Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Sci Rep ; 14(1): 19863, 2024 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-39191834

RESUMO

The significant advances in the differentiation of human pluripotent stem (hPS) cells into pancreatic endocrine cells, including functional ß-cells, have been based on a detailed understanding of the underlying developmental mechanisms. However, the final differentiation steps, leading from endocrine progenitors to mono-hormonal and mature pancreatic endocrine cells, remain to be fully understood and this is reflected in the remaining shortcomings of the hPS cell-derived islet cells (SC-islet cells), which include a lack of ß-cell maturation and variability among different cell lines. Additional signals and modifications of the final differentiation steps will have to be assessed in a combinatorial manner to address the remaining issues and appropriate reporter lines would be useful in this undertaking. Here we report the generation and functional validation of hPS cell reporter lines that can monitor the generation of INS+ and GCG+ cells and their resolution into mono-hormonal cells (INSeGFP, INSeGFP/GCGmCHERRY) as well as ß-cell maturation (INSeGFP/MAFAmCHERRY) and function (INSGCaMP6). The reporter hPS cell lines maintained strong and widespread expression of pluripotency markers and differentiated efficiently into definitive endoderm and pancreatic progenitor (PP) cells. PP cells from all lines differentiated efficiently into islet cell clusters that robustly expressed the corresponding reporters and contained glucose-responsive, insulin-producing cells. To demonstrate the applicability of these hPS cell reporter lines in a high-content live imaging approach for the identification of optimal differentiation conditions, we adapted our differentiation procedure to generate SC-islet clusters in microwells. This allowed the live confocal imaging of multiple SC-islets for a single condition and, using this approach, we found that the use of the N21 supplement in the last stage of the differentiation increased the number of monohormonal ß-cells without affecting the number of α-cells in the SC-islets. The hPS cell reporter lines and the high-content live imaging approach described here will enable the efficient assessment of multiple conditions for the optimal differentiation and maturation of SC-islets.


Assuntos
Diferenciação Celular , Genes Reporter , Células Secretoras de Insulina , Ilhotas Pancreáticas , Células-Tronco Pluripotentes , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Linhagem Celular , Insulina/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Fluorescência Verde/genética
2.
J Pathol ; 263(4-5): 429-441, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38837231

RESUMO

The Ppy gene encodes pancreatic polypeptide (PP) secreted by PP- or γ-cells, which are a subtype of endocrine cells localised mainly in the islet periphery. For a detailed characterisation of PP cells, we aimed to establish PP cell lines. To this end, we generated a mouse model harbouring the SV40 large T antigen (TAg) in the Rosa26 locus, which is expressed upon Ppy-promoter-mediated Cre-loxP recombination. Whereas Insulin1-CreERT-mediated TAg expression in beta cells resulted in insulinoma, surprisingly, Ppy-Cre-mediated TAg expression resulted in the malignant transformation of Ppy-lineage cells. These mice showed distorted islet structural integrity at 5 days of age compared with normal islets. CK19+ duct-like lesions contiguous with the islets were observed at 2 weeks of age, and mice developed aggressive pancreatic ductal adenocarcinoma (PDAC) at 4 weeks of age, suggesting that PDAC can originate from the islet/endocrine pancreas. This was unexpected as PDAC is believed to originate from the exocrine pancreas. RNA-sequencing analysis of Ppy-lineage islet cells from 7-day-old TAg+ mice showed a downregulation and an upregulation of endocrine and exocrine genes, respectively, in addition to the upregulation of genes and pathways associated with PDAC. These results suggest that the expression of an oncogene in Ppy-lineage cells induces a switch from endocrine cell fate to PDAC. Our findings demonstrate that Ppy-lineage cells may be an origin of PDAC and may provide novel insights into the pathogenesis of pancreatic cancer, as well as possible therapeutic strategies. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Assuntos
Carcinoma Ductal Pancreático , Linhagem da Célula , Neoplasias Pancreáticas , Animais , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Camundongos , Camundongos Transgênicos , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Transformação Celular Neoplásica/metabolismo , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/metabolismo , Antígenos Transformantes de Poliomavirus/genética , Antígenos Transformantes de Poliomavirus/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos
3.
Physiol Rep ; 12(9): e16040, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38725080

RESUMO

The endocrine pancreas is composed of clusters of cell groups called pancreatic islets. These cells are responsible for the synthesis and secretion of hormones crucial for glycemic homeostasis, such as insulin and glucagon. Therefore, these cells were the targets of many studies. One method to study and/or understand endocrine pancreatic physiology is the isolation of these islets and stimulation of hormone production using different concentrations of glucose, agonists, and/or antagonists of specific secretagogues and mimicking the stimulation of hormonal synthesis and secretion. Many researchers studied pancreatic physiology in murine models due to their ease of maintenance and rapid development. However, the isolation of pancreatic islets involves meticulous processes that may vary between rodent species. The present study describes a simple and effective technical protocol for isolating intact islets from mice and rats for use as a practical guide for researchers. The method involves digestion of the acinar parenchyma by intraductal collagenase. Isolated islets are suitable for in vitro endocrine secretion analyses, microscopy techniques, and biochemical analyses.


Assuntos
Ilhotas Pancreáticas , Animais , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/citologia , Camundongos , Ratos , Masculino , Camundongos Endogâmicos C57BL , Separação Celular/métodos
4.
Biomedicines ; 12(2)2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38397919

RESUMO

Posttransplant diabetes mellitus (PTDM) is a common complication after kidney transplantation. Pathophysiologically, whether beta-cell dysfunction rather than insulin resistance may be the predominant defect in PTDM has been a matter of debate. The aim of the present analysis was to compare glucometabolism in kidney transplant recipients with and without PTDM. To this aim, we included 191 patients from a randomized controlled trial who underwent oral glucose tolerance tests (OGTTs) 6 months after transplantation. We derived several basic indices of beta-cell function and insulin resistance as well as variables from mathematical modeling for a more robust beta-cell function assessment. Mean ± standard deviation of the insulin sensitivity parameter PREDIM was 3.65 ± 1.68 in PTDM versus 5.46 ± 2.57 in NON-PTDM. Model-based glucose sensitivity (indicator of beta-cell function) was 68.44 ± 57.82 pmol∙min-1∙m-2∙mM-1 in PTDM versus 143.73 ± 112.91 pmol∙min-1∙m-2∙mM-1 in NON-PTDM, respectively. Both basic indices and model-based parameters of beta-cell function were more than 50% lower in patients with PTDM, indicating severe beta-cell impairment. Nonetheless, some defects in insulin sensitivity were also present, although less marked. We conclude that in PTDM, the prominent defect appears to be beta-cell dysfunction. From a pathophysiological point of view, patients at high risk for developing PTDM may benefit from intensive treatment of hyperglycemia over the insulin secretion axis.

5.
Perfusion ; 39(3): 593-602, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36757374

RESUMO

INTRODUCTION: Glycemic control is essential for improving the prognosis of cardiac surgery, although precise recommendations have not yet been established. Under a constant blood glucose level, the insulin infusion rate correlates with insulin resistance during glycemic control using an artificial pancreas (AP). We conducted this retrospective study to elucidate changes in intraoperative insulin sensitivity as a first step to creating glycemic control guidelines. METHODS: Fifty-five cardiac surgery patients at our hospital who underwent intraoperative glycemic control using an AP were enrolled. Twenty-three patients undergoing surgical procedures requiring cardiac arrest under hypothermic cardiopulmonary bypass (CPB) with minimum rectal temperatures lower than 32°C, 13 patients undergoing surgical procedures requiring cardiac arrest under hypothermic CPB with minimum rectal temperatures of 32°C, eight patients undergoing on-pump beating coronary artery bypass grafting and 11 patients undergoing off-pump coronary artery bypass were assigned to groups A, B, C and D, respectively. We analyzed the time course of changes in the data derived from glycemic control using the AP. RESULTS: Significant time course changes were observed in groups A and B, but not in groups C and D. Insulin resistance was induced after the start of hypothermic CPB in groups A and B, and the induced change was not resolved by the rewarming procedure, remaining sustained until the end of surgery. CONCLUSIONS: Hypothermia is the predominant factor of the induced insulin resistance during cardiac surgery. Thus, careful glycemic management during hypothermic CPB is important. Prospective clinical studies are required to confirm the findings of this study.


Assuntos
Ponte de Artéria Coronária sem Circulação Extracorpórea , Parada Cardíaca , Hipotermia Induzida , Resistência à Insulina , Pâncreas Artificial , Humanos , Estudos Retrospectivos , Estudos Prospectivos , Ponte Cardiopulmonar/métodos
6.
J Nutr Biochem ; 123: 109490, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37865384

RESUMO

Maternal high-fat diet (HFD) is related to an increased risk of glucose metabolism disorders throughout the whole life of offspring. The pancreas is a glucose homeostasis regulator. Accumulating evidence has revealed that maternal HFD affects offspring pancreas structure and function. However, the potential mechanism remains unclear. In this study, the mouse dam was fed with HFD or control diet (CD) during prepregnancy, pregnancy and lactation. The pancreatic insulin secretion function and islet genome methylome of offspring were analyzed. Pancreatic islet specific gene methylation was detected by using MeDIP qPCR. The results showed that body weight, blood glucose after oral glucose loads, fasting serum insulin, and HOMA-IR index values were significantly higher in male 12-week-old offspring from HFD dams than in the offspring from CD dams. Maternal HFD induced insulin secretion defects in male offspring. Compared with that in maternal CD group, methylation of the Abcc8 and Kcnj11 genes was increased in maternal HFD group in male offspring pancreatic islets. Furthermore, the expression levels of Abcc8 and Kcnj11 were downregulated by intrauterine exposure to a maternal HFD. In summary, maternal HFD results in a long-term functional disorder of the pancreas that is involved in insulin secretion-related gene DNA hypermethylation.


Assuntos
Dieta Hiperlipídica , Efeitos Tardios da Exposição Pré-Natal , Gravidez , Feminino , Camundongos , Masculino , Animais , Humanos , Dieta Hiperlipídica/efeitos adversos , Metilação de DNA , Glucose/metabolismo , Expressão Gênica , Pâncreas/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Fenômenos Fisiológicos da Nutrição Materna
7.
Adv Healthc Mater ; : e2302104, 2023 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-37751946

RESUMO

With the increasing demand for low-sugar, low-calorie healthy diets, artificial sweeteners are widely used as substitutes for sugar in the food industry. Therefore, developing models that can better predict the effects of sugar substitutes on the human body is necessary. Here, a new type of endocrine pancreas-on-a-chip is developed based on a microfiber assembly and its stimulation of pancreatic secretion by glucose or sugar substitutes is evaluated. This new endocrine pancreas-on-a-chip is assembled using two components: (1) a cell-loaded hollow methacrylate gelatin (GelMA)/calcium alginate (CaA) composite microfiber prepared by microfluidic spinning to achieve vascular simulation and material transport, and (2) a 3D pancreatic islet culture layer, which also serves as a fiber assembly microchip. Using this established organ chip, the effects of five sweeteners (glucose, erythritol, xylitol, sodium cyclamate, and sucralose) were investigated on pancreatic islet cell viability and insulin and glucagon secretion. The constructed endocrine pancreas-on-a-chip has potential for the safety evaluation of sugar-substituted food additives, which can expand the application of organ chips in the field of food safety and provide a new platform for evaluating various food additives.

8.
J Endocrinol ; 259(1)2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37523232

RESUMO

Since the discovery of glucagon 100 years ago, the hormone and the pancreatic islet alpha cells that produce it have remained enigmatic relative to insulin-producing beta cells. Canonically, alpha cells have been described in the context of glucagon's role in glucose metabolism in liver, with glucose as the primary nutrient signal regulating alpha cell function. However, current data reveal a more holistic model of metabolic signalling, involving glucagon-regulated metabolism of multiple nutrients by the liver and other tissues, including amino acids and lipids, providing reciprocal feedback to regulate glucagon secretion and even alpha cell mass. Here we describe how various nutrients are sensed, transported and metabolised in alpha cells, providing an integrative model for the metabolic regulation of glucagon secretion and action. Importantly, we discuss where these nutrient-sensing pathways intersect to regulate alpha cell function and highlight key areas for future research.


Assuntos
Células Secretoras de Glucagon , Glucagon , Glucagon/metabolismo , Células Secretoras de Glucagon/metabolismo , Glucose/metabolismo , Transdução de Sinais , Fígado/metabolismo , Insulina/metabolismo
9.
Data Brief ; 48: 109213, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37383828

RESUMO

The datasets of this article present the experimental parameters resulting from the assessment of δ-cells in the islet organs of the endocrine pancreas as a potential biomarker of endocrine disruption (ED) mediated by graphene oxide (GO), using Japanese medaka fish as the model. These datasets support the article "Evaluation of pancreatic δ-cells as a potential target site of graphene oxide toxicity in Japanese medaka (Oryzias latipes) fish". GO used in the experiments was either obtained from a commercial source or synthesized in the laboratory by us. GO was sonicated for 5 min in ice temperature before application. The experiments were conducted on reproductively active adult fish maintained as a breeding pair (one male and one female) in 500 ml balanced salt solution (BSS) either by immersion (IMR) in GO (20 mg/L) continuously for 96 h with the refreshing of media once in every 24 h, or by a single intraperitoneal (IP) administration of GO (100 µg/g) to both male and female partners. Control fish were maintained in BSS only (IMR experiment), or nanopure water (vehicle) was injected into the peritoneal cavity (IP experiment). The IP experimental fish were anesthetized in MS-222 (100 mg/L in BSS); the injected volume (0.5 µL/10 mg fish) never exceeds 50 µl/fish. After injection, the injected fish were allowed for recovery in clean BSS and after recovery both partners were transferred to 1 L glass jars with 500 mL BSS. During depuration, the media of the breeders refreshed once every 24 h and the eggs were collected. After 21 days, the survived fish were anaesthetized, and the trunk region was preserved in 4% paraformaldehyde in PBS (20 mM) containing 0.05% Tween 20. The phenotypic sex of adult fish was assessed externally by secondary sex characters (fin features) and internally by gonad (testis and ovary) histology. Once the location of pancreas was determined after HE stains, immunohistochemical technique was applied on next few slides using rabbit derived polyclonal antisomatostatin antibody as primary antibody and a commercial kit for colorimetric determination of δ-cells in the islet organs was used. Images were captured using an Olympus CKX53 inverted microscope with DP22 camera and CellSens software. Using imagej software, a minimum 3 images of principal islets and one image of secondary islets were assessed. The immunoreactivity of δ-cells, due to neuron-like appearance and filopodia like processes, enabled us to separate them from other cell types found in the pancreatic islets of medaka. Based on immunoreactivity, we have classified islet cells into three categories; noncommunicating delta cells (NCDC), communicating cells (CC), and non-delta cells (NDC), and expressed as number of cells (NCDC/CC/NDC)/mm2 of islet organs. The nuclear area (µm2) and the linear length of filopodia of NCDCs were also considered for evaluation. Numerical data were analysed by Kruskal-Wallis test followed by Mann-Whitney's test as post hoc test and presented as means  ±  SEM. Statistically significant differences were considered for p ≤ 0.05.

10.
Cell Tissue Res ; 393(1): 63-81, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37126142

RESUMO

Peroxisomal dysfunction unhinges cellular metabolism by causing the accumulation of toxic metabolic intermediates (e.g. reactive oxygen species, very -chain fatty acids, phytanic acid or eicosanoids) and the depletion of important lipid products (e.g. plasmalogens, polyunsaturated fatty acids), leading to various proinflammatory and devastating pathophysiological conditions like metabolic syndrome and age-related diseases including diabetes. Because the peroxisomal antioxidative marker enzyme catalase is low abundant in Langerhans islet cells, peroxisomes were considered scarcely present in the endocrine pancreas. Recently, studies demonstrated that the peroxisomal metabolism is relevant for pancreatic cell functionality. During the postnatal period, significant changes occur in the cell structure and the metabolism to trigger the final maturation of the pancreas, including cell proliferation, regulation of energy metabolism, and activation of signalling pathways. Our aim in this study was to (i) morphometrically analyse the density of peroxisomes in mouse endocrine versus exocrine pancreas and (ii) investigate how the distribution and the abundance of peroxisomal proteins involved in biogenesis, antioxidative defence and fatty acid metabolism change during pancreatic maturation in the postnatal period. Our results prove that endocrine and exocrine pancreatic cells contain high amounts of peroxisomes with heterogeneous protein content indicating that distinct endocrine and exocrine cell types require a specific set of peroxisomal proteins depending on their individual physiological functions. We further show that significant postnatal changes occur in the peroxisomal compartment of different pancreatic cells that are most probably relevant for the metabolic maturation and differentiation of the pancreas during the development from birth to adulthood.


Assuntos
Pâncreas Exócrino , Peroxissomos , Camundongos , Animais , Peroxissomos/metabolismo , Pâncreas Exócrino/metabolismo , Antioxidantes/metabolismo , Ácidos Graxos/metabolismo , Espécies Reativas de Oxigênio/metabolismo
11.
Eur J Endocrinol ; 188(4): R73-R87, 2023 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-36929837

RESUMO

Successful pancreas or islet transplantation is currently the only cure for type 1 diabetes mellitus. Since the first pancreas transplant in 1966, there have been various refinements of surgical technique along with improved immunosuppressive regimens, resulting in significantly improved outcomes, with contemporary research into graft monitoring and newer biomarkers, potentially lengthening graft survival rates further. Patients with insulin-dependent diabetes mellitus who are eligible for pancreas or islet transplantation represent a select group, the tip of the iceberg for a significant global diabetes disease burden. In the last 50 years, there have been quantum advances in alternative technologies in diabetes therapy, both experimental and translational. Further development and improved access are required to treat the larger proportion of people suffering from diabetes. Emerging stem cell therapy is still experimental whereas alternatives including automated insulin delivery systems and islet cell transplantation are already used in some countries. Whilst automated insulin delivery systems have increased in efficacy, they still do not achieve the near physiological control of blood sugar, which can be achieved by successful pancreas or islet transplantation. This state-of-the-art review provides a summary of pancreas and islet transplantation to its current place in diabetes therapy, along with alternative and future therapies, including the obstacles associated with the dissemination of these new therapies. With the advent of these modern cellular and technological advances, this review addresses the question: are we entering an era where whole organ pancreas transplantation could be replaced entirely by modern technological advances in diabetes therapy?


Assuntos
Diabetes Mellitus Tipo 1 , Insulinas , Transplante das Ilhotas Pancreáticas , Transplante de Pâncreas , Humanos , Transplante de Pâncreas/métodos , Transplante das Ilhotas Pancreáticas/métodos , Diabetes Mellitus Tipo 1/terapia , Imunossupressores
12.
Stem Cell Rev Rep ; 19(4): 1082-1097, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36749553

RESUMO

Recently, we reported that forkhead box A2 (FOXA2) is required for the development of human pancreatic α- and ß-cells. However, whether miRNAs play a role in regulating pancreatic genes during pancreatic development in the absence of FOXA2 expression is largely unknown. Here, we aimed to capture the dysregulated miRNAs and to identify their pancreatic-specific gene targets in pancreatic progenitors (PPs) derived from wild-type induced pluripotent stem cells (WT-iPSCs) and from iPSCs lacking FOXA2 (FOXA2-/-iPSCs). To identify differentially expressed miRNAs (DEmiRs), and genes (DEGs), two different FOXA2-/-iPSC lines were differentiated into PPs. FOXA2-/- PPs showed a significant reduction in the expression of the main PP transcription factors (TFs) in comparison to WT-PPs. RNA sequencing analysis demonstrated significant reduction in the mRNA expression of genes involved in the development and function of exocrine and endocrine pancreas. Furthermore, miRNA profiling identified 107 downregulated and 111 upregulated DEmiRs in FOXA2-/- PPs compared to WT-PPs. Target prediction analysis between DEmiRs and DEGs identified 92 upregulated miRNAs, predicted to target 1498 downregulated genes in FOXA2-/- PPs. Several important pancreatic TFs essential for pancreatic development were targeted by multiple DEmiRs. Selected DEmiRs and DEGs were further validated using RT-qPCR. Our findings revealed that FOXA2 expression is crucial for pancreatic development through regulating the expression of pancreatic endocrine and exocrine genes targeted by a set of miRNAs at the pancreatic progenitor stage. These data provide novel insights of the effect of FOXA2 deficiency on miRNA-mRNA regulatory networks controlling pancreatic development and differentiation.


Assuntos
Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Fator 3-beta Nuclear de Hepatócito , Células-Tronco Pluripotentes Induzidas , Ilhotas Pancreáticas , MicroRNAs , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/fisiologia , MicroRNAs/genética , Humanos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/crescimento & desenvolvimento , Ilhotas Pancreáticas/metabolismo , Diferenciação Celular/genética , Linhagem Celular
13.
Expert Rev Endocrinol Metab ; 18(1): 19-32, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36692892

RESUMO

INTRODUCTION: Diabetes of the exocrine pancreas (DEP; a.k.a. pancreatic diabetes or pancreatogenic diabetes or type 3c diabetes mellitus or T3cDM) refers to different diabetes types resulting from disorders of the exocrine pancreas. DEP is characterized by the structural and functional loss of glucose-normalizing insulin secretion in the context of exocrine pancreatic dysfunction. Among these forms, new-onset diabetes mellitus secondary to total pancreatectomy (TP) has unique pathophysiological and clinical features, for which we propose a new nomenclature such as post-total pancreatectomy diabetes mellitus (PTPDM). AREAS COVERED: TP results in the complete loss of pancreatic parenchyma, with subsequent absolute insulinopenia and lifelong need for exogenous insulin therapy. Patients with PTPDM also exhibit deficiency of glucagon, amylin and pancreatic polypeptide. These endocrine abnormalities, coupled with increased peripheral insulin sensitivity, deficiency of pancreatic enzymes and TP-related modifications of gastrointestinal anatomy, can lead to marked glucose variability and increased risk of iatrogenic (insulin-induced) severe hypoglycemic episodes ('brittle diabetes'). EXPERT OPINION: We believe that diabetes mellitus secondary to TP should not be included in the DEP spectrum in light of its peculiar pathophysiological and clinical features. Therefore, we propose a new classification for this entity, that would likely provide more accurate prognosis and treatment strategies.


Assuntos
Diabetes Mellitus , Pâncreas Exócrino , Humanos , Pancreatectomia/efeitos adversos , Pancreatectomia/métodos , Pâncreas , Insulina/uso terapêutico , Glucose , Diabetes Mellitus/terapia
14.
Int J Mol Sci ; 24(2)2023 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-36674599

RESUMO

Obesogenic endocrine-disrupting chemicals (EDCs) belong to the group of environmental contaminants, which can adversely affect human health. A growing body of evidence supports that chronic exposure to EDCs can contribute to a rapid increase in obesity among adults and children, especially in wealthy industrialized countries with a high production of widely used industrial chemicals such as plasticizers (bisphenols and phthalates), parabens, flame retardants, and pesticides. The main source of human exposure to obesogenic EDCs is through diet, particularly with the consumption of contaminated food such as meat, fish, fruit, vegetables, milk, and dairy products. EDCs can promote obesity by stimulating adipo- and lipogenesis of target cells such as adipocytes and hepatocytes, disrupting glucose metabolism and insulin secretion, and impacting hormonal appetite/satiety regulation. In vitro models still play an essential role in investigating potential environmental obesogens. The review aimed to provide information on currently available two-dimensional (2D) in vitro animal and human cell models applied for studying the mechanisms of obesogenic action of various industrial chemicals such as food contaminants. The advantages and limitations of in vitro models representing the crucial endocrine tissue (adipose tissue) and organs (liver and pancreas) involved in the etiology of obesity and metabolic diseases, which are applied to evaluate the effects of obesogenic EDCs and their disruption activity, were thoroughly and critically discussed.


Assuntos
Disruptores Endócrinos , Criança , Animais , Humanos , Disruptores Endócrinos/farmacologia , Tecido Adiposo/metabolismo , Adipócitos , Obesidade/induzido quimicamente , Obesidade/metabolismo , Leite
15.
Transpl Int ; 35: 10555, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36090775

RESUMO

Intrahepatic islet transplantation is a promising ß-cell replacement strategy for the treatment of type 1 diabetes. Instant blood-mediated inflammatory reactions, acute inflammatory storm, and graft revascularization delay limit islet engraftment in the peri-transplant phase, hampering the success rate of the procedure. Growing evidence has demonstrated that islet engraftment efficiency may take advantage of several bioengineering approaches aimed to recreate both vascular and endocrine compartments either ex vivo or in vivo. To this end, endocrine pancreas bioengineering is an emerging field in ß-cell replacement, which might provide endocrine cells with all the building blocks (vascularization, ECM composition, or micro/macro-architecture) useful for their successful engraftment and function in vivo. Studies on reshaping either the endocrine cellular composition or the islet microenvironment have been largely performed, focusing on a single building block element, without, however, grasping that their synergistic effect is indispensable for correct endocrine function. Herein, the review focuses on the minimum building blocks that an ideal vascularized endocrine scaffold should have to resemble the endocrine niche architecture, composition, and function to foster functional connections between the vascular and endocrine compartments. Additionally, this review highlights the possibility of designing bioengineered scaffolds integrating alternative endocrine sources to overcome donor organ shortages and the possibility of combining novel immune-preserving strategies for long-term graft function.


Assuntos
Insulinas , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Bioengenharia , Matriz Extracelular , Humanos , Ilhotas Pancreáticas/irrigação sanguínea , Transplante das Ilhotas Pancreáticas/métodos
16.
FASEB J ; 36(10): e22574, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36165227

RESUMO

In this study, the caprine pancreas has been presented as an alternative to the porcine organ for pancreatic xenotransplantation with lesser risk factors. The obtained caprine pancreas underwent a systematic cycle of detergent perfusion for decellularization. It was perfused using anionic (0.5% w/v sodium dodecyl sulfate) as well as non-ionic (0.1% v/v triton X-100, t-octyl phenoxy polyethoxy ethanol) detergents and washed intermittently with 1XPBS supplemented with 0.1% v/v antibiotic and nucleases in a gravitation-driven set-up. After 48 h, a white decellularized pancreas was obtained, and its extracellular matrix (ECM) content was examined for scaffold-like properties. The ECM content was assessed for removal of cellular content, and nuclear material was evaluated with temporal H&E staining. Quantified DNA was found to be present in a negligible amount in the resultant decellularized pancreas tissue (DPT), thus prohibiting it from triggering any immunogenicity. Collagen and fibronectin were confirmed to be preserved upon trichrome and immunohistochemical staining, respectively. SEM and AFM images reveal interconnected collagen fibril networks in the DPT, confirming that collagen was unaffected. sGAG was visualized using Prussian blue staining and quantified with DMMB assay, where DPT has effectively retained this ECM component. Uniaxial tensile analysis revealed that DPT possesses better elasticity than NPT (native pancreatic tissue). Physical parameters like tensile strength, stiffness, biodegradation, and swelling index were retained in the DPT with negligible loss. The cytocompatibility analysis of DPT has shown no cytotoxic effect for up to 72 h on normal insulin-producing cells (MIN-6) and cancerous glioblastoma (LN229) cells in vitro. The scaffold was recellularized using isolated mouse islets, which have established in vitro cell proliferation for up to 9 days. The scaffold received at the end of the decellularization cycle was found to be non-toxic to the cells, retained biological and physical properties of the native ECM, suitable for recellularization, and can be used as a safer and better alternative as a transplantable organ from a xenogeneic source.


Assuntos
Detergentes , Insulinas , Animais , Antibacterianos/farmacologia , Colágeno/metabolismo , DNA/metabolismo , Matriz Extracelular Descelularizada , Detergentes/química , Detergentes/metabolismo , Detergentes/farmacologia , Etanol/farmacologia , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Cabras , Insulinas/análise , Insulinas/metabolismo , Insulinas/farmacologia , Camundongos , Octoxinol/análise , Octoxinol/metabolismo , Octoxinol/farmacologia , Pâncreas , Estudos Prospectivos , Dodecilsulfato de Sódio/análise , Dodecilsulfato de Sódio/metabolismo , Dodecilsulfato de Sódio/farmacologia , Suínos , Engenharia Tecidual/métodos , Alicerces Teciduais/química
17.
Front Endocrinol (Lausanne) ; 13: 981723, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36147573

RESUMO

Although important for digestion and metabolism in repose, the healthy endocrine pancreas also plays a key role in facilitating energy transduction around physical exercise. During exercise, decrements in pancreatic ß-cell mediated insulin release opposed by increments in α-cell glucagon secretion stand chief among the hierarchy of glucose-counterregulatory responses to decreasing plasma glucose levels. As a control hub for several major glucose regulatory hormones, the endogenous pancreas is therefore essential in ensuring glucose homeostasis. Type 1 diabetes (T1D) is pathophysiological condition characterised by a destruction of pancreatic ß-cells resulting in pronounced aberrations in glucose control. Yet beyond the beta-cell perhaps less considered is the impact of T1D on all other pancreatic endocrine cell responses during exercise and whether they differ to those observed in healthy man. For physicians, understanding how the endocrine pancreas responds to exercise in people with and without T1D may serve as a useful model from which to identify whether there are clinically relevant adaptations that need consideration for glycaemic management. From a physiological perspective, delineating differences or indeed similarities in such responses may help inform appropriate exercise test interpretation and subsequent program prescription. With more complex advances in automated insulin delivery (AID) systems and emerging data on exercise algorithms, a timely update is warranted in our understanding of the endogenous endocrine pancreatic responses to physical exercise in people with and without T1D. By placing our focus here, we may be able to offer a nexus of better understanding between the clinical and engineering importance of AIDs requirements during physical exercise.


Assuntos
Diabetes Mellitus Tipo 1 , Células Secretoras de Glucagon , Ilhotas Pancreáticas , Glicemia/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Exercício Físico/fisiologia , Glucagon , Células Secretoras de Glucagon/metabolismo , Glucose/metabolismo , Humanos , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino
18.
Vet Clin North Am Exot Anim Pract ; 25(3): 631-661, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36122944

RESUMO

Endocrine disease in exotic species is less common than in small animals. Nevertheless, the diagnostic principles used in small animals can be adapted to evaluate endocrine disease in many of the exotic species although species-specific aspects need to be considered. This article covers important diseases such as thyroid dysfunction in reptiles and birds, hyperthyroidism in guinea pigs, and hyperadrenocorticism in ferrets. Glucose metabolism in neoplasms affecting normal physiology, such as insulinoma in ferrets and gastric neuroendocrine carcinoma in bearded dragons, is discussed. Calcium abnormalities, including metabolic bone disease in reptiles and hypocalcemia in birds, are also covered.


Assuntos
Animais Exóticos , Doenças do Sistema Endócrino , Animais , Aves , Cálcio , Doenças do Sistema Endócrino/veterinária , Furões , Glucose , Cobaias
19.
Tissue Cell ; 78: 101902, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36007379

RESUMO

Lelio Orci, chairman of the Department of Morphology at the University of Geneva Medical School in Switzerland, was one of the most eminent morphologists of the last century, author of fundamental contributions to the study of microanatomy, especially regarding the ultrastructure of the endocrine pancreas and of the molecular mechanisms of cell secretion. In his work, Orci transformed EM-ultrastructure into a creative art form. The aim of this article is to demonstrate, through a few examples of the scientific work of this extraordinary scientist the reality of this assertion.


Assuntos
Ilhotas Pancreáticas
20.
Cell Reprogram ; 24(5): 252-258, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35838597

RESUMO

One major goal of regenerative medicine is the production of pancreatic endocrine islets to treat insulin-dependent diabetic patients. Among the different methods developed to achieve this goal, a particularly promising approach is direct lineage reprogramming, in which non-ß-cells are directly converted to glucose-responsive, insulin-secreting ß-like cells. Efforts by different research groups have led to critical insights in the inducing factors necessary and types of somatic tissues suitable for direct conversion to ß-like cells. Nevertheless, there is limited understanding of the molecular mechanisms underlying direct cell fate conversion. Significant challenges also remain in translating discoveries into therapeutics that will eventually benefit diabetic patients. This review aims to cover the advances made in the direct reprogramming of somatic cells into ß-like cells and discuss the remaining challenges.


Assuntos
Diabetes Mellitus , Células Secretoras de Insulina , Linhagem da Célula , Reprogramação Celular , Diabetes Mellitus/metabolismo , Glucose , Humanos , Insulina/genética , Células Secretoras de Insulina/metabolismo , Pâncreas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA