Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Int J Biol Sci ; 19(13): 4157-4165, 2023.
Article in English | MEDLINE | ID: mdl-37705740

ABSTRACT

Prostaglandins are lipid mediators involved in physiological processes, such as constriction or dilation of blood vessels, but also pathophysiological processes, which include inflammation, pain and fever. They are produced by almost all cell types in the organism by activation of Prostaglandin endoperoxide synthases/Cyclooxygenases. The inducible Prostaglandin Endoperoxide Synthase 2/Cyclooxygenase 2 (PTGS2/COX2) plays an important role in pathologies associated with inflammatory signaling. The main product derived from PTGS2/COX2 expression and activation is Prostaglandin E2 (PGE2), which promotes a wide variety of tissue-specific effects, pending environmental inputs. One of the major sources of PGE2 are infiltrating inflammatory cells - the production of this molecule increases drastically in damaged tissues. Immune infiltration is a hallmark of type 1 diabetes mellitus, a multifactorial disease that leads to autoimmune-mediated pancreatic beta cell destruction. Controversial effects for the PTGS2/COX2-PGE2 signaling cascade in pancreatic islet cells subjected to diabetogenic conditions have been reported, allocating PGE2 as both, cause and consequence of inflammation. Herein, we review the main effects of this molecular pathway in a tissue-specific manner, with a special emphasis on beta cell mass protection/destruction and its potential role in the prevention or development of T1DM. We also discuss strategies to target this pathway for future therapies.


Subject(s)
Diabetes Mellitus, Type 1 , Dinoprostone , Humans , Cyclooxygenase 2/genetics , Signal Transduction , Inflammation
2.
iScience ; 25(5): 104345, 2022 May 20.
Article in English | MEDLINE | ID: mdl-35602948

ABSTRACT

LRH-1/NR5A2 is implicated in islet morphogenesis postnatally, and its activation using the agonist BL001 protects islets against apoptosis, reverting hyperglycemia in mouse models of Type 1 Diabetes Mellitus. Islet transcriptome profiling revealed that the expression of PTGS2/COX2 is increased by BL001. Herein, we sought to define the role of LRH-1 in postnatal islet morphogenesis and chart the BL001 mode of action conferring beta cell protection. LRH-1 ablation within developing beta cells impeded beta cell proliferation, correlating with mouse growth retardation, weight loss, and hypoglycemia leading to lethality. LRH-1 deletion in adult beta cells abolished the BL001 antidiabetic action, correlating with beta cell destruction and blunted Ptgs2 induction. Islet PTGS2 inactivation led to reduced PGE2 levels and loss of BL001 protection against cytokines as evidenced by increased cytochrome c release and cleaved-PARP. The PTGER1 antagonist-ONO-8130-negated BL001-mediated islet survival. Our results define the LRH-1/PTGS2/PGE2/PTGER1 signaling axis as a key pathway mediating BL001 survival properties.

3.
Theranostics ; 11(14): 6983-7004, 2021.
Article in English | MEDLINE | ID: mdl-34093866

ABSTRACT

Rationale: We recently demonstrated that the 'Metabesity' factor HMG20A regulates islet beta-cell functional maturity and adaptation to physiological stress such as pregnancy and pre-diabetes. HMG20A also dictates central nervous system (CNS) development via inhibition of the LSD1-CoREST complex but its expression pattern and function in adult brain remains unknown. Herein we sought to determine whether HMG20A is expressed in the adult CNS, specifically in hypothalamic astrocytes that are key in glucose homeostasis and whether similar to islets, HMG20A potentiates astrocyte function in response to environmental cues. Methods: HMG20A expression profile was assessed by quantitative PCR (QT-PCR), Western blotting and/or immunofluorescence in: 1) the hypothalamus of mice exposed or not to either a high-fat diet or a high-fat high-sucrose regimen, 2) human blood leukocytes and adipose tissue obtained from healthy or diabetic individuals and 3) primary mouse hypothalamic astrocytes exposed to either high glucose or palmitate. RNA-seq and cell metabolic parameters were performed on astrocytes treated or not with a siHMG20A. Astrocyte-mediated neuronal survival was evaluated using conditioned media from siHMG20A-treated astrocytes. The impact of ORY1001, an inhibitor of the LSD1-CoREST complex, on HMG20A expression, reactive astrogliosis and glucose metabolism was evaluated in vitro and in vivo in high-fat high-sucrose fed mice. Results: We show that Hmg20a is predominantly expressed in hypothalamic astrocytes, the main nutrient-sensing cell type of the brain. HMG20A expression was upregulated in diet-induced obesity and glucose intolerant mice, correlating with increased transcript levels of Gfap and Il1b indicative of inflammation and reactive astrogliosis. Hmg20a transcript levels were also increased in adipose tissue of obese non-diabetic individuals as compared to obese diabetic patients. HMG20A silencing in astrocytes resulted in repression of inflammatory, cholesterol biogenesis and epithelial-to-mesenchymal transition pathways which are hallmarks of reactive astrogliosis. Accordingly, HMG20A depleted astrocytes exhibited reduced mitochondrial bioenergetics and increased susceptibility to apoptosis. Neuron viability was also hindered in HMG20A-depleted astrocyte-derived conditioned media. ORY1001 treatment rescued expression of reactive astrogliosis-linked genes in HMG20A ablated astrocytes while enhancing cell surface area, GFAP intensity and STAT3 expression in healthy astrocytes, mimicking the effect of HMG20A. Furthermore, ORY1001 treatment protected against obesity-associated glucose intolerance in mice correlating with a regression of hypothalamic HMG20A expression, indicative of reactive astrogliosis attenuation with improved health status. Conclusion: HMG20A coordinates the astrocyte polarization state. Under physiological pressure such as obesity and insulin resistance that induces low grade inflammation, HMG20A expression is increased to induce reactive astrogliosis in an attempt to preserve the neuronal network and re-establish glucose homeostasis. Nonetheless, a chronic metabesity state or functional mutations will result in lower levels of HMG20A, failure to promote reactive astrogliosis and increase susceptibility of neurons to stress-induced apoptosis. Such effects could be reversed by ORY1001 treatment both in vitro and in vivo, paving the way for a new therapeutic approach for Type 2 Diabetes Mellitus.


Subject(s)
Astrocytes/metabolism , Diabetes Mellitus, Type 2/metabolism , Gliosis/metabolism , High Mobility Group Proteins/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Obesity/metabolism , Adipose Tissue/metabolism , Adult , Animals , Cell Survival/drug effects , Co-Repressor Proteins/antagonists & inhibitors , Diet, High-Fat , Glial Fibrillary Acidic Protein/metabolism , Glucose/metabolism , High Mobility Group Proteins/antagonists & inhibitors , High Mobility Group Proteins/genetics , Histone Demethylases/antagonists & inhibitors , Humans , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mitochondria/genetics , Mitochondria/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , RNA, Small Interfering , RNA-Seq
4.
Int J Mol Sci ; 22(8)2021 Apr 19.
Article in English | MEDLINE | ID: mdl-33921851

ABSTRACT

Diabetes is a chronic metabolic disease caused by an absolute or relative deficiency in functional pancreatic ß-cells that leads to defective control of blood glucose. Current treatments for diabetes, despite their great beneficial effects on clinical symptoms, are not curative treatments, leading to a chronic dependence on insulin throughout life that does not prevent the secondary complications associated with diabetes. The overwhelming increase in DM incidence has led to a search for novel antidiabetic therapies aiming at the regeneration of the lost functional ß-cells to allow the re-establishment of the endogenous glucose homeostasis. Here we review several aspects that must be considered for the development of novel and successful regenerative therapies for diabetes: first, the need to maintain the heterogeneity of islet ß-cells with several subpopulations of ß-cells characterized by different transcriptomic profiles correlating with differences in functionality and in resistance/behavior under stress conditions; second, the existence of an intrinsic islet plasticity that allows stimulus-mediated transcriptome alterations that trigger the transdifferentiation of islet non-ß-cells into ß-cells; and finally, the possibility of using agents that promote a fully functional/mature ß-cell phenotype to reduce and reverse the process of dedifferentiation of ß-cells during diabetes.


Subject(s)
Islets of Langerhans/metabolism , Regenerative Medicine/methods , Animals , Cell Transdifferentiation/physiology , Diabetes Mellitus, Type 1/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism
5.
Cancers (Basel) ; 14(1)2021 Dec 24.
Article in English | MEDLINE | ID: mdl-35008251

ABSTRACT

During metastasis, invading tumor cells and circulating tumor cells (CTC) face multiple mechanical challenges during migration through narrow pores and cell squeezing. However, little is known on the importance and consequences of mechanical stress for tumor progression and success in invading a new organ. Recently, several studies have shown that cell constriction can lead to nuclear envelope rupture (NER) during interphase. This loss of proper nuclear compartmentalization has a profound effect on the genome, being a key driver for the genome evolution needed for tumor progression. More than just being a source of genomic alterations, the transient nuclear envelope collapse can also support metastatic growth by several mechanisms involving the innate immune response cGAS/STING pathway. In this review we will describe the importance of the underestimated role of cellular squeezing in the progression of tumorigenesis. We will describe the complexity and difficulty for tumor cells to reach the metastatic site, detail the genomic aberration diversity due to NER, and highlight the importance of the activation of the innate immune pathway on cell survival. Cellular adaptation and nuclear deformation can be the key to the metastasis success in many unsuspected aspects.

6.
Int J Mol Sci ; 20(24)2019 12 06.
Article in English | MEDLINE | ID: mdl-31817798

ABSTRACT

Gestational diabetes mellitus (GDM), a metabolic disease that develops with the increase in insulin resistance during late pregnancy, is currently one of the most common complications affecting pregnancy. The polygenic nature of GDM, together with the interplay between different genetic variants with nutritional and environmental factors has hindered the full understanding of the etiology of this disease. However, an important genetic overlap has been found with type 2 diabetes mellitus (T2DM) and, as in the case of T2DM, most of the identified loci are associated with ß-cell function. Early detection of GDM and adequate interventions to control the maternal glycemia are necessary to avoid the adverse outcomes for both the mother and the offspring. The in utero exposure to the diabetic milieu predispose these children for future diseases, among them T2DM, originating a vicious circle implicated in the increased prevalence of both GDM and T2DM. The involvement of inflammatory processes in the development of GDM highlights the importance of pancreatic ß-cell factors able to favor the adaptation processes required during gestation, concomitantly with the protection of the islets from an inflammatory milieu. In this regard, two members of the Pax family of transcription factors, PAX4 and PAX8, together with the chromatin remodeler factor HMG20A, have gained great relevance due to their involvement in ß-cell mass adaptation together with their anti-inflammatory properties. Mutations in these factors have been associated with GDM, highlighting these as novel candidates for genetic screening analysis in the identification of women at risk of developing GDM.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Diabetes, Gestational/metabolism , Diabetes, Gestational/physiopathology , Islets of Langerhans/physiology , Blood Glucose/metabolism , Female , High Mobility Group Proteins/metabolism , Homeodomain Proteins/metabolism , Humans , PAX8 Transcription Factor/metabolism , Paired Box Transcription Factors/metabolism , Pregnancy
7.
Genes (Basel) ; 10(5)2019 05 08.
Article in English | MEDLINE | ID: mdl-31072002

ABSTRACT

The high prevalence of type 2 diabetes mellitus (T2DM), together with the fact that current treatments are only palliative and do not avoid major secondary complications, reveals the need for novel approaches to treat the cause of this disease. Efforts are currently underway to identify therapeutic targets implicated in either the regeneration or re-differentiation of a functional pancreatic islet ß-cell mass to restore insulin levels and normoglycemia. However, T2DM is not only caused by failures in ß-cells but also by dysfunctions in the central nervous system (CNS), especially in the hypothalamus and brainstem. Herein, we review the physiological contribution of hypothalamic neuronal and glial populations, particularly astrocytes, in the control of the systemic response that regulates blood glucose levels. The glucosensing capacity of hypothalamic astrocytes, together with their regulation by metabolic hormones, highlights the relevance of these cells in the control of glucose homeostasis. Moreover, the critical role of astrocytes in the response to inflammation, a process associated with obesity and T2DM, further emphasizes the importance of these cells as novel targets to stimulate the CNS in response to metabesity (over-nutrition-derived metabolic dysfunctions). We suggest that novel T2DM therapies should aim at stimulating the CNS astrocytic response, as well as recovering the functional pancreatic ß-cell mass. Whether or not a common factor expressed in both cell types can be feasibly targeted is also discussed.


Subject(s)
Brain/metabolism , Glucose/metabolism , Islets of Langerhans/metabolism , Metabolic Diseases/metabolism , Animals , Astrocytes/metabolism , Energy Metabolism , Homeostasis , Humans
8.
Diabetes ; 68(1): 109-118, 2019 01.
Article in English | MEDLINE | ID: mdl-30352879

ABSTRACT

Transient Pax8 expression was reported in mouse islets during gestation, whereas a genome-wide linkage and admixture mapping study highlighted PAX8 as a candidate gene for diabetes mellitus (DM). We sought the significance of PAX8 expression in mouse and human islet biology. PAX8 was induced in gestating mouse islets and in human islets treated with recombinant prolactin. Global gene expression profiling of human and mouse islets overexpressing the corresponding species-specific PAX8 revealed the modulation of distinct genetic pathways that converge on cell survival. Accordingly, apoptosis was reduced in PAX8-overexpressing islets. These findings support that PAX8 could be a candidate gene for the study of gestational DM (GDM). PAX8 was genotyped in patients with GDM and gestational thyroid dysfunction (GTD), a pathology commonly found in patients with mutations on PAX8 A novel missense PAX8 mutation (p.T356M, c.1067C>T) was identified in a female diagnosed with GDM and GTD as well as in her father with type 2 DM but was absent in control patients. The p.T356M variant did not alter protein stability or cellular localization, whereas its transactivation activity was hindered. In parallel, a retrospective clinical analysis uncovered that a pregnant female harboring a second PAX8 mutation (p.P25R, c.74C>G) previously reported to cause congenital hypothyroidism also developed GDM. These data indicate that increased expression of PAX8 affects islet viability and that PAX8 could be considered as a candidate gene for the study of GDM.


Subject(s)
Diabetes, Gestational/metabolism , PAX8 Transcription Factor/metabolism , Animals , Cell Survival/genetics , Cell Survival/physiology , Diabetes, Gestational/genetics , Female , Genotype , Glucose Tolerance Test , Humans , Immunohistochemistry , Mice, Inbred C57BL , Mutation/genetics , Mutation, Missense/genetics , PAX8 Transcription Factor/genetics , Pedigree , Pregnancy , Retrospective Studies
9.
Curr Opin Pharmacol ; 43: 1-10, 2018 12.
Article in English | MEDLINE | ID: mdl-30048825

ABSTRACT

The high prevalence of diabetes mellitus (DM) in our society, together with the fact that current treatments are only palliative and do not prevent the development of life threatening side effects, highlights the urgent need for novel therapies targeting the root cause of the disease. Independent of the etiology of DM, the definitive therapeutic approach will imply the restitution of an adequate functional ß-cell mass capable of compensating for the insulin demand of the organism. The recent demonstration of heterogeneity within the islets as well as their innate plasticity has encouraged the development of studies aiming at potentiation of the regenerative capacity of islets. In this regard, factors implicated in pancreas ontogeny as well as in the adaptation processes that take place in the islets under situations of increased insulin demand have gained much interest. One of these factors is the transcription factor PAX4, required for ß-cell formation during embryonic development and implicated in adult ß-cell adaptation under stress situations. Here we review the therapeutic potential of PAX4 as well as its downstream targets for the development of novel treatments for DM.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus/drug therapy , Drug Design , Homeodomain Proteins/metabolism , Hypoglycemic Agents/therapeutic use , Insulin-Secreting Cells/drug effects , Paired Box Transcription Factors/metabolism , Animals , Biomarkers/blood , Blood Glucose/metabolism , Cell Plasticity , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Diffusion of Innovation , Humans , Hypoglycemic Agents/adverse effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Molecular Targeted Therapy , Phenotype , Signal Transduction/drug effects
10.
Nat Commun ; 9(1): 1488, 2018 04 16.
Article in English | MEDLINE | ID: mdl-29662071

ABSTRACT

Type 1 diabetes mellitus (T1DM) is due to the selective destruction of islet beta cells by immune cells. Current therapies focused on repressing the immune attack or stimulating beta cell regeneration still have limited clinical efficacy. Therefore, it is timely to identify innovative targets to dampen the immune process, while promoting beta cell survival and function. Liver receptor homologue-1 (LRH-1) is a nuclear receptor that represses inflammation in digestive organs, and protects pancreatic islets against apoptosis. Here, we show that BL001, a small LRH-1 agonist, impedes hyperglycemia progression and the immune-dependent inflammation of pancreas in murine models of T1DM, and beta cell apoptosis in islets of type 2 diabetic patients, while increasing beta cell mass and insulin secretion. Thus, we suggest that LRH-1 agonism favors a dialogue between immune and islet cells, which could be druggable to protect against diabetes mellitus.


Subject(s)
Cell Communication/drug effects , Diabetes Mellitus, Experimental/therapy , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Phenalenes/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/pathology , Female , Gene Expression Regulation , Humans , Immunity, Innate , Insulin/metabolism , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Islets of Langerhans Transplantation , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/immunology , Streptozocin , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Transplantation, Heterologous
11.
Cell Death Dis ; 9(3): 279, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29449530

ABSTRACT

HMG20A (also known as iBRAF) is a chromatin factor involved in neuronal differentiation and maturation. Recently small nucleotide polymorphisms (SNPs) in the HMG20A gene have been linked to type 2 diabetes mellitus (T2DM) yet neither expression nor function of this T2DM candidate gene in islets is known. Herein we demonstrate that HMG20A is expressed in both human and mouse islets and that levels are decreased in islets of T2DM donors as compared to islets from non-diabetic donors. In vitro studies in mouse and human islets demonstrated that glucose transiently increased HMG20A transcript levels, a result also observed in islets of gestating mice. In contrast, HMG20A expression was not altered in islets from diet-induced obese and pre-diabetic mice. The T2DM-associated rs7119 SNP, located in the 3' UTR of the HMG20A transcript reduced the luciferase activity of a reporter construct in the human beta 1.1E7 cell line. Depletion of Hmg20a in the rat INS-1E cell line resulted in decreased expression levels of its neuronal target gene NeuroD whereas Rest and Pax4 were increased. Chromatin immunoprecipitation confirmed the interaction of HMG20A with the Pax4 gene promoter. Expression levels of Mafa, Glucokinase, and Insulin were also inhibited. Furthermore, glucose-induced insulin secretion was blunted in HMG20A-depleted islets. In summary, our data demonstrate that HMG20A expression in islet is essential for metabolism-insulin secretion coupling via the coordinated regulation of key islet-enriched genes such as NeuroD and Mafa and that depletion induces expression of genes such as Pax4 and Rest implicated in beta cell de-differentiation. More importantly we assign to the T2DM-linked rs7119 SNP the functional consequence of reducing HMG20A expression likely translating to impaired beta cell mature function.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/metabolism , High Mobility Group Proteins/metabolism , Insulin-Secreting Cells/metabolism , Polymorphism, Single Nucleotide , 3' Untranslated Regions , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blood Glucose/metabolism , Cell Line, Tumor , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Female , Genetic Predisposition to Disease , High Mobility Group Proteins/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Insulin-Secreting Cells/pathology , Lipids/blood , Male , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Phenotype , Rats
12.
Cell Stress ; 2(6): 141-143, 2018 May 22.
Article in English | MEDLINE | ID: mdl-31225479

ABSTRACT

Type 1 diabetes mellitus (T1DM) is defined as an autoimmune disease that targets the selective destruction of islet insulin-producing beta cells by infiltrating immune cells (insulitis). As a result, the organism is no longer able to produce insulin and develops hyperglycaemia and, if untreated, death. Despite advances in medical device technology and insulin analogues as well as strives in generating in vitro insulin-producing cells, there is still no robust therapy to substitute and protect beta cells that are lost in T1DM. Clinical trials aimed at blocking the immune-mediated beta cell destruction have had moderate success leaving a gap in our understanding of disease aetiology. Such breach in knowledge may stem from the oversight that inhibiting the immune attack likely impairs beta cell regeneration and emphasizes a fundamental paradigm in the approach to treat the disease: A non-mutually exclusive strategy in which the uncontrolled self-directed inflammatory immune response (and not the global immune system) as well as beta cell regeneration are exquisitely fine tuned in order to successfully regain immunological tolerance and restoration of a functional beta cell mass. As such, defining factors that can guide a pro-inflammatory immune cell destructive environment towards an anti-inflammatory environment facilitating beta cell survival and stimulate regeneration would define an unprecedented class of immune-regenerative therapeutic agents for T1DM. In our recent study we identify the liver receptor homolog 1 (LRH-1, also known as NR5A2) as a 'druggable' target that fulfills these criteria restoring glycemic control in various mouse models of T1DM as well as improving human islet survival and function both in vitro and in vivo (Nat Comms, 9:1488).

14.
Genes (Basel) ; 8(3)2017 Mar 09.
Article in English | MEDLINE | ID: mdl-28282933

ABSTRACT

Paired box 4 (PAX4) is a key factor in the generation of insulin producing ß-cells during embryonic development. In adult islets, PAX4 expression is sequestered to a subset of ß-cells that are prone to proliferation and more resistant to stress-induced apoptosis. The importance of this transcription factor for adequate pancreatic islets functionality has been manifested by the association of mutations in PAX4 with the development of diabetes, independently of its etiology. Overexpression of this factor in adult islets stimulates ß-cell proliferation and increases their resistance to apoptosis. Additionally, in an experimental model of autoimmune diabetes, a novel immunomodulatory function for this factor has been suggested. Altogether these data pinpoint at PAX4 as an important target for novel regenerative therapies for diabetes treatment, aiming at the preservation of the remaining ß-cells in parallel to the stimulation of their proliferation to replenish the ß-cell mass lost during the progression of the disease. However, the adequate development of such therapies requires the knowledge of the molecular mechanisms controlling the expression of PAX4 as well as the downstream effectors that could account for PAX4 action.

15.
Expert Opin Ther Targets ; 21(1): 77-89, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27841034

ABSTRACT

INTRODUCTION: Four members of the PAX family, PAX2, PAX4, PAX6 and PAX8 are known to be expressed in the pancreas. Accumulated evidences indicate that several pancreatic expressed PAX genes play a significant role in pancreatic development/functionality and alterations in these genes are involved in the pathogenesis of pancreatic diseases. Areas covered: In this review, we summarize the ongoing research related to pancreatic PAX genes in diabetes mellitus and pancreatic neuroendocrine tumors. We dissect the current knowledge at different levels; from mechanistic studies in cell lines performed to understand the molecular processes controlled by pancreatic PAX genes, to in vivo studies using rodent models that over-express or lack specific PAX genes. Finally, we describe human studies associating variants on pancreatic-expressed PAX genes with pancreatic diseases. Expert opinion: Based on the current literature, we propose that future interventions to treat pancreatic neuroendocrine tumors and diabetes mellitus could be developed via the modulation of PAX4 and/or PAX6 regulated pathways.


Subject(s)
Diabetes Mellitus/genetics , Neuroendocrine Tumors/genetics , Paired Box Transcription Factors/genetics , Pancreatic Neoplasms/genetics , Animals , Cell Line , Diabetes Mellitus/physiopathology , Diabetes Mellitus/therapy , Disease Models, Animal , Gene Expression Regulation , Humans , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/therapy , Pancreas/physiopathology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy
16.
Sci Rep ; 5: 15672, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26503027

ABSTRACT

PAX4 is a key regulator of pancreatic islet development whilst in adult acute overexpression protects ß-cells against stress-induced apoptosis and stimulates proliferation. Nonetheless, sustained PAX4 expression promotes ß-cell dedifferentiation and hyperglycemia, mimicking ß-cell failure in diabetic patients. Herein, we study mechanisms that allow stringent PAX4 regulation endowing favorable ß-cell adaptation in response to changing environment without loss of identity. To this end, PAX4 expression was monitored using a mouse bearing the enhanced green fluorescent protein (GFP) and cre recombinase construct under the control of the islet specific pax4 promoter. GFP was detected in 30% of islet cells predominantly composed of PAX4-enriched ß-cells that responded to glucose-induced insulin secretion. Lineage tracing demonstrated that all islet cells were derived from PAX4(+) progenitor cells but that GFP expression was confined to a subpopulation at birth which declined with age correlating with reduced replication. However, this GFP(+) subpopulation expanded during pregnancy, a state of active ß-cell replication. Accordingly, enhanced proliferation was exclusively detected in GFP(+) cells consistent with cell cycle genes being stimulated in PAX4-overexpressing islets. Under stress conditions, GFP(+) cells were more resistant to apoptosis than their GFP(-) counterparts. Our data suggest PAX4 defines an expandable ß-cell sub population within adult islets.


Subject(s)
Apoptosis/physiology , Gene Expression Regulation/physiology , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/cytology , Paired Box Transcription Factors/metabolism , Animals , Cell Dedifferentiation/physiology , Cell Lineage , Cell Proliferation/physiology , Diabetes Mellitus/pathology , Green Fluorescent Proteins/genetics , Homeodomain Proteins/genetics , Hyperglycemia/pathology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/classification , Mice , Mice, Inbred C57BL , Mice, Transgenic , Paired Box Transcription Factors/genetics , Promoter Regions, Genetic/genetics
17.
Dis Model Mech ; 7(3): 351-62, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24487409

ABSTRACT

Human kidney predominant protein, NCU-G1, is a highly conserved protein with an unknown biological function. Initially described as a nuclear protein, it was later shown to be a bona fide lysosomal integral membrane protein. To gain insight into the physiological function of NCU-G1, mice with no detectable expression of this gene were created using a gene-trap strategy, and Ncu-g1(gt/gt) mice were successfully characterized. Lysosomal disorders are mainly caused by lack of or malfunctioning of proteins in the endosomal-lysosomal pathway. The clinical symptoms vary, but often include liver dysfunction. Persistent liver damage activates fibrogenesis and, if unremedied, eventually leads to liver fibrosis/cirrhosis and death. We demonstrate that the disruption of Ncu-g1 results in spontaneous liver fibrosis in mice as the predominant phenotype. Evidence for an increased rate of hepatic cell death, oxidative stress and active fibrogenesis were detected in Ncu-g1(gt/gt) liver. In addition to collagen deposition, microscopic examination of liver sections revealed accumulation of autofluorescent lipofuscin and iron in Ncu-g1(gt/gt) Kupffer cells. Because only a few transgenic mouse models have been identified with chronic liver injury and spontaneous liver fibrosis development, we propose that the Ncu-g1(gt/gt) mouse could be a valuable new tool in the development of novel treatments for the attenuation of fibrosis due to chronic liver damage.


Subject(s)
Iron/metabolism , Kupffer Cells/metabolism , Lipofuscin/metabolism , Liver Cirrhosis/metabolism , Lysosomes/metabolism , Membrane Proteins/metabolism , Animals , Cathepsin D/metabolism , Cell Death , Collagen/metabolism , Female , Fluorescence , Gene Targeting , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Inflammation/pathology , Kupffer Cells/pathology , Kupffer Cells/ultrastructure , Liver/metabolism , Liver/pathology , Liver Cirrhosis/pathology , Male , Mice, Inbred C57BL , Oxidative Stress , Phenotype , Reproducibility of Results , Splenomegaly/metabolism , Splenomegaly/pathology
19.
Histochem Cell Biol ; 136(5): 595-607, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21932072

ABSTRACT

The paired box transcription factor Pax8 is critical for development of the eye, thyroid gland as well as the urinary and reproductive organs. In adult, Pax8 overexpression is associated with kidney, ovarian and thyroid tumors and has emerged as a specific marker for these cancers. Recently, Pax8 expression was also reported in human pancreatic islets and in neuroendocrine tumors, identifying Pax8 as a novel member of the Pax family expressed in the pancreas. Herein, we sought to provide a comprehensive analysis of Pax8 expression during pancreogenesis and in adult islets. Immunohistochemical analysis using the most employed Pax8 polyclonal antibody revealed strong nuclear staining in the developing mouse pancreas and in mature human and mouse islets. Astonishingly, Pax8 mRNA in mouse islets was undetectable while human islets exhibited low levels. These discrepancies raised the possibility of antibody cross-reactivity. This premise was confirmed by demonstrating that the polyclonal Pax8 antibody also recognized the islet-enriched Pax6 protein both by Western blotting and immunohistochemistry. Thus, in islets polyclonal Pax8 staining corresponds mainly to Pax6. In order to circumvent this caveat, a novel Pax8 monoclonal antibody was used to re-evaluate whether Pax8 was indeed expressed in islets. Surprisingly, Pax8 was not detected in neither the developing pancreas or in mature islets. Reappraisal of pancreatic neuroendocrine tumors using this Pax8 monoclonal antibody exhibited no immunostaining as compared to the Pax8 polyclonal antibody. In conclusion, Pax8 is not expressed in the pancreas and cast doubts on the value of Pax8 as a pancreatic neuroendocrine tumor marker.


Subject(s)
Islets of Langerhans/embryology , Neuroendocrine Tumors/metabolism , Paired Box Transcription Factors/metabolism , Pancreatic Neoplasms/metabolism , Animals , Animals, Newborn , Biomarkers, Tumor/metabolism , Cell Nucleus/metabolism , Cross Reactions , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Humans , Islets of Langerhans/metabolism , Kidney/embryology , Kidney/metabolism , Liver/embryology , Liver/metabolism , Mice , Neuroendocrine Tumors/pathology , PAX8 Transcription Factor , Paired Box Transcription Factors/genetics , Pancreas/embryology , Pancreas/metabolism , Pancreatic Neoplasms/pathology , RNA, Messenger/metabolism
20.
Hum Mol Genet ; 20(14): 2823-33, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21536586

ABSTRACT

Liver receptor homolog (LRH-1) is an orphan nuclear receptor (NR5A2) that regulates cholesterol homeostasis and cell plasticity in endodermal-derived tissues. Estrogen increases LRH-1 expression conveying cell protection and proliferation. Independently, estrogen also protects isolated human islets against cytokine-induced apoptosis. Herein, we demonstrate that LRH-1 is expressed in islets, including ß-cells, and that transcript levels are modulated by 17ß-estradiol through the estrogen receptor (ER)α but not ERß signaling pathway. Repression of LRH-1 by siRNA abrogated the protective effect conveyed by estrogen on rat islets against cytokines. Adenoviral-mediated overexpression of LRH-1 in human islets did not alter proliferation but conferred protection against cytokines and streptozotocin-induced apoptosis. Expression levels of the cell cycle genes cyclin D1 and cyclin E1 as well as the antiapoptotic gene bcl-xl were unaltered in LRH-1 expressing islets. In contrast, the steroidogenic enzymes CYP11A1 and CYP11B1 involved in glucocorticoid biosynthesis were both stimulated in transduced islets. In parallel, graded overexpression of LRH-1 dose-dependently impaired glucose-induced insulin secretion. Our results demonstrate the crucial role of the estrogen target gene nr5a2 in protecting human islets against-stressed-induced apoptosis. We postulate that this effect is mediated through increased glucocorticoid production that blunts the pro-inflammatory response of islets.


Subject(s)
Apoptosis , Gene Expression Regulation , Insulin-Secreting Cells/metabolism , Receptors, Cytoplasmic and Nuclear/biosynthesis , Stress, Physiological , Adenoviridae , Animals , Cell Line, Tumor , Cholesterol/biosynthesis , Cholesterol/genetics , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin E/genetics , Cyclin E/metabolism , Cytokines/genetics , Cytokines/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/genetics , Estrogens/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Mice , Mice, Knockout , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/genetics , Steroid 11-beta-Hydroxylase/genetics , Steroid 11-beta-Hydroxylase/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...