Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Diabetes ; 67(7): 1310-1321, 2018 07.
Article in English | MEDLINE | ID: mdl-29728363

ABSTRACT

In type 2 diabetes (T2D), hepatic insulin resistance is strongly associated with nonalcoholic fatty liver disease (NAFLD). In this study, we hypothesized that the DNA methylome of livers from patients with T2D compared with livers of individuals with normal plasma glucose levels can unveil some mechanism of hepatic insulin resistance that could link to NAFLD. Using DNA methylome and transcriptome analyses of livers from obese individuals, we found that hypomethylation at a CpG site in PDGFA (encoding platelet-derived growth factor α) and PDGFA overexpression are both associated with increased T2D risk, hyperinsulinemia, increased insulin resistance, and increased steatohepatitis risk. Genetic risk score studies and human cell modeling pointed to a causative effect of high insulin levels on PDGFA CpG site hypomethylation, PDGFA overexpression, and increased PDGF-AA secretion from the liver. We found that PDGF-AA secretion further stimulates its own expression through protein kinase C activity and contributes to insulin resistance through decreased expression of insulin receptor substrate 1 and of insulin receptor. Importantly, hepatocyte insulin sensitivity can be restored by PDGF-AA-blocking antibodies, PDGF receptor inhibitors, and by metformin, opening therapeutic avenues. Therefore, in the liver of obese patients with T2D, the increased PDGF-AA signaling contributes to insulin resistance, opening new therapeutic avenues against T2D and possibly NAFLD.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin Resistance , Liver/metabolism , Obesity/metabolism , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Adult , Case-Control Studies , Cells, Cultured , DNA Methylation , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Epigenesis, Genetic/physiology , Female , Genetic Predisposition to Disease , Humans , Insulin Resistance/genetics , Liver/pathology , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Obesity/complications , Obesity/genetics , Obesity/pathology , Signal Transduction/genetics , Up-Regulation/genetics
2.
Nat Genet ; 50(2): 175-179, 2018 02.
Article in English | MEDLINE | ID: mdl-29311637

ABSTRACT

Study of monogenic forms of obesity has demonstrated the pivotal role of the central leptin-melanocortin pathway in controlling energy balance, appetite and body weight 1 . The majority of loss-of-function mutations (mostly recessive or co-dominant) have been identified in genes that are directly involved in leptin-melanocortin signaling. These genes, however, only explain obesity in <5% of cases, predominantly from outbred populations 2 . We previously showed that, in a consanguineous population in Pakistan, recessive mutations in known obesity-related genes explain ~30% of cases with severe obesity3-5. These data suggested that new monogenic forms of obesity could also be identified in this population. Here we identify and functionally characterize homozygous mutations in the ADCY3 gene encoding adenylate cyclase 3 in children with severe obesity from consanguineous Pakistani families, as well as compound heterozygous mutations in a severely obese child of European-American descent. These findings highlight ADCY3 as an important mediator of energy homeostasis and an attractive pharmacological target in the treatment of obesity.


Subject(s)
Adenylyl Cyclases/genetics , Loss of Function Mutation , Obesity, Morbid/genetics , Adenylyl Cyclases/chemistry , Adolescent , Animals , Case-Control Studies , Cells, Cultured , Child , Cohort Studies , Consanguinity , Cricetinae , Energy Metabolism/genetics , Female , Gene Frequency , Genetic Predisposition to Disease , Homozygote , Humans , Male , Mice , Mice, Knockout , Models, Molecular , Obesity, Morbid/epidemiology , Obesity, Morbid/metabolism , Pakistan/epidemiology , Pedigree
3.
J Control Release ; 246: 164-173, 2017 01 28.
Article in English | MEDLINE | ID: mdl-27984105

ABSTRACT

On-demand delivery of therapeutics plays an essential role in simplifying and improving patient care. The high loading capacity of reduced graphene oxide (rGO) for drugs has made this matrix of particular interest for its hybridization with therapeutics. In this work, we describe the formulation of rGO impregnated poly(ethylene glycol) dimethacrylate based hydrogels (PEGDMA-rGO) and their efficient loading with insulin. Near-infrared (NIR) light induced heating of the PEGDMA-rGO hydrogels allows for highly efficient insulin release. Most importantly, we validate that the NIR irradiation of the hydrogel has no effect on the biological and metabolic activities of the released insulin. The ease of insulin loading/reloading makes this photothermally triggered release strategy of interest for diabetic patients. Additionally, the rGO-based protein releasing platform fabricated here can be expanded towards 'on demand' release of various other therapeutically relevant biomolecules.


Subject(s)
Delayed-Action Preparations/chemistry , Graphite/chemistry , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Administration, Cutaneous , Animals , Caco-2 Cells , Hep G2 Cells , Hot Temperature , Humans , Hypoglycemic Agents/pharmacokinetics , Insulin/pharmacokinetics , Light , Methacrylates/chemistry , Oxidation-Reduction , Polyethylene Glycols/chemistry , Skin Absorption , Swine
4.
J Control Release ; 245: 137-146, 2017 01 10.
Article in English | MEDLINE | ID: mdl-27914995

ABSTRACT

The development of a skin-mounted patch capable of controlled transcutaneous delivery of therapeutics through thermal activation provides a unique solution for the controlled release of active principles over long-term periods. Here, we report on a flexible transdermal patch for photothermal triggered release of ondansetron (ODS), a commonly used drug for the treatment of chemotherapy-induced nausea and vomiting and used as model compound here. To achieve this, a dispersion of ODS-loaded reduced graphene oxide (rGO-ODS) nanosheets were deposited onto Kapton to produce a flexible polyimide-based patch. It is demonstrated that ODS loaded Kapton/rGO patches have a high drug delivery performance upon irradiation with a continuous laser beam at 980nm for 10min due to an induced photothermal heating effect. The ability of ODS impregnated Kapton/rGO patches as transdermal delivery scaffolds for ODS across the skin is in addition investigated using porcine ear skin as a model. We show that the cumulative quantity and flux of ODS passing the skin are highly depending on the laser power density used. At 5Wcm-2 irradiation, the ODS flux across pig skin was determined to be 1.6µgcm-2h-1 comparable to other approaches. The use of tween 20 as skin enhancer could significantly increase the ODS flux to 13.2µgcm-2h-1. While the skin penetration enhancement is comparable to that obtained using other well-known permeation enhancers, the actual superiority and interest of the proposed approach is that the Kapton/rGO photoactivatable skin patch can be loaded with any drugs and therapeutics of interest, making the approach extremely versatile. The on demand delivery of drugs upon local laser irradiation and the possibility to reload the interface with the drug makes this new drug administration route very appealing.


Subject(s)
Antiemetics/administration & dosage , Graphite/administration & dosage , Ondansetron/administration & dosage , Oxides/administration & dosage , Skin Absorption/radiation effects , Transdermal Patch , Administration, Cutaneous , Animals , Antiemetics/radiation effects , Graphite/chemistry , Graphite/radiation effects , In Vitro Techniques , Lasers , Light , Ondansetron/radiation effects , Oxidation-Reduction , Oxides/chemistry , Oxides/radiation effects , Skin/metabolism , Swine
5.
Int J Gynecol Cancer ; 25(7): 1194-200, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26244757

ABSTRACT

OBJECTIVE: Ovarian cancer prognosis remains dire after primary therapy. Recurrence rates are disappointingly high as 60% of women with advanced epithelial ovarian cancer considered in remission will develop recurrent disease within 5 years. Special attention to undetected peritoneal metastasis and residual tumorous cells during surgery is necessary as they are the main predictive factors of recurrences. Folate receptor α (FRα) shows promising prospects in targeting ovarian cancerous cells. Our aim was to determine if the Fischer model described by Rose et al could be used to evaluate folate-targeted therapies in preclinical studies. METHODS: NuTu-19 epithelial ovarian cancer cell line was used to induce peritoneal carcinomatosis in female Fischer 344 rats. FRα expression by NuTu-19 cells was assessed in vitro by immunofluorescence using "Cytospin®" protocol. In vitro folate-targeted compound uptake by NuTu-19 cells was evaluated by incubation of FRα-positive ovarian cancer cell lines (NuTu-19/SKOV-3/OVCAR-3/IGROV-1) with or without (control) a folate-targeted photosensitizer. Intracellular incorporation was assessed by confocal microscopy. Determination of in vivo FRα tissue expression by several organs of the peritoneal cavity was studied by immunohistochemistry. RESULTS: NuTu-19 cells express FRα which allows intracellular incorporation of folate-targeted compound by endocytosis. FRα is expressed in tumor tissue, ovary, and liver. Peritoneum, colon, small intestine, and kidney do not express the receptor. CONCLUSIONS: Female Fischer 344 rat is an inexpensive reproducible and efficient preclinical model to study ovarian peritoneal carcinomatosis folate-targeted therapies.


Subject(s)
Disease Models, Animal , Folate Receptor 1/antagonists & inhibitors , Folic Acid/metabolism , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Peritoneal Neoplasms/drug therapy , Photosensitizing Agents/pharmacology , Animals , Apoptosis/drug effects , Carcinoma, Ovarian Epithelial , Cell Proliferation/drug effects , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/secondary , Rats , Rats, Inbred F344 , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Nat Med ; 21(5): 512-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25894829

ABSTRACT

Type 2 diabetes (T2D) is characterized by chronic hyperglycemia resulting from a deficiency in insulin signaling, because of insulin resistance and/or defects in insulin secretion; it is also associated with increases in glucagon and endogenous glucose production (EGP). Gliflozins, including dapagliflozin, are a new class of approved oral antidiabetic agents that specifically inhibit sodium-glucose co-transporter 2 (SGLT2) function in the kidney, thus preventing renal glucose reabsorption and increasing glycosuria in diabetic individuals while reducing hyperglycemia. However, gliflozin treatment in subjects with T2D increases both plasma glucagon and EGP by unknown mechanisms. In spite of the rise in EGP, T2D patients treated with gliflozin have lower blood glucose levels than those receiving placebo, possibly because of increased glycosuria; however, the resulting increase in plasma glucagon levels represents a possible concerning side effect, especially in a patient population already affected by hyperglucagonemia. Here we demonstrate that SGLT2 is expressed in glucagon-secreting alpha cells of the pancreatic islets. We further found that expression of SLC5A2 (which encodes SGLT2) was lower and glucagon (GCG) gene expression was higher in islets from T2D individuals and in normal islets exposed to chronic hyperglycemia than in islets from non-diabetics. Moreover, hepatocyte nuclear factor 4-α (HNF4A) is specifically expressed in human alpha cells, in which it controls SLC5A2 expression, and its expression is downregulated by hyperglycemia. In addition, inhibition of either SLC5A2 via siRNA-induced gene silencing or SGLT2 via dapagliflozin treatment in human islets triggered glucagon secretion through KATP channel activation. Finally, we found that dapagliflozin treatment further promotes glucagon secretion and hepatic gluconeogenesis in healthy mice, thereby limiting the decrease of plasma glucose induced by fasting. Collectively, these results identify a heretofore unknown role of SGLT2 and designate dapagliflozin an alpha cell secretagogue.


Subject(s)
Benzhydryl Compounds/chemistry , Gene Expression Regulation , Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Glucosides/chemistry , Sodium-Glucose Transporter 2/metabolism , Administration, Oral , Adult , Animals , Blood Glucose/chemistry , Cell Separation , Female , Flow Cytometry , Gene Expression Profiling , Gene Silencing , Glucagon/blood , Gluconeogenesis , Hepatocyte Nuclear Factor 4/metabolism , Humans , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , RNA, Small Interfering/metabolism , Sodium-Glucose Transporter 1/metabolism
7.
Cell Transplant ; 24(1): 1-9, 2015.
Article in English | MEDLINE | ID: mdl-23683575

ABSTRACT

Reliable assessment of islet viability, mass, and purity must be met prior to transplanting an islet preparation into patients with type 1 diabetes. The standard method for quantifying human islet preparations is by direct microscopic analysis of dithizone-stained islet samples, but this technique may be susceptible to inter-/intraobserver variability, which may induce false positive/negative islet counts. Here we describe a simple, reliable, automated digital image analysis (ADIA) technique for accurately quantifying islets into total islet number, islet equivalent number (IEQ), and islet purity before islet transplantation. Islets were isolated and purified from n = 42 human pancreata according to the automated method of Ricordi et al. For each preparation, three islet samples were stained with dithizone and expressed as IEQ number. Islets were analyzed manually by microscopy or automatically quantified using Nikon's inverted Eclipse Ti microscope with built-in NIS-Elements Advanced Research (AR) software. The AIDA method significantly enhanced the number of islet preparations eligible for engraftment compared to the standard manual method (p < 0.001). Comparisons of individual methods showed good correlations between mean values of IEQ number (r(2) = 0.91) and total islet number (r(2) = 0.88) and thus increased to r(2) = 0.93 when islet surface area was estimated comparatively with IEQ number. The ADIA method showed very high intraobserver reproducibility compared to the standard manual method (p < 0.001). However, islet purity was routinely estimated as significantly higher with the manual method versus the ADIA method (p < 0.001). The ADIA method also detected small islets between 10 and 50 µm in size. Automated digital image analysis utilizing the Nikon Instruments software is an unbiased, simple, and reliable teaching tool to comprehensively assess the individual size of each islet cell preparation prior to transplantation. Implementation of this technology to improve engraftment may help to advance the therapeutic efficacy and accessibility of islet transplantation across centers.


Subject(s)
Image Processing, Computer-Assisted/instrumentation , Image Processing, Computer-Assisted/methods , Islets of Langerhans Transplantation , Islets of Langerhans/cytology , Software , Adult , Female , Humans , Male , Middle Aged , Reproducibility of Results
8.
J Endocrinol ; 216(1): 21-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23092880

ABSTRACT

Regulated associated protein of mTOR (Raptor) and rapamycin-insensitive companion of mTOR (rictor) are two proteins that delineate two different mTOR complexes, mTORC1 and mTORC2 respectively. Recent studies demonstrated the role of rictor in the development and function of ß-cells. mTORC1 has long been known to impact ß-cell function and development. However, most of the studies evaluating its role used either drug treatment (i.e. rapamycin) or modification of expression of proteins known to modulate its activity, and the direct role of raptor in insulin secretion is unclear. In this study, using siRNA, we investigated the role of raptor and rictor in insulin secretion and production in INS-1 cells and the possible cross talk between their respective complexes, mTORC1 and mTORC2. Reduced expression of raptor is associated with increased glucose-stimulated insulin secretion and intracellular insulin content. Downregulation of rictor expression leads to impaired insulin secretion without affecting insulin content and is able to correct the increased insulin secretion mediated by raptor siRNA. Using dominant-negative or constitutively active forms of Akt, we demonstrate that the effect of both raptor and rictor is mediated through alteration of Akt signaling. Our finding shed new light on the mechanism of control of insulin secretion and production by the mTOR, and they provide evidence for antagonistic effect of raptor and rictor on insulin secretion in response to glucose by modulating the activity of Akt, whereas only raptor is able to control insulin biosynthesis.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Multiprotein Complexes/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Substitution , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Humans , Hyperglycemia/metabolism , Insulin Secretion , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/genetics , Mutant Proteins/metabolism , Phosphorylation , Protein Processing, Post-Translational , Proteins/antagonists & inhibitors , Proteins/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , RNA, Small Interfering , Rapamycin-Insensitive Companion of mTOR Protein , Regulatory-Associated Protein of mTOR , Serine/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics
9.
J Endocrinol ; 214(2): 225-32, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22582094

ABSTRACT

Zinc ions are essential for the formation of insulin crystals in pancreatic ß cells, thereby contributing to packaging efficiency of stored insulin. Zinc fluxes are regulated through the SLC30A (zinc transporter, ZNT) family. Here, we investigated the effect of metabolic stress associated with the prediabetic state (zinc depletion, glucotoxicity, and lipotoxicity) on ZNT expression and human pancreatic islet function. Both zinc depletion and lipotoxicity (but not glucotoxicity) downregulated ZNT8 (SLC30A8) expression and altered the glucose-stimulated insulin secretion index (GSIS). ZNT8 overexpression in human islets protected them from the decrease in GSIS induced by tetrakis-(2-pyridylmethyl) ethylenediamine and palmitate but not from cell death. In addition, zinc supplementation decreased palmitate-induced human islet cell death without restoring GSIS. Altogether, we showed that ZNT8 expression responds to variation in zinc and lipid levels in human ß cells, with repercussions on insulin secretion. Prospects for increasing ZNT8 expression and/or activity may prove beneficial in type 2 diabetes in humans.


Subject(s)
Cation Transport Proteins/genetics , Cation Transport Proteins/physiology , Islets of Langerhans/metabolism , Adult , Cation Transport Proteins/metabolism , Cell Death/drug effects , Cell Death/genetics , Cells, Cultured , Chelating Agents/pharmacology , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Humans , Islets of Langerhans/drug effects , Islets of Langerhans/physiology , Lipids/pharmacology , Lipids/toxicity , Palmitic Acid/pharmacology , Palmitic Acid/toxicity , Transfection , Zinc/pharmacology , Zinc Transporter 8
SELECTION OF CITATIONS
SEARCH DETAIL
...