166 results on '"human pancreatic islets"'
Search Results
2. Simulating the Loss of -cell Mass in a Human Pancreatic Islet: Structural and Functional Implications
- Author
-
Ruiz-Santiago, Sergio, Godínez-Fernández, José Rafael, Félix-Martínez, Gerardo Jorge, Magjarevic, Ratko, Series Editor, Ładyżyński, Piotr, Associate Editor, Ibrahim, Fatimah, Associate Editor, Lackovic, Igor, Associate Editor, Rock, Emilio Sacristan, Associate Editor, Trujillo-Romero, Citlalli Jessica, editor, Gonzalez-Landaeta, Rafael, editor, Chapa-González, Christian, editor, Dorantes-Méndez, Guadalupe, editor, Flores, Dora-Luz, editor, Flores Cuautle, J. J. Agustin, editor, Ortiz-Posadas, Martha R., editor, Salido Ruiz, Ricardo A., editor, and Zuñiga-Aguilar, Esmeralda, editor
- Published
- 2023
- Full Text
- View/download PDF
3. Human Pancreatic Islets React to Glucolipotoxicity by Secreting Pyruvate and Citrate.
- Author
-
Perrier, Johan, Nawrot, Margaux, Madec, Anne-Marie, Chikh, Karim, Chauvin, Marie-Agnès, Damblon, Christian, Sabatier, Julia, Thivolet, Charles H., Rieusset, Jennifer, Rautureau, Gilles J. P., and Panthu, Baptiste
- Abstract
Progressive decline in pancreatic beta-cell function is central to the pathogenesis of type 2 diabetes (T2D). Here, we explore the relationship between the beta cell and its nutritional environment, asking how an excess of energy substrate leads to altered energy production and subsequent insulin secretion. Alterations in intracellular metabolic homeostasis are key markers of islets with T2D, but changes in cellular metabolite exchanges with their environment remain unknown. We answered this question using nuclear magnetic resonance-based quantitative metabolomics and evaluated the consumption or secretion of 31 extracellular metabolites from healthy and T2D human islets. Islets were also cultured under high levels of glucose and/or palmitate to induce gluco-, lipo-, and glucolipotoxicity. Biochemical analyses revealed drastic alterations in the pyruvate and citrate pathways, which appear to be associated with mitochondrial oxoglutarate dehydrogenase (OGDH) downregulation. We repeated these manipulations on the rat insulinoma-derived beta-pancreatic cell line (INS-1E). Our results highlight an OGDH downregulation with a clear effect on the pyruvate and citrate pathways. However, citrate is directed to lipogenesis in the INS-1E cells instead of being secreted as in human islets. Our results demonstrate the ability of metabolomic approaches performed on culture media to easily discriminate T2D from healthy and functional islets. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
4. Fluorescein-based sensors to purify human α-cells for functional and transcriptomic analyses
- Author
-
Sevim Kahraman, Kimitaka Shibue, Dario F De Jesus, Hyunki Kim, Jiang Hu, Debasish Manna, Bridget Wagner, Amit Choudhary, and Rohit N Kulkarni
- Subjects
α-cells ,pseudoislets ,human pancreatic islets ,diabetes ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Pancreatic α-cells secrete glucagon, an insulin counter-regulatory peptide hormone critical for the maintenance of glucose homeostasis. Investigation of the function of human α-cells remains a challenge due to the lack of cost-effective purification methods to isolate high-quality α-cells from islets. Here, we use the reaction-based probe diacetylated Zinpyr1 (DA-ZP1) to introduce a novel and simple method for enriching live α-cells from dissociated human islet cells with ~95% purity. The α-cells, confirmed by sorting and immunostaining for glucagon, were cultured up to 10 days to form α-pseudoislets. The α-pseudoislets could be maintained in culture without significant loss of viability, and responded to glucose challenge by secreting appropriate levels of glucagon. RNA-sequencing analyses (RNA-seq) revealed that expression levels of key α-cell identity genes were sustained in culture while some of the genes such as DLK1, GSN, SMIM24 were altered in α-pseudoislets in a time-dependent manner. In conclusion, we report a method to sort human primary α-cells with high purity that can be used for downstream analyses such as functional and transcriptional studies.
- Published
- 2023
- Full Text
- View/download PDF
5. Long-term cultures of human pancreatic islets in self-assembling peptides hydrogels
- Author
-
Amanda Marchini, Maria Gessica Ciulla, Barbara Antonioli, Alessandro Agnoli, Umberto Bovio, Virginia Visnoviz, Federico Bertuzzi, and Fabrizio Gelain
- Subjects
human pancreatic islets ,self-assembling peptides ,biomimetic nanomaterial ,three-dimensional cell cultures ,type 1 diabetes mellitus ,Biotechnology ,TP248.13-248.65 - Abstract
Human pancreatic islets transplantation is an experimental therapeutic treatment for Type I Diabetes. Limited islets lifespan in culture remains the main drawback, due to the absence of native extracellular matrix as mechanical support after their enzymatic and mechanical isolation procedure. Extending the limited islets lifespan by creating a long-term in vitro culture remains a challenge. In this study, three biomimetic self-assembling peptides were proposed as potential candidates to recreate in vitro a pancreatic extracellular matrix, with the aim to mechanically and biologically support human pancreatic islets, by creating a three-dimensional culture system. The embedded human islets were analyzed for morphology and functionality in long-term cultures (14-and 28-days), by evaluating β-cells content, endocrine component, and extracellular matrix constituents. The three-dimensional support provided by HYDROSAP scaffold, and cultured into MIAMI medium, displayed a preserved islets functionality, a maintained rounded islets morphology and an invariable islets diameter up to 4 weeks, with results analogues to freshly-isolated islets. In vivo efficacy studies of the in vitro 3D cell culture system are ongoing; however, preliminary data suggest that human pancreatic islets pre-cultured for 2 weeks in HYDROSAP hydrogels and transplanted under subrenal capsule may restore normoglycemia in diabetic mice. Therefore, engineered self-assembling peptide scaffolds may provide a useful platform for long-term maintenance and preservation of functional human pancreatic islets in vitro.
- Published
- 2023
- Full Text
- View/download PDF
6. Sex Differences in the Molecular Programs of Pancreatic Cells Contribute to the Differential Risks of Type 2 Diabetes.
- Author
-
Yong, Hyo Jeong, Toledo, Maria Pilar, Nowakowski, Richard S, and Wang, Yue J
- Abstract
Epidemiology studies demonstrate that women are at a significantly lower risk of developing type 2 diabetes (T2D) compared to men. However, the molecular basis of this risk difference is not well understood. In this study, we examined the sex differences in the genetic programs of pancreatic endocrine cells. We combined pancreas perifusion data and single-cell genomic data from our laboratory and from publicly available data sets to investigate multiple axes of the sex differences in the human pancreas at the single-cell type and single-cell level. We systematically compared female and male islet secretion function, gene expression program, and regulatory principles of pancreatic endocrine cells. The perifusion data indicate that female endocrine cells have a higher secretion capacity than male endocrine cells. Single-cell RNA-sequencing analysis suggests that endocrine cells in male controls have molecular signatures that resemble T2D. In addition, we identified genomic elements associated with genome-wide association study T2D loci to have differential accessibility between female and male delta cells. These genomic elements may play a sex-specific causal role in the pathogenesis of T2D. We provide molecular mechanisms that explain the differential risk of T2D between women and men. Knowledge gained from our study will accelerate the development of diagnostics and therapeutics in sex-aware precision medicine for diabetes. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
7. Enhanced BMP Signaling Alters Human β-Cell Identity and Function.
- Author
-
Dekker E, Triñanes J, Muñoz Garcia A, de Graaf N, de Koning E, and Carlotti F
- Abstract
Inflammation contributes to the pathophysiology of diabetes. Identifying signaling pathways involved in pancreatic β-cell failure and identity loss can give insight into novel potential treatment strategies to prevent the loss of functional β-cell mass in diabetes. It is reported earlier that the immunosuppressive drug tacrolimus has a detrimental effect on human β-cell identity and function by activating bone morphogenetic protein (BMP) signaling. Here it is hypothesized that enhanced BMP signaling plays a role in inflammation-induced β-cell failure. Single-cell transcriptomics analyses of primary human islets reveal that IL-1β+IFNγ and IFNα treatment activated BMP signaling in β-cells. These findings are validated by qPCR. Furthermore, enhanced BMP signaling with recombinant BMP2 or 4 triggers a reduced expression of key β-cell maturity genes, associated with increased ER stress, and impaired β-cell function. Altogether, these results indicate that inflammation-activated BMP signaling is detrimental to pancreatic β-cells and that BMP-signaling can be a target to preserve β-cell identity and function in a pro-inflammatory environment., (© 2024 The Author(s). Advanced Biology published by Wiley‐VCH GmbH.)
- Published
- 2024
- Full Text
- View/download PDF
8. Microwell culture platform maintains viability and mass of human pancreatic islets
- Author
-
Hiroyuki Kato, Tatsuaki Miwa, Janine Quijano, Leonard Medrano, Jose Ortiz, Akiko Desantis, Keiko Omori, Aya Wada, Kentaro Tatsukoshi, Fouad Kandeel, Yoko Mullen, Hsun Teresa Ku, and Hirotake Komatsu
- Subjects
microwells ,long-term culture ,human pancreatic islets ,diabetes ,type 1 diabetes ,islet transplantation ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
BackgroundTransplantation of the human pancreatic islets is a promising approach for specific types of diabetes to improve glycemic control. Although effective, there are several issues that limit the clinical expansion of this treatment, including difficulty in maintaining the quality and quantity of isolated human islets prior to transplantation. During the culture, we frequently observe the multiple islets fusing together into large constructs, in which hypoxia-induced cell damage significantly reduces their viability and mass. In this study, we introduce the microwell platform optimized for the human islets to prevent unsolicited fusion, thus maintaining their viability and mass in long-term cultures.MethodHuman islets are heterogeneous in size; therefore, two different-sized microwells were prepared in a 35 mm-dish format: 140 µm × 300 µm-microwells for 160 µm-islets. Human islets (2,000 islet equivalent) were filtered through a 160 µm-mesh to prepare two size categories for subsequent two week-cultures in each microwell dish. Conventional flat-bottomed 35 mm-dishes were used for non-filtered islets (2,000 islet equivalent/2 dishes). Post-cultured islets are collected to combine in each condition (microwells and flat) for the comparisons in viability, islet mass, morphology, function and metabolism. Islets from three donors were independently tested.ResultsThe microwell platform prevented islet fusion during culture compared to conventional flat bottom dishes, which improved human islet viability and mass. Islet viability and mass on the microwells were well-maintained and comparable to those in pre-culture, while flat bottom dishes significantly reduced islet viability and mass in two weeks. Morphology assessed by histology, insulin-secreting function and metabolism by oxygen consumption did not exhibit the statistical significance among the three different conditions.ConclusionMicrowell-bottomed dishes maintained viability and mass of human islets for two weeks, which is significantly improved when compared to the conventional flat-bottomed dishes.
- Published
- 2022
- Full Text
- View/download PDF
9. Nanovesicles From Lactobacillus johnsonii N6.2 Reduce Apoptosis in Human Beta Cells by Promoting AHR Translocation and IL10 Secretion.
- Author
-
Teixeira, Leandro D., Harrison, Natalie A., da Silva, Danilo R., Mathews, Clayton E., Gonzalez, Claudio F., and Lorca, Graciela L.
- Subjects
PANCREATIC beta cells ,ARYL hydrocarbon receptors ,ISLANDS of Langerhans ,SECRETION ,LACTOBACILLUS - Abstract
L. johnsonii N6.2 releases nano-sized vesicles (NVs) with distinct protein and lipid contents. We hypothesized that these NVs play a central role in the delivery of bioactive molecules that may act as mechanistic effectors in immune modulation. In this report, we observed that addition of NVs to the human pancreatic cell line βlox5 reduced cytokine-induced apoptosis. Through RNAseq analyses, increased expression of CYP1A1, CYP1B1, AHRR , and TIPARP genes in the aryl hydrocarbon receptor (AHR) pathways were found to be significantly induced in presence of NVs. AHR nuclear translocation was confirmed by confocal microscopy. The role of NVs on beta cell function was further evaluated using primary human pancreatic islets. It was found that NVs significantly increased insulin secretion in presence of high glucose concentrations. These increases positively correlated with increased GLUT6 and SREBF1 mRNA and coincided with reduced oxidative stress markers. Furthermore, incubation of NVs with THP-1 macrophages promoted the M2 tolerogenic phenotype through STAT3 activation, expression of AHR-dependent genes and secretion of IL10. Altogether, our findings indicate that bacterial NVs have the potential to modulate glucose homeostasis in the host by directly affecting insulin secretion by islets and through the induction of a tolerogenic immune phenotype. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
10. Nanovesicles From Lactobacillus johnsonii N6.2 Reduce Apoptosis in Human Beta Cells by Promoting AHR Translocation and IL10 Secretion
- Author
-
Leandro D. Teixeira, Natalie A. Harrison, Danilo R. da Silva, Clayton E. Mathews, Claudio F. Gonzalez, and Graciela L. Lorca
- Subjects
AHR ,lactobacillus johnsonii N6.2 ,human beta cell line βlox5 ,human monocyte cell line THP-1 ,human pancreatic islets ,IL10 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
L. johnsonii N6.2 releases nano-sized vesicles (NVs) with distinct protein and lipid contents. We hypothesized that these NVs play a central role in the delivery of bioactive molecules that may act as mechanistic effectors in immune modulation. In this report, we observed that addition of NVs to the human pancreatic cell line βlox5 reduced cytokine-induced apoptosis. Through RNAseq analyses, increased expression of CYP1A1, CYP1B1, AHRR, and TIPARP genes in the aryl hydrocarbon receptor (AHR) pathways were found to be significantly induced in presence of NVs. AHR nuclear translocation was confirmed by confocal microscopy. The role of NVs on beta cell function was further evaluated using primary human pancreatic islets. It was found that NVs significantly increased insulin secretion in presence of high glucose concentrations. These increases positively correlated with increased GLUT6 and SREBF1 mRNA and coincided with reduced oxidative stress markers. Furthermore, incubation of NVs with THP-1 macrophages promoted the M2 tolerogenic phenotype through STAT3 activation, expression of AHR-dependent genes and secretion of IL10. Altogether, our findings indicate that bacterial NVs have the potential to modulate glucose homeostasis in the host by directly affecting insulin secretion by islets and through the induction of a tolerogenic immune phenotype.
- Published
- 2022
- Full Text
- View/download PDF
11. Pancreatic Islet Biobanking Facilities in India: The Need of the Hour to Deal with Diabetes?
- Author
-
Gandasi, Nikhil R., Rangarajan, Annapoorni, Rao, Harshavardhan, Singh, Meetu, and Kothegala, Lakshmi
- Published
- 2023
- Full Text
- View/download PDF
12. Persistent or Transient Human β Cell Dysfunction Induced by Metabolic Stress: Specific Signatures and Shared Gene Expression with Type 2 Diabetes
- Author
-
Lorella Marselli, Anthony Piron, Mara Suleiman, Maikel L. Colli, Xiaoyan Yi, Amna Khamis, Gaelle R. Carrat, Guy A. Rutter, Marco Bugliani, Laura Giusti, Maurizio Ronci, Mark Ibberson, Jean-Valery Turatsinze, Ugo Boggi, Paolo De Simone, Vincenzo De Tata, Miguel Lopes, Daniela Nasteska, Carmela De Luca, Marta Tesi, Emanuele Bosi, Pratibha Singh, Daniela Campani, Anke M. Schulte, Michele Solimena, Peter Hecht, Brian Rady, Ivona Bakaj, Alessandro Pocai, Lisa Norquay, Bernard Thorens, Mickaël Canouil, Philippe Froguel, Decio L. Eizirik, Miriam Cnop, and Piero Marchetti
- Subjects
type 2 diabetes ,lipoglucotoxicity ,glucolipotoxicity ,human pancreatic islets ,beta cells ,damage ,Biology (General) ,QH301-705.5 - Abstract
Summary: Pancreatic β cell failure is key to type 2 diabetes (T2D) onset and progression. Here, we assess whether human β cell dysfunction induced by metabolic stress is reversible, evaluate the molecular pathways underlying persistent or transient damage, and explore the relationships with T2D islet traits. Twenty-six islet preparations are exposed to several lipotoxic/glucotoxic conditions, some of which impair insulin release, depending on stressor type, concentration, and combination. The reversal of dysfunction occurs after washout for some, although not all, of the lipoglucotoxic insults. Islet transcriptomes assessed by RNA sequencing and expression quantitative trait loci (eQTL) analysis identify specific pathways underlying β cell failure and recovery. Comparison of a large number of human T2D islet transcriptomes with those of persistent or reversible β cell lipoglucotoxicity show shared gene expression signatures. The identification of mechanisms associated with human β cell dysfunction and recovery and their overlap with T2D islet traits provide insights into T2D pathogenesis, fostering the development of improved β cell-targeted therapeutic strategies.
- Published
- 2020
- Full Text
- View/download PDF
13. SARS-CoV-2 Receptor Angiotensin I-Converting Enzyme Type 2 (ACE2) Is Expressed in Human Pancreatic β-Cells and in the Human Pancreas Microvasculature
- Author
-
Daniela Fignani, Giada Licata, Noemi Brusco, Laura Nigi, Giuseppina E. Grieco, Lorella Marselli, Lut Overbergh, Conny Gysemans, Maikel L. Colli, Piero Marchetti, Chantal Mathieu, Decio L. Eizirik, Guido Sebastiani, and Francesco Dotta
- Subjects
diabetes ,COVID-19 ,angiotensin I-converting enzyme type 2 (ACE2) ,beta-cell ,human pancreatic islets ,SARS-CoV-2 ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Increasing evidence demonstrated that the expression of Angiotensin I-Converting Enzyme type 2 (ACE2) is a necessary step for SARS-CoV-2 infection permissiveness. In light of the recent data highlighting an association between COVID-19 and diabetes, a detailed analysis aimed at evaluating ACE2 expression pattern distribution in human pancreas is still lacking. Here, we took advantage of INNODIA network EUnPOD biobank collection to thoroughly analyze ACE2, both at mRNA and protein level, in multiple human pancreatic tissues and using several methodologies. Using multiple reagents and antibodies, we showed that ACE2 is expressed in human pancreatic islets, where it is preferentially expressed in subsets of insulin producing β-cells. ACE2 is also highly expressed in pancreas microvasculature pericytes and moderately expressed in rare scattered ductal cells. By using different ACE2 antibodies we showed that a recently described short-ACE2 isoform is also prevalently expressed in human β-cells. Finally, using RT-qPCR, RNA-seq and High-Content imaging screening analysis, we demonstrated that pro-inflammatory cytokines, but not palmitate, increase ACE2 expression in the β-cell line EndoC-βH1 and in primary human pancreatic islets. Taken together, our data indicate a potential link between SARS-CoV-2 and diabetes through putative infection of pancreatic microvasculature and/or ductal cells and/or through direct β-cell virus tropism.
- Published
- 2020
- Full Text
- View/download PDF
14. Single-Cell Transcriptomics Links Loss of Human Pancreatic β-Cell Identity to ER Stress
- Author
-
Nathalie Groen, Floris Leenders, Ahmed Mahfouz, Amadeo Munoz-Garcia, Mauro J. Muraro, Natascha de Graaf, Ton. J. Rabelink, Rob Hoeben, Alexander van Oudenaarden, Arnaud Zaldumbide, Marcel J. T. Reinders, Eelco J. P. de Koning, and Françoise Carlotti
- Subjects
human pancreatic islets ,β-cells ,ER stress ,islet integrity ,single-cell RNAseq ,type 2 diabetes ,Cytology ,QH573-671 - Abstract
The maintenance of pancreatic islet architecture is crucial for proper β-cell function. We previously reported that disruption of human islet integrity could result in altered β-cell identity. Here we combine β-cell lineage tracing and single-cell transcriptomics to investigate the mechanisms underlying this process in primary human islet cells. Using drug-induced ER stress and cytoskeleton modification models, we demonstrate that altering the islet structure triggers an unfolding protein response that causes the downregulation of β-cell maturity genes. Collectively, our findings illustrate the close relationship between endoplasmic reticulum homeostasis and β-cell phenotype, and strengthen the concept of altered β-cell identity as a mechanism underlying the loss of functional β-cell mass.
- Published
- 2021
- Full Text
- View/download PDF
15. SARS-CoV-2 Receptor Angiotensin I-Converting Enzyme Type 2 (ACE2) Is Expressed in Human Pancreatic β -Cells and in the Human Pancreas Microvasculature.
- Author
-
Fignani, Daniela, Licata, Giada, Brusco, Noemi, Nigi, Laura, Grieco, Giuseppina E., Marselli, Lorella, Overbergh, Lut, Gysemans, Conny, Colli, Maikel L., Marchetti, Piero, Mathieu, Chantal, Eizirik, Decio L., Sebastiani, Guido, and Dotta, Francesco
- Subjects
ANGIOTENSIN receptors ,SARS-CoV-2 ,ISLANDS of Langerhans ,VIRAL tropism ,PANCREAS ,LOSARTAN ,ANGIOTENSIN-receptor blockers - Abstract
Increasing evidence demonstrated that the expression of Angiotensin I-Converting Enzyme type 2 (ACE2) is a necessary step for SARS-CoV-2 infection permissiveness. In light of the recent data highlighting an association between COVID-19 and diabetes, a detailed analysis aimed at evaluating ACE2 expression pattern distribution in human pancreas is still lacking. Here, we took advantage of INNODIA network EUnPOD biobank collection to thoroughly analyze ACE2, both at mRNA and protein level, in multiple human pancreatic tissues and using several methodologies. Using multiple reagents and antibodies, we showed that ACE2 is expressed in human pancreatic islets, where it is preferentially expressed in subsets of insulin producing β -cells. ACE2 is also highly expressed in pancreas microvasculature pericytes and moderately expressed in rare scattered ductal cells. By using different ACE2 antibodies we showed that a recently described short-ACE2 isoform is also prevalently expressed in human β -cells. Finally, using RT-qPCR, RNA-seq and High-Content imaging screening analysis, we demonstrated that pro-inflammatory cytokines, but not palmitate, increase ACE2 expression in the β -cell line EndoC- β H1 and in primary human pancreatic islets. Taken together, our data indicate a potential link between SARS-CoV-2 and diabetes through putative infection of pancreatic microvasculature and/or ductal cells and/or through direct β -cell virus tropism. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
16. CYTOPROTECTIVE EFFECTS OF GINSENOSIDE RD ON APOPTOSIS-ASSOCIATED CELL DEATH IN THE ISOLATED HUMAN PANCREATIC ISLETS.
- Author
-
Kaviani, Maryam, Keshtkar, Somayeh, Azarpira, Negar, Aghdaei, Mahdokht Hossein, Geramizadeh, Bita, Karimi, Mohammad Hossein, Yaghobi, Ramin, Esfandiari, Elaheh, Shamsaeefar, Alireza, Nikeghbalian, Saman, and Al-Abdullahd, Ismail H.
- Subjects
- *
ISLANDS of Langerhans , *CELL death , *PROTEIN expression , *GENE expression - Abstract
Ginsenoside Rd (GS-Rd), one of the main pharmacologically active components of ginseng, has shown the potential to stabilize mitochondrial membrane integrity and decrease apoptotic death in neuronal and non-neuronal cells. The present study aimed to evaluate the effect of this bioactive molecule on the apoptosis-associated cell death in human pancreatic islets. In this regard human pancreatic islets were isolated and grouped for the treatment with GS-Rd. The isolated islets were treated with different concentrations of GS-Rd. After 24 and 72 h of incubation, the islets were evaluated in terms of viability, BAX, BCL2, and insulin gene expression, BAX, BCL2, and caspase- 3 protein expression, apoptosis, and glucose-induced insulin/C-peptide secretion. Our results revealed the islet survival was significantly decreased in the control group after 72 h of incubation. However, GS-Rd inhibited the progress of the islet death in the treated groups. TUNEL staining revealed that the preventive effect of this molecule was caused by the inhibition of apoptosis-associated death. In this regard, the activation of caspase-3 was down-regulated in the presence of GS-Rd. GS-Rd did not exhibit undesirable effects on glucose-induced insulin and C-peptide stimulation secretion. In conclusion, GS-Rd inhibited the progress of death of cultured human pancreatic islets by diminishing the apoptosis of the islet cells. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
17. MC1568 improves insulin secretion in islets from type 2 diabetes patients and rescues β-cell dysfunction caused by Hdac7 upregulation.
- Author
-
Daneshpajooh, Mahboubeh, Eliasson, Lena, Bacos, Karl, and Ling, Charlotte
- Subjects
- *
INSULIN , *TYPE 2 diabetes , *HISTONE deacetylase , *MITOCHONDRIA , *APOPTOSIS - Abstract
Aims: It has in recent years been established that epigenetic changes contribute to β-cell dysfunction and type 2 diabetes (T2D). For example, we have showed that the expression of histone deacetylase 7 (HDAC7) is increased in pancreatic islets of individuals with T2D and that increased levels of Hdac7 in β-cells impairs insulin secretion. The HDAC inhibitor MC1568 rescued this secretory impairment, suggesting that inhibitors specific for HDAC7 may be useful clinically in the treatment of T2D. The aim of the current study was to further explore HDAC7 as a novel therapeutic target in T2D.Methods: Hdac7 was overexpressed in clonal β-cells followed by the analysis of insulin secretion, mitochondrial function, as well as cell number and apoptosis in the presence or absence of MC1568. Furthermore, the effect of MC1568 on insulin secretion in human pancreatic islets from non-diabetic donors and donors with T2D was also studied.Results: Overexpression of Hdac7 in clonal β-cells significantly reduced insulin secretion, mitochondrial respiration, and ATP content, while it increased apoptosis. These impairments were all rescued by treatment with MC1568. The inhibitor also increased glucose-stimulated insulin secretion in islets from donors with T2D, while having no effect on islets from non-diabetic donors.Conclusions: HDAC7 inhibition protects β-cells from mitochondrial dysfunction and apoptosis, and increases glucose-stimulated insulin secretion in islets from human T2D donors. Our study supports specific HDAC7 inhibitors as novel options in the treatment of T2D. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
18. A Versatile Model of Microfluidic Perifusion System for the Evaluation of C-Peptide Secretion Profiles: Comparison Between Human Pancreatic Islets and HLSC-Derived Islet-Like Structures
- Author
-
Yonathan Gomez, Victor Navarro-Tableros, Ciro Tetta, Giovanni Camussi, and Maria Felice Brizzi
- Subjects
microfluidic ,perifusion ,c-peptide ,human pancreatic islets ,obese ,t2dm ,hlsc-ils ,Biology (General) ,QH301-705.5 - Abstract
A robust and easy-to-use tool for the ex vivo dynamic evaluation of pancreatic islet (PI) function is essential for further development of novel cell-based therapeutic approaches to treating diabetes. Here, we developed four different glucose perifusion protocols (GPPs) in a microfluidic perifusion system (MPS), based entirely on commercially available components. After validation, the GPPs were used to evaluate C-peptide secretion profiles of PIs derived from different donors (healthy, obese, and type 2 diabetic) and from human liver stem-cell-derived islet-like structures (HLSC-ILS). Using this device, we demonstrated that PIs derived from healthy donors displayed a physiological C-peptide secretion profile as characterized by the response to (a) different glucose concentrations, (b) consecutive pulses of high-glucose concentrations, (c) a glucose threshold ranging from 5−8 mM, and (d) a constant high-glucose perifusion in a biphasic manner. Moreover, we were able to detect a dysregulated secretion profile in PIs derived from both obese and type 2 diabetes mellitus (T2DM) donors. Finally, we also evaluated the kinetic secretion profiles of HLSC-ILS, demonstrating that, nonetheless, with a lower amplitude of secretion compared to PI derived from healthy donors, they were already glucose-responsive on day seven post-differentiation. In conclusion, we have provided evidence that our MPS is a versatile device and may represent a valuable tool to study insulin-producing cells in vitro.
- Published
- 2020
- Full Text
- View/download PDF
19. Integration of human pancreatic islet genomic data refines regulatory mechanisms at Type 2 Diabetes susceptibility loci
- Author
-
Matthias Thurner, Martijn van de Bunt, Jason M Torres, Anubha Mahajan, Vibe Nylander, Amanda J Bennett, Kyle J Gaulton, Amy Barrett, Carla Burrows, Christopher G Bell, Robert Lowe, Stephan Beck, Vardhman K Rakyan, Anna L Gloyn, and Mark I McCarthy
- Subjects
Type 2 Diabetes ,human pancreatic islets ,GWAS ,epigenetics ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Human genetic studies have emphasised the dominant contribution of pancreatic islet dysfunction to development of Type 2 Diabetes (T2D). However, limited annotation of the islet epigenome has constrained efforts to define the molecular mechanisms mediating the, largely regulatory, signals revealed by Genome-Wide Association Studies (GWAS). We characterised patterns of chromatin accessibility (ATAC-seq, n = 17) and DNA methylation (whole-genome bisulphite sequencing, n = 10) in human islets, generating high-resolution chromatin state maps through integration with established ChIP-seq marks. We found enrichment of GWAS signals for T2D and fasting glucose was concentrated in subsets of islet enhancers characterised by open chromatin and hypomethylation, with the former annotation predominant. At several loci (including CDC123, ADCY5, KLHDC5) the combination of fine-mapping genetic data and chromatin state enrichment maps, supplemented by allelic imbalance in chromatin accessibility pinpointed likely causal variants. The combination of increasingly-precise genetic and islet epigenomic information accelerates definition of causal mechanisms implicated in T2D pathogenesis.
- Published
- 2018
- Full Text
- View/download PDF
20. Fluorescein-based sensors to purify human α-cells for functional and transcriptomic analyses.
- Author
-
Kahraman S, Shibue K, De Jesus DF, Kim H, Hu J, Manna D, Wagner B, Choudhary A, and Kulkarni RN
- Subjects
- Humans, Glucagon metabolism, Transcriptome, Insulin metabolism, Glucose metabolism, Fluoresceins metabolism, Islets of Langerhans metabolism, Glucagon-Secreting Cells metabolism, Insulin-Secreting Cells metabolism
- Abstract
Pancreatic α-cells secrete glucagon, an insulin counter-regulatory peptide hormone critical for the maintenance of glucose homeostasis. Investigation of the function of human α-cells remains a challenge due to the lack of cost-effective purification methods to isolate high-quality α-cells from islets. Here, we use the reaction-based probe diacetylated Zinpyr1 (DA-ZP1) to introduce a novel and simple method for enriching live α-cells from dissociated human islet cells with ~95% purity. The α-cells, confirmed by sorting and immunostaining for glucagon, were cultured up to 10 days to form α-pseudoislets. The α-pseudoislets could be maintained in culture without significant loss of viability, and responded to glucose challenge by secreting appropriate levels of glucagon. RNA-sequencing analyses (RNA-seq) revealed that expression levels of key α-cell identity genes were sustained in culture while some of the genes such as DLK1 , GSN , SMIM24 were altered in α-pseudoislets in a time-dependent manner. In conclusion, we report a method to sort human primary α-cells with high purity that can be used for downstream analyses such as functional and transcriptional studies., Competing Interests: SK is an employee of Boehringer Ingelheim Pharmaceuticals, Inc, KS, DD, HK, JH, DM, BW, AC No competing interests declared, RK is on the Scientific Advisory Board of Novo Nordisk, Biomea and Inversago Therapeutics, (© 2023, Kahraman et al.)
- Published
- 2023
- Full Text
- View/download PDF
21. MicroRNA Expression Analysis of In Vitro Dedifferentiated Human Pancreatic Islet Cells Reveals the Activation of the Pluripotency-Related MicroRNA Cluster miR-302s.
- Author
-
Sebastiani, Guido, Grieco, Giuseppina Emanuela, Brusco, Noemi, Ventriglia, Giuliana, Formichi, Caterina, Marselli, Lorella, Marchetti, Piero, and Dotta, Francesco
- Subjects
- *
MICRORNA , *CELL differentiation , *CELL populations , *CELL communication , *PHENOTYPES - Abstract
β-cell dedifferentiation has been recently suggested as an additional mechanism contributing to type-1 and to type-2 diabetes pathogenesis. Moreover, several studies demonstrated that in vitro culture of native human pancreatic islets derived from non-diabetic donors resulted in the generation of an undifferentiated cell population. Additional evidence from in vitro human β-cell lineage tracing experiments, demonstrated that dedifferentiated cells derive from β-cells, thus representing a potential in vitro model of β-cell dedifferentiation. Here, we report the microRNA expression profiles analysis of in vitro dedifferentiated islet cells in comparison to mature human native pancreatic islets. We identified 13 microRNAs upregulated and 110 downregulated in islet cells upon in vitro dedifferentiation. Interestingly, among upregulated microRNAs, we observed the activation of microRNA miR-302s cluster, previously defined as pluripotency-associated. Bioinformatic analysis indicated that miR-302s are predicted to target several genes involved in the control of β-cell/epithelial phenotype maintenance; accordingly, such genes were downregulated upon human islet in vitro dedifferentiation. Moreover, we uncovered that cell-cell contacts are needed to maintain low/null expression levels of miR-302. In conclusion, we showed that miR-302 microRNA cluster genes are involved in in vitro dedifferentiation of human pancreatic islet cells and inhibits the expression of multiple genes involved in the maintenance of β-cell mature phenotype. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
22. 3D-Models of Insulin-Producing β-Cells: from Primary Islet Cells to Stem Cell-Derived Islets.
- Author
-
Ribeiro, Diana, Kvist, Alexander J., Wittung-Stafshede, Pernilla, Hicks, Ryan, and Forslöw, Anna
- Subjects
- *
PANCREATIC beta cells , *ISLANDS of Langerhans , *DRUG development , *EXTRACELLULAR matrix , *MOLECULAR memory - Abstract
There is a need for physiologically relevant assay platforms to provide functionally relevant models of diabetes, to accelerate the discovery of new treatment options and boost developments in drug discovery. In this review, we compare several 3D-strategies that have been used to increase the functional relevance of ex vivo human primary pancreatic islets and developments into the generation of stem cell derived pancreatic beta-cells (β-cells). Special attention will be given to recent approaches combining the use of extracellular matrix (ECM) scaffolds with pancreatic molecular memory, which can be used to improve yield and functionality of in vitro stem cell-derived pancreatic models. The ultimate goal is to develop scalable cell-based platforms for diabetes research and drug screening. This article will critically assess key aspects related to in vitro pancreatic 3D-ECM models and highlight the most promising approaches for future research. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
23. Ultrastructural alterations of pancreatic beta cells in human diabetes mellitus.
- Author
-
Masini, Matilde, Martino, Luisa, Marselli, Lorella, Bugliani, Marco, Boggi, Ugo, Filipponi, Franco, Marchetti, Piero, and De Tata, Vincenzo
- Subjects
AUTOPSY ,DIABETES ,PANCREATIC beta cells - Abstract
Background: Both types of diabetes are characterized by beta-cell failure and death, leading to insulin insufficiency. Very limited information is currently available about the ultrastructural alterations of beta cells in human diabetes. Our aim was to provide a comprehensive ultrastructural analysis of human pancreatic islets in type 1 (T1D) and type 2 (T2D) diabetic patients.Methods: We performed a morphometric electron microscopy evaluation of beta cells obtained from the pancreas of 8 nondiabetic (ND), 5 T1D, and 8 T2D organ donors.Results: A lower amount of beta cells was found in both T1D and T2D than in ND islets, whereas alpha cells were increased only in T2D. An increased number of bi-hormonal cells (showing both insulin and glucagon granules in their cytoplasm) were found in T1D. Insulin granules were less represented in T2D than in ND beta cells, whereas no significant changes were found in T1D. Volume density of the endoplasmic reticulum was increased in T2D and unchanged in T1D; mitochondria number and volume were significantly higher in T2D than in ND beta cells, whereas no significant differences were found in T1D. In both T1D and T2D, more beta cells showed signs of apoptosis than in ND.Conclusions: Our results show that in each type of diabetes, beta cells exhibit specific ultrastructural alterations, whose better understanding might improve therapeutic strategies. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
24. Effects of bone marrow on the microenvironment of the human pancreatic islet: A Protein Profile Approach.
- Author
-
Kim, Joseph W, Vang, Souriya, Luo, John ZQ, Newton, William C, and Luo, Luguang
- Subjects
- *
BONE marrow , *ISLANDS of Langerhans , *STEM cells , *CARRIER proteins , *ENDOPEPTIDASES , *MASS spectrometry - Abstract
Stem cells are a new therapeutic modality that may support the viability and function of human organs and tissue. Our previous studies have revealed that human allogeneic bone marrow (BM) sustains pancreatic β cell function and survival. This paper examines whether BM creates a microenvironment that supports human pancreatic islets in vitro by evaluating 107 proteins in culture media from BM, islet, and islet/bone marrow (IB) with mass spectrometry. Proteins were considered up- or down-regulated if p-values < 0.05 and fold change was greater than 2 fold I VS. IB. In addition, proteins identified that were uniquely found in islets co-cultured with bone marrow, but not in islets or bone marrow. A 95% protein probability was used as a threshold. Twenty three proteins were upregulated, and sixteen proteins were downregulated. The function of each protein is listed based on the protein database, which include structural proteins (9 upregulated, 4 downregulated); anti-protease and anti-endopeptidase enzymes (8 upregulated); cation binding proteins (6 up-regulated). Six proteins were uniquely identified in islet co-cultured with bone marrow. Three are anti-proteases or anti-endopeptidases, and 1 is a structural protein. These findings suggest that BM, by changing culture media proteins, may be one of mechanisms to maintain human islet function and survival. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
25. Characterization of Acyl-CoA synthetase isoforms in pancreatic beta cells: Gene silencing shows participation of ACSL3 and ACSL4 in insulin secretion.
- Author
-
Ansari, Israr-ul H., Longacre, Melissa J., Stoker, Scott W., Kendrick, Mindy A., O'Neill, Lucas M., Zitur, Laura J., Fernandez, Luis A., Ntambi, James M., and MacDonald, Michael J.
- Subjects
- *
ACYL-CoA synthetase , *PANCREATIC beta cells , *GENE silencing , *INSULIN , *LABORATORY rats - Abstract
Long-chain acyl-CoA synthetases (ACSLs) convert fatty acids to fatty acyl-CoAs to regulate various physiologic processes. We characterized the ACSL isoforms in a cell line of homogeneous rat beta cells (INS-1 832/13 cells) and human pancreatic islets. ACSL4 and ACSL3 proteins were present in the beta cells and human and rat pancreatic islets and concentrated in insulin secretory granules and less in mitochondria and negligible in other intracellular organelles. ACSL1 and ACSL6 proteins were not seen in INS-1 832/13 cells or pancreatic islets. ACSL5 protein was seen only in INS-1 832/13 cells. With shRNA-mediated gene silencing we developed stable ACSL knockdown cell lines from INS-1 832/13 cells. Glucose-stimulated insulin release was inhibited ∼50% with ACSL4 and ACSL3 knockdown and unaffected in cell lines with knockdown of ACSL5, ACLS6 and ACSL1. Lentivirus shRNA-mediated gene silencing of ACSL4 and ACSL3 in human pancreatic islets inhibited glucose-stimulated insulin release. ACSL4 and ACSL3 knockdown cells showed inhibition of ACSL enzyme activity more with arachidonate than with palmitate as a substrate, consistent with their preference for unsaturated fatty acids as substrates. ACSL4 knockdown changed the patterns of fatty acids in phosphatidylserines and phosphatidylethanolamines. The results show the involvement of ACLS4 and ACLS3 in insulin secretion. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
26. Proteomic profiling of human islets collected from frozen pancreata using laser capture microdissection.
- Author
-
Zhang, Lina, Lanzoni, Giacomo, Battarra, Matteo, Inverardi, Luca, and Zhang, Qibin
- Subjects
- *
PROTEOMICS , *MICRODISSECTION , *ETIOLOGY of diabetes , *ISLANDS of Langerhans , *CELL culture - Abstract
The etiology of Type 1 Diabetes (T1D) remains elusive. Enzymatically isolated and cultured (EIC) islets cannot fully reflect the natural protein composition and disease process of in vivo islets, because of the stress from isolation procedures. In order to study islet protein composition in conditions close to the natural environment, we performed proteomic analysis of EIC islets, and laser capture microdissected (LCM) human islets and acinar tissue from fresh-frozen pancreas sections of three cadaveric donors. 1104 and 706 proteins were identified from 6 islets equivalents (IEQ) of LCM islets and acinar tissue, respectively. The proteomic profiles of LCM islets were reproducible within and among cadaveric donors. The endocrine hormones were only detected in LCM islets, whereas catalytic enzymes were significantly enriched in acinar tissue. Furthermore, high overlap (984 proteins) and similar function distribution were found between LCM and EIC islets proteomes, except that EIC islets had more acinar contaminants and stress-related signal transducer activity proteins. The comparison among LCM islets, LCM acinar tissue and EIC islets proteomes indicates that LCM combined with proteomic methods enables accurate and unbiased profiling of islet proteome from frozen pancreata. This paves the way for proteomic studies on human islets during the progression of T1D. Significance The etiological agent triggering autoimmunity against beta cells in Type 1 diabetes (T1D) remains obscure. The in vitro models available (enzymatically isolated and cultured islets, EIC islets) do not accurately reflect what happens in vivo due to lack of the natural environment where islets exist and the preparation-induced changes in cell physiology. The importance of this study is that we investigated the feasibility of laser capture microdissection (LCM) for the isolation of intact islets from frozen cadaveric pancreatic tissue sections. We compared the protein profile of LCM islets (9 replicates from 3 cadaveric donors) with that of both LCM acinar tissues (6 replicates from the same 3 cadaveric donor as LCM islets) and EIC islets (at least 4 replicates for each sample with the same islets equivalents) by using proteomics techniques with advanced instrumentation, nanoLC-Q Exactive HF Orbitrap mass spectrometry (nano LC-MS/MS). The results demonstrate that the LCM method is reliable in isolating islets with an intact environment. LCM-based islet proteomics is a feasible approach to obtain good proteome coverage for assessing the pathology of T1D using cadaveric pancreatic samples, even from very small sample amounts. Future applications of this LCM-based proteomic method may help us understand the pathogenesis of T1D and identify potential biomarkers for T1D diagnosis at an early stage. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
27. HDAC7 is overexpressed in human diabetic islets and impairs insulin secretion in rat islets and clonal beta cells.
- Author
-
Daneshpajooh, Mahboubeh, Bacos, Karl, Bysani, Madhusudhan, Bagge, Annika, Ottosson Laakso, Emilia, Vikman, Petter, Eliasson, Lena, Mulder, Hindrik, and Ling, Charlotte
- Abstract
Aims/hypothesis: Pancreatic beta cell dysfunction is a prerequisite for the development of type 2 diabetes. Histone deacetylases (HDACs) may affect pancreatic endocrine function and glucose homeostasis through alterations in gene regulation. Our aim was to investigate the role of HDAC7 in human and rat pancreatic islets and clonal INS-1 beta cells (INS-1 832/13). Methods: To explore the role of HDAC7 in pancreatic islets and clonal beta cells, we used RNA sequencing, mitochondrial functional analyses, microarray techniques, and HDAC inhibitors MC1568 and trichostatin A. Results: Using RNA sequencing, we found increased HDAC7 expression in human pancreatic islets from type 2 diabetic compared with non-diabetic donors. HDAC7 expression correlated negatively with insulin secretion in human islets. To mimic the situation in type 2 diabetic islets, we overexpressed Hdac7 in rat islets and clonal beta cells. In both, Hdac7 overexpression resulted in impaired glucose-stimulated insulin secretion. Furthermore, it reduced insulin content, mitochondrial respiration and cellular ATP levels in clonal beta cells. Overexpression of Hdac7 also led to changes in the genome-wide gene expression pattern, including increased expression of Tcf7l2 and decreased expression of gene sets regulating DNA replication and repair as well as nucleotide metabolism. In accordance, Hdac7 overexpression reduced the number of beta cells owing to enhanced apoptosis. Finally, we found that inhibiting HDAC7 activity with pharmacological inhibitors or small interfering RNA-mediated knockdown restored glucose-stimulated insulin secretion in beta cells that were overexpressing Hdac7. Conclusions/interpretation: Taken together, these results indicate that increased HDAC7 levels caused beta cell dysfunction and may thereby contribute to defects seen in type 2 diabetic islets. Our study supports HDAC7 inhibitors as a therapeutic option for the treatment of type 2 diabetes. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
28. The Integrated Islet Distribution Program answers the call for improved human islet phenotyping and reporting of human islet characteristics in research articles.
- Author
-
Brissova, Marcela, Niland, Joyce C., Cravens, James, Olack, Barbara, Sowinski, Janice, and Evans-Molina, Carmella
- Published
- 2019
- Full Text
- View/download PDF
29. Fatty acids stimulate insulin secretion from human pancreatic islets at fasting glucose concentrations via mitochondria-dependent and -independent mechanisms.
- Author
-
Jing Cen, Sargsyan, Ernest, and Bergsten, Peter
- Subjects
- *
FATTY acids , *INSULIN , *ISLANDS of Langerhans , *MICROBIOLOGICAL techniques , *MITOCHONDRIA , *RESEARCH funding , *DATA analysis software , *DESCRIPTIVE statistics , *IN vitro studies , *ONE-way analysis of variance - Published
- 2016
- Full Text
- View/download PDF
30. Decreased Expression of Cilia Genes in Pancreatic Islets as a Risk Factor for Type 2 Diabetes in Mice and Humans
- Author
-
Anett Seelig, O Kluth, Charlotte Ling, Ulrika Krus, Annette Schürmann, Heja Aga, Markus Jähnert, Heike Vogel, Jantje M. Gerdes, Stephan Scherneck, Mandy Stadion, and Pascal Gottmann
- Subjects
Male ,0301 basic medicine ,endocrine system ,endocrine system diseases ,Kinesins ,Type 2 diabetes ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,ddc:570 ,Cell Line, Tumor ,Insulin-Secreting Cells ,Ciliogenesis ,medicine ,Animals ,KIF3A ,Cilia ,Gene ,lcsh:QH301-705.5 ,Cells, Cultured ,geography ,geography.geographical_feature_category ,Cilium ,Pancreatic islets ,Cell Cycle ,New Zealand Obese Mouse ,Beta-cell Proliferation ,Cilia Genes ,Human Pancreatic Islets ,Islet Transcriptomics ,Pathway Enrichment Analysis ,Primary Cilia ,Type 2 Diabetes ,Islet ,medicine.disease ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Diabetes Mellitus, Type 2 ,lcsh:Biology (General) ,Institut für Ernährungswissenschaft ,Transcriptome ,030217 neurology & neurosurgery - Abstract
Summary: An insufficient adaptive beta-cell compensation is a hallmark of type 2 diabetes (T2D). Primary cilia function as versatile sensory antennae regulating various cellular processes, but their role on compensatory beta-cell replication has not been examined. Here, we identify a significant enrichment of downregulated, cilia-annotated genes in pancreatic islets of diabetes-prone NZO mice as compared with diabetes-resistant B6-ob/ob mice. Among 327 differentially expressed mouse cilia genes, 81 human orthologs are also affected in islets of diabetic donors. Islets of nondiabetic mice and humans show a substantial overlap of upregulated cilia genes that are linked to cell-cycle progression. The shRNA-mediated suppression of KIF3A, essential for ciliogenesis, impairs division of MIN6 beta cells as well as in dispersed primary mouse and human islet cells, as shown by decreased BrdU incorporation. These findings demonstrate the substantial role of cilia-gene regulation on islet function and T2D risk. : Kluth et al. identify a significant enrichment of cilia-annotated genes that are differentially expressed in pancreatic islets of obese mice. Many of the genes were also linked to human T2D, suggesting that dysregulation of cilia-associated genes may participate in T2D risk. Keywords: beta-cell proliferation, cilia genes, human pancreatic islets, islet transcriptomics, New Zealand Obese mouse, pathway enrichment analysis, primary cilia, type 2 diabetes
- Published
- 2019
31. Microencapsulation and tissue engineering as an alternative treatment of diabetes
- Author
-
S.S. Maria-Engler, M. Mares-Guia, M.L.C. Correa, E.M.C. Oliveira, C.A.M. Aita, K. Krogh, T. Genzini, M.P. Miranda, M. Ribeiro, L. Vilela, I.L. Noronha, F.G. Eliaschewitz, and M.C. Sogayar
- Subjects
human pancreatic islets ,pancreas ,extracellular matrix ,stem cells ,islet microencapsulation ,transplantation ,ductal cells ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
In the 70's, pancreatic islet transplantation arose as an attractive alternative to restore normoglycemia; however, the scarcity of donors and difficulties with allotransplants, even under immunosuppressive treatment, greatly hampered the use of this alternative. Several materials and devices have been developed to circumvent the problem of islet rejection by the recipient, but, so far, none has proved to be totally effective. A major barrier to transpose is the highly organized islet architecture and its physical and chemical setting in the pancreatic parenchyma. In order to tackle this problem, we assembled a multidisciplinary team that has been working towards setting up the Human Pancreatic Islets Unit at the Chemistry Institute of the University of São Paulo, to collect and process pancreas from human donors, upon consent, in order to produce purified, viable and functional islets to be used in transplants. Collaboration with the private enterprise has allowed access to the latest developed biomaterials for islet encapsulation and immunoisolation. Reasoning that the natural islet microenvironment should be mimicked for optimum viability and function, we set out to isolate extracellular matrix components from human pancreas, not only for analytical purposes, but also to be used as supplementary components of encapsulating materials. A protocol was designed to routinely culture different pancreatic tissues (islets, parenchyma and ducts) in the presence of several pancreatic extracellular matrix components and peptide growth factors to enrich the beta cell population in vitro before transplantation into patients. In addition to representing a therapeutic promise, this initiative is an example of productive partnership between the medical and scientific sectors of the university and private enterprises.
- Published
- 2001
- Full Text
- View/download PDF
32. Long-term cultures of human pancreatic islets in self-assembling peptides hydrogels.
- Author
-
Marchini A, Ciulla MG, Antonioli B, Agnoli A, Bovio U, Visnoviz V, Bertuzzi F, and Gelain F
- Abstract
Human pancreatic islets transplantation is an experimental therapeutic treatment for Type I Diabetes. Limited islets lifespan in culture remains the main drawback, due to the absence of native extracellular matrix as mechanical support after their enzymatic and mechanical isolation procedure. Extending the limited islets lifespan by creating a long-term in vitro culture remains a challenge. In this study, three biomimetic self-assembling peptides were proposed as potential candidates to recreate in vitro a pancreatic extracellular matrix, with the aim to mechanically and biologically support human pancreatic islets, by creating a three-dimensional culture system. The embedded human islets were analyzed for morphology and functionality in long-term cultures (14-and 28-days), by evaluating β-cells content, endocrine component, and extracellular matrix constituents. The three-dimensional support provided by HYDROSAP scaffold, and cultured into MIAMI medium, displayed a preserved islets functionality, a maintained rounded islets morphology and an invariable islets diameter up to 4 weeks, with results analogues to freshly-isolated islets. In vivo efficacy studies of the in vitro 3D cell culture system are ongoing; however, preliminary data suggest that human pancreatic islets pre-cultured for 2 weeks in HYDROSAP hydrogels and transplanted under subrenal capsule may restore normoglycemia in diabetic mice. Therefore, engineered self-assembling peptide scaffolds may provide a useful platform for long-term maintenance and preservation of functional human pancreatic islets in vitro ., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Marchini, Ciulla, Antonioli, Agnoli, Bovio, Visnoviz, Bertuzzi and Gelain.)
- Published
- 2023
- Full Text
- View/download PDF
33. Isolation and Purification of Human Pancreatic Islets.
- Author
-
Wei L
- Subjects
- Humans, Insulin, Serum Albumin, Human, Cell Separation, Islets of Langerhans, Organ Preservation Solutions, Islets of Langerhans Transplantation
- Abstract
Successful islet isolation is the key to islet transplantation in diabetic patients. However, islet isolation is a technically complex and time-consuming manual process. Optimizing the islet isolation process can improve islet yield and quality, reduce operators, and thus reduce costs.The isolation and purification of human islets include pancreas acquisition and preservation, pancreas digestion, islet purification, islet culture, and islet quality identification. Briefly, after the duodenum was removed, the pancreas was trimmed, the main pancreatic duct was intubated at the distal end of the pancreatic head, collagenase was injected into the pancreatic duct, and the perfused pancreatic tissue was cut and then digested in a Ricordi chamber. A digestion temperature of 37 °C was continuously used to assess the number of samples and the integrity of the lysed and released islets. At the end of the digestion process, collect the digested tissue in a 500 mL centrifuge tube prefilled with 25 mL of cold (4 °C) human serum albumin and centrifuge twice at 150 g for 3 min. After mixing with UW solution as islet storage solution, put it on ice (shake occasionally to prevent clumping) after 30 min. Digested pancreatic tissue was centrifuged at 2200 rpm for 5 min in a COBE 2991 cell processor to isolate islets from exocrine tissue using a continuous density gradient. The purified islet fractions were washed twice in HBSS supplemented with 10% human serum albumin and finally collected in CMRL1066 medium supplemented with the corresponding liquid. The purity of purified islets was calculated by DTZ staining, the survival rate of islets was calculated by FDA/PI staining, and islet function was determined by in vitro glucose-stimulated insulin secretion test., (© 2023. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
34. Les rôles de l'oscillateur circadien dans la fonction, le dysfonctionnement et la régénération des îlots pancréatiques
- Author
-
Petrenko, Volodymyr, Dibner, Charna, and Golay, Alain
- Subjects
ddc:616 ,Metabolism ,Beta cells ,Circadian clock ,ddc:612 ,Human pancreatic islets ,Type 2 Diabetes - Abstract
Nos travaux proposent un lien fonctionnel entre les horloges moléculaires du pancréas endocrine, sa fonction en condition physiologique et la pathogenèse du diabète type 2 (DT2) chez l'être humain. Nos expériences démontrent que la perturbation des horloges dans les cellules α- et β- des îlots humains altère l'accumulation et l'exocytose des granules d'insuline et de glucagon. Nous avons également observé que les îlots de donneurs atteints de DT2 possèdent des horloges dont l'amplitude circadienne est diminuée et dont la capacité de synchronisation in vitro est affaiblie. De manière intéressante, le modulateur d'horloges, Nobilétine, stimule l'amplitude des oscillations circadiennes ainsi que la sécrétion de l'insuline de ces îlots pathologiques. Enfin, nous rapportons que le mécanisme de régénération des cellules β chez les souris diabétiques est sous l'influence des horloges moléculaires via le rôle essentiel du composant clé BMAL1 dans ce mécanisme.
- Published
- 2021
35. Cathepsin c regulates cytokine-induced apoptosis in β-cell model systems
- Author
-
Joachim Størling, Caroline Frørup, Tina Fløyel, and Flemming Pociot
- Subjects
Chemokine ,type 1 diabetes ,p38 mitogen-activated protein kinases ,Pro-inflammatory cytokines ,Apoptosis ,QH426-470 ,pro-inflammatory cytokines ,Models, Biological ,Article ,Cathepsin C ,Proinflammatory cytokine ,Islets of Langerhans ,lysosomal proteases ,Cathepsin H ,Insulin-Secreting Cells ,Genetics ,medicine ,CXCL10 ,Animals ,Humans ,Human pancreatic islets ,Genetics (clinical) ,Cells, Cultured ,Cathepsin ,Inflammation ,biology ,Chemistry ,Pancreatic islets ,Lysosomal proteases ,MAPK ,Cell biology ,Rats ,human pancreatic islets ,medicine.anatomical_structure ,Diabetes Mellitus, Type 1 ,Type 1 diabetes ,inflammation ,biology.protein ,β-cell death ,Cytokines ,CTSC - Abstract
Emerging evidence suggests that several of the lysosomal cathepsin proteases are genetically associated with type 1 diabetes (T1D) and participate in immune-mediated destruction of the pancreatic β cells. We previously reported that the T1D candidate gene cathepsin H is downregulated by pro-inflammatory cytokines in human pancreatic islets and regulates β-cell function, apoptosis, and disease progression in children with new-onset T1D. In the present study, the objective was to investigate the expression patterns of all 15 known cathepsins in β-cell model systems and examine their role in the regulation of cytokine-induced apoptosis. Real-time qPCR screening of the cathepsins in human islets, 1.1B4 and INS-1E β-cell models identified several cathepsins that were expressed and regulated by pro-inflammatory cytokines. Using small interfering RNAs to knock down (KD) the cytokine-regulated cathepsins, we identified an anti-apoptotic function of cathepsin C as KD increased cytokine-induced apoptosis. KD of cathepsin C correlated with increased phosphorylation of JNK and p38 mitogen-activated protein kinases, and elevated chemokine CXCL10/IP-10 expression. This study suggests that cathepsin C is a modulator of β-cell survival, and that immune modulation of cathepsin expression in islets may contribute to immune-mediated β-cell destruction in T1D.
- Published
- 2021
- Full Text
- View/download PDF
36. Infection of human islets of langerhans with two strains of coxsackie B virus serotype 1: Assessment of virus replication, degree of cell death and induction of genes involved in the innate immunity pathway.
- Author
-
Anagandula, Mahesh, Richardson, Sarah J., Oberste, M. Steven, Sioofy‐Khojine, Amir‐Babak, Hyöty, Heikki, Morgan, Noel G., Korsgren, Olle, and Frisk, Gun
- Abstract
Type 1 diabetes mellitus is believed to be triggered, in part, by one or more environmental factors and human enteroviruses (HEVs) are among the candidates. Therefore, this study has examined whether two strains of HEV may differentially affect the induction of genes involved in pathways leading to the synthesis of islet hormones, chemokines and cytokines in isolated, highly purified, human islets. Isolated, purified human pancreatic islets were infected with strains of Coxsackievirus B1.Viral replication and the degree of CPE/islet dissociation were monitored. The expression of insulin, glucagon, CXCL10, TLR3, IF1H1, CCL5, OAS-1, IFNβ, and DDX58 was analyzed. Both strains replicated in islets but only one of strain caused rapid islet dissociation/CPE. Expression of the insulin gene was reduced during infection of islets with either viral strain but the gene encoding glucagon was unaffected. All genes analyzed which are involved in viral sensing and the development of innate immunity were induced by Coxsackie B viruses, with the notable exception of TLR3. There was no qualitative difference in the expression pattern between each strain but the magnitude of the response varied between donors. The lack of virus induced expression of TLR3, together with the differential regulation of IF1H1, OAS1 and IFNβ, (each of which has polymorphic variants influence the predisposition to type 1 diabetes), that might result in defective clearance of virus from islet cells. The reduced expression of the insulin gene and the unaffected expression of the gene encoding glucagon by Coxsackie B1 infection is consistent with the preferential β-cell tropism of the virus. J. Med. Virol. 86:1402-1411, 2014. © 2013 Wiley Periodicals, Inc. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
37. Effects of palmitate on genome-wide mRNA expression and DNA methylation patterns in human pancreatic islets.
- Author
-
Hall, Elin, Volkov, Petr, Dayeh, Tasnim, Bacos, Karl, Rönn, Tina, Nitert, Marloes Dekker, and Ling, Charlotte
- Abstract
Background: Circulating free fatty acids are often elevated in patients with type 2 diabetes (T2D) and obese individuals. Chronic exposure to high levels of saturated fatty acids has detrimental effects on islet function and insulin secretion. Altered gene expression and epigenetics may contribute to T2D and obesity. However, there is limited information on whether fatty acids alter the genome-wide transcriptome profile in conjunction with DNA methylation patterns in human pancreatic islets. To dissect the molecular mechanisms linking lipotoxicity to impaired insulin secretion, we investigated the effects of a 48 h palmitate treatment in vitro on genome-wide mRNA expression and DNA methylation patterns in human pancreatic islets. Methods: Genome-wide mRNA expression was analyzed using Affymetrix GeneChip® Human Gene 1.0 ST whole transcript-based array (n = 13) and genome-wide DNA methylation was analyzed using Infinium HumanMethylation450K BeadChip (n = 13) in human pancreatic islets exposed to palmitate or control media for 48 h. A non-parametric paired Wilcoxon statistical test was used to analyze mRNA expression. Apoptosis was measured using Apo-ONE® Homogeneous Caspase-3/7 Assay (n = 4). Results: While glucose-stimulated insulin secretion was decreased, there was no significant effect on apoptosis in human islets exposed to palmitate. We identified 1,860 differentially expressed genes in palmitate-treated human islets. These include candidate genes for T2D, such as TCF7L2, GLIS3, HNF1B and SLC30A8. Additionally, genes in glycolysis/gluconeogenesis, pyruvate metabolism, fatty acid metabolism, glutathione metabolism and one carbon pool by folate were differentially expressed in palmitate-treated human islets. Palmitate treatment altered the global DNA methylation level and DNA methylation levels of CpG island shelves and shores, 5′UTR, 3′UTR and gene body regions in human islets. Moreover, 290 genes with differential expression had a corresponding change in DNA methylation, for example, TCF7L2 and GLIS3. Importantly, out of the genes differentially expressed due to palmitate treatment in human islets, 67 were also associated with BMI and 37 were differentially expressed in islets from T2D patients. Conclusion: Our study demonstrates that palmitate treatment of human pancreatic islets gives rise to epigenetic modifications that together with altered gene expression may contribute to impaired insulin secretion and T2D. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
38. St. John's wort extract and hyperforin protect rat and human pancreatic islets against cytokine toxicity.
- Author
-
Novelli, Michela, Beffy, Pascale, Menegazzi, Marta, Tata, Vincenzo, Martino, Luisa, Sgarbossa, Anna, Porozov, Svetlana, Pippa, Anna, Masini, Matilde, Marchetti, Piero, and Masiello, Pellegrino
- Subjects
- *
HYPERICUM , *ISLANDS of Langerhans , *CYTOKINES , *IMMUNOREGULATION ,THERAPEUTIC use of plant extracts - Abstract
The extract of Hypericum perforatum (St. John's wort, SJW) and its component hyperforin (HPF) were previously shown to inhibit cytokine-induced activation of signal transducer and activator of transcription-1 and nuclear factor κB and prevent apoptosis in a cultured β-cell line. Objective of this study was to assess the protection exerted by SJW and HPF on isolated rat and human islets exposed to cytokines in vitro. Functional, ultrastructural, biomolecular and cell death evaluation studies were performed. In both rat and human islets, SJW and HPF counteracted cytokine-induced functional impairment and down-regulated mRNA expression of pro-inflammatory target genes, such as iNOS, CXCL9, CXCL10, COX2. Cytokine-induced NO production from cultured islets, evaluated by nitrites measurement in the medium, was significantly reduced in the presence of the vegetal compounds. Noteworthy, the increase in apoptosis and necrosis following 48-h exposure to cytokines was fully prevented by SJW and partially by HPF. Ultrastructural morphometric analysis in human islets exposed to cytokines for 20 h showed that SJW or HPF avoided early β-cell damage (e.g., mitochondrial alterations and loss of insulin granules). In conclusion, SJW compounds protect rat and human islets against cytokine effects by counteracting key mechanisms of cytokine-mediated β-cell injury and represent promising pharmacological tools for prevention or limitation of β-cell dysfunction and loss in type 1 diabetes. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
39. A Versatile Model of Microfluidic Perifusion System for the Evaluation of C-Peptide Secretion Profiles: Comparison Between Human Pancreatic Islets and HLSC-Derived Islet-Like Structures
- Author
-
Victor Navarro-Tableros, Ciro Tetta, Maria Felice Brizzi, Giovanni Camussi, and Yonathan Gomez
- Subjects
0301 basic medicine ,Cell ,microfluidic ,Medicine (miscellaneous) ,T2DM ,Pharmacology ,01 natural sciences ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Diabetes mellitus ,medicine ,Secretion ,lcsh:QH301-705.5 ,geography ,geography.geographical_feature_category ,obese ,Chemistry ,C-peptide ,Pancreatic islets ,010401 analytical chemistry ,HLSC-ILS ,Islet ,medicine.disease ,In vitro ,0104 chemical sciences ,human pancreatic islets ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,Ex vivo ,perifusion - Abstract
A robust and easy-to-use tool for the ex vivo dynamic evaluation of pancreatic islet (PI) function is essential for further development of novel cell-based therapeutic approaches to treating diabetes. Here, we developed four different glucose perifusion protocols (GPPs) in a microfluidic perifusion system (MPS), based entirely on commercially available components. After validation, the GPPs were used to evaluate C-peptide secretion profiles of PIs derived from different donors (healthy, obese, and type 2 diabetic) and from human liver stem-cell-derived islet-like structures (HLSC-ILS). Using this device, we demonstrated that PIs derived from healthy donors displayed a physiological C-peptide secretion profile as characterized by the response to (a) different glucose concentrations, (b) consecutive pulses of high-glucose concentrations, (c) a glucose threshold ranging from 5&ndash, 8 mM, and (d) a constant high-glucose perifusion in a biphasic manner. Moreover, we were able to detect a dysregulated secretion profile in PIs derived from both obese and type 2 diabetes mellitus (T2DM) donors. Finally, we also evaluated the kinetic secretion profiles of HLSC-ILS, demonstrating that, nonetheless, with a lower amplitude of secretion compared to PI derived from healthy donors, they were already glucose-responsive on day seven post-differentiation. In conclusion, we have provided evidence that our MPS is a versatile device and may represent a valuable tool to study insulin-producing cells in vitro.
- Published
- 2020
- Full Text
- View/download PDF
40. Microwell culture platform maintains viability and mass of human pancreatic islets.
- Author
-
Kato H, Miwa T, Quijano J, Medrano L, Ortiz J, Desantis A, Omori K, Wada A, Tatsukoshi K, Kandeel F, Mullen Y, Ku HT, and Komatsu H
- Subjects
- Humans, Insulin, Glycemic Control, Hypoxia, Oxygen Consumption, Islets of Langerhans
- Abstract
Background: Transplantation of the human pancreatic islets is a promising approach for specific types of diabetes to improve glycemic control. Although effective, there are several issues that limit the clinical expansion of this treatment, including difficulty in maintaining the quality and quantity of isolated human islets prior to transplantation. During the culture, we frequently observe the multiple islets fusing together into large constructs, in which hypoxia-induced cell damage significantly reduces their viability and mass. In this study, we introduce the microwell platform optimized for the human islets to prevent unsolicited fusion, thus maintaining their viability and mass in long-term cultures., Method: Human islets are heterogeneous in size; therefore, two different-sized microwells were prepared in a 35 mm-dish format: 140 µm × 300 µm-microwells for <160 µm-islets and 200 µm × 370 µm-microwells for >160 µm-islets. Human islets (2,000 islet equivalent) were filtered through a 160 µm-mesh to prepare two size categories for subsequent two week-cultures in each microwell dish. Conventional flat-bottomed 35 mm-dishes were used for non-filtered islets (2,000 islet equivalent/2 dishes). Post-cultured islets are collected to combine in each condition (microwells and flat) for the comparisons in viability, islet mass, morphology, function and metabolism. Islets from three donors were independently tested., Results: The microwell platform prevented islet fusion during culture compared to conventional flat bottom dishes, which improved human islet viability and mass. Islet viability and mass on the microwells were well-maintained and comparable to those in pre-culture, while flat bottom dishes significantly reduced islet viability and mass in two weeks. Morphology assessed by histology, insulin-secreting function and metabolism by oxygen consumption did not exhibit the statistical significance among the three different conditions., Conclusion: Microwell-bottomed dishes maintained viability and mass of human islets for two weeks, which is significantly improved when compared to the conventional flat-bottomed dishes., Competing Interests: This study was performed as a collaborative study between AGC Techno Glass and Arthur Riggs Diabetes & Metabolism Research Institute of City of Hope. TM, AW, and KT are the employees at AGC Techno Glass. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Kato, Miwa, Quijano, Medrano, Ortiz, Desantis, Omori, Wada, Tatsukoshi, Kandeel, Mullen, Ku and Komatsu.)
- Published
- 2022
- Full Text
- View/download PDF
41. Identification of CpG-SNPs associated with type 2 diabetes and differential DNA methylation in human pancreatic islets.
- Author
-
Dayeh, T., Olsson, A., Volkov, P., Almgren, P., Rönn, T., and Ling, C.
- Abstract
Aims/hypothesis: To date, the molecular function of most of the reported type 2 diabetes-associated loci remains unknown. The introduction or removal of cytosine-phosphate-guanine (CpG) dinucleotides, which are possible sites of DNA methylation, has been suggested as a potential mechanism through which single-nucleotide polymorphisms (SNPs) can affect gene function via epigenetics. The aim of this study was to examine if any of 40 SNPs previously associated with type 2 diabetes introduce or remove a CpG site and if these CpG-SNPs are associated with differential DNA methylation in pancreatic islets of 84 human donors. Methods: DNA methylation was analysed using pyrosequencing. Results: We found that 19 of 40 (48%) type 2 diabetes-associated SNPs introduce or remove a CpG site. Successful DNA methylation data were generated for 16 of these 19 CpG-SNP loci, representing the candidate genes TCF7L2, KCNQ1, PPARG, HHEX, CDKN2A, SLC30A8, DUSP9, CDKAL1, ADCY5, SRR, WFS1, IRS1, DUSP8, HMGA2, TSPAN8 and CHCHD9. All analysed CpG-SNPs were associated with differential DNA methylation of the CpG-SNP site in human islets. Moreover, six CpG-SNPs, representing TCF7L2, KCNQ1, CDKN2A, ADCY5, WFS1 and HMGA2, were also associated with DNA methylation of surrounding CpG sites. Some of the type 2 diabetes CpG-SNP sites that exhibit differential DNA methylation were further associated with gene expression, alternative splicing events determined by splice index, and hormone secretion in the human islets. The 19 type 2 diabetes-associated CpG-SNPs are in strong linkage disequilibrium ( r > 0.8) with a total of 295 SNPs, including 91 CpG-SNPs. Conclusions/interpretation: Our results suggest that the introduction or removal of a CpG site may be a molecular mechanism through which some of the type 2 diabetes SNPs affect gene function via differential DNA methylation and consequently contributes to the phenotype of the disease. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
42. Autonomous and self-sustained circadian oscillators displayed in human islet cells.
- Author
-
Pulimeno, P., Mannic, T., Sage, D., Giovannoni, L., Salmon, P., Lemeille, S., Giry-Laterriere, M., Unser, M., Bosco, D., Bauer, C., Morf, J., Halban, P., Philippe, J., and Dibner, C.
- Abstract
Aims/hypothesis: Following on from the emerging importance of the pancreas circadian clock on islet function and the development of type 2 diabetes in rodent models, we aimed to examine circadian gene expression in human islets. The oscillator properties were assessed in intact islets as well as in beta cells. Methods: We established a system for long-term bioluminescence recording in cultured human islets, employing lentivector gene delivery of the core clock gene Bmal1 (also known as Arntl)-luciferase reporter. Beta cells were stably labelled using a rat insulin2 promoter fluorescent construct. Single-islet/cell oscillation profiles were measured by combined bioluminescence-fluorescence time-lapse microscopy. Results: Human islets synchronised in vitro exhibited self-sustained circadian oscillations of Bmal1-luciferase expression at both the population and single-islet levels, with period lengths of 23.6 and 23.9 h, respectively. Endogenous BMAL1 and CRY1 transcript expression was circadian in synchronised islets over 48 h, and antiphasic to REV-ERBα (also known as NR1D1), PER1, PER2, PER3 and DBP transcript circadian profiles. HNF1A and PDX1 exhibited weak circadian oscillations, in phase with the REV-ERBα transcript. Dispersed islet cells were strongly oscillating as well, at population and single-cell levels. Importantly, beta and non-beta cells revealed oscillatory profiles that were well synchronised with each other. Conclusions/interpretation: We provide for the first time compelling evidence for high-amplitude cell-autonomous circadian oscillators displayed in human pancreatic islets and in dispersed human islet cells. Moreover, these clocks are synchronised between beta and non-beta cells in primary human islet cell cultures. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
43. Meta-analysis and functional effects of the SLC30A8 rs13266634 polymorphism on isolated human pancreatic islets
- Author
-
Cauchi, Stéphane, Guerra, Silvia Del, Choquet, Hélène, D’Aleo, Valentina, Groves, Christopher J., Lupi, Roberto, McCarthy, Mark I., Froguel, Philippe, and Marchetti, Piero
- Subjects
- *
ISLANDS of Langerhans , *GENETIC polymorphisms , *META-analysis , *INSULIN , *TYPE 2 diabetes , *DISEASE susceptibility , *GENE expression , *MESSENGER RNA - Abstract
Abstract: Background: The C-allele of rs13266634 located in SLC30A8 (ZNT8) has been strongly associated with decreased insulin release and with type 2 diabetes (T2D) susceptibility in some but not all studies. To shed further light on this issue, we performed a meta-analysis of the association between rs13266634 and T2D in different ethnic groups and assessed the relationships between SLC30A8 genotypes and some properties of isolated human islets. Methods: From 32 original articles, a total of 77,234 control individuals and 44,945 subjects with T2D were studied in meta-analysis. To assess the relationships between SLC30A8 genotype and islet cell phenotype, insulin secretion in response to glucose, glucose plus arginine and glucose plus glibenclamide was determined in pancreatic islets isolated from 82 multiorgan donors genotyped for the rs13266634 polymorphism. Quantitative expression of SLC30A8, Insulin and Glucagon mRNA was also measured. Results: Overall, each SLC30A8 risk allele was associated with a 14% increased risk for T2D (P =2.78×10−34). The population risk of T2D attributable to this polymorphism was estimated at 9.5% in Europeans and 8.1% in East Asians. Basal and stimulated insulin secretion from human islets as well as islet expressions of SLC30A8, Insulin and Glucagon were not affected by the presence of the polymorphism. However, SLC30A8 expression was positively correlated with Insulin (r =0.75, P =6.43×10−6) and Glucagon (r: 0.70, P =4.89×10−5) levels. Conclusions: The SLC30A8 rs13266634 polymorphism is among the most confirmed genetic markers of T2D in Europeans and East Asians. In isolated human islets, the risk C-allele does not affect ex-vivo insulin secretion and SLC30A8 expression, which is correlated with that of insulin and glucagon. [Copyright &y& Elsevier]
- Published
- 2010
- Full Text
- View/download PDF
44. G-protein-coupled receptor 40 (GPR40) expression and its regulation in human pancreatic islets: The role of type 2 diabetes and fatty acids.
- Author
-
Del Guerra, S., Bugliani, M., D'Aleo, V., Del Prato, S., Boggi, U., Mosca, F., Filipponi, F., and Lupi, R.
- Abstract
Abstract: Background and aims: GPR40 is a membrane-bound receptor paired with medium and long-chain fatty acids (FFA) as endogenous ligands. Its acute activation potentiates insulin secretion from beta cells, whereas prolonged binding might contribute to the deleterious effects of chronic exposure to FFA. Little information is available on the expression of GPR40 and its regulation in human islets (HI). Material and methods: HI were prepared by enzymatic digestion and gradient separation from the pancreas of 20 non-diabetic (Ctrl) and 13 type 2 diabetic (T2DM) multiorgan donors, and functional and molecular studies were then performed. Results: By qualitative and quantitative PCR experiments, mRNA expression was shown in HI. Both in T2DM islets and in Ctrl islets pre-exposed for 24h to 1.0mmol/l FFA (palmitate:oleate, 2:1), GPR40 mRNA expression was significantly reduced (p <0.01) in the T2DM cells as compared to Ctrl cells. A significant positive correlation was found between glucose-stimulated insulin secretion and GPR40 expression. Conclusions: These results show the expression of GPR40 in human pancreatic islets which are regulated by FFA. The finding that T2DM islets have a lower GPR40 expression, and the correlation of these genes with insulin secretion, raises the possibility of an involvement of GPR40 in human diabetes beta-cell dysfunction. [Copyright &y& Elsevier]
- Published
- 2010
- Full Text
- View/download PDF
45. Single-Cell Transcriptomics Links Loss of Human Pancreatic β-Cell Identity to ER Stress.
- Author
-
Groen, Nathalie, Leenders, Floris, Mahfouz, Ahmed, Munoz-Garcia, Amadeo, Muraro, Mauro J., de Graaf, Natascha, Rabelink, Ton. J., Hoeben, Rob, van Oudenaarden, Alexander, Zaldumbide, Arnaud, Reinders, Marcel J. T., Koning, Eelco J. P. de, and Carlotti, Françoise
- Subjects
- *
DENATURATION of proteins , *ENDOPLASMIC reticulum , *ISLANDS of Langerhans , *EIGENFUNCTIONS - Abstract
The maintenance of pancreatic islet architecture is crucial for proper β-cell function. We previously reported that disruption of human islet integrity could result in altered β-cell identity. Here we combine β-cell lineage tracing and single-cell transcriptomics to investigate the mechanisms underlying this process in primary human islet cells. Using drug-induced ER stress and cytoskeleton modification models, we demonstrate that altering the islet structure triggers an unfolding protein response that causes the downregulation of β-cell maturity genes. Collectively, our findings illustrate the close relationship between endoplasmic reticulum homeostasis and β-cell phenotype, and strengthen the concept of altered β-cell identity as a mechanism underlying the loss of functional β-cell mass. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
46. PPARγ-dependent and -independent effects of Rosiglitazone on lipotoxic human pancreatic islets
- Author
-
Vandewalle, Brigitte, Moerman, Ericka, Lefebvre, Bruno, Defrance, Frédérique, Gmyr, Valéry, Lukowiak, Bruno, Kerr Conte, Julie, and Pattou, François
- Subjects
- *
PANCREATIC secretions , *GLUTATHIONE , *SUPEROXIDE dismutase , *APOPTOSIS - Abstract
Abstract: We explored the in vitro effects of Rosiglitazone (RZG), a PPARγ agonist, on human pancreatic islet dysfunctions induced by chronic free fatty acid exposure. We demonstrated that RZG beneficial effects on insulin secretion and apoptosis did not imply PDX-1 or insulin gene modulation. It rather involved, through a PPARγ-dependent mechanism, a reduction of iNOS overexpressed in lipotoxic islets. This reduction likely led to the restoration of ATP level and insulin secretion as well as the decrease in apoptosis. More interestingly, we also demonstrated that RZG beneficial effects involved PPARγ-independent mechanisms. RZG treatment led to a limitation of oxidative stress exemplified by an increase of GPx and SOD expression. It also increased UCP2 expression that seemed to display antioxidant action in this model. Thus, RZG did not appear to exert a direct action on insulin expression but rather an indirect action on insulin secretion and apoptosis, through PPARγ-dependent and -independent mechanisms, via regulation of nitrogen and oxygen reactive species injury. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
47. Beneficial effects of prolactin and laminin on human pancreatic islet-cell cultures
- Author
-
Labriola, Leticia, Montor, Wagner R., Krogh, Karin, Lojudice, Fernando H., Genzini, Tércio, Goldberg, Anna C., Eliaschewitz, Freddy G., and Sogayar, Mari C.
- Subjects
- *
PITUITARY hormones , *ISLANDS of Langerhans , *CELL division , *CELL proliferation - Abstract
Abstract: The problem of pancreas donor shortage could be addressed through in vitro islet-cell proliferation prior to transplantation into diabetic patients. Therefore, we set out to evaluate the effects of prolactin (rhPRL) and laminin on primary cultures of human pancreatic islets. Our results showed that rhPRL induced an increase in islet-cell number and in cumulative insulin secretion (p <0.01). However, glucose-induced insulin secretion was enhanced only in the presence of both laminin and rhPRL. In addition, we describe, for the first time in human islets, the PRL-induced activation of JAK2, and signal transducer and activator of transcription (STAT) 1, 3 and 5. Our results demonstrate a significant beneficial effect of rhPRL and laminin on human islets and support widely held notion that the closer physiological stimuli and environment of beta cells are mimicked, the better are the results in cell proliferation and secretory function, both essential for successful islet transplantation. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
48. 1,25-dihydroxyvitamin D3 protects human pancreatic islets against cytokine-induced apoptosis via down-regulation of the fas receptor.
- Author
-
Riachy, R., Vandewalle, B., Moerman, E., Belaich, S., Lukowiak, B., Gmyr, V., Muharram, G., Kerr Conte, J., and Pattou, F.
- Subjects
PANCREATIC beta cells ,DIABETES ,APOPTOSIS ,CELL membranes ,AUTOIMMUNE diseases ,CYTOKINES - Abstract
Beta cell loss occurs at the onset of type 1 diabetes and after islet graft. It results from the dysfunction and destruction of beta cells mainly achieved by apoptosis. One of the mediators believed to be involved in beta cell apoptosis is Fas, a transmembrane cell surface receptor transducing an apoptotic death signal and contributing to the pathogenesis of several autoimmune diseases. Fas expression is particularly induced in beta cells by inflammatory cytokines secreted by islet-infiltrating mononuclear cells and makes cells susceptible to apoptosis by interaction with Fas-ligand expressing cells. We have previously demonstrated that 1,25 (OH)
2 D3 , the active metabolite of vitamin D, known to exhibit immunomodulatory properties and prevent the development of type 1 diabetes in NOD mice, is efficient against apoptosis induced by cytokines in human pancreatic islets in vitro. The effects were mainly mediated by the inactivation of NF-kappa-B. In this study we demonstrated that 1,25 (OH)2 D3 was also able to counteract cytokine-induced Fas expression in human islets both at the mRNA and protein levels. These results were reinforced by our microarray analysis highlighting the beneficial effects of 1,25 (OH)2 D3 on death signals induced by Fas activation. Our results provides additional evidence that 1,25 (OH)2 D3 may be an interesting tool to help prevent the onset of type 1 diabetes and improve islet graft survival. [ABSTRACT FROM AUTHOR]- Published
- 2006
- Full Text
- View/download PDF
49. Inflammatory gene expression in Coxsackievirus B-4-infected human islets of Langerhans
- Author
-
Olsson, Annika, Johansson, Ulrika, Korsgren, Olle, and Frisk, Gun
- Subjects
- *
CYTOKINES , *IMMUNOREGULATION , *INFECTION , *PEPTIDES - Abstract
Abstract: The event that triggers the autoimmune destruction of insulin-producing β-cells in type 1 diabetes mellitus (T1DM) is still unknown. Enterovirus, especially Coxsackievirus, infections have long been associated with this disease. Cytokines and chemokines induced by an enterovirus infection may act to trigger the autoimmune reactions that produce T1DM. Gene expression was examined in isolated human islets infected with a Coxsackievirus-B4 (CBV-4) strain causing lytic infection (V89-4557) and in islets infected with a CBV-4 strain establishing persistent infection (VD2921). Microarray analysis indicated that infection with the CBV-4 strains resulted in specific induction of a number of inflammatory genes, including IL-1β, IL-6, IL-8, MCP-1, and RANTES. Importantly, the inflammatory genes induced by the CBV-4 infections differed in the two strains, with more cytokines being induced by the non-lytic CBV-4 strain than by the lytic strain. These cytokines and chemokines have the potential to rapidly induce inflammatory reactions when expressed in vivo and could contribute to the autoimmune reactions associated with the development of T1DM. [Copyright &y& Elsevier]
- Published
- 2005
- Full Text
- View/download PDF
50. Cathepsin C Regulates Cytokine-Induced Apoptosis in β-Cell Model Systems.
- Author
-
Fløyel, Tina, Frørup, Caroline, Størling, Joachim, and Pociot, Flemming
- Subjects
- *
JNK mitogen-activated protein kinases , *SMALL interfering RNA , *TYPE 1 diabetes , *CATHEPSINS , *ISLANDS of Langerhans , *ELASTASES - Abstract
Emerging evidence suggests that several of the lysosomal cathepsin proteases are genetically associated with type 1 diabetes (T1D) and participate in immune-mediated destruction of the pancreatic β cells. We previously reported that the T1D candidate gene cathepsin H is downregulated by pro-inflammatory cytokines in human pancreatic islets and regulates β-cell function, apoptosis, and disease progression in children with new-onset T1D. In the present study, the objective was to investigate the expression patterns of all 15 known cathepsins in β-cell model systems and examine their role in the regulation of cytokine-induced apoptosis. Real-time qPCR screening of the cathepsins in human islets, 1.1B4 and INS-1E β-cell models identified several cathepsins that were expressed and regulated by pro-inflammatory cytokines. Using small interfering RNAs to knock down (KD) the cytokine-regulated cathepsins, we identified an anti-apoptotic function of cathepsin C as KD increased cytokine-induced apoptosis. KD of cathepsin C correlated with increased phosphorylation of JNK and p38 mitogen-activated protein kinases, and elevated chemokine CXCL10/IP-10 expression. This study suggests that cathepsin C is a modulator of β-cell survival, and that immune modulation of cathepsin expression in islets may contribute to immune-mediated β-cell destruction in T1D. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.