Double-Blind Method , C-Peptide/blood , C-Peptide/metabolism , Prospective Studies
5.
Diabetes Care ; 47(6): 1048-1055, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38621411

ABSTRACT

OBJECTIVE: Mixed-meal tolerance test-stimulated area under the curve (AUC) C-peptide at 12-24 months represents the primary end point for nearly all intervention trials seeking to preserve ß-cell function in recent-onset type 1 diabetes. We hypothesized that participant benefit might be detected earlier and predict outcomes at 12 months posttherapy. Such findings would support shorter trials to establish initial efficacy. RESEARCH DESIGN AND METHODS: We examined data from six Type 1 Diabetes TrialNet immunotherapy randomized controlled trials in a post hoc analysis and included additional stimulated metabolic indices beyond C-peptide AUC. We partitioned the analysis into successful and unsuccessful trials and analyzed the data both in the aggregate as well as individually for each trial. RESULTS: Among trials meeting their primary end point, we identified a treatment effect at 3 and 6 months when using C-peptide AUC (P = 0.030 and P < 0.001, respectively) as a dynamic measure (i.e., change from baseline). Importantly, no such difference was seen in the unsuccessful trials. The use of C-peptide AUC as a 6-month dynamic measure not only detected treatment efficacy but also suggested long-term C-peptide preservation (R2 for 12-month C-peptide AUC adjusted for age and baseline value was 0.80, P < 0.001), and this finding supported the concept of smaller trial sizes down to 54 participants. CONCLUSIONS: Early dynamic measures can identify a treatment effect among successful immune therapies in type 1 diabetes trials with good long-term prediction and practical sample size over a 6-month period. While external validation of these findings is required, strong rationale and data exist in support of shortening early-phase clinical trials.


Subject(s)
C-Peptide , Diabetes Mellitus, Type 1 , Immunotherapy , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Humans , C-Peptide/blood , C-Peptide/metabolism , Immunotherapy/methods , Female , Male , Adolescent , Treatment Outcome , Randomized Controlled Trials as Topic , Child , Adult , Area Under Curve
6.
Diabetes ; 73(6): 953-963, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38506952

ABSTRACT

Normal-weight individuals with usual-onset type 2 diabetes have reduced ß-cell function and greater insulin sensitivity compared with their obese counterparts. The relative contribution of ß-cell dysfunction and insulin resistance to young-onset type 2 diabetes (YOD) among normal-weight individuals is not well established. In 44 individuals with YOD (24 with normal weight and 20 with obesity) and 24 healthy control individuals with normoglycemia (12 with normal weight and 12 with obesity), we conducted 2-h 12 mmol/L hyperglycemic clamps to measure acute (0-10 min) and steady-state (100-120 min) insulin and C-peptide responses, as well as insulin sensitivity index. Normal-weight individuals with YOD had lower acute insulin response, steady-state insulin and C-peptide responses, and a higher insulin sensitivity index compared with their obese counterparts with YOD. Compared with BMI-matched healthy control individuals, normal-weight individuals with YOD had lower acute and steady-state insulin and C-peptide responses but a similar insulin sensitivity index. The impairment of steady-state ß-cell response relative to healthy control individuals was more pronounced in normal-weight versus obese individuals with YOD. In conclusion, normal-weight Chinese with YOD exhibited worse ß-cell function but preserved insulin sensitivity relative to obese individuals with YOD and BMI-matched healthy individuals with normoglycemia. The selection of glucose-lowering therapy should account for pathophysiological differences underlying YOD between normal-weight and obese individuals.


Subject(s)
C-Peptide , Diabetes Mellitus, Type 2 , Insulin Resistance , Insulin-Secreting Cells , Insulin , Obesity , Humans , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Insulin Resistance/physiology , Obesity/metabolism , Obesity/physiopathology , Male , Female , Adult , C-Peptide/blood , C-Peptide/metabolism , Insulin/metabolism , Insulin/blood , Glucose Clamp Technique , Blood Glucose/metabolism , Body Mass Index , China/epidemiology , Age of Onset , Asian People , Young Adult , Case-Control Studies , East Asian People
7.
Am J Physiol Endocrinol Metab ; 326(5): E673-E680, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38446636

ABSTRACT

Residual beta cells are present in most patients with longstanding type 1 diabetes but it is unknown whether these beta cells react normally to different stimuli. Moreover a defect in proinsulin conversion and abnormal alpha cell response are also part of the islet dysfunction. A three-phase [euglycemia, hyperglycemia, and hyperglycemia + glucagon-like peptide 1 (GLP-1)] clamp was performed in patients with longstanding type 1 diabetes. Intravenous arginine boluses were administered at the end of each phase. On another day, a mixed meal stimulation test with a subsequent intravenous arginine bolus was performed. C-peptide was detectable in a subgroup of subjects at baseline (2/15) or only after stimulation (3/15). When detectable, C-peptide increased 2.9-fold [95% CI: 1.2-7.1] during the hyperglycemia phase and 14.1-fold [95% CI: 3.1-65.2] during the hyperglycemia + GLP-1 phase, and 22.3-fold [95% CI: 5.6-89.1] during hyperglycemia + GLP-1 + arginine phase when compared with baseline. The same subset of patients with a C-peptide response were identified during the mixed meal stimulation test as during the clamp. There was an inhibition of glucagon secretion (0.72-fold, [95% CI: 0.63-0.84]) during the glucose clamp irrespective of the presence of detectable beta cell function. Proinsulin was only present in a subset of subjects with detectable C-peptide (3/15) and proinsulin mimicked the C-peptide response to the different stimuli when detectable. Residual beta cells in longstanding type 1 diabetes respond adequately to different stimuli and could be of clinical benefit.NEW & NOTEWORTHY If beta cell function is detectable, the beta cells react relatively normal to the different stimuli except for the first phase response to intravenous glucose. An oral mixed meal followed by an intravenous arginine bolus can identify residual beta cell function/mass as well as the more commonly used glucose potentiated arginine-induced insulin secretion during a hyperglycemic clamp.


Subject(s)
Arginine , C-Peptide , Diabetes Mellitus, Type 1 , Food, Formulated , Glucagon-Like Peptide 1 , Glucose , Islets of Langerhans , Adult , Female , Humans , Male , Middle Aged , Arginine/administration & dosage , Arginine/pharmacology , Blood Glucose/metabolism , C-Peptide/blood , C-Peptide/metabolism , Diabetes Mellitus, Type 1/metabolism , Glucagon/metabolism , Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide 1/metabolism , Glucose/administration & dosage , Glucose/metabolism , Glucose Clamp Technique , Hyperglycemia/metabolism , Insulin/metabolism , Insulin/administration & dosage , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Islets of Langerhans/metabolism , Islets of Langerhans/drug effects
8.
J Cell Physiol ; 239(5): e31212, 2024 May.
Article in English | MEDLINE | ID: mdl-38308646

ABSTRACT

C-peptide, a byproduct of insulin synthesis believed to be biologically inert, is emerging as a multifunctional molecule. C-peptide serves an anti-inflammatory and anti-atherogenic role in type 1 diabetes mellitus (T1DM) and early T2DM. C-peptide protects endothelial cells by activating AMP-activated protein kinase α, thus suppressing the activity of NAD(P)H oxidase activity and reducing reactive oxygen species (ROS) generation. It also prevents apoptosis by regulating hyperglycemia-induced p53 upregulation and mitochondrial adaptor p66shc overactivation, as well as reducing caspase-3 activity and promoting expression of B-cell lymphoma-2. Additionally, C-peptide suppresses platelet-derived growth factor (PDGF)-beta receptor and p44/p42 mitogen-activated protein (MAP) kinase phosphorylation to inhibit vascular smooth muscle cells (VSMC) proliferation. It also diminishes leukocyte adhesion by virtue of its capacity to abolish nuclear factor kappa B (NF-kB) signaling, a major pro-inflammatory cascade. Consequently, it is envisaged that supplementation of C-peptide in T1DM might ameliorate or even prevent end-organ damage. In marked contrast, C-peptide increases monocyte recruitment and migration through phosphoinositide 3-kinase (PI-3 kinase)-mediated pathways, induces lipid accumulation via peroxisome proliferator-activated receptor γ upregulation, and stimulates VSMC proliferation and CD4+ lymphocyte migration through Src-kinase and PI-3K dependent pathways. Thus, it promotes atherosclerosis and microvascular damage in late T2DM. Indeed, C-peptide is now contemplated as a potential biomarker for insulin resistance in T2DM and linked to increased coronary artery disease risk. This shift in the understanding of the pathophysiology of diabetes from being a single hormone deficiency to a dual hormone disorder warrants a careful consideration of the role of C-peptide as a unique molecule with promising diagnostic, prognostic, and therapeutic applications.


Subject(s)
C-Peptide , Humans , C-Peptide/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Signal Transduction
9.
Diabetes ; 73(6): 823-833, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38349844

ABSTRACT

Type 1 diabetes is a chronic autoimmune disease in which destruction of pancreatic ß-cells causes life-threatening metabolic dysregulation. Numerous approaches are envisioned for new therapies, but limitations of current clinical outcome measures are significant disincentives to development efforts. C-peptide, a direct byproduct of proinsulin processing, is a quantitative biomarker of ß-cell function that is not cleared by the liver and can be measured in the peripheral blood. Studies of quantitative measures of ß-cell function have established a predictive relationship between stimulated C-peptide as a measure of ß-cell function and clinical benefits. C-peptide levels at diagnosis are often high enough to afford glycemic control benefits associated with protection from end-organ complications of diabetes, and even lower levels offer protection from severe hypoglycemia in type 1 diabetes, as observed in large prospective cohort studies and interventional trials of islet transplantation. These observations support consideration of C-peptide not just as a biomarker of ß-cell function but also as a specific, sensitive, feasible, and clinically meaningful outcome defining ß-cell preservation or restoration for clinical trials of disease-modifying therapies. Regulatory acceptance of C-peptide as a validated surrogate for demonstration of efficacy would greatly facilitate development of disease-modifying therapies for type 1 diabetes.


Subject(s)
Biomarkers , C-Peptide , Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Diabetes Mellitus, Type 1/drug therapy , C-Peptide/metabolism , C-Peptide/blood , Humans , Biomarkers/blood , Biomarkers/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Clinical Trials as Topic
10.
Nutr Metab Cardiovasc Dis ; 34(5): 1142-1145, 2024 May.
Article in English | MEDLINE | ID: mdl-38220504

ABSTRACT

BACKGROUND AND AIMS: Bariatric surgery is effective for treating type 2 diabetes (T2D) in patients with obesity, although a significant proportion of these patients do not achieve diabetes remission after the surgery even after significant weight loss and metabolic improvement. C-peptide is a valuable marker of beta cell function and insulin secretion, but renal function must be considered when interpreting measurements in patients with T2D. The study aims to investigate the association of serum levels of C-peptide adjusted for creatinine with diabetes remission and glycemic target achievement after bariatric surgery in patients with obesity and T2D. METHODS AND RESULTS: Prospective data from a cohort of 84 patients with obesity and T2D submitted to Roux-en-Y gastric bypass (RYGB) were collected at baseline and at least a 6-month follow up. A multivariate binomial regression model showed that Ln(C-peptide/creatinine) and age were significantly associated with 6-month T2D remission. The area under the curve for the receiver operating characteristic analysis (AUROC) to predict remission was 0.87, and more accurate than the AUROC based on C-peptide levels alone (0.75). The same model was also able to predict achieving an HbA1c target of 7 % (53 mmol/mol) (AUROC 0.96). CONCLUSION: In conclusion, Ln(C-peptide/creatinine) ratio could be a useful tool in predicting T2D remission and target achievement after RYGB surgery, providing a more accurate reflection of beta cell function in bariatric patients.


Subject(s)
Bariatric Surgery , Diabetes Mellitus, Type 2 , Gastric Bypass , Humans , C-Peptide/metabolism , Creatinine , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/surgery , Diabetes Mellitus, Type 2/complications , Obesity/diagnosis , Obesity/surgery , Obesity/complications , Pilot Projects , Prospective Studies , Remission Induction
11.
Am J Transplant ; 24(1): 20-29, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37659605

ABSTRACT

Strong xenorejection limits the clinical application of porcine islet transplantation in type 1 diabetes. Targeting T cell-mediated rejection is one of the main approaches to improve long-term graft survival. Here we study engraftment and survival of porcine islet cells expressing human programmed cell death ligand-1 (hPD-L1) in a humanized mouse model. Neonatal islet-like clusters (NPICCs) from transgenic hPD-L1 (hPD-L1-Tg) and wild-type (Wt) pigs were transplanted into nonobese diabetic-scid IL2rγnull mice stably reconstituted with human immune cells (hPD-L1 n = 10; Wt n = 6). Primary endpoint was development of normoglycemia during a 16-week observation period after transplantation. Secondary endpoints were porcine C-peptide levels and immune cell infiltration. Animals transplanted with hPD-L1-Tg neonatal islet-like clusters achieved a superior normoglycemic rate (50% versus 0%) and significantly higher plasma C-peptide levels as compared to the Wt group, indicating long-term beta cell function. Intracytoplasmic fluorescence-activated cell sorting analysis and immunohistochemistry revealed significantly decreased frequencies of interferonγ-expressing splenic hCD8-positive T cells and reduced intragraft-infiltrating immune cells. We here demonstrate that expression of hPD-L1 provides strong islet xenograft protection without administration of immunosuppressive drugs. These findings support the hypothesis that hPD-L1 has the capacity to control cellular rejection and therefore represents a very promising transgene candidate for clinical porcine islet xenotransplantation.


Subject(s)
Diabetes Mellitus , Islets of Langerhans Transplantation , Islets of Langerhans , Mice , Animals , Humans , Swine , B7-H1 Antigen/metabolism , C-Peptide/metabolism , Islets of Langerhans/metabolism , Mice, Knockout , Transplantation, Heterologous , Mice, SCID , Graft Rejection/etiology
12.
Tissue Cell ; 86: 102280, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38029457

ABSTRACT

This investigation aimed to establish the promising role of insulin-producing cells (IPCs) growing from bone marrow-mesenchymal stem cells (BM-MSCs) in relieving hyperglycemia induced in rats. BM-MSCs were differentiated into IPCs using three different protocols. The efficiency of BM-MSCs differentiation into IPCs in vitro was confirmed by detecting IPCs specific gene expression (Foxa-2, PDX-1 and Ngn-3) and insulin release assay. The in vivo study design included 3 groups of male Wistar rats; negative control group, diabetic group and IPCs-transfused group (5 ×106 cells of the most functional IPCs/rat). One month after IPCs infusion, serum glucose, insulin, c-peptide and visfatin levels as well as pancreatic glucagon level were quantified. Gene expression analysis of pancreatic Foxa-2 and Sox-17, IGF-1 and FGF-10 was done. Additionally, histological investigation of pancreatic tissue sections was performed. Our data clarified that, the most functional IPCs are those generated from BM-MSCs using differentiation protocol 3 as indicated by the significant up-regulation of Foxa-2, PDX-1 and Ngn-3 gene expression levels. These findings were further emphasized by releasing of a significant amount of insulin in response to glucose load. The transplantation of the IPCs in diabetic rats elicited significant decline in serum glucose, visfatin and pancreatic glucagon levels along with significant rise in serum insulin and c-peptide levels. Moreover, it triggered significant up-regulation in the expression levels of pancreatic Foxa-2, Sox-17, IGF-1 and FGF-10 genes versus the untreated diabetic counterpart. The histopathological examination of pancreatic tissue almost assisted the biochemical and molecular genetic analyses. These results disclose that the cell therapy holds potential to develop a new cure for DM based on the capability of BM-MSCs to generate ß-cell phenotype using specific protocol.


Subject(s)
Diabetes Mellitus, Experimental , Insulin-Secreting Cells , Male , Rats , Animals , Insulin-Like Growth Factor I/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Glucagon/metabolism , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Experimental/metabolism , C-Peptide/metabolism , Rats, Wistar , Insulin/metabolism , Cell Differentiation/genetics , Glucose/metabolism , Cell- and Tissue-Based Therapy , Bone Marrow Cells
13.
J Clin Endocrinol Metab ; 109(2): 413-423, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-37671625

ABSTRACT

BACKGROUND: Macroencapsulated pancreatic endoderm cells (PECs) can reverse diabetes in rodents and preclinical studies revealed that thyroid hormones in vitro and in vivo bias PECs to differentiate into insulin-producing cells. In an ongoing clinical trial, PECs implanted in macroencapsulation devices into patients with type 1 diabetes were safe but yielded heterogeneous outcomes. Though most patients developed meal responsive C-peptide, levels were heterogeneous and explanted grafts had variable numbers of surviving cells with variable distribution of endocrine cells. METHODS: We measured circulating triiodothyronine and thyroxine levels in all patients treated at 1 of the 7 sites of the ongoing clinical trial and determined if thyroid hormone levels were associated with the C-peptide or glucagon levels and cell fate of implanted PECs. RESULTS: Both triiodothyronine and thyroxine levels were significantly associated with the proportion of cells that adopted an insulin-producing fate with a mature phenotype. Thyroid hormone levels were inversely correlated to circulating glucagon levels after implantation, suggesting that thyroid hormones lead PECs to favor an insulin-producing fate over a glucagon-producing fate. In mice, hyperthyroidism led to more rapid maturation of PECs into insulin-producing cells similar in phenotype to PECs in euthyroid mice. CONCLUSION: These data highlight the relevance of thyroid hormones in the context of PEC therapy in patients with type 1 diabetes and suggest that a thyroid hormone adjuvant therapy may optimize cell outcomes in some PEC recipients.


Subject(s)
Diabetes Mellitus, Type 1 , Humans , Mice , Animals , Diabetes Mellitus, Type 1/metabolism , C-Peptide/metabolism , Thyroxine/metabolism , Triiodothyronine/metabolism , Endoderm/metabolism , Endoderm/transplantation , Glucagon/metabolism
14.
J Diabetes Investig ; 15(4): 437-448, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38151917

ABSTRACT

AIMS/INTRODUCTION: Endogenous insulin secretion could be recovered by improving hyperglycemia in patients with type 2 diabetes. This study aimed to investigate the association between short-term recovery of insulin secretion during hospitalization and clinical background or future glycemic control in patients with type 2 diabetes. MATERIALS AND METHODS: A total of 127 patients with type 2 diabetes were included. The recovery of endogenous insulin secretion was determined using the following indices: index A: fasting C-peptide index (CPI) at discharge - fasting CPI on admission; index B: postprandial CPI at discharge - postprandial CPI on admission; and index C: Δ C-peptide immunoreactivity (CPR) (postprandial CPR - fasting CPR) at discharge - ΔCPR on admission. We examined the associations of each index with clinical background and future glycemic control measured by glycosylated hemoglobin and continuous glucose monitoring. RESULTS: Using index A and B, the age was significantly younger, whereas BMI and visceral fat area were significantly higher in the high-recovery group than in the low-recovery group. Changes in glycosylated hemoglobin levels were significantly greater at 6 and 12 months in the high-recovery group in the analysis of index C. The receiver operating characteristic curve analysis identified the index B and index C as indicators to predict glycosylated hemoglobin <7.0% at 6 months after discharge. Furthermore, index C was positively correlated with the time in the target glucose range, and inversely correlated with the standard deviation of glucose at 3 and 12 months after discharge. CONCLUSIONS: Short-term recovery of insulin secretion in response to a meal during hospitalization, evaluated with the index-C, might predict future glycemic control.


Subject(s)
Diabetes Mellitus, Type 2 , Humans , Insulin Secretion , Insulin/metabolism , Glycated Hemoglobin , C-Peptide/metabolism , Blood Glucose/analysis , Blood Glucose Self-Monitoring , Glycemic Control , Glucose , Postprandial Period/physiology
15.
Front Endocrinol (Lausanne) ; 14: 1181744, 2023.
Article in English | MEDLINE | ID: mdl-37916149

ABSTRACT

Objective: To determine changes in incretins, systemic inflammation, intestinal permeability and microbiome modifications 12 months after metabolic RYGB (mRYGB) in patients with type 2 diabetes (T2D) and their relationship with metabolic improvement. Materials and methods: Prospective single-center non-randomized controlled study, including patients with class II-III obesity and T2D undergoing mRYGB. At baseline and one year after surgery we performed body composition measurements, biochemical analysis, a meal tolerance test (MTT) and lipid test (LT) with determination of the area under the curve (AUC) for insulin, C-peptide, GLP-1, GLP-2, and fasting determinations of succinate, zonulin, IL-6 and study of gut microbiota. Results: Thirteen patients aged 52.6 ± 6.5 years, BMI 39.3 ± 1.4 kg/m2, HbA1c 7.62 ± 1.5% were evaluated. After mRYGB, zonulin decreased and an increase in AUC after MTT was observed for GLP-1 (pre 9371 ± 5973 vs post 15788 ± 8021 pM, P<0.05), GLP-2 (pre 732 ± 182 vs post 1190 ± 447 ng/ml, P<0.001) and C- peptide, as well as after LT. Species belonging to Streptococaceae, Akkermansiacea, Rickenellaceae, Sutterellaceae, Enterobacteriaceae, Oscillospiraceae, Veillonellaceae, Enterobacterales_uc, and Fusobacteriaceae families increased after intervention and correlated positively with AUC of GLP-1 and GLP-2, and negatively with glucose, HbA1c, triglycerides and adiposity markers. Clostridium perfringens and Roseburia sp. 40_7 behaved similarly. In contrast, some species belonging to Lachnospiraceae, Erysipelotricaceae, and Rumnicocaceae families decreased and showed opposite correlations. Higher initial C-peptide was the only predictor for T2D remission, which was achieved in 69% of patients. Conclusions: Patients with obesity and T2D submitted to mRYGB show an enhanced incretin response, a reduced gut permeability and a metabolic improvement, associated with a specific microbiota signature.


Subject(s)
Diabetes Mellitus, Type 2 , Gastric Bypass , Gastrointestinal Microbiome , Humans , Glucagon-Like Peptide 1/metabolism , Diabetes Mellitus, Type 2/complications , C-Peptide/metabolism , Prospective Studies , Obesity/metabolism , Incretins/metabolism , Glucagon-Like Peptide 2
16.
Cell Rep Methods ; 3(10): 100602, 2023 Oct 23.
Article in English | MEDLINE | ID: mdl-37820726

ABSTRACT

First-phase glucose-stimulated insulin secretion is mechanistically linked to type 2 diabetes, yet the underlying metabolism is difficult to discern due to significant islet-to-islet variability. Here, we miniaturize a fluorescence anisotropy immunoassay onto a microfluidic device to measure C-peptide secretion from individual islets as a surrogate for insulin (InsC-chip). This method measures secretion from up to four islets at a time with ∼7 s resolution while providing an optical window for real-time live-cell imaging. Using the InsC-chip, we reveal two glucose-dependent peaks of insulin secretion (i.e., a double peak) within the classically defined 1st phase (<10 min). By combining real-time secretion and live-cell imaging, we show islets transition from glycolytic to oxidative phosphorylation (OxPhos)-driven metabolism at the nadir of the peaks. Overall, these data validate the InsC-chip to measure glucose-stimulated insulin secretion while revealing new dynamics in secretion defined by a shift in glucose metabolism.


Subject(s)
Diabetes Mellitus, Type 2 , Islets of Langerhans , Humans , Islets of Langerhans/metabolism , Diabetes Mellitus, Type 2/metabolism , C-Peptide/metabolism , Glucose/pharmacology , Lab-On-A-Chip Devices
17.
Int J Mol Sci ; 24(20)2023 Oct 21.
Article in English | MEDLINE | ID: mdl-37895113

ABSTRACT

This study aimed to investigate the effects of C-peptide on C2C12 myotubes and a mouse model. Both in vitro and in vivo experiments were conducted to elucidate the role of C-peptide in muscle atrophy. Various concentrations (0, 0.01, 0.1, 1, 10, and 100 nM) of C-peptide were used on the differentiated C2C12 myotubes with or without dexamethasone (DEX). C57BL/6J mice were administered with C-peptide and DEX for 8 days, followed by C-peptide treatment for 12 days. Compared to the DEX group, C-peptide increased the fusion and differentiation indices and suppressed atrophic factor expression in C2C12 myotubes. However, 100 nM C-peptide decreased the fusion and differentiation indices and increased atrophic factor expression regardless of DEX treatment. In C57BL/6J mice, DEX + C-peptide co-treatment significantly attenuated the body and muscle weight loss and improved the grip strength and cross-sectional area of the gastrocnemius (Gas) and quadriceps (Quad) muscles. C-peptide downregulated the mRNA and protein levels of muscle degradation-related markers, particularly Atrogin-1, in Gas and Quad muscles. This study underscores the potential of C-peptides in mitigating muscle weight reduction and preserving muscle function during muscle atrophy via molecular regulation. In addition, the work presents basic data for future studies on the effect of C-peptide on diabetic muscular dystrophy.


Subject(s)
Dexamethasone , Muscular Atrophy , Mice , Animals , C-Peptide/metabolism , Dexamethasone/therapeutic use , Mice, Inbred C57BL , Muscular Atrophy/metabolism , Muscle, Skeletal/metabolism , Muscle Fibers, Skeletal/metabolism
18.
Sci Rep ; 13(1): 11434, 2023 07 15.
Article in English | MEDLINE | ID: mdl-37454238

ABSTRACT

Acetylcholinesterase (AChE) is a highly conserved enzyme responsible for the regulation of acetylcholine signaling within the brain and periphery. AChE has also been shown to participate in non-enzymatic activity and contribute to cellular development and aging. In particular, enzymatic cleavage of the synaptic AChE isoform, AChE-T, is shown to generate a bioactive T30 peptide that binds to the ⍺7 nicotinic acetylcholine receptor (nAChR) at synapses. Here, we explore intracellular mechanisms of T30 signaling within the human cholinergic neural cell line SH-SY5Y using high performance liquid chromatography (HPLC) coupled to electrospray ionization mass spectrometry (ESI-MS/MS). Proteomic analysis of cells exposed to (100 nM) T30 for 3-days reveals significant changes within proteins important for cell growth. Specifically, bioinformatic analysis identifies proteins that converge onto the mammalian target of rapamycin (mTOR) pathway signaling. Functional experiments confirm that T30 regulates neural cell growth via mTOR signaling and ⍺7 nAChR activation. T30 was found promote mTORC1 pro-growth signaling through an increase in phosphorylated elF4E and S6K1, and a decrease in the autophagy LC3B-II protein. These findings are corroborated in hippocampal neurons and show that T30 promotes dendritic arborization. Taken together, our findings define mTOR as a novel pathway activated by T30 interaction with the nAChR and suggest a role for this process in human disease.


Subject(s)
Neuroblastoma , Receptors, Nicotinic , Humans , Receptors, Nicotinic/metabolism , Acetylcholinesterase/metabolism , Proteomics , Tandem Mass Spectrometry , Peptides/metabolism , TOR Serine-Threonine Kinases/metabolism , C-Peptide/metabolism
19.
Hum Mol Genet ; 32(19): 2929-2939, 2023 09 16.
Article in English | MEDLINE | ID: mdl-37498167

ABSTRACT

Human disease-associated genetic variations often map to long non-coding RNA (lncRNA) genes; however, elucidation of their functional impact is challenging. We previously identified a new genetic variant rs4454083 (A/G) residing in exon of an uncharacterized lncRNA ARBAG that strongly associates with plasma levels of C-peptide, a hormone that regulates insulin bioavailability. On the opposite strand, rs4454083 also corresponds to an intron of a cerebellum-specific GABA receptor subunit gene GABRA6 that mediates strengthening of inhibitory synapses by insulin. Here, we show that alleles of rs4454083 modulate transcript levels of the antisense gene, ARBAG, which then controls the expression of the sense gene, GABRA6. Predisposing to low C-peptide, GG (a minor allele genotype across ethnicities) stabilizes ARBAG lncRNA causing higher transcript levels in cerebellum. ARBAG lncRNA abundance leads to cleavage of GABRA6 mRNA at the complementary region, resulting in a dysfunctional GABRA6 protein that would not be recruited for synapse strengthening. Together, our findings in human cerebellar cell-line and induced Pluripotent Stem Cells (iPSCs) demonstrate biological role of a novel lncRNA in determining the ratio of mRNA isoforms of a protein-coding gene and the ability of an embedded variant in modulating lncRNA stability leading to inter-individual differences in protein expression.


Subject(s)
RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , C-Peptide/genetics , C-Peptide/metabolism , Genome-Wide Association Study , Cerebellum/metabolism , RNA, Antisense/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism
20.
Front Endocrinol (Lausanne) ; 14: 1175640, 2023.
Article in English | MEDLINE | ID: mdl-37409229

ABSTRACT

Aim: In a recent randomized, multicenter trial (NCT02814838) a short-term anti-inflammatory treatment with ladarixin (LDX; an inhibitor of the CXCR1/2 chemokine receptors) did not show benefit on preserving residual beta cell function in new-onset type 1 diabetes. We present a post hoc analysis of trial patients in the predefined subgroup analysis developed according to baseline daily insulin requirement (DIR) tertiles. Method: A double-blind, randomized (2:1), placebo-controlled study was conducted in 45 men and 31 women (aged 18-46 years) within 100 days of the first insulin administration. Patients received LDX (400 mg twice daily) for three cycles of 14 days on/14 days off, or placebo. The primary endpoint was the area under the curve for C-peptide [AUC (0-120 min)] in response to a 2-h mixed meal tolerance test (MMTT) at week 13 ± 1. Seventy-five patients completed the week 13 MMTT and were divided into three groups according to the DIR tertiles: lower, ≤ 0.23U/kg/die (n = 25); middle, 0.24-0.40 U/kg/die (n = 24); upper, ≥ 0.41 U/kg/die (n = 26). Results: When considering the patients in the upper tertile (HIGH-DIR), C-peptide AUC (0-120 min) at 13 weeks was higher in the LDX group (n = 16) than in the placebo (n = 10) group [difference: 0.72 nmol/L (95% CI 0.9-1.34), p = 0.027]. This difference reduced over time (0.71 nmol/L at 26 weeks, p = 0.04; 0.42 nmol/L at 52 weeks, p = 0.29), while it has never been significant at any time in patients in the lower and/or middle tertile (LOW-DIR). We characterized at baseline the HIGH-DIR and found that endo-metabolic (HOMA-B, adiponectin, and glucagon-to-C-peptide ratio) and immunologic (chemokine (C-C motif) ligand 2 (CCL2)/monocyte chemoattractant protein 1 (MCP1) and Vascular Endothelial Growth Factor (VEGF)) features distinguished this group from LOW-DIR. Conclusion: While LDX did not prevent the progressive loss of beta-cell function in the majority of treated subjects, the post hoc analysis suggests that it could work in subjects with HIGH-DIR at baseline. As we found differences in endo-metabolic and immunologic parameters within this subgroup, this generates the hypothesis that the interactions between host factors and drug action can contribute to its efficacy. Further research is needed to evaluate this hypothesis.


Subject(s)
Diabetes Mellitus, Type 1 , Male , Humans , Female , Diabetes Mellitus, Type 1/drug therapy , C-Peptide/metabolism , Prospective Studies , Vascular Endothelial Growth Factor A , Insulin/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...