Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Physiol Regul Integr Comp Physiol ; 321(3): R303-R316, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34259034

ABSTRACT

Neonatal antibiotics administered to human infants initiate gut microbiota dysbiosis that may have long-term effects on body weight and metabolism. We examined antibiotic-induced adaptations in pancreatic islets of the piglet, a well-accepted model of human infant microbiota and pancreas development. Neonatal piglets randomized to amoxicillin [30 mg/kg body wt/day; n = 7, antibiotic (ANTI)] or placebo [vehicle control; n = 7, control (CON)] from postnatal day (PND)0-13 were euthanized at PND7, 14, and 49. The metabolic phenotype along with functional, immunohistological, and transcriptional phenotypes of the pancreatic islets were studied. The gut microbiome was characterized by 16S rRNA gene sequencing, and microbial metabolites and microbiome-sensitive host molecules were measured. Compared with CON, ANTI PND7 piglets had elevated transcripts of genes involved in glucagon-like peptide 1 ((GLP-1) synthesis or signaling in islets (P < 0.05) coinciding with higher plasma GLP-1 (P = 0.11), along with increased tumor necrosis factor α (Tnf) (P < 0.05) and protegrin 1 (Npg1) (P < 0.05). Antibiotic-induced relative increases in Escherichia, Coprococcus, Ruminococcus, Dehalobacterium, and Oscillospira of the ileal microbiome at PND7 normalized after antibiotic withdrawal. In ANTI islets at PND14, the expression of key regulators pancreatic and duodenal homeobox 1 (Pdx1), insulin-like growth factor-2 (Igf2), and transcription factor 7-like 2 (Tcf7l2) was downregulated, preceding a 40% reduction of ß-cell area (P < 0.01) and islet insulin content at PND49 (P < 0.05). At PND49, a twofold elevated plasma insulin concentration (P = 0.07) was observed in ANTI compared with CON. We conclude that antibiotic treatment of neonatal piglets elicited gut microbial changes accompanied by phasic alterations in key regulatory genes in pancreatic islets at PND7 and 14. By PND49, reduced ß-cell area and islet insulin content were accompanied by elevated nonfasted insulin despite normoglycemia, indicative of islet stress.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gastrointestinal Microbiome/drug effects , Glucagon-Like Peptide 1/metabolism , Insulin-Secreting Cells/drug effects , Animals , Gastrointestinal Microbiome/physiology , Glucagon/drug effects , Glucagon/metabolism , Insulin/blood , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Swine
2.
J Biol Chem ; 294(13): 4889-4897, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30696767

ABSTRACT

The Nr4a subfamily of nuclear receptor comprises three members in mammalian cells: Nur77/Nr4a1, Nurr1/Nr4a2, and Nor1/Nr4a3. Nr4a proteins play key roles in the regulation of glucose homeostasis in peripheral metabolic tissues. However, their biological functions in ß-cells remain relatively uncharacterized. Here we sought to investigate the potential role of Nor1 in the regulation of ß-cell mass and, in particular, ß-cell survival/apoptosis. We used histological analysis to examine the consequences of genetic deletion of either Nur77 and Nor1 on ß-cell mass, investigated the expression patterns of Nr4as in human islets and INS cells and performed gain- and loss-of-function experiments to further characterize the role of Nor1 in ß-cell apoptosis. Surprisingly, Nor1 knockout mice displayed increased ß-cell mass, whereas mice with genetic deletion of Nur77 did not exhibit any significant differences compared with their WT littermates. The increase in ß-cell mass in Nor1 knockout mice was accompanied by improved glucose tolerance. A gene expression study performed in both human islets and INS cells revealed that Nor1 expression is significantly increased by pro-inflammatory cytokines and, to a lesser extent, by elevated concentrations of glucose. Nor1 overexpression in both INS and human islet cells caused apoptosis, whereas siRNA-mediated Nor1 knockdown prevented cytokine-induced ß-cell death. Finally, Nor1 expression was up-regulated in islets of individuals with type 2 diabetes. Altogether, our results uncover that Nor1 negatively regulates ß-cell mass. Nor1 represents a promising molecular target in diabetes treatment to prevent ß-cell destruction.


Subject(s)
Apoptosis , DNA-Binding Proteins/biosynthesis , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Nerve Tissue Proteins/biosynthesis , Receptors, Steroid/biosynthesis , Receptors, Thyroid Hormone/biosynthesis , Up-Regulation , Animals , Cytokines , DNA-Binding Proteins/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Humans , Insulin-Secreting Cells/pathology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics
3.
J Lipid Res ; 59(9): 1695-1708, 2018 09.
Article in English | MEDLINE | ID: mdl-30007917

ABSTRACT

De novo phosphatidylcholine (PC) synthesis via CTP:phosphocholine cytidylyltransferase-α (CTα) is required for VLDL secretion. To determine the precise role of de novo PC synthesis in intestinal lipid metabolism, we deleted CTα exclusively in the intestinal epithelium of mice (CTαIKO mice). When fed a chow diet, CTαIKO mice showed normal fat absorption despite a ∼30% decrease in intestinal PC concentrations relative to control mice, suggesting that biliary PC can fully support chylomicron secretion under these conditions. However, when fed a high-fat diet, CTαIKO mice showed impaired passage of FAs and cholesterol from the intestinal lumen into enterocytes. Impaired intestinal lipid uptake in CTαIKO mice was associated with lower plasma triglyceride concentrations, higher plasma glucagon-like peptide 1 and peptide YY, and disruption of intestinal membrane lipid transporters after a high-fat meal relative to control mice. Unexpectedly, biliary bile acid and PC secretion was enhanced in CTαIKO mice due to a shift in expression of bile-acid transporters to the proximal intestine, indicative of accelerated enterohepatic cycling. These data show that intestinal de novo PC synthesis is required for dietary lipid absorption during high-fat feeding and that the reacylation of biliary lyso-PC cannot compensate for loss of CTα under these conditions.


Subject(s)
Dietary Fats/metabolism , Homeostasis/drug effects , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Phosphatidylcholines/biosynthesis , Animals , Biological Transport/drug effects , Body Weight/drug effects , Cholesterol/metabolism , Choline-Phosphate Cytidylyltransferase/deficiency , Choline-Phosphate Cytidylyltransferase/genetics , Choline-Phosphate Cytidylyltransferase/metabolism , Diet, High-Fat/adverse effects , Dietary Fats/adverse effects , Gene Knockout Techniques , Mice , Mice, Inbred C57BL
4.
Diabetologia ; 60(8): 1432-1441, 2017 08.
Article in English | MEDLINE | ID: mdl-28547133

ABSTRACT

AIMS/HYPOTHESIS: Heat shock factor protein 1 (HSF1) is a transcription factor that regulates the expression of key molecular chaperones, thereby orchestrating the cellular response to stress. This system was recently implicated in the control of insulin sensitivity and is therefore being scrutinised as a novel therapeutic avenue for type 2 diabetes. However, the regulation and biological actions of HSF1 in beta cells remain elusive. Herein, we sought to investigate the regulation of HSF1 in pancreatic beta cells and to study its potential role in cell survival. METHODS: We exposed human islets and beta cell lines to glucolipotoxicity and thapsigargin. HSF1 activity was evaluated by gel shift assay. HSF1 acetylation and interaction with the protein acetylase cAMP response element binding protein (CBP) were investigated by western blot. We measured the expression of HSF1 and its canonical targets in islets from Goto-Kakizaki (GK) rat models of diabetes and delineated the effects of HSF1 acetylation using mutants mimicking constitutive acetylation and deacetylation of the protein. RESULTS: Glucolipotoxicity promoted HSF1 acetylation and interaction with CBP. Glucolipotoxicity-induced HSF1 acetylation inhibited HSF1 DNA binding activity and decreased the expression of its target genes. Restoration of HSF1 activity in beta cells prevented glucolipotoxicity-induced endoplasmic reticulum stress and apoptosis. However, overexpression of a mutant protein (K80Q) mimicking constitutive acetylation of HSF1 failed to confer protection against glucolipotoxicity. Finally, we showed that expression of HSF1 and its target genes were altered in islets from diabetic GK rats, suggesting that this pathway could participate in the pathophysiology of diabetes and constitutes a potential site for therapeutic intervention. CONCLUSIONS/INTERPRETATION: Our results unravel a new mechanism by which HSF1 inhibition is required for glucolipotoxicity-induced beta cell apoptosis. Restoring HSF1 activity may represent a novel strategy for the maintenance of a functional beta cell mass. Our study supports the therapeutic potential of HSF1/heat shock protein-targeting agents in diabetes treatment.


Subject(s)
Glucose/pharmacology , Heat Shock Transcription Factors/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Palmitic Acid/pharmacology , Transcription Factors/metabolism , Acetylation/drug effects , Animals , Apoptosis/genetics , Apoptosis/physiology , Chromatin Immunoprecipitation , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 2/metabolism , Electrophoretic Mobility Shift Assay , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Heat Shock Transcription Factors/genetics , Humans , Male , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Rats
5.
J Biol Chem ; 289(12): 8413-9, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24509857

ABSTRACT

The tumor suppressor gene ST18 was originally characterized as the third member of the neural zinc finger transcription factor family. However, little is known about its biological functions. Herein, we demonstrate that, in the pancreas, ST18 expression is restricted to endocrine cells. The detection of ST18 expression in pancreatic ß-cells prompted us to investigate its regulation and its role in ß-cell mass and function. We show that ST18 expression and activity are increased by cytotoxic concentrations of fatty acids and cytokines in INS832/13 cells. Furthermore, ST18 is also increased in islets of diet-induced obese animals. Overexpression and RNA interference knockdown studies demonstrate that ST18 induces ß-cell apoptosis and curtails ß-cell replication. Finally, our data suggest that ST18 impairs insulin secretion. Taken together, our findings indicate that ST18 could represent a novel transcriptional mediator of lipotoxicity and cytokine-induced ß-cell death. We suggest that genetic or pharmacologic manipulations of ST18 could help maintain a functional ß-cell mass.


Subject(s)
Apoptosis , DNA-Binding Proteins/metabolism , Insulin-Secreting Cells/cytology , Nerve Tissue Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Cells, Cultured , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Gene Expression Regulation , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Obesity/metabolism , Rats , Rats, Wistar , Repressor Proteins/analysis , Repressor Proteins/genetics , Transcription Factors/analysis , Transcription Factors/genetics , Zinc Fingers
6.
Blood Purif ; 39(1-3): 145-50, 2015.
Article in English | MEDLINE | ID: mdl-25672966

ABSTRACT

According to the World Health Organization reports, nowadays burden of chronic kidney diseases (CKD) is well documented. The high prevalence of noncommunicable diseases (NCD) such as hypertension, diabetes, and obesity, which are the main causes of CKD, is a big concern in the world health scenario. These NCD can progress slowly to end-stage renal disease (ESRD) and the low-middle income countries (LMIC) like Haiti are not left unscathed by this worldwide scourge. Several well-known public health issues prevalent in Haiti such as acute diarrheal infections, malaria, tuberculosis, cholera, and acquired immunodeficiency syndrome (AIDS), can also impair the function of the kidney. Dialysis, a form of renal replacement therapy (RRT), represents a life-saving therapy for all patients affected with impaired kidney. In Haiti, few patients have access to health insurance or disability financial support. Considering that seventy-two percent (72%) of Haitians live with less than USD 2 per day, survival with CKD can be quite stressful for them. Data on the weight of the dialysis and its management are scarce. Addressing the need for dialysis in Haiti is an important component in decision-making and planning processes in the health sector. This paper is intended to bring forth discussion on the use of this type of renal replacement therapy in Haiti: the past, the present, and the challenges it presents. We will also make some recommendations in order to manage this serious problem.


Subject(s)
Kidney Failure, Chronic/economics , Kidney Failure, Chronic/therapy , Poverty , Renal Dialysis/statistics & numerical data , Adult , Child , Communicable Diseases/complications , Communicable Diseases/economics , Communicable Diseases/epidemiology , Diarrhea/complications , Diarrhea/economics , Diarrhea/epidemiology , Female , Haiti/epidemiology , Health Services Accessibility/statistics & numerical data , Humans , Kidney Failure, Chronic/epidemiology , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/pathology , Male , Medically Uninsured/statistics & numerical data , Prevalence , Public Health/economics , Public Health/statistics & numerical data , Renal Dialysis/economics
7.
Am J Physiol Gastrointest Liver Physiol ; 306(11): G938-46, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24699330

ABSTRACT

The leucine-to-glutamate (Leu→Glu) pathway, which metabolizes the carbon atoms of l-leucine to form l-glutamate, was studied by incubation of rat tissue segments with l-[U-(14)C]leucine and estimation of the [(14)C]glutamate formed. Metabolism of the leucine carbon chain occurs in most rat tissues, but maximal activity of the Leu→Glu pathway for glutamate formation is limited to the thoracic aorta and pancreas. In rat aorta, the Leu→Glu pathway functions to relax the underlying smooth muscle; its functions in the pancreas are unknown. This report characterizes the Leu→Glu pathway of rat pancreas and develops methods to examine its functions. Pancreatic segments effect net formation of glutamate on incubation with l-leucine, l-glutamine, or a mix of 18 other plasma amino acids at their concentrations in normal rat plasma. Glutamate formed from leucine remains mainly in the tissue, whereas that from glutamine enters the medium. The pancreatic Leu→Glu pathway uses the leucine carbons for net glutamate formation; the α-amino group is not used; the stoichiometry is as follows: 1 mol of leucine yields 2 mol of glutamate (2 leucine carbons per glutamate) plus 2 mol of CO2. Comparison of the Leu→Glu pathway in preparations of whole pancreatic segments, isolated acini, and islets of Langerhans localizes it in the acini; relatively high activity is found in cultures of the AR42J cell line and very little in the INS-1 832/13 cell line. Pancreatic tissue glutamate concentration is homeostatically regulated in the range of ∼1-3 µmol/g wet wt. l-Valine and leucine ethyl, benzyl, and tert-butyl esters inhibit the Leu→Glu pathway without decreasing tissue total glutamate.


Subject(s)
Glutamic Acid/biosynthesis , Leucine/metabolism , Pancreas/metabolism , Animals , Aorta/metabolism , Carbon Radioisotopes , Cell Line, Tumor , Male , Rats , Rats, Sprague-Dawley
8.
Nat Med ; 13(10): 1203-10, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17873882

ABSTRACT

Gain-of-function mutations in NOTCH1 are common in T-cell lymphoblastic leukemias and lymphomas (T-ALL), making this receptor a promising target for drugs such as gamma-secretase inhibitors, which block a proteolytic cleavage required for NOTCH1 activation. However, the enthusiasm for these therapies has been tempered by tumor resistance and the paucity of information on the oncogenic programs regulated by oncogenic NOTCH1. Here we show that NOTCH1 regulates the expression of PTEN (encoding phosphatase and tensin homolog) and the activity of the phosphoinositol-3 kinase (PI3K)-AKT signaling pathway in normal and leukemic T cells. Notch signaling and the PI3K-AKT pathway synergize in vivo in a Drosophila melanogaster model of Notch-induced tumorigenesis, and mutational loss of PTEN is associated with human T-ALL resistance to pharmacological inhibition of NOTCH1. Overall, these findings identify transcriptional control of PTEN and regulation of the PI3K-AKT pathway as key elements of the leukemogenic program activated by NOTCH1 and provide the basis for the design of new therapeutic strategies for T-ALL.


Subject(s)
Drosophila Proteins/genetics , Gene Expression Regulation, Leukemic/genetics , Leukemia, T-Cell/metabolism , PTEN Phosphohydrolase/genetics , Receptor, Notch1/antagonists & inhibitors , Animals , DNA Mutational Analysis , Disease Models, Animal , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/metabolism , Female , Humans , Leukemia, T-Cell/genetics , Mice , Models, Genetic , Mutation , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Pregnancy , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Signal Transduction , Transgenes
10.
JBJS Rev ; 12(8)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39102470

ABSTRACT

BACKGROUND: Bone radiation-induced sarcomas (B-RIS) are secondary neoplasms with reportedly worse overall survival than de novo bone sarcoma. Treatment strategy for these neoplasms remains uncertain. Our systematic review sought to assess overall survival based on histology and surgical intervention. METHODS: A systemic review was conducted following Preferred Reporting Items for Systematic reviews and Meta-Analyses guidelines and registered in PROSPERO (438415). Studies describing oncologic outcomes of patients with B-RIS in the appendicular and axial skeleton were included. The Strengthening the Reporting of Observational Studies in Epidemiology checklist was used for quality assessment. Survival analysis by histologic subtype and surgery type was performed in a subset of 234 patients from 11 articles with individualized data. A total of 20 articles with a total of 566 patients were included. The most frequent location was the pelvis (27.7%), and the main histological types were osteosarcoma (69.4%), undifferentiated pleomorphic sarcoma (14.1%), and fibrosarcoma (9.2%). Limb-salvage and amputation were performed in 68.5% and 31.5% of cases, respectively. RESULTS: Local recurrence was 13%, without difference between limb-salvage surgery and amputation (p = 0.51). The metastasis rate was 42.3%. Five-year OS was 43.7% (95% confidence interval [CI], 33.3%-53.5%) for osteosarcoma, 31.5% (95% CI, 11.3%-54.2%) for UPS, and 28.1% (95% CI, 10.6%-48.8%) for fibrosarcoma. Five-year OS was 49.2% (95% CI, 35.3%-61.6%) for limb-salvage and 46.9% (95% CI, 29.1%-62.9%) for amputation. There was no difference in 5-year OS between histologic subtypes (p = 0.18) or treatment type (p = 0.86). CONCLUSION: B-RIS demonstrated poor OS at 5 years after initial management regardless of histology. Limb-salvage surgery was not associated with lower 5-year OS compared with amputation. Future studies should compare both groups while controlling for confounders. LEVEL OF EVIDENCE: Level III. See Instructions for Authors for a complete description of levels of evidence.


Subject(s)
Bone Neoplasms , Neoplasms, Radiation-Induced , Sarcoma , Humans , Bone Neoplasms/radiotherapy , Bone Neoplasms/surgery , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Sarcoma/radiotherapy , Sarcoma/pathology , Sarcoma/surgery , Sarcoma/mortality , Neoplasms, Radiation-Induced/pathology , Neoplasms, Radiation-Induced/surgery , Neoplasms, Radiation-Induced/etiology , Limb Salvage , Male , Female , Osteosarcoma/pathology , Osteosarcoma/mortality , Osteosarcoma/surgery , Osteosarcoma/radiotherapy , Adult , Treatment Outcome , Middle Aged , Adolescent
11.
Article in English | MEDLINE | ID: mdl-35007755

ABSTRACT

Cytidine triphosphate:phosphocholine cytidylyltransferase-α (CTα) is the rate limiting enzyme in the major pathway for de novo phosphatidylcholine (PC) synthesis. When CTα is deleted specifically in intestinal epithelial cells of adult mice (CTαIKO mice) fed a high-fat diet they present with weight loss, lipid malabsorption, and high postprandial GLP-1 levels. The current study aimed to characterize the changes that occur in the small intestines of CTαIKO mice using transcriptomics and to determine whether intestinal function could be rescued in CTαIKO mice. We found that impaired de novo PC synthesis in the gut is linked to lower abundance of transcripts related to lipid metabolism and higher abundance of transcripts related to ER stress and cell death, together with loss of goblet cells from the small intestinal epithelium. Furthermore, impaired movement of fatty acids from the intestinal lumen into enterocytes was observed in isolated intestinal sacs derived from CTαIKO mice, a model that excludes factors such as bile, gastric emptying, the nervous system, and circulating hormones. Antibiotic treatment prevented acute weight loss and normalized jejunum TG concentrations after refeeding but did not prevent ER stress or loss of goblet cells in CTαIKO mice. Dietary PC supplementation partially prevented loss of goblet cells but was unable to normalize jejunal TG concentrations after refeeding in CTαIKO mice. High postprandial plasma GLP-1 levels were present in CTαIKO mice regardless of antibiotic treatment, dietary PC content, or dietary fat content. Together, these data show that there is a specific requirement from de novo PC synthesis in maintaining small intestinal homeostasis, including dietary lipid uptake, normal hormone secretion, and barrier function.


Subject(s)
Dietary Fats , Phosphatidylcholines , Animals , Anti-Bacterial Agents , Dietary Fats/metabolism , Glucagon-Like Peptide 1/metabolism , Intestinal Mucosa/metabolism , Mice , Phosphatidylcholines/metabolism , Weight Loss
12.
Int Med Case Rep J ; 14: 533-538, 2021.
Article in English | MEDLINE | ID: mdl-34408503

ABSTRACT

OBJECTIVE: Frasier syndrome is a rare genetic nephropathy characterized by the presence of progressive glomerulopathy with proteinuria associated with male pseudo hermaphroditism. This case study described a picture of a young boy where the clinical suspicion context reminded the Frasier syndrome. To our knowledge, this case is the first described in Haiti. CASE STUDY: This is a 19-year-old young phenotypically male, born with a genital anomaly, was seen on referral at the nephrology/dialysis unit of the internal medicine department of the State University Hospital of Haiti for evaluation and follow-up. Insidious progression of symptoms had occurred over 3 years. Over three months of outpatient follow-up, he had four sets of renal labs drawn, and all showed impaired renal function. At the ultrasound, a bilateral cryptorchidism is described in the inguinal, and presence of functional ovaries with follicles of variable size scattered in the parenchyma. So, in the light of these anamnestic, clinical and paraclinical findings, we concluded to the diagnosis of end-stage renal failure by progressive glomerulopathy in a context of Frasier's syndrome. CONCLUSION: With any clinical picture consisting of genital anomalies at birth, renal symptomatology during childhood and the diagnosis of renal failure during adolescence, rare genetic nephropathies, such as Frasier syndrome must be considered.

13.
Cell Mol Gastroenterol Hepatol ; 11(4): 999-1021, 2021.
Article in English | MEDLINE | ID: mdl-33238221

ABSTRACT

BACKGROUND & AIMS: Patients with ulcerative colitis have low concentrations of the major membrane lipid phosphatidylcholine (PC) in gastrointestinal mucus, suggesting that defects in colonic PC metabolism might be involved in the development of colitis. To determine the precise role that PC plays in colonic barrier function, we examined mice with intestinal epithelial cell (IEC)-specific deletion of the rate-limiting enzyme in the major pathway for PC synthesis: cytidine triphosphate:phosphocholine cytidylyltransferase-α (CTαIKO mice). METHODS: Colonic tissue of CTαIKO mice and control mice was analyzed by histology, immunofluorescence, electron microscopy, quantitative polymerase chain reaction, Western blot, and thin-layer chromatography. Histopathologic colitis scores were assigned by a pathologist blinded to the experimental groupings. Intestinal permeability was assessed by fluorescein isothiocyanate-dextran gavage and fecal microbial composition was analyzed by sequencing 16s ribosomal RNA amplicons. Subsets of CTαIKO mice and control mice were treated with dietary PC supplementation, antibiotics, or 4-phenylbutyrate. RESULTS: Inducible loss of CTα in the intestinal epithelium reduced colonic PC concentrations and resulted in rapid and spontaneous colitis with 100% penetrance in adult mice. Colitis development in CTαIKO mice was traced to a severe and unresolving endoplasmic reticulum stress response in IECs with altered membrane phospholipid composition. This endoplasmic reticulum stress response was linked to the necroptotic death of IECs, leading to excessive loss of goblet cells, formation of a thin mucus barrier, increased intestinal permeability, and infiltration of the epithelium by microbes. CONCLUSIONS: Maintaining the PC content of IEC membranes protects against colitis development in mice, showing a crucial role for IEC phospholipid equilibrium in colonic homeostasis. SRA accession number: PRJNA562603.


Subject(s)
Choline-Phosphate Cytidylyltransferase/pharmacology , Colitis/pathology , Endoplasmic Reticulum Stress , Goblet Cells/pathology , Intestinal Mucosa/pathology , Necroptosis , Phosphatidylcholines/metabolism , Animals , Colitis/chemically induced , Colitis/immunology , Colitis/metabolism , Dextran Sulfate/toxicity , Female , Gastrointestinal Microbiome , Homeostasis , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Permeability
14.
Diabetes ; 70(11): 2626-2638, 2021 11.
Article in English | MEDLINE | ID: mdl-34462260

ABSTRACT

SUMOylation reduces oxidative stress and preserves islet mass at the expense of robust insulin secretion. To investigate a role for the deSUMOylating enzyme sentrin-specific protease 1 (SENP1) following metabolic stress, we put pancreas/gut-specific SENP1 knockout (pSENP1-KO) mice on a high-fat diet (HFD). Male pSENP1-KO mice were more glucose intolerant following HFD than littermate controls but only in response to oral glucose. A similar phenotype was observed in females. Plasma glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide 1 (GLP-1) responses were identical in pSENP1-KO and wild-type littermates, including the HFD-induced upregulation of GIP responses. Islet mass was not different, but insulin secretion and ß-cell exocytotic responses to the GLP-1 receptor agonist exendin-4 (Ex4) and GIP were impaired in islets lacking SENP1. Glucagon secretion from pSENP1-KO islets was also reduced, so we generated ß-cell-specific SENP1 KO mice. These phenocopied the pSENP1-KO mice with selective impairment in oral glucose tolerance following HFD, preserved islet mass expansion, and impaired ß-cell exocytosis and insulin secretion to Ex4 and GIP without changes in cAMP or Ca2+ levels. Thus, ß-cell SENP1 limits oral glucose intolerance following HFD by ensuring robust insulin secretion at a point downstream of incretin signaling.


Subject(s)
Cysteine Endopeptidases/metabolism , Diet, High-Fat/adverse effects , Gene Expression Regulation/drug effects , Insulin-Secreting Cells/metabolism , Animals , Cysteine Endopeptidases/genetics , Glucose/pharmacology , Glucose Intolerance , Glucose Tolerance Test , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Incretins , Insulin, Regular, Human/pharmacology , Mice , Mice, Knockout , Trans-Activators/genetics , Trans-Activators/metabolism
15.
Cells ; 9(1)2020 01 09.
Article in English | MEDLINE | ID: mdl-31936632

ABSTRACT

Nor1, the third member of the Nr4a subfamily of nuclear receptor, is garnering increased interest in view of its role in the regulation of glucose homeostasis. Our previous study highlighted a proapoptotic role of Nor1 in pancreatic beta cells and showed that Nor1 expression was increased in islets isolated from type 2 diabetic individuals, suggesting that Nor1 could mediate the deterioration of islet function in type 2 diabetes. However, the mechanism remains incompletely understood. We herein investigated the subcellular localization of Nor1 in INS832/13 cells and dispersed human beta cells. We also examined the consequences of Nor1 overexpression on mitochondrial function and morphology. Our results show that, surprisingly, Nor1 is mostly cytoplasmic in beta cells and undergoes mitochondrial translocation upon activation by proinflammatory cytokines. Mitochondrial localization of Nor1 reduced glucose oxidation, lowered ATP production rates, and inhibited glucose-stimulated insulin secretion. Western blot and microscopy images revealed that Nor1 could provoke mitochondrial fragmentation via mitophagy. Our study unveils a new mode of action for Nor1, which affects beta-cell viability and function by disrupting mitochondrial networks.


Subject(s)
Insulin-Secreting Cells/metabolism , Mitochondria/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 3/metabolism , Cell Line , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Insulin Secretion , Insulin-Secreting Cells/ultrastructure , Mitochondria/ultrastructure , Mitophagy , Oxidation-Reduction
16.
Child Abuse Negl ; 89: 178-191, 2019 03.
Article in English | MEDLINE | ID: mdl-30685625

ABSTRACT

OBJECTIVE: Interpersonal violence affecting women and children is increasingly recognized as a public health priority in humanitarian emergencies. Yet, research and intervention efforts have been fragmented across gender-based violence and child protection sectors. Using data from the Transforming Households: Reducing Incidence of Violence in Emergencies (THRIVE) project, this study sought to qualitatively investigate the intersecting drivers of multiple forms of violence in Côteaux, Haiti, while obtaining insight on how these drivers may be influenced by a humanitarian emergency. METHODS: This analysis used transcripts obtained using a photo elicitation approach over the course of three sessions per person. Thirty-six individuals participated in the study: eight adult females, ten adult males, eight adolescent females, ten adolescent males. Participants were given cameras to capture images related to family relationships, family safety, and changes to family dynamics due to Hurricane Matthew and its aftermath. In subsequent sessions, these photographs were used as prompts for qualitative interviews. RESULTS: Multiple and converging drivers of interpersonal violence were identified including the accumulation of daily stressors, loss of power/control, learned behavior (intergenerational cycle of abuse), and inequitable gender norms, all of which were influenced by the humanitarian context caused by Hurricane Matthew. CONCLUSIONS: Our findings suggest multiple and converging drivers of violence may be exacerbated in times of crises, requiring interdisciplinary responses. In order to comprehensively address the drivers of violence, practitioners and policy makers should consider the needs of individuals and their families holistically, integrating community-led, gender transformative efforts and positive parenting with basic needs provision.


Subject(s)
Altruism , Child Abuse/psychology , Cyclonic Storms , Violence/psychology , Adolescent , Adult , Adverse Childhood Experiences , Child , Family Characteristics , Female , Haiti , Humans , Interpersonal Relations , Male , Qualitative Research , Violence/statistics & numerical data
17.
Neurochem Res ; 33(9): 1845-51, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18351462

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is a glucoincretin hormone most intensively studied for its actions on insulin secreting beta-cells. GLP-1 and its receptor are also found in brain and accumulating evidence indicates that GLP-1 has neuroprotective actions. Here, we investigated whether GLP-1 protects neuronal cells from death evoked by nerve growth factor (NGF) withdrawal. Compromised trophic factor signaling may underlie neurodegenerative diseases ranging from Alzheimer disease to diabetic neuropathies. We report that GLP-1 provides sustained protection of cultured neuronal PC12 cells and sympathetic neurons from degeneration and death caused by NGF deprivation. Past work shows that NGF deprivation induces the pro-apoptotic protein Bim which contributes to neuron death. Here, we find that GLP-1 suppresses Bim induction promoted by NGF deprivation. Thus, GLP-1 may protect neurons, at least in part, by suppressing Bim induction. Our findings support the idea that drugs that mimic or elevate GLP-1 represent potential therapeutics for neurodegenerative diseases.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cell Death/physiology , Glucagon-Like Peptide 1/metabolism , Membrane Proteins/metabolism , Nerve Growth Factor/metabolism , Neurons/physiology , Neuroprotective Agents/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Bcl-2-Like Protein 11 , Cells, Cultured , Humans , Neurons/cytology , Neurons/pathology , PC12 Cells , Rats
18.
Islets ; 10(6): 213-220, 2018.
Article in English | MEDLINE | ID: mdl-30311843

ABSTRACT

Hyperinsulinemic hypoglycemia syndrome (HIHG) is a rare complication of roux-en-Y gastric bypass surgery. The pathology is associated with an excessive function of pancreatic beta-cells, and requires pancreas resection in patients that are recalcitrant to nutritional and pharmacological interventions. The exact prevalence is not clearly understood and the underlying mechanisms not yet fully characterized. We herein sought to perform histological and molecular examination of pancreatic sections obtained from a patient who developed HIHG as a complication of gastric bypass compared to 3 weight-matched controls. We studied markers of cellular replication and beta-cell differentiation by immunohistochemistry and immunofluorescence. HIHG after gastric bypass was characterized by a profound increase in beta-cell mass. Cellular proliferation was increased in islets and ducts compared to controls, suggesting unrestrained proliferation in HIHG. We also detected beta-cell differentiation markers in duct cells and occasional duct cells displaying both insulin and glucagon immunoreactivity. These histological observations suggest that beta-cell differentiation from ductal progenitor cells could also underly beta-cell mass expansion in HIHG. Altogether, our results can be construed to demonstrate that HIHG after gastric bypass is characterized by abnormal beta-cell mass expansion, resulting from both unrestrained beta-cell replication and neogenesis.


Subject(s)
Cell Proliferation/physiology , Gastric Bypass/adverse effects , Hyperinsulinism/pathology , Hypoglycemia/pathology , Insulin-Secreting Cells/pathology , Obesity, Morbid/surgery , Adult , Humans , Hyperinsulinism/etiology , Hyperinsulinism/surgery , Hypoglycemia/etiology , Hypoglycemia/surgery , Male , Obesity, Morbid/pathology , Postoperative Complications/pathology
19.
Diabetes ; 55(5): 1190-6, 2006 May.
Article in English | MEDLINE | ID: mdl-16644672

ABSTRACT

The glucoincretin hormone glucagon-like peptide-1 (GLP-1) increases pancreatic beta-cell proliferation and survival through sequential activation of the epidermal growth factor receptor (EGFR), phosphatidylinositol-3 kinase (PI 3-kinase), and Akt. We investigated the role of transcription factor FoxO1 in the proliferative and antiapoptotic actions of GLP-1 in beta-cells. GLP-1 inhibited FoxO1 through phosphorylation-dependent nuclear exclusion in pancreatic beta (INS832/13) cells. The effect of GLP-1 was suppressed by inhibitors of EGFR (AG1478) and PI 3-kinase (LY294002). In contrast, LY294002 but not AG1478 suppressed insulin-induced FoxO1 phosphorylation. Expression of constitutively nuclear FoxO1 in beta-cells prevented the proliferative and antiapoptotic actions of GLP-1 in cultured beta-cells and the increase in pancreatic beta-cell mass in response to Exendin4 in transgenic mice. Gene expression and chromatin immunoprecipitation assays demonstrated that GLP-1 increases pancreatic and duodenal homeobox gene-1 and Foxa2 expression and inhibits FoxO1 binding to both promoters. We propose that FoxO1 mediates the pleiotropic effects of the glucoincretin hormone on cell proliferation and survival.


Subject(s)
Forkhead Transcription Factors/physiology , Glucagon-Like Peptides/pharmacology , Glucagon/pharmacology , Peptide Fragments/pharmacology , Animals , Cell Division , Cell Line , Cell Survival , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Glucagon-Like Peptide 1 , Glucose/physiology , Green Fluorescent Proteins/genetics , Insulin/pharmacology , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/physiology , Mice , Mice, Transgenic
20.
Mol Cell Biol ; 24(13): 5721-32, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15199129

ABSTRACT

Activating transcription factor 3 (ATF3) is a stress-inducible gene and encodes a member of the ATF/CREB family of transcription factors. However, the physiological significance of ATF3 induction by stress signals is not clear. In this report, we describe several lines of evidence supporting a role of ATF3 in stress-induced beta-cell apoptosis. First, ATF3 is induced in beta cells by signals relevant to beta-cell destruction: proinflammatory cytokines, nitric oxide, and high concentrations of glucose and palmitate. Second, induction of ATF3 is mediated in part by the NF-kappaB and Jun N-terminal kinase/stress-activated protein kinase signaling pathways, two stress-induced pathways implicated in both type 1 and type 2 diabetes. Third, transgenic mice expressing ATF3 in beta cells develop abnormal islets and defects secondary to beta-cell deficiency. Fourth, ATF3 knockout islets are partially protected from cytokine- or nitric oxide-induced apoptosis. Fifth, ATF3 is expressed in the islets of patients with type 1 or type 2 diabetes, and in the islets of nonobese diabetic mice that have developed insulitis or diabetes. Taken together, our results suggest ATF3 to be a novel regulator of stress-induced beta-cell apoptosis.


Subject(s)
Apoptosis , Islets of Langerhans/pathology , Stress, Physiological , Transcription Factors/physiology , Activating Transcription Factor 3 , Animals , Apoptosis/genetics , Cytokines/pharmacology , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Gene Expression Regulation/drug effects , Glucagon/analysis , Insulin/analysis , Islets of Langerhans/chemistry , Mice , Mice, Knockout , Mice, Transgenic , Pancreas/chemistry , Pancreas/pathology , Signal Transduction , Transcription Factors/analysis , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL