Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Front Endocrinol (Lausanne) ; 15: 1426175, 2024.
Article in English | MEDLINE | ID: mdl-39286266

ABSTRACT

Background: Testosterone (T) therapy increases lean mass and reduces total body and truncal fat mass in hypogonadal men. However, the underlying molecular mechanisms for the reciprocal changes in fat and lean mass in humans are not entirely clear. Methods: Secondary analysis of specimens obtained from a single-arm, open-label clinical trial on pharmacogenetics of response to T therapy in men with late-onset hypogonadism, conducted between 2011 and 2016 involving 105 men (40-74 years old), who were given intramuscular T cypionate 200 mg every 2 weeks for 18 months. Subcutaneous fat (SCF), peripheral blood mononuclear cells (PBMC) and serum were obtained from the participants at different time points of the study. We measured transcription factors for adipogenesis and myogenesis in the SCF, and PBMC, respectively, by real-time quantitative PCR at baseline and 6 months. Serum levels of FOLLISTATIN, PAX7, MYOSTATIN, ADIPSIN, and PRDM16 were measured by ELISA. Results: As expected, there was a significant increase in T and estradiol levels after 6 months of T therapy. There was also a reduction in fat mass and an increase in lean mass after 6 months of T therapy. Gene-protein studies showed a significant reduction in the expression of the adipogenic markers PPARγ in SCF and ADIPSIN levels in the serum, together with a concomitant significant increase in the expression of myogenic markers, MYOD in PBMC and PAX7 and FOLLISTATIN levels in the serum after 6 months of T therapy compared to baseline. Interestingly, there was a significant increase in the adipo-myogenic switch, PRDM16, expression in SCF and PBMC, and in circulating protein levels in the serum after 6 months of T therapy, which is likely from increased estradiol. Conclusion: Our study supports that molecular shift from the adipogenic to the myogenic pathway in men with hypogonadism treated with T could be mediated directly or indirectly by enhanced PRDM16 activity, in turn a result from increased estradiol level. This might have led to the reduction in body fat and increase in lean mass commonly seen in hypogonadal men treated with T.


Subject(s)
Body Composition , DNA-Binding Proteins , Hypogonadism , Testosterone , Transcription Factors , Humans , Male , Testosterone/blood , Middle Aged , Hypogonadism/drug therapy , Hypogonadism/metabolism , Hypogonadism/blood , Adult , Aged , Transcription Factors/metabolism , Transcription Factors/genetics , Body Composition/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Signal Transduction/drug effects , Hormone Replacement Therapy
2.
Elife ; 122024 Jul 31.
Article in English | MEDLINE | ID: mdl-39082939

ABSTRACT

The kidneys facilitate energy conservation through reabsorption of nutrients including glucose. Almost all the filtered blood glucose is reabsorbed by the kidneys. Loss of glucose in urine (glycosuria) is offset by an increase in endogenous glucose production to maintain normal energy supply in the body. How the body senses this glucose loss and consequently enhances glucose production is unclear. Using renal Slc2a2 (also known as Glut2) knockout mice, we demonstrate that elevated glycosuria activates the hypothalamic-pituitary-adrenal axis, which in turn drives endogenous glucose production. This phenotype was attenuated by selective afferent renal denervation, indicating the involvement of the afferent nerves in promoting the compensatory increase in glucose production. In addition, through plasma proteomics analyses we observed that acute phase proteins - which are usually involved in the body's defense mechanisms against a threat - were the top candidates which were either upregulated or downregulated in renal Slc2a2 KO mice. Overall, afferent renal nerves contribute to promoting endogenous glucose production in response to elevated glycosuria and loss of glucose in urine is sensed as a biological threat in mice. These findings may be useful in improving the efficiency of drugs like SGLT2 inhibitors that are intended to treat hyperglycemia by enhancing glycosuria but are met with a compensatory increase in endogenous glucose production.


Subject(s)
Glucose Transporter Type 2 , Glucose , Glycosuria , Hypothalamus , Kidney , Mice, Knockout , Animals , Mice , Glucose/metabolism , Kidney/metabolism , Glycosuria/metabolism , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 2/genetics , Hypothalamus/metabolism , Male , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiology , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiology
3.
bioRxiv ; 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-37790458

ABSTRACT

The kidneys facilitate energy conservation through reabsorption of nutrients including glucose. Almost all of the filtered blood glucose is reabsorbed by the kidneys. Loss of glucose in urine (glycosuria) is offset by an increase in endogenous glucose production to maintain normal energy supply in the body. How the body senses this glucose loss and consequently enhances glucose production is unclear. Using renal Glut2 knockout mice, we demonstrate that elevated glycosuria activates the hypothalamic-pituitary-adrenal axis, which in turn drives endogenous glucose production. This phenotype was attenuated by selective afferent renal denervation, indicating the involvement of the afferent nerves in promoting the compensatory increase in glucose production. In addition, through plasma proteomics analyses we observed that acute phase proteins - which are usually involved in body's defense mechanisms against a threat - were the top candidates which were either upregulated or downregulated in renal Glut2 KO mice. Overall, afferent renal nerves contribute to promoting endogenous glucose production in response to elevated glycosuria and loss of glucose in urine is sensed as a biological threat in mice. These findings may be useful in improving efficiency of drugs like SGLT2 inhibitors that are intended to treat hyperglycemia by enhancing glycosuria, but are met with a compensatory increase in endogenous glucose production.

4.
Diabetologia ; 67(1): 170-189, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37712955

ABSTRACT

AIMS/HYPOTHESIS: The brain is a major consumer of glucose as an energy source and regulates systemic glucose as well as energy balance. Although glucose transporters such as GLUT2 and sodium-glucose cotransporter 2 (SGLT2) are known to regulate glucose homeostasis and metabolism, the identity of a receptor that binds glucose to activate glucose signalling pathways in the brain is unknown. In this study, we aimed to discover a glucose receptor in the mouse hypothalamus. METHODS: Here we used a high molecular mass glucose-biotin polymer to enrich glucose-bound mouse hypothalamic neurons through cell-based affinity chromatography. We then subjected the enriched neurons to proteomic analyses and identified adhesion G-protein coupled receptor 1 (ADGRL1) as a top candidate for a glucose receptor. We validated glucose-ADGRL1 interactions using CHO cells stably expressing human ADGRL1 and ligand-receptor binding assays. We generated and determined the phenotype of global Adgrl1-knockout mice and hypothalamus-specific Adgrl1-deficient mice. We measured the variables related to glucose and energy homeostasis in these mice. We also generated an Adgrl1Cre mouse model to investigate the role of ADGRL1 in sensing glucose using electrophysiology. RESULTS: Adgrl1 is highly expressed in the ventromedial nucleus of the hypothalamus (VMH) in mice. Lack of Adgrl1 in the VMH in mice caused fasting hyperinsulinaemia, enhanced glucose-stimulated insulin secretion and insulin resistance. In addition, the Adgrl1-deficient mice had impaired feeding responses to glucose and fasting coupled with abnormal glucose sensing and decreased physical activity before development of obesity and hyperglycaemia. In female mice, ovariectomy was necessary to reveal the contribution of ADGRL1 to energy and glucose homeostasis. CONCLUSIONS/INTERPRETATION: Altogether, our findings demonstrate that ADGRL1 binds glucose and is involved in energy as well as glucose homeostasis in a sex-dependent manner. Targeting ADGRL1 may introduce a new class of drugs for the treatment of type 2 diabetes and obesity.


Subject(s)
Diabetes Mellitus, Type 2 , Animals , Cricetinae , Female , Humans , Mice , Cricetulus , Diabetes Mellitus, Type 2/complications , Energy Metabolism/genetics , Glucose/metabolism , Homeostasis/physiology , Mice, Knockout , Obesity/metabolism , Proteomics
5.
Discov Med ; 35(178): 653-663, 2023 10.
Article in English | MEDLINE | ID: mdl-37811607

ABSTRACT

Mitochondria-associated membranes (MAMs) play a significant role in multiple cellular processes including lipid metabolism and neuronal survival. Fatty acids constitute 80% of the dry mass of the brain and are vital for life. Apart from mitochondrial ß-oxidation, fatty acids are metabolized in part by peroxisomes to regulate the generation of acyl Coenzyme A and adenosine triphosphate (ATP). Ablation of mitochondria and its associated genes tether endoplasmic reticulum (ER)-Mitochondria contact and results in loss of function leading to aberrant lipid metabolism. Additionally, an increase in reactive oxygen species (ROS) levels along with free radicals' generation may lead to alteration in the integrity of membrane phospholipids, proteins, and DNA. Hence, it is critical to understand the effect of structural and functional aspects of mitochondria on lipid homeostasis. This review explains the role of mitochondrial dysfunction in lipid metabolism and its impact on various neurodegenerative diseases and metabolic disorders.


Subject(s)
Fatty Acids , Mitochondria , Humans , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Homeostasis , Fatty Acids/metabolism , Lipids
6.
Front Endocrinol (Lausanne) ; 14: 1168687, 2023.
Article in English | MEDLINE | ID: mdl-37576965

ABSTRACT

Fractures associated with Type2 diabetes (T2DM) are major public health concerns in an increasingly obese and aging population. Patients with obesity or T2DM have normal or better than normal bone mineral density but at an increased risk for fractures. Hence it is crucial to understand the pathophysiology and mechanism of how T2DM and obesity result in altered bone physiology leading to increased fracture risk. Although enhanced osteoclast mediated bone resorption has been reported for these patients, the most notable observation among patients with T2DM is the reduction in bone formation from mostly dysfunction in osteoblast differentiation and survival. Studies have shown that obesity and T2DM are associated with increased adipogenesis which is most likely at the expense of reduced osteogenesis and myogenesis considering that adipocytes, osteoblasts, and myoblasts originate from the same progenitor cells. Furthermore, emerging data point to an inter-relationship between bone and metabolic homeostasis suggesting that these physiologic processes could be under the control of common regulatory pathways. Thus, this review aims to explore the complex mechanisms involved in lineage differentiation and their effect on bone pathophysiology in patients with obesity and T2DM along with an examination of potential novel pharmacological targets or a re-evaluation of existing drugs to improve bone homeostasis.


Subject(s)
Diabetes Mellitus, Type 2 , Fractures, Bone , Humans , Aged , Osteogenesis/physiology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Bone and Bones/metabolism , Bone Density , Fractures, Bone/etiology , Obesity/complications
7.
Biomolecules ; 13(3)2023 03 16.
Article in English | MEDLINE | ID: mdl-36979475

ABSTRACT

OBJECTIVE: Glucose transporter 2 (GLUT2) is expressed in the pancreatic ß-cell, intestine, liver, and kidney in mice. Although GLUT2 is considered as a major regulator of insulin secretion, in vivo contribution of ß-cell Glut2 to glucose-stimulated insulin secretion and systemic glucose homeostasis is undefined. Therefore, the main objective of this study is to determine the role of ß-cell Glut2 in regulating insulin secretion and blood glucose levels in mice. METHODS: We produced mice in which we can knock down Glut2 at a desired time specifically in ß-cells (ß-Glut2 KD) by crossing Glut2LoxP/LoxP mice with Ins1CreERT2 mouse strain and using the Cre-Lox recombination technique. We measured fasting blood glucose levels, glucose tolerance, and glucose-stimulated insulin secretion in the ß-Glut2 KD mice. We used qRT-PCR and immunofluorescence to validate the deficiency of ß-cell Glut2 in ß-Glut2 KD mice. RESULTS: We report that both male and female ß-Glut2 KD mice have normal glucose-stimulated insulin secretion. Moreover, the ß-Glut2 KD mice exhibit normal fasting blood glucose levels and glucose tolerance. The ß-Glut2 KD mice have upregulated GLUT1 in islets. CONCLUSIONS: Our findings demonstrate that normal ß-cell Glut2 expression is not essential for regulating glucose-stimulated insulin secretion and systemic glucose homeostasis in mice. Therefore, the currently assumed role of ß-cell GLUT2 in regulating insulin secretion and blood glucose levels needs to be recalibrated. This will allow an opportunity to determine the contribution of other ß-cell glucose transporters or factors whose normal expression may be necessary for mediating glucose stimulated insulin secretion.


Subject(s)
Glucose Transporter Type 2 , Insulin-Secreting Cells , Animals , Female , Male , Mice , Blood Glucose/metabolism , Glucose/pharmacology , Glucose/metabolism , Homeostasis , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Glucose Transporter Type 2/metabolism
8.
Biomedicines ; 11(2)2023 Feb 20.
Article in English | MEDLINE | ID: mdl-36831180

ABSTRACT

The growing burden of obesity and osteoporosis is a major public health concern. Emerging evidence of the role of adipokines on bone metabolism has led to the discovery of novel adipokines over the last decade. Obesity is recognized as a state of adipose tissue inflammation that adversely affects bone health. Adipokines secreted from white adipose tissue (WAT) and bone marrow adipose tissue (BMAT) exerts endocrine and paracrine effects on the survival and function of osteoblasts and osteoclasts. An increase in marrow fat is implicated in osteoporosis and, hence, it is crucial to understand the complex interplay between adipocytes and bone. The objective of this review is to summarize recent advances in our understanding of the role of different adipokines on bone metabolism. METHODS: This is a comprehensive review of the literature available in PubMED and Cochrane databases, with an emphasis on the last five years using the keywords. RESULTS: Leptin has shown some positive effects on bone metabolism; in contrast, both adiponectin and chemerin have consistently shown a negative association with BMD. No significant association was found between resistin and BMD. Novel adipokines such as visfatin, LCN-2, Nesfatin-1, RBP-4, apelin, and vaspin have shown bone-protective and osteoanabolic properties that could be translated into therapeutic targets. CONCLUSION: New evidence suggests the potential role of novel adipokines as biomarkers to predict osteoporosis risk, and as therapeutic targets for the treatment of osteoporosis.

9.
Front Endocrinol (Lausanne) ; 13: 936159, 2022.
Article in English | MEDLINE | ID: mdl-36171900

ABSTRACT

Introduction: Type 2 diabetes mellitus (T2DM) is well-known to be associated with normal bone density but, concurrently, low bone turnover and increased risk for fracture. One of the proposed mechanisms is possible derangement in bone precursor cells, which could be represented by deficiencies in circulating osteogenic progenitor (COP) cells and osteoclast precursors (OCP). The objective of our study is to understand whether extent of glycemic control has an impact on these cells, and to identify other factors that may as well. Methods: This was a secondary analysis of baseline data from 51 male participants, aged 37-65 in an ongoing clinical trial at Michael E. DeBakey VA Medical Center, Houston, Texas, USA. At study entry serum Hemoglobin A1c was measured by high-performance liquid chromatography osteocalcin (OCN) and C-terminal telopeptide of type 1 collagen (CTx) were measured by ELISA, and testosterone and estradiol by liquid-chromatography/mass-spectrometry. Areal bone mineral density (BMD), trabecular bone score and body composition were measured by dual energy x-ray absorptiometry, while COP and OCP were measured by flow cytometry. Results: When adjusted for serum testosterone, parathyroid hormone, and 25-hydroxyvitamin D, those with poor long-term glycemic control had significantly higher percentage of COP (p = 0.04). COP correlated positively with visceral adipose tissue (VAT) volume (r = 0.37, p = 0.01) and negatively with free testosterone (r = -0.28, p = 0.05) and OCN (r = -0.28, p = 0.07), although only borderline for the latter. OCP correlated positively with age, FSH, lumbar spine BMD, and COP levels, and negatively with glucose, triglycerides, and free estradiol. Multivariable regression analyses revealed that, in addition to being predictors for each other, another independent predictor for COP was VAT volume while age, glucose, and vitamin D for OCP. Conclusion: Our results suggest that high COP could be a marker of poor metabolic control. However, given the complex nature and the multitude of factors influencing osteoblastogenesis/adipogenesis, it is possible that the increase in COP is a physiologic response of the bone marrow to increased osteoblast apoptosis from poor glycemic control. Alternatively, it is also likely that a metabolically unhealthy profile may retard the development of osteogenic precursors to fully mature osteoblastic cells.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperglycemia , Adult , Aged , Collagen Type I/metabolism , Diabetes Mellitus, Type 2/metabolism , Estradiol , Follicle Stimulating Hormone/metabolism , Glucose , Glycated Hemoglobin/metabolism , Glycemic Control , Humans , Intra-Abdominal Fat/metabolism , Male , Middle Aged , Osteocalcin , Osteoclasts/metabolism , Parathyroid Hormone/metabolism , Testosterone , Triglycerides , Vitamin D
10.
Int J Mol Sci ; 22(4)2021 Feb 03.
Article in English | MEDLINE | ID: mdl-33546300

ABSTRACT

Type 1 diabetes mellitus is an autoimmune disease characterized by increased production of pro-inflammatory cytokines secreted by infiltrating macrophages and T cells that destroy pancreatic ß cells in a free radical-dependent manner that causes decrease or absence of insulin secretion and consequent hyperglycemia. Hence, suppression of pro-inflammatory cytokines and oxidative stress may ameliorate or decrease the severity of diabetes mellitus. To investigate the effect and mechanism(s) of action of RVD1, an anti-inflammatory metabolite derived from docosahexaenoic acid (DHA), on STZ-induced type 1 DM in male Wistar rats, type 1 diabetes was induced by single intraperitoneal (i.p) streptozotocin (STZ-65 mg/kg) injection. RVD1 (60 ng/mL, given intraperitoneally) was administered from day 1 along with STZ for five consecutive days. Plasma glucose, IL-6, TNF-α, BDNF (brain-derived neurotrophic factor that has anti-diabetic actions), LXA4 (lipoxin A4), and RVD1 levels and BDNF concentrations in the pancreas, liver, and brain tissues were measured. Apoptotic (Bcl2/Bax), inflammatory (COX-1/COX-2/Nf-κb/iNOS/PPAR-γ) genes and downstream insulin signaling proteins (Gsk-3ß/Foxo1) were measured in the pancreatic tissue along with concentrations of various antioxidants and lipid peroxides. RVD1 decreased severity of STZ-induced type 1 DM by restoring altered plasma levels of TNF-α, IL-6, and BDNF (p < 0.001); expression of pancreatic COX-1/COX-2/PPAR-γ genes and downstream insulin signaling proteins (Gsk-3ß/Foxo1) and the concentrations of antioxidants and lipid peroxides to near normal. RVD1 treatment restored expression of Bcl2/Pdx genes, plasma LXA4 (p < 0.001) and RVD1 levels and increased brain, pancreatic, intestine, and liver BDNF levels to near normal. The results of the present study suggest that RVD1 can prevent STZ-induced type 1 diabetes by its anti-apoptotic, anti-inflammatory, and antioxidant actions and by activating the Pdx gene that is needed for pancreatic ß cell proliferation.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Diabetes Mellitus, Type 1/drug therapy , Docosahexaenoic Acids/pharmacology , Inflammation , Oxidative Stress , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/chemically induced , Diabetes Mellitus, Type 1/metabolism , Docosahexaenoic Acids/therapeutic use , Gene Expression Regulation , Insulin-Secreting Cells , Male , Rats , Rats, Wistar , Streptozocin/toxicity
11.
Arch Med Res ; 51(6): 492-503, 2020 08.
Article in English | MEDLINE | ID: mdl-32451116

ABSTRACT

OBJECTIVE: To study whether resolvin D1 (RvD1), a metabolite of docosahexaenoic acid (DHA), prevents NA-STZ-induced type 2 diabetes mellitus (type 2 DM) in vivo and if so, what could be the mechanism of this action. MATERIAL AND METHODS: Single intra-peritoneal (i.p) injection of NA-STZ (175 mg/kg body weight of NA and 65 mg/kg of STZ) was injected simultaneously with RvD1 (60 ng/animal) (injected for 5 consecutive days) to Wistar rats. The effect of RvD1 on plasma glucose levels and apoptotic (Bcl2/Bax) and inflammatory (NF-κB/iNOS) protein expression, plasma lipoxin A4 and BDNF (brain-derived neurotrophic factor) were studied. Protein expressions of PI3k-Akt-mTOR pathway along with histopathological studies of brain were also evaluated. RESULTS: NA-STZ-induced type 2 DM rats showed hyperglycemia, enhanced plasma IL-6/TNF-α (p ≤0.01), reduced plasma BDNF (p ≤0.01) and LXA4 (p ≤0.01) levels and low BDNF in pancreatic, hepatic and brain tissues (p <0.001), which were restored to near normal (p ≤0.01) in RvD1 treated group. RvD1 increased insulin sensitivity by suppressing inflammation (NF-κB/iNOS) (p ≤0.01) and decreasing apoptosis (Bcl2/Bax) and restoring BDNF and LXA4 levels to near normal. RvD1 treatment increased phosphorylation of Akt (Ser473), and subsequent activation (phosphorylation) of downstream signaling molecules of PI3K and mTOR indicating that RvD1 acts through PI3K/Akt/mTOR axis. DISCUSSION: RvD1 is effective in preventing NA-STZ-induced type 2 DM in vivo by suppressing oxidative damage, enhancing the production of anti-inflammatory LXA4 and enhancing neuronal cell survival by augmenting the production of BDNF. Thus, RvD1 may be of benefit not only in preventing diabetes mellitus but also diabetes associated Alzheimer's disease and memory loss.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Brain/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Docosahexaenoic Acids/therapeutic use , Niacinamide/adverse effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis , Diabetes Mellitus, Type 2/chemically induced , Humans , Male , Rats , Rats, Wistar
12.
Lipids Health Dis ; 18(1): 214, 2019 Dec 10.
Article in English | MEDLINE | ID: mdl-31823816

ABSTRACT

OBJECTIVE: To study whether minimal doses of arachidonic acid (AA), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) and lipoxin A4 (LXA4) and brain-derived neurotrophic factor (BDNF), when used in combination can protect RIN5F cells from chemical-induced cytotoxicity. As a corollary, to know whether plasma BDNF and LXA4 are altered in STZ-induced type 2 DM animals. MATERIALS AND METHODS: RIN5F cells, alloxan (AL), streptozotocin (STZ), doxorubicin (DB), and benzo(a)pyrene (BP) were used in this study. Chemical-induced apoptosis and changes in antioxidants, lipid peroxides and nitric oxide (NO) and LXA4 and BDNF levels in RIN5F cells were studied. Alterations in plasma concentrations of BDNF and LXA4 in STZ-induced type 2 diabetes animals was estimated. RESULTS: BDNF, LXA4 and AA, EPA and DHA protected (P < 0.001 and P < 0.01 respectively) against AL/STZ/DB/BP-induced toxicity to RIN5F cells in vitro. AL/ STZ/DB/BP inhibited BDNF and LXA4 production by RIN5F cells and were restored to normal by AA, EPA and DHA. Sub-optimal doses of BDNF, LXA4, AA and EPA when used in combination protected against cytotoxic action of AL/STZ/DB/BP on RIN5F cells in vitro by restoring LXA4/BDNF levels and altered antioxidant/lipid peroxides/NO levels (P < 0.01) to normal. STZ (65 mg/kg)-induced type 2 diabetes mellitus animals showed reduced plasma BDNF and LXA4 levels (P < 0.001). DISCUSSION: AL/STZ/DB/BP-induced cytotoxicity to RIN5F cells in vitro can be prevented by BDNF, LXA4 and AA. AL/STZ/DB/BP are cytotoxic, possibly, by suppressing the production of LXA4 and BDNF in RIN5F cells. STZ-induced type 2 DM animals have decreased plasma levels of LXA4 and BDNF. CONCLUSION: The results of the present study suggest that BDNF, LXA4, EPA, DHA, AA, GLA and BDNF protect pancreatic ß cells from the cytotoxic action of various chemicals and prevent development of diabetes mellitus. LXA4 seems to be the mediator of these cytoprotective actions of BDNF and PUFAs suggesting a close interaction exists among these molecules (BDNF, PUFAs and LXA4). Hence, methods developed to deliver a combination of PUFAs (especially AA), LXA4 and BDNF may prevent development of diabetes mellitus (both type 1 and type 2).


Subject(s)
Brain-Derived Neurotrophic Factor/administration & dosage , Cell Death/drug effects , Diabetes Mellitus, Type 2/pathology , Fatty Acids, Unsaturated/administration & dosage , Insulin-Secreting Cells/pathology , Lipoxins/administration & dosage , Alloxan/pharmacology , Animals , Arachidonic Acid/pharmacology , Benzo(a)pyrene/pharmacology , Brain-Derived Neurotrophic Factor/blood , Cell Line, Tumor , Cytotoxins , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/blood , Doxorubicin/pharmacology , Drug Interactions , Insulin-Secreting Cells/drug effects , Insulinoma , Lipoxins/blood , Pancreatic Neoplasms , Rats , Rats, Wistar , Streptozocin/pharmacology
13.
Lipids Health Dis ; 17(1): 168, 2018 Jul 24.
Article in English | MEDLINE | ID: mdl-30041644

ABSTRACT

BACKGROUND: Proteins of the insulin signaling pathway are needed for cell proliferation and development and glucose homeostasis. It is not known whether insulin signalling markers (Foxo1, Gsk3ß) can be correlated with the expression on PI3K-Akt-mTOR pathway, which are needed for cell survival and maintenance of glucose homeostasis. In the present study, we studied the expression of Foxo1, Gsk3ß and PI3K-Akt-mTOR in the brain of streptozotocin-induced type 2 diabetes mellitus Wistar rats. METHODS: The study was performed both in vitro (RIN5F cells) and in vivo (male Wistar rats). Gene expression of Nf-kB, IkB, Bax, Bcl-2 and Pdx1 gene was studied invitro by qRT-PCR in RIN5F cells. In STZ (65 mg/kg i.p.)-induced type 2 DM Wistar rats, blood glucose and insulin levels, iNOS, Foxo1, NF-κB, pGsk3ß and PPAR-γ1 levels along with PI3k-Akt-mTOR were measured in brain tissue. RESULTS: RIN5F cells treated with STZ showed increase in the expression of NF-kB and Bax and decrease in IkB, Bcl-2 and PDX1. Brain tissue of STZ-induced type 2 DM animals showed a significant reduction in secondary messengers of insulin signalling (Foxo1) (P < 0.001) and Gsk3ß (P < 0.01) and a significant alteration in the expression of phosphorylated-Akt (P < 0.001) mTOR (P < 0.01) and PI3K. CONCLUSION: These results suggest that STZ induces pancreatic ß cell apoptosis by enhancing inflammation. Significant alterations in the expression brain insulin signaling and cell survival pathways seen in brain of STZ-treated animals implies that alterations neuronal apoptosis may have a role in altered glucose homeostasis seen in type 2 DM that may also explain the increased incidence of cognitive dysfunction seen in them.


Subject(s)
Brain/metabolism , Diabetes Mellitus, Type 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Brain/physiopathology , Brain-Derived Neurotrophic Factor/pharmacology , Cell Line , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/genetics , Gene Expression Regulation/drug effects , Lipoxins/blood , Lipoxins/metabolism , Male , Phosphorylation , Rats, Wistar , Serine/metabolism , Streptozocin/administration & dosage , Streptozocin/toxicity
14.
Biochem Biophys Res Commun ; 486(2): 406-413, 2017 04 29.
Article in English | MEDLINE | ID: mdl-28315336

ABSTRACT

BACKGROUND: Neurodegenerative disorders, such as deficits in learning, memory and cognition and Alzheimer's disease are associated with diabetes mellitus. Brain-derived neurotrophic factor (BDNF) is a neurotrophic factor and is known to possess anti-obesity, anti-diabetic actions and is believed to have a role in memory and Alzheimer's disease. OBJECTIVE: To investigate whether STZ can reduce BDNF production by rat insulinoma (RIN5F) cells in vitro and decrease BDNF levels in the pancreas, liver and brain in vivo. METHODS: Streptozotocin (STZ)-induced cytotoxicity to RIN5F cells in vitro and type 2 DM in Wistar rats was employed in the present study. Cell viability, activities of various anti-oxidants and secretion of BDNF by RIN5F cells in vitro were measured using MTT assay, biochemical methods and ELISA respectively. In STZ-induced type 2 DM rats: plasma glucose, interleukin-6 and tumor necrosis factor-α levels and BDNF protein expression in the pancreas, liver and brain tissues were measured. In addition, neuronal count and morphology in the hippocampus and hypothalamus areas was assessed. RESULTS: STZ-induced suppression of RIN5F cell viability was abrogated by BDNF. STZ suppressed BDNF secretion by RIN5F cells in vitro. STZ-induced type 2 DM rats showed hyperglycemia, enhanced plasma IL-6 and TNF-αlevels and reduced plasma and pancreas, liver and brain tissues (P < 0.001) and increased oxidative stress compared to untreated control. Hypothalamic and hippocampal neuron in STZ-treated animals showed a decrease in the number of neurons and morphological changes suggesting of STZ cytotoxicity. CONCLUSIONS: The results of the present study suggest that STZ is not only cytotoxic to pancreatic beta cells but also to hypothalamic and hippocampal neurons by inducing oxidative stress. STZ ability to suppress BDNF production by pancreas, liver and brain tissues suggests that impaired memory, learning, and cognitive dysfunction seen in diabetes mellitus could be due to BDNF deficiency.


Subject(s)
Apoptosis/drug effects , Brain-Derived Neurotrophic Factor/genetics , Diabetes Mellitus, Experimental/genetics , Insulin-Secreting Cells/drug effects , Streptozocin/administration & dosage , Animals , Blood Glucose/metabolism , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Brain-Derived Neurotrophic Factor/metabolism , Cell Line , Cell Survival/drug effects , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Gene Expression Regulation , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypothalamus/pathology , Inflammation , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Interleukin-6/genetics , Interleukin-6/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Niacinamide/administration & dosage , Oxidative Stress/drug effects , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
15.
Metabolism ; 65(5): 667-684, 2016 May.
Article in English | MEDLINE | ID: mdl-27085775

ABSTRACT

OBJECTIVE: The study was conducted to observe whether brain-derived neurotrophic factor (BDNF) has cytoprotective actions against alloxan (AL), streptozotocin (STZ), doxorubicin (DB) and benzo(a)pyrene (BP) compounds in vitro that may account for its beneficial action in diabetes mellitus. MATERIALS AND METHODS: This in vitro study was performed using rat insulinoma (RIN5F) cells. Possible cytoprotective action of BDNF (using pre-treatment, simultaneous and post-treatment schedules of RIN5F cells with BDNF) against the four chemicals tested was evaluated using MTT and apoptosis assays. Possible mechanism of cytoprotective action of BDNF was assessed by measuring BCl2/IKB-ß/Pdx mRNA transcripts and anti-oxidant levels in RIN5F cells. Effect of alloxan, STZ, doxorubicin and BP on the production of BDNF by RIN5F cells was also studied. RESULTS: Results of the present study revealed that BDNF in the doses (100ng>50ng>10ng/ml) has significant cytoprotection (P<0.001, P<0.01) on cytotoxic action of AL, STZ, DB and BP against rat insulinoma RIN5F (5×10(4) cells/100µl) cells in vitro. It was observed that AL, STZ, DB and BP inhibited BDNF production significantly (P<0.001) in a dose-dependent manner by RIN5F cells (0.5×10(6) cells/500µl) in vitro, while BDNF not only prevented apoptosis induced by these four chemicals but also significantly increased (P<0.001) BCl2/IKB-ß/Pdx mRNA transcripts and restored anti-oxidant levels (P<0.01) in RIN5F cells to normal. DISCUSSION: These results suggest that BDNF has potent cytoprotective actions, restores anti-oxidant defenses to normal and thus, prevents apoptosis and preserves insulin secreting capacity of ß cells. In addition, BDNF enhanced viability of RIN 5F in vitro. Thus, BDNF not only has anti-diabetic actions but also preserves pancreatic ß cells integrity and enhances their viability. These results imply that BDNF functions as an endogenous cytoprotective molecule that may explain its beneficial actions in some neurological conditions as well.


Subject(s)
Alloxan/antagonists & inhibitors , Apoptosis/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Insulin-Secreting Cells/drug effects , Models, Biological , Streptozocin/antagonists & inhibitors , Alloxan/toxicity , Animals , Benzo(a)pyrene/antagonists & inhibitors , Benzo(a)pyrene/toxicity , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Brain-Derived Neurotrophic Factor/genetics , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/antagonists & inhibitors , Doxorubicin/toxicity , Drug Resistance , Gene Expression Regulation/drug effects , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Kinetics , Poisons/chemistry , Poisons/toxicity , Rats , Recombinant Proteins/metabolism , Streptozocin/toxicity
17.
Arch Med Sci ; 11(6): 1164-78, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26788077

ABSTRACT

Brain-derived neurotrophic factor (BDNF) plays an important role in neuronal survival and growth, serves as a neurotransmitter modulator, and participates in neuronal plasticity, which is essential for learning and memory. It is widely expressed in the CNS, gut and other tissues. BDNF binds to its high affinity receptor TrkB (tyrosine kinase B) and activates signal transduction cascades (IRS1/2, PI3K, Akt), crucial for CREB and CBP production, that encode proteins involved in ß cell survival. BDNF and insulin-like growth factor-1 have similar downstream signaling mechanisms incorporating both p-CAMK and MAPK that increase the expression of pro-survival genes. Brain-derived neurotrophic factor regulates glucose and energy metabolism and prevents exhaustion of ß cells. Decreased levels of BDNF are associated with neurodegenerative diseases with neuronal loss, such as Parkinson's disease, Alzheimer's disease, multiple sclerosis and Huntington's disease. Thus, BDNF may be useful in the prevention and management of several diseases including diabetes mellitus.

18.
PLoS One ; 9(12): e114766, 2014.
Article in English | MEDLINE | ID: mdl-25536345

ABSTRACT

In the present study, we noted that bleomycin induced growth inhibitory action was augmented by all the polyunsaturated fatty acids (PUFAs) tested on human neuroblastoma IMR-32 (0.5 × 10(4) cells/100 µl of IMR) cells (EPA > DHA > ALA = GLA = AA > DGLA = LA: ∼ 60, 40, 30, 10-20% respectively) at the maximum doses used. Of all the prostaglandins (PGE1, PGE2, PGF2α, and PGI2) and leukotrienes (LTD4 and LTE4) tested; PGE1, PGE2 and LTD4 inhibited the growth of IMR-32 cells to a significant degree at the highest doses used. Lipoxin A4 (LXA4), 19,20-dihydroxydocosapentaenoate (19, 20 DiHDPA) and 10(S),17(S)-dihydroxy-4Z,7Z,11E,13Z,15E,19Z-docosahexaenoic acid (protectin: 10(S),17(S)DiHDoHE), metabolites of DHA, significantly inhibited the growth of IMR-32 cells. Pre-treatment with AA, GLA, DGLA and EPA and simultaneous treatment with all PUFAs used in the study augmented growth inhibitory action of bleomycin. Surprisingly, both indomethacin and nordihydroguaiaretic acid (NDGA) at 60 and 20 µg/ml respectively enhanced the growth of IMR-32 cells even in the presence of bleomycin. AA enhanced oxidant stress in IMR-32 cells as evidenced by an increase in lipid peroxides, superoxide dismutase levels and glutathione peroxidase activity. These results suggest that PUFAs suppress growth of human neuroblastoma cells, augment growth inhibitory action of bleomycin by enhancing formation of lipid peroxides and altering the status of anti-oxidants and, in all probability, increase the formation of lipoxins, resolvins and protectins from their respective precursors that possess growth inhibitory actions.


Subject(s)
Bleomycin/toxicity , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/pharmacology , Neuroblastoma/pathology , Antioxidants/pharmacology , Arachidonic Acid/pharmacology , CD59 Antigens/pharmacology , Cell Death/drug effects , Cell Extracts , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclooxygenase Inhibitors/pharmacology , Docosahexaenoic Acids/pharmacology , Humans , Leukotrienes/pharmacology , Lipid Peroxidation/drug effects , Lipoxins/pharmacology , Lipoxygenase Inhibitors/pharmacology , Nitric Oxide/metabolism , Prostaglandins/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL