1
|
Tomaszewska E, Rudyk H, Muszyński S, Hułas-Stasiak M, Leszczyński N, Mielnik-Błaszczak M, Donaldson J, Dobrowolski P. Prenatal Fumonisin Exposure Impairs Bone Development via Disturbances in the OC/Leptin and RANKL/RANK/OPG Systems in Weaned Rat Offspring. Int J Mol Sci 2023; 24:ijms24108743. [PMID: 37240089 DOI: 10.3390/ijms24108743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/10/2023] [Accepted: 05/13/2023] [Indexed: 05/28/2023] Open
Abstract
The goal of the current study was to examine the effects of prenatal exposure to fumonisins (FBs) on bone properties and metabolism in weaned rat offspring divided into groups intoxicated with FBs at either 0 (the 0 FB group), 60 (the 60 FB group), or 90 mg/kg b.w. 0 (the 90 FB group). Female and male offspring exposed to FBs at a dose of 60 mg/kg b.w. had heavier femora. Mechanical bone parameters changed in a sex and FBs dose-dependent manner. Growth hormone and osteoprotegerin decreased in both sexes, regardless of FBs dose. In males osteocalcin decreased, while receptor activator for nuclear factor kappa-Β ligand increased regardless of FBs dose; while in females changes were dose dependent. Leptin decreased in both male FBs-intoxicated groups, bone alkaline phosphatase decreased only in the 60 FB group. Matrix metalloproteinase-8 protein expression increased in both female FBs-intoxicated groups and decreased in male 90 FB group. Osteoprotegerin and tissue inhibitor of metalloproteinases 2 protein expression decreased in males, regardless of FBs dose, while nuclear factor kappa-Β ligand expression increased only in the 90 FB group. The disturbances in bone metabolic processes seemed to result from imbalances in the RANKL/RANK/OPG and the OC/leptin systems.
Collapse
Affiliation(s)
- Ewa Tomaszewska
- Department of Animal Physiology, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, 20-950 Lublin, Poland
| | - Halyna Rudyk
- Department of Animal Physiology, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, 20-950 Lublin, Poland
- Laboratory of Feed Additives and Premixtures Control, State Research Control Institute of Veterinary Drugs and Feed Additives, 79000 Lviv, Ukraine
| | - Siemowit Muszyński
- Department of Biophysics, Faculty of Environmental Biology, University of Life Sciences in Lublin, 20-950 Lublin, Poland
| | - Monika Hułas-Stasiak
- Department of Functional Anatomy and Cytobiology, Faculty of Biology and Biotechnology, Maria Curie-Sklodowska University, 20-033 Lublin, Poland
| | - Norbert Leszczyński
- Department of Agricultural, Forest and Transport Machinery, Faculty of Production Engineering, University of Life Sciences in Lublin, 20-612 Lublin, Poland
| | - Maria Mielnik-Błaszczak
- Chair and Department of Developmental Dentistry, Medical University of Lublin, 20-081 Lublin, Poland
| | - Janine Donaldson
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Parktown, Johannesburg 2193, South Africa
| | - Piotr Dobrowolski
- Department of Functional Anatomy and Cytobiology, Faculty of Biology and Biotechnology, Maria Curie-Sklodowska University, 20-033 Lublin, Poland
| |
Collapse
|
2
|
Clark KA, Shin AC, Sirivelu MP, MohanKumar RC, Maddineni SR, Ramachandran R, MohanKumar PS, MohanKumar SMJ. Evaluation of the Central Effects of Systemic Lentiviral-Mediated Leptin Delivery in Streptozotocin-Induced Diabetic Rats. Int J Mol Sci 2021; 22:ijms222413197. [PMID: 34947993 PMCID: PMC8703968 DOI: 10.3390/ijms222413197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/23/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022] Open
Abstract
Type 1 diabetes (T1D) is characterized by hyperphagia, hyperglycemia and activation of the hypothalamic-pituitary-adrenal (HPA) axis. We have reported previously that daily leptin injections help to alleviate these symptoms. Therefore, we hypothesized that leptin gene therapy could help to normalize the neuroendocrine dysfunction seen in T1D. Adult male Sprague Dawley rats were injected i.v. with a lentiviral vector containing the leptin gene or green fluorescent protein. Ten days later, they were injected with the vehicle or streptozotocin (STZ). HPA function was assessed by measuring norepinephrine (NE) levels in the paraventricular nucleus (PVN) and serum corticosterone (CS). Treatment with the leptin lentiviral vector (Lepvv) increased leptin and insulin levels in non-diabetic rats, but not in diabetic animals. There was a significant reduction in blood glucose levels in diabetic rats due to Lepvv treatment. Both NE levels in the PVN and serum CS were reduced in diabetic rats treated with Lepvv. Results from this study provide evidence that leptin gene therapy in STZ-induced diabetic rats was able to partially normalize some of the neuroendocrine abnormalities, but studies with higher doses of the Lepvv are needed to develop this into a viable option for treating T1D.
Collapse
MESH Headings
- Animals
- Corticosterone/blood
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/therapy
- Diabetes Mellitus, Type 1/chemically induced
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/therapy
- Disease Models, Animal
- Genetic Therapy
- Genetic Vectors/administration & dosage
- Injections, Intravenous
- Lentivirus/genetics
- Leptin/genetics
- Male
- Norepinephrine/metabolism
- Paraventricular Hypothalamic Nucleus/metabolism
- Rats
- Rats, Sprague-Dawley
Collapse
Affiliation(s)
- Kimberly A. Clark
- Neuroscience Graduate Program, Michigan State University, E. Lansing, MI 48824, USA; (K.A.C.); (P.S.M.)
| | - Andrew C. Shin
- Neurobiology of Nutrition Laboratory, Department of Nutritional Sciences, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA;
| | - Madhu P. Sirivelu
- Pathobiology and Diagnostic Investigation, Michigan State University, E. Lansing, MI 48824, USA;
| | - Ramya C. MohanKumar
- Neuroendocrine Research Laboratory, University of Georgia, Athens, GA 30602, USA;
| | - Sreenivasa R. Maddineni
- Department of Poultry Science, College of Agricultural Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (S.R.M.); (R.R.)
| | - Ramesh Ramachandran
- Department of Poultry Science, College of Agricultural Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (S.R.M.); (R.R.)
| | - Puliyur S. MohanKumar
- Neuroscience Graduate Program, Michigan State University, E. Lansing, MI 48824, USA; (K.A.C.); (P.S.M.)
- Pathobiology and Diagnostic Investigation, Michigan State University, E. Lansing, MI 48824, USA;
- Neuroendocrine Research Laboratory, University of Georgia, Athens, GA 30602, USA;
- Department of Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
| | - Sheba M. J. MohanKumar
- Neuroscience Graduate Program, Michigan State University, E. Lansing, MI 48824, USA; (K.A.C.); (P.S.M.)
- Neuroendocrine Research Laboratory, University of Georgia, Athens, GA 30602, USA;
- Department of Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
- Correspondence: ; Tel.: +1-706-542-1945
| |
Collapse
|
3
|
Corbin KD, Driscoll KA, Pratley RE, Smith SR, Maahs DM, Mayer-Davis EJ. Obesity in Type 1 Diabetes: Pathophysiology, Clinical Impact, and Mechanisms. Endocr Rev 2018; 39:629-663. [PMID: 30060120 DOI: 10.1210/er.2017-00191] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 06/21/2018] [Indexed: 02/07/2023]
Abstract
There has been an alarming increase in the prevalence of obesity in people with type 1 diabetes in recent years. Although obesity has long been recognized as a major risk factor for the development of type 2 diabetes and a catalyst for complications, much less is known about the role of obesity in the initiation and pathogenesis of type 1 diabetes. Emerging evidence suggests that obesity contributes to insulin resistance, dyslipidemia, and cardiometabolic complications in type 1 diabetes. Unique therapeutic strategies may be required to address these comorbidities within the context of intensive insulin therapy, which promotes weight gain. There is an urgent need for clinical guidelines for the prevention and management of obesity in type 1 diabetes. The development of these recommendations will require a transdisciplinary research strategy addressing metabolism, molecular mechanisms, lifestyle, neuropsychology, and novel therapeutics. In this review, the prevalence, clinical impact, energy balance physiology, and potential mechanisms of obesity in type 1 diabetes are described, with a special focus on the substantial gaps in knowledge in this field. Our goal is to provide a framework for the evidence base needed to develop type 1 diabetes-specific weight management recommendations that account for the competing outcomes of glycemic control and weight management.
Collapse
Affiliation(s)
- Karen D Corbin
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida
| | - Kimberly A Driscoll
- Department of Pediatrics, School of Medicine, University of Colorado Denver, Aurora, Colorado.,Barbara Davis Center for Diabetes, Aurora, Colorado
| | - Richard E Pratley
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida
| | - Steven R Smith
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida
| | - David M Maahs
- Division of Pediatric Endocrinology, Department of Pediatrics, Stanford University, Stanford, California
| | - Elizabeth J Mayer-Davis
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | |
Collapse
|
4
|
Philbrick KA, Martin SA, Colagiovanni AR, Branscum AJ, Turner RT, Iwaniec UT. Effects of hypothalamic leptin gene therapy on osteopetrosis in leptin-deficient mice. J Endocrinol 2018; 236:57-68. [PMID: 29191939 PMCID: PMC5771473 DOI: 10.1530/joe-17-0524] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 11/20/2017] [Indexed: 12/26/2022]
Abstract
Impaired resorption of cartilage matrix deposited during endochondral ossification is a defining feature of juvenile osteopetrosis. Growing, leptin-deficient ob/ob mice exhibit a mild form of osteopetrosis. However, the extent to which the disease is (1) self-limiting and (2) reversible by leptin treatment is unknown. We addressed the first question by performing histomorphometric analysis of femurs in rapidly growing (2-month-old), slowly growing (4-month-old) and skeletally mature (6-month-old) wild-type (WT) and ob/ob male mice. Absent by 6 months of age in WT mice, cartilage matrix persisted to varying extents in distal femur epiphysis, metaphysis and diaphysis in ob/ob mice, suggesting that the osteopetrotic phenotype is not entirely self-limiting. To address the second question, we employed hypothalamic recombinant adeno-associated virus (rAAV) gene therapy to restore leptin signaling in ob/ob mice. Two-month-old mice were randomized to one of the three groups: (1) untreated control, (2) rAAV-Leptin or (3) control vector rAAV-green fluorescent protein and vectors injected intracerebroventricularly. Seven months later, rAAV-leptin-treated mice exhibited no cartilage in the metaphysis and greatly reduced cartilage in the epiphysis and diaphysis. At the cellular level, the reduction in cartilage was associated with increased bone turnover. These findings (1) support the concept that leptin is important for normal replacement of cartilage by bone, and (2) demonstrate that osteopetrosis in ob/ob mice is bone-compartment-specific and reversible by leptin at skeletal sites capable of undergoing robust bone turnover.
Collapse
Affiliation(s)
- Kenneth A Philbrick
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Stephen A Martin
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Amy R Colagiovanni
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Adam J Branscum
- Biostatistics ProgramSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Russell T Turner
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
- Center for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USA
| | - Urszula T Iwaniec
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
- Center for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USA
| |
Collapse
|
5
|
D'souza AM, Neumann UH, Glavas MM, Kieffer TJ. The glucoregulatory actions of leptin. Mol Metab 2017; 6:1052-1065. [PMID: 28951828 PMCID: PMC5605734 DOI: 10.1016/j.molmet.2017.04.011] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/18/2017] [Accepted: 04/24/2017] [Indexed: 12/28/2022] Open
Abstract
Background The hormone leptin is an important regulator of metabolic homeostasis, able to inhibit food intake and increase energy expenditure. Leptin can also independently lower blood glucose levels, particularly in hyperglycemic models of leptin or insulin deficiency. Despite significant efforts and relevance to diabetes, the mechanisms by which leptin acts to regulate blood glucose levels are not fully understood. Scope of review Here we assess literature relevant to the glucose lowering effects of leptin. Leptin receptors are widely expressed in multiple cell types, and we describe both peripheral and central effects of leptin that may be involved in lowering blood glucose. In addition, we summarize the potential clinical application of leptin in regulating glucose homeostasis. Major conclusions Leptin exerts a plethora of metabolic effects on various tissues including suppressing production of glucagon and corticosterone, increasing glucose uptake, and inhibiting hepatic glucose output. A more in-depth understanding of the mechanisms of the glucose-lowering actions of leptin may reveal new strategies to treat metabolic disorders.
Collapse
Affiliation(s)
- Anna M D'souza
- Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | - Ursula H Neumann
- Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | - Maria M Glavas
- Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | - Timothy J Kieffer
- Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada.,Department of Surgery, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada
| |
Collapse
|
6
|
Ladyman SR, Grattan DR. Central Effects of Leptin on Glucose Homeostasis are Modified during Pregnancy in the Rat. J Neuroendocrinol 2016; 28. [PMID: 27623562 DOI: 10.1111/jne.12431] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 08/03/2016] [Accepted: 09/10/2016] [Indexed: 01/03/2023]
Abstract
Despite increased leptin concentrations during pregnancy, fat mass and food intake are increased. The satiety response to central leptin is suppressed, indicating a state of leptin insensitivity in the hypothalamus. Although the regulation of food intake is a major function of leptin, this hormone also influences a wide range of functions within the body. These actions include the regulation of glucose homeostasis, which undergoes major adaptation in the maternal body to generate optimal conditions for foetal development and growth. The present study aimed to investigate the effects of central leptin treatment on glucose homeostasis in pregnant rats to determine whether pregnancy-induced leptin insensitivity is functionally specific, and to further investigate changes in glucose homeostasis during pregnancy. After an overnight fast, nonpregnant and day 14 pregnant rats received an i.c.v. injection of leptin (100 ng or 4 μg) or vehicle then underwent a glucose tolerance test (GTT). Further groups of nonpregnant and day 14 pregnant rats were killed 30 min after leptin (doses ranging from 40 ng-4 μg) or vehicle i.c.v. injections for western blot analysis of phospho-signal transducer and activator of transcription 3 (STAT3) and phospho-Akt in various hypothalamic nuclei. Central leptin injection prior to a GTT lead to lowered basal insulin concentrations and impaired glucose tolerance in nonpregnant female rats, whereas the same doses of leptin had no significant effect on glucose tolerance in day 14 pregnant rats, indicating that, similar to the satiety actions of leptin, the effects of leptin on glucose homeostasis are suppressed during pregnancy. Furthermore, in the arcuate nucleus and ventromedial and dorsomedial nuclei of the hypothalamus, comprising three leptin-sensitive areas, there was no evidence that leptin induced Akt phosphorylation despite significant increases in phospho-STAT3, suggesting that leptin does not act through phospho-Akt in these areas in female rats.
Collapse
Affiliation(s)
- S R Ladyman
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago, Dunedin, New Zealand.
| | - D R Grattan
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
7
|
Denroche HC, Glavas MM, Tudurí E, Karunakaran S, Quong WL, Philippe M, Britton HM, Clee SM, Kieffer TJ. Disrupted Leptin Signaling in the Lateral Hypothalamus and Ventral Premammillary Nucleus Alters Insulin and Glucagon Secretion and Protects Against Diet-Induced Obesity. Endocrinology 2016; 157:2671-85. [PMID: 27183315 DOI: 10.1210/en.2015-1998] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Leptin signaling in the central nervous system, and particularly the arcuate hypothalamic nucleus, is important for regulating energy and glucose homeostasis. However, the roles of extra-arcuate leptin responsive neurons are less defined. In the current study, we generated mice with widespread inactivation of the long leptin receptor isoform in the central nervous system via Synapsin promoter-driven Cre (Lepr(flox/flox) Syn-cre mice). Within the hypothalamus, leptin signaling was disrupted in the lateral hypothalamic area (LHA) and ventral premammillary nucleus (PMV) but remained intact in the arcuate hypothalamic nucleus and ventromedial hypothalamic nucleus, dorsomedial hypothalamic nucleus, and nucleus of the tractus solitarius. To investigate the role of LHA/PMV neuronal leptin signaling, we examined glucose and energy homeostasis in Lepr(flox/flox) Syn-cre mice and Lepr(flox/flox) littermates under basal and diet-induced obese conditions and tested the role of LHA/PMV neurons in leptin-mediated glucose lowering in streptozotocin-induced diabetes. Lepr(flox/flox) Syn-cre mice did not have altered body weight or blood glucose levels but were hyperinsulinemic and had enhanced glucagon secretion in response to experimental hypoglycemia. Surprisingly, when placed on a high-fat diet, Lepr(flox/flox) Syn-cre mice were protected from weight gain, glucose intolerance, and diet-induced hyperinsulinemia. Peripheral leptin administration lowered blood glucose in streptozotocin-induced diabetic Lepr(flox/flox) Syn-cre mice as effectively as in Lepr(flox/flox) littermate controls. Collectively these findings suggest that leptin signaling in LHA/PMV neurons is not critical for regulating glucose levels but has an indispensable role in the regulation of insulin and glucagon levels and, may promote the development of diet-induced hyperinsulinemia and weight gain.
Collapse
Affiliation(s)
- Heather C Denroche
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Maria M Glavas
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Eva Tudurí
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Subashini Karunakaran
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Whitney L Quong
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Marion Philippe
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Heidi M Britton
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Susanne M Clee
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Timothy J Kieffer
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| |
Collapse
|
8
|
Denroche HC, Kwon MM, Glavas MM, Tudurí E, Philippe M, Quong WL, Kieffer TJ. The role of autonomic efferents and uncoupling protein 1 in the glucose-lowering effect of leptin therapy. Mol Metab 2016; 5:716-724. [PMID: 27656409 PMCID: PMC5021671 DOI: 10.1016/j.molmet.2016.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 06/19/2016] [Indexed: 01/06/2023] Open
Abstract
Objective Leptin reverses hyperglycemia in rodent models of type 1 diabetes (T1D). Direct application of leptin to the brain can lower blood glucose in diabetic rodents, and can activate autonomic efferents and non-shivering thermogenesis in brown adipose tissue (BAT). We investigated whether leptin reverses hyperglycemia through a mechanism that requires autonomic innervation, or uncoupling protein 1 (UCP1)-mediated thermogenesis. Methods To examine the role of parasympathetic and sympathetic efferents in the glucose-lowering action of leptin, mice with a subdiaphragmatic vagotomy or 6-hydroxydopamine induced chemical sympathectomy were injected with streptozotocin (STZ) to induce hyperglycemia, and subsequently leptin treated. To test whether the glucose-lowering action of leptin requires activation of UCP1-mediated thermogenesis in BAT, we administered leptin in STZ-diabetic Ucp1 knockout (Ucp1−/−) mice and wildtype controls. Results Leptin ameliorated STZ-induced hyperglycemia in both intact and vagotomised mice. Similarly, mice with a partial chemical sympathectomy did not have an attenuated response to leptin-mediated glucose lowering relative to sham controls, and showed intact leptin-induced Ucp1 expression in BAT. Although leptin activated BAT thermogenesis in STZ-diabetic mice, the anti-diabetic effect of leptin was not blunted in Ucp1−/− mice. Conclusions These results suggest that leptin lowers blood glucose in insulin-deficient diabetes through a manner that does not require parasympathetic or sympathetic innervation, and thus imply that leptin lowers blood glucose through an alternative CNS-mediated mechanism or redundant target tissues. Furthermore, we conclude that the glucose lowering action of leptin is independent of UCP1-dependent thermogenesis. Leptin does not require vagal innervation to reverse hyperglycemia. Leptin therapy reverses hyperglycemia in mice with a partial chemical sympathectomy. Leptin reverses hyperglycemia independent of uncoupling protein 1.
Collapse
Key Words
- 6OHDA, 6-hydroxydopamine
- ANS, autonomic nervous system
- BAT, brown adipose tissue
- Brown adipose tissue
- CCK, cholecystokinin
- CNS, central nervous system
- Glucose
- STZ, streptozotocin
- Streptozotocin
- Sympathectomy
- T1D, type 1 diabetes
- TH, tyrosine hydroxylase
- Type 1 diabetes
- UCP1, uncoupling protein 1
- Vagotomy
- iBAT, interscapular BAT
Collapse
Affiliation(s)
- Heather C Denroche
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michelle M Kwon
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Maria M Glavas
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Eva Tudurí
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Marion Philippe
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Whitney L Quong
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Timothy J Kieffer
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada; Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
9
|
Coleman SK, Rebalka IA, D’Souza DM, Hawke TJ. Skeletal muscle as a therapeutic target for delaying type 1 diabetic complications. World J Diabetes 2015; 6:1323-1336. [PMID: 26674848 PMCID: PMC4673386 DOI: 10.4239/wjd.v6.i17.1323] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/01/2015] [Accepted: 11/25/2015] [Indexed: 02/05/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic autoimmune disease targeting the pancreatic beta-cells and rendering the person hypoinsulinemic and hyperglycemic. Despite exogenous insulin therapy, individuals with T1DM will invariably develop long-term complications such as blindness, kidney failure and cardiovascular disease. Though often overlooked, skeletal muscle is also adversely affected in T1DM, with both physical and metabolic derangements reported. As the largest metabolic organ in the body, impairments to skeletal muscle health in T1DM would impact insulin sensitivity, glucose/lipid disposal and basal metabolic rate and thus affect the ability of persons with T1DM to manage their disease. In this review, we discuss the impact of T1DM on skeletal muscle health with a particular focus on the proposed mechanisms involved. We then identify and discuss established and potential adjuvant therapies which, in association with insulin therapy, would improve the health of skeletal muscle in those with T1DM and thereby improve disease management- ultimately delaying the onset and severity of other long-term diabetic complications.
Collapse
|
10
|
Kalani A, Kamat PK, Tyagi N. Diabetic Stroke Severity: Epigenetic Remodeling and Neuronal, Glial, and Vascular Dysfunction. Diabetes 2015; 64:4260-71. [PMID: 26470785 PMCID: PMC4876759 DOI: 10.2337/db15-0422] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 08/28/2015] [Indexed: 01/02/2023]
Abstract
We determined the mechanism of severity during type 1 diabetic (T1D) stroke (ischemia-reperfusion [IR] injury) that affects potential markers associated with epigenetics, neuronal, glial, and vascular components of the brain with regard to nondiabetic stroke. The study used male genetic T1D Ins2(+/-) Akita and wild-type (C57BL/6J) mice. The experimental mice groups were 1) sham, 2) IR, 3) sham(Akita), and 4) IR(Akita). Mice were subjected to middle cerebral artery occlusion for 40 min, followed by reperfusion for 24 h. Brain tissues were analyzed for inflammation, neuro-glio-vascular impairments, matrix metalloproteinase (MMP)-9 expression, and epigenetic alterations (DNA methyltransferase-3a [DNMT-3a]; DNA methyltransferase-1 [DNMT-1]; 5-methylcytosine [5-mC]; and 5-hydroxymethylcytosine [5-hmC]). Intracarotid fluorescein isothiocyanate-BSA infusion was used to determine pial-venular permeability. IR(Akita) mice showed more infarct volume, edema, inflammation, and vascular MMP-9 expression compared with IR and sham groups. Sham(Akita) mice showed the highest DNMT-1 and DNMT-3a levels compared with the other groups. Reduced tight and adherent junction expressions and severe venular leakage exemplified intense cerebrovascular impairment in IR(Akita) mice compared with the other groups. Interestingly, we found differential regulations (downregulated expression) of epigenetic (5-mC, DNMTs), vascular (endothelial nitric oxide synthase), glial (connexin-43, glial fibrillary acidic protein, CD11b), and neuronal (neuron-specific enolase, neuronal nitric oxide synthase) markers in IR(Akita) compared with the IR group. These findings suggest that IR injury in T1D is more severe because it intensifies differential epigenetic markers and neuro-glio-vascular changes compared with nondiabetic mice.
Collapse
Affiliation(s)
- Anuradha Kalani
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, KY
| | - Pradip Kumar Kamat
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, KY
| | - Neetu Tyagi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, KY
| |
Collapse
|
11
|
Denroche HC, Kwon MM, Quong WL, Neumann UH, Kulpa JE, Karunakaran S, Clee SM, Brownsey RW, Covey SD, Kieffer TJ. Leptin induces fasting hypoglycaemia in a mouse model of diabetes through the depletion of glycerol. Diabetologia 2015; 58:1100-8. [PMID: 25715699 DOI: 10.1007/s00125-015-3529-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 01/28/2015] [Indexed: 12/27/2022]
Abstract
AIMS/HYPOTHESIS Leptin has profound glucose-lowering effects in rodent models of type 1 diabetes, and is currently being tested clinically to treat this disease. In addition to reversing hyperglycaemia, leptin therapy corrects multiple lipid, energy and neuroendocrine imbalances in rodent models of type 1 diabetes, yet the precise mechanism has not been fully defined. Thus, we performed metabolic analyses to delineate the downstream metabolic pathway mediating leptin-induced glucose lowering in diabetic mice. METHODS Mice were injected with streptozotocin (STZ) to induce insulin-deficient diabetes, and were subsequently treated with 20 μg/day recombinant murine leptin or vehicle for 5 to 14 days. Energy-yielding substrates were measured in the liver and plasma, and endogenous glucose production was assessed by tolerance to extended fasting. RESULTS STZ-leptin-treated mice developed severe hypoketotic hypoglycaemia during prolonged fasting, indicative of suppressed endogenous ketone and glucose production. STZ-leptin mice displayed normal gluconeogenic and glycogenolytic capacity, but had depleted circulating glycerol and NEFA. The depletion of glycerol and NEFA correlated tightly with the kinetics of glucose lowering in response to chronic leptin administration, and was not mimicked by single leptin injection. Administration of glycerol acutely reversed fasting-induced hypoglycaemia in leptin-treated mice. CONCLUSIONS/INTERPRETATION The findings of this study suggest that the diminution of circulating glycerol reduces endogenous glucose production, contributing to severe fasting-induced hypoglycaemia in leptin-treated rodent models of type 1 diabetes, and support that depletion of glycerol contributes to the glucose-lowering action of leptin.
Collapse
Affiliation(s)
- Heather C Denroche
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada, V6T 1Z3
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Bhaswant M, Poudyal H, Brown L. Mechanisms of enhanced insulin secretion and sensitivity with n-3 unsaturated fatty acids. J Nutr Biochem 2015; 26:571-84. [PMID: 25841249 DOI: 10.1016/j.jnutbio.2015.02.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 02/09/2015] [Accepted: 02/11/2015] [Indexed: 12/16/2022]
Abstract
The widespread acceptance that increased dietary n-3 polyunsaturated fatty acids (PUFAs), especially α-linolenic acid (ALA), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), improve health is based on extensive studies in animals, isolated cells and humans. Visceral adiposity is part of the metabolic syndrome, together with insulin resistance, dyslipidemia, hypertension and inflammation. Alleviation of metabolic syndrome requires normalization of insulin release and responses. This review assesses our current knowledge of the mechanisms that allow n-3 PUFAs to improve insulin secretion and sensitivity. EPA has been more extensively studied than either ALA or DHA. The complex actions of EPA include increased G-protein-receptor-mediated release of glucagon-like peptide 1 (GLP-1) from enteroendocrine L-cells in the intestine, up-regulation of the apelin pathway and down-regulation of other control pathways to promote insulin secretion by the pancreatic β-cells, together with suppression of inflammatory responses to adipokines, inhibition of peroxisome proliferator-activated receptor α actions and prevention of decreased insulin-like growth factor-1 secretion to improve peripheral insulin responses. The receptors involved and the mechanisms of action probably differ for ALA and DHA, with antiobesity effects predominating for ALA and anti-inflammatory effects for DHA. Modifying both GLP-1 release and the actions of adipokines by n-3 PUFAs could lead to additive improvements in both insulin secretion and sensitivity.
Collapse
Affiliation(s)
- Maharshi Bhaswant
- Centre for Chronic Disease Prevention & Management, College of Health and Biomedicine, Victoria University, Melbourne VIC 3021, Australia; School of Health and Wellbeing, University of Southern Queensland, Toowoomba QLD 4350, Australia
| | - Hemant Poudyal
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine and The Hakubi Center for Advanced Research, Kyoto University, Kyoto 606-8302, Japan
| | - Lindsay Brown
- School of Health and Wellbeing, University of Southern Queensland, Toowoomba QLD 4350, Australia.
| |
Collapse
|
13
|
Davis C, Mudd J, Hawkins M. Neuroprotective effects of leptin in the context of obesity and metabolic disorders. Neurobiol Dis 2014; 72 Pt A:61-71. [DOI: 10.1016/j.nbd.2014.04.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 04/09/2014] [Accepted: 04/21/2014] [Indexed: 12/16/2022] Open
|
14
|
Odle AK, Haney A, Allensworth-James M, Akhter N, Childs GV. Adipocyte versus pituitary leptin in the regulation of pituitary hormones: somatotropes develop normally in the absence of circulating leptin. Endocrinology 2014; 155:4316-28. [PMID: 25116704 PMCID: PMC4197982 DOI: 10.1210/en.2014-1172] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Leptin is a cytokine produced by white fat cells, skeletal muscle, the placenta, and the pituitary gland among other tissues. Best known for its role in regulating appetite and energy expenditure, leptin is produced largely by and in proportion to white fat cells. Leptin is also important to the maintenance and function of the GH cells of the pituitary. This was shown when the deletion of leptin receptors on somatotropes caused decreased numbers of GH cells, decreased circulating GH, and adult-onset obesity. To determine the source of leptin most vital to GH cells and other pituitary cell types, we compared two different leptin knockout models with Cre-lox technology. The global Lep-null model is like the ob/ob mouse, whereby only the entire exon 3 is deleted. The selective adipocyte-Lep-null model lacks adipocyte leptin but retains pituitary leptin, allowing us to investigate the pituitary as a potential source of circulating leptin. Male and female mice lacking adipocyte leptin (Adipocyte-lep-null) did not produce any detectable circulating leptin and were infertile, suggesting that the pituitary does not contribute to serum levels. In the presence of only pituitary leptin, however, these same mutants were able to maintain somatotrope numbers and GH mRNA levels. Serum GH trended low, but values were not significant. However, hypothalamic GHRH mRNA was significantly reduced in these animals. Other serum hormone and pituitary mRNA differences were observed, some of which varied from previous results reported in ob/ob animals. Whereas pituitary leptin is capable of maintaining somatotrope numbers and GH mRNA production, the decreased hypothalamic GHRH mRNA and low (but not significant) serum GH levels indicate an important role for adipocyte leptin in the regulation of GH secretion in the mouse. Thus, normal GH secretion may require the coordinated actions of both adipocyte and pituitary leptin.
Collapse
Affiliation(s)
- Angela K Odle
- Department of Neurobiology and Developmental Sciences, College of Medicine, Center for Translational Neurosciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | | | | | | | | |
Collapse
|
15
|
Abstract
The fat‐derived hormone, leptin, is well known to regulate body weight. However, there is now substantial evidence that leptin also plays a primary role in the regulation of glucose homeostasis, independent of actions on food intake, energy expenditure or body weight. As such, leptin might have clinical utility in treating hyperglycemia, particularly in conditions of leptin deficiency, such as lipodystrophy and diabetes mellitus. The mechanisms through which leptin modulates glucose metabolism have not been fully elucidated. Leptin receptors are widely expressed in peripheral tissues, including the endocrine pancreas, liver, skeletal muscle and adipose, and both direct and indirect leptin action on these tissues contributes to the control of glucose homeostasis. Here we review the role of leptin in glucose homeostasis, along with our present understanding of the mechanisms involved. (J Diabetes Invest, doi: 10.1111/j.2040‐1124.2012.00203.x, 2012)
Collapse
Affiliation(s)
- Heather C Denroche
- Department of Cellular and Physiological Sciences, The Life Sciences Institute
| | - Frank K Huynh
- Department of Cellular and Physiological Sciences, The Life Sciences Institute
| | - Timothy J Kieffer
- Department of Cellular and Physiological Sciences, The Life Sciences Institute ; Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
16
|
Amitani M, Asakawa A, Amitani H, Inui A. The role of leptin in the control of insulin-glucose axis. Front Neurosci 2013; 7:51. [PMID: 23579596 PMCID: PMC3619125 DOI: 10.3389/fnins.2013.00051] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 03/18/2013] [Indexed: 12/21/2022] Open
Abstract
Obesity and diabetes mellitus are great public health concerns throughout the world because of their increasing incidence and prevalence. Leptin, the adipocyte hormone, is well known for its role in the regulation of food intake and energy expenditure. In addition to the regulation of appetite and satiety that recently has attracted much attentions, insight has also been gained into the critical role of leptin in the control of the insulin-glucose axis, peripheral glucose and insulin responsiveness. Since the discovery of leptin, leptin has been taken for its therapeutic potential to obesity and diabetes. Recently, the therapeutic effects of central leptin gene therapy have been reported in insulin-deficient diabetes in obesity animal models such as ob/ob mise, diet-induced obese mice, and insulin-deficient type 1 diabetes mice, and also in patients with inactivating mutations in the leptin gene. Herein, we review the role of leptin in regulating feeding behavior and glucose metabolism and also the therapeutic potential of leptin in obesity and diabetes mellitus.
Collapse
Affiliation(s)
- Marie Amitani
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima, Japan
| | | | | | | |
Collapse
|
17
|
Minokoshi Y, Toda C, Okamoto S. Regulatory role of leptin in glucose and lipid metabolism in skeletal muscle. Indian J Endocrinol Metab 2012; 16:S562-S568. [PMID: 23565491 PMCID: PMC3602985 DOI: 10.4103/2230-8210.105573] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Leptin is a hormone secreted by adipocytes that plays a pivotal role in regulation of food intake, energy expenditure, and neuroendocrine function. Several lines of evidences indicate that independent of the anorexic effect, leptin regulates glucose and lipid metabolism in peripheral tissues in rodents and humans. It has been shown that leptin improves the diabetes phenotype in lipodystrophic patients and rodents. Moreover, leptin suppresses the development of severe, progressive impairment of glucose metabolism in insulin-deficient diabetes in rodents. We found that leptin increases glucose uptake and fatty acid oxidation in skeletal muscle in rats and mice in vivo. Leptin increases glucose uptake in skeletal muscle via the hypothalamic-sympathetic nervous system axis and β-adrenergic mechanism, while leptin stimulates fatty acid oxidation in muscle via AMP-activated protein kinase (AMPK). Leptin-induced fatty acid oxidation results in the decrease of lipid accumulation in muscle, which can lead to functional impairments called as "lipotoxicity." Activation of AMPK occurs by direct action of leptin on muscle and through the medial hypothalamus-sympathetic nervous system and α-adrenergic mechanism. Thus, leptin plays an important role in the regulation of glucose and fatty acid metabolism in skeletal muscle.
Collapse
Affiliation(s)
- Yasuhiko Minokoshi
- Division of Endocrinology and Metabolism, National Institute for Physiological Sciences, Myodaiji, Okazaki, Aichi - 444-8787, Japan
| | - Chitoku Toda
- Division of Endocrinology and Metabolism, National Institute for Physiological Sciences, Myodaiji, Okazaki, Aichi - 444-8787, Japan
| | - Shiki Okamoto
- Division of Endocrinology and Metabolism, National Institute for Physiological Sciences, Myodaiji, Okazaki, Aichi - 444-8787, Japan
| |
Collapse
|
18
|
Nakano M, Asakawa A, Inui A. Long-term correction of type 1 and 2 diabetes by central leptin gene therapy independent of effects on appetite and energy expenditure. Indian J Endocrinol Metab 2012; 16:S556-S561. [PMID: 23565490 PMCID: PMC3602984 DOI: 10.4103/2230-8210.105572] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Adipocyte-derived leptin is a hormone associated with the regulation of energy homeostasis, including glucose metabolism. Hyperleptinemia, induced by the consumption of energy-enriched diets, inhibits leptin transport across the blood-brain barrier, and thereby produces leptin insufficiency in the hypothalamus. As a result of sustained leptin insufficiency, the hypothalamic restraint on pancreatic insulin secretion is lost. Additionally, both glucose metabolism and energy expenditure are also diminished, and both type 1 and type 2 diabetes are induced. A replication-deficient recombinant adeno-associated virus vector engineered to encode the leptin gene (rAVV-LEP) has been used in models of diabetes as a novel therapeutic approach. After rAVV-LEP injection in ob/ob mice, hypothalamic leptin expression was increased, body weight was suppressed, and hyperinsulinemia was ameliorated. Additionally injection of rAVV-LEP into the hypothalamus suppressed the expression of orexigenic neuropeptide Y (NPY) and enhanced anorexigenic pro-opiomelanocortin (POMC) in the arcuate nucleus (ARC) in rats. It is proposed that central leptin gene therapy should be tested clinically to reduce the worldwide epidemic of obesity, diabetes, and shortened life span. In this article, the information has been assembled from published review articles on this topic.
Collapse
Affiliation(s)
- Masako Nakano
- Department of Social and Behavioral Medicine, Kagoshima University Graduate Medical and Dental Sciences, Kagoshima, Japan
| | - Akihiro Asakawa
- Department of Social and Behavioral Medicine, Kagoshima University Graduate Medical and Dental Sciences, Kagoshima, Japan
| | - Akio Inui
- Department of Social and Behavioral Medicine, Kagoshima University Graduate Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
19
|
Affiliation(s)
- Satya P. Kalra
- Department of Neuroscience and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, 32610, USA
| |
Collapse
|
20
|
Naito M, Fujikura J, Ebihara K, Miyanaga F, Yokoi H, Kusakabe T, Yamamoto Y, Son C, Mukoyama M, Hosoda K, Nakao K. Therapeutic impact of leptin on diabetes, diabetic complications, and longevity in insulin-deficient diabetic mice. Diabetes 2011; 60:2265-73. [PMID: 21810600 PMCID: PMC3161331 DOI: 10.2337/db10-1795] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE The aim of the current study was to evaluate the long-term effects of leptin on glucose metabolism, diabetes complications, and life span in an insulin-dependent diabetes model, the Akita mouse. RESEARCH DESIGN AND METHODS We cross-mated Akita mice with leptin-expressing transgenic (LepTg) mice to produce Akita mice with physiological hyperleptinemia (LepTg:Akita). Metabolic parameters were monitored for 10 months. Pair-fed studies and glucose and insulin tolerance tests were performed. The pancreata and kidneys were analyzed histologically. The plasma levels and pancreatic contents of insulin and glucagon, the plasma levels of lipids and a marker of oxidative stress, and urinary albumin excretion were measured. Survival rates were calculated. RESULTS Akita mice began to exhibit severe hyperglycemia and hyperphagia as early as weaning. LepTg:Akita mice exhibited normoglycemia after an extended fast even at 10 months of age. The 6-h fasting blood glucose levels in LepTg:Akita mice remained about half the level of Akita mice throughout the study. Food intake in LepTg:Akita mice was suppressed to a level comparable to that in WT mice, but pair feeding did not affect blood glucose levels in Akita mice. LepTg:Akita mice maintained insulin hypersensitivity and displayed better glucose tolerance than did Akita mice throughout the follow-up. LepTg:Akita mice had normal levels of plasma glucagon, a marker of oxidative stress, and urinary albumin excretion rates. All of the LepTg:Akita mice survived for >12 months, the median mortality time of Akita mice. CONCLUSIONS These results indicate that leptin is therapeutically useful in the long-term treatment of insulin-deficient diabetes.
Collapse
|
21
|
Iwaniec UT, Boghossian S, Trevisiol CH, Wronski TJ, Turner RT, Kalra SP. Hypothalamic leptin gene therapy prevents weight gain without long-term detrimental effects on bone in growing and skeletally mature female rats. J Bone Miner Res 2011; 26:1506-16. [PMID: 21328617 PMCID: PMC3129999 DOI: 10.1002/jbmr.365] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Hypothalamic leptin gene therapy normalizes the mosaic skeletal phenotype of leptin-deficient ob/ob mice. However, it is not clear whether increased hypothalamic leptin alters bone metabolism in animals already producing the hormone. The objective of this study was to evaluate the long duration effects of recombinant adeno-associated virus-rat leptin (rAAV-Lep) hypothalamic gene therapy on weight gain and bone metabolism in growing and skeletally mature leptin-replete female Sprague-Dawley rats. Rats were either unoperated or implanted with cannulas in the third ventricle of the hypothalamus and injected with either rAAV-Lep or rAAV-GFP (control vector encoding green fluorescent protein) and maintained on standard rat chow fed ad libitum for either 5 or 10 weeks (starting at 3 months of age) or 18 weeks (starting at 9 months of age). Tibias, femurs, or lumbar vertebrae were analyzed by micro-computed tomography and/or histomorphometry. In comparison with age-matched rAAV-GFP rats, rAAV-Lep rats maintained a lower body weight for the duration of studies. At 5 weeks after vector administration, rAAV-Lep rats had lower cancellous bone volume and bone marrow adiposity but higher osteoblast perimeter compared with nonoperated controls. However, these values did not differ between the two groups at 10 weeks after vector administration. Differences in cancellous bone volume and architecture were not detected between the rAAV-Lep and rAAV-GFP groups at either time point. Also, rAAV-Lep had no negative effects on bone in the 9-month-old skeletally mature rats at 18 weeks after vector administration. We hypothesize that the transient reductions in bone mass and bone marrow adiposity at 5 weeks after vector administration were due to hypothalamic surgery. We conclude that increased hypothalamic leptin, sufficient to prevent weight gain, has minimal specific effects (rAAV-Lep versus rAAV-GFP) on bone metabolism in normal female rats.
Collapse
Affiliation(s)
- Urszula T Iwaniec
- Department of Nutrition and Exercise Sciences, Oregon State University, Corvallis, OR 97331, USA.
| | | | | | | | | | | |
Collapse
|
22
|
Marino JS, Xu Y, Hill JW. Central insulin and leptin-mediated autonomic control of glucose homeostasis. Trends Endocrinol Metab 2011; 22:275-85. [PMID: 21489811 PMCID: PMC5154334 DOI: 10.1016/j.tem.2011.03.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 02/25/2011] [Accepted: 03/05/2011] [Indexed: 12/17/2022]
Abstract
Largely as a result of rising obesity rates, the incidence of type 2 diabetes is escalating rapidly. Type 2 diabetes results from multi-organ dysfunctional glucose metabolism. Recent publications have highlighted hypothalamic insulin- and adipokine-sensing as a major determinant of peripheral glucose and insulin responsiveness. The preponderance of evidence indicates that the brain is the master regulator of glucose homeostasis, and that hypothalamic insulin and leptin signaling in particular play a crucial role in the development of insulin resistance. This review discusses the neuronal crosstalk between the hypothalamus, autonomic nervous system, and tissues associated with the pathogenesis of type 2 diabetes, and how hypothalamic insulin and leptin signaling are integral to maintaining normal glucose homeostasis.
Collapse
Affiliation(s)
- Joseph S Marino
- Center for Diabetes and Endocrine Research, College of Medicine, The University of Toledo, Toledo, OH 43614, USA
| | | | | |
Collapse
|
23
|
Gautron L, Elmquist JK. Sixteen years and counting: an update on leptin in energy balance. J Clin Invest 2011; 121:2087-93. [PMID: 21633176 DOI: 10.1172/jci45888] [Citation(s) in RCA: 245] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cloned in 1994, the ob gene encodes the protein hormone leptin, which is produced and secreted by white adipose tissue. Since its discovery, leptin has been found to have profound effects on behavior, metabolic rate, endocrine axes, and glucose fluxes. Leptin deficiency in mice and humans causes morbid obesity, diabetes, and various neuroendocrine anomalies, and replacement leads to decreased food intake, normalized glucose homeostasis, and increased energy expenditure. Here, we provide an update on the most current understanding of leptin-sensitive neural pathways in terms of both anatomical organization and physiological roles.
Collapse
Affiliation(s)
- Laurent Gautron
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | |
Collapse
|
24
|
Wang Y, Asakawa A, Inui A, Kosai KI. Leptin gene therapy in the fight against diabetes. Expert Opin Biol Ther 2011; 10:1405-14. [PMID: 20690892 DOI: 10.1517/14712598.2010.512286] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
IMPORTANCE OF THE FIELD The incidence of diabetes is increasing worldwide, yet current treatments are not always effective for all patient or disease types. AREAS COVERED IN THIS REVIEW Here, we summarize the biologic and clinical roles of leptin in diabetes, and discuss candidate viral vectors that may be employed in the clinical use of central leptin gene therapy for diabetes. WHAT THE READER WILL GAIN We discuss how studies on leptin, a regulator of the insulin-glucose axis, have significantly advanced our understanding of the roles of energy homeostasis and insulin resistance in the pathogeneses of metabolic syndrome and diabetes. Recent studies have demonstrated the long-term therapeutic effects of central leptin gene therapy in obesity and diabetes via decreased insulin resistance and increased glucose metabolism. Many of these studies have employed viral vectors, which afford high in vivo gene transduction efficiencies compared with non-viral vectors. TAKE HOME MESSAGE Adeno-associated viral vectors are particularly well suited for central leptin gene therapy owing to their low toxicity and ability to drive transgene expression for extended periods.
Collapse
Affiliation(s)
- Yuqing Wang
- Kagoshima University Graduate School of Medical and Dental Sciences, Department of Gene Therapy and Regenerative Medicine, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | | | | | | |
Collapse
|
25
|
Kalra SP. Pivotal role of leptin-hypothalamus signaling in the etiology of diabetes uncovered by gene therapy: a new therapeutic intervention? Gene Ther 2011; 18:319-25. [PMID: 21209624 DOI: 10.1038/gt.2010.164] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The incidence of diabetes mellitus has soared to epidemic proportion worldwide. The debilitating chronic hyperglycemia is caused by either lack of insulin as in diabetes type 1 or its ineffectiveness as in diabetes type 2. Frequent replacement of insulin with or without insulin analogs for optimum glycemic control are the conventional cumbersome therapies. Recent application of leptin gene transfer technology has uncovered the participation of adipocytes-derived leptin-dependent hypothalamic neural signaling in glucose homeostasis and demonstrated that a breakdown in this communication due to leptin insufficiency in the hypothalamus underlies the etiology of chronic hyperglycemia. Reinstatement of central leptin sufficiency by hyperleptinemia produced either by intravenous leptin infusion or a single systemic injection of recombinant adenovirus vector encoding leptin gene suppressed hyperglycemia and evoked euglycemia only transiently in rodent models of diabetes type 1. In contrast, stable restoration of leptin sufficiency, solely in the hypothalamus, with biologically active leptin transduced by an intracerebroventicular injection of recombinant adeno-associated virus vector encoding leptin gene (rAAV-lep) abolished hyperglycemia and imposed euglycemia through the extended duration of experiment by stimulating glucose disposal in the periphery in models of diabetes type 1. Further, similar hypothalamic leptin transgene expression abrogated chronic hyperglycemia and hyperinsulinemia, the predisposing risk factors of the age and environmentally acquired diabetes type 2, and instituted euglycemia by independently activating relays that stimulate glucose metabolism and repress hyperinsulinemia and improve insulin sensitivity in the periphery. Consequently, this durable antidiabetic efficacy of one time rAAV-lep neurotherapy offers a potential novel substitute for insulin therapy following preclinical trials in subhuman primates and humans.
Collapse
Affiliation(s)
- S P Kalra
- Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610-0244, USA.
| |
Collapse
|
26
|
Turner R, Iwaniec U. Moderate weight gain does not influence bone metabolism in skeletally mature female rats. Bone 2010; 47:631-5. [PMID: 20601291 PMCID: PMC2926277 DOI: 10.1016/j.bone.2010.06.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2010] [Accepted: 06/12/2010] [Indexed: 01/07/2023]
Abstract
Bone mass is correlated with body weight during growth. However, it is unclear how bone mass is influenced by weight gain following skeletal maturity. The purpose of this study was to determine the effects of weight maintenance and two rates of weight gain on bone metabolism using skeletally mature female rats. Eight-month-old female rats were fed one of 3 diets for 13 weeks: Lieber-DeCarli liquid diet ad lib (control diet), the same diet with caloric restriction to maintain initial body weight (calorie-restricted diet), and the same diet fed ad lib with the exception that appetite was enhanced (calorie-increased diet) by replacing a small quantity of maltose-dextran isocalorically with ethanol (0.5% caloric intake). Compared to baseline, rats fed the calorie-restricted, control, and calorie-increased diets changed in weight by -1+/-2% (mean+/-SE), 10+/-3%, and 21+/-2%, respectively. Weight gain was associated with a significant increase in serum leptin, a putative regulator of bone formation. In contrast, significant differences in tibial bone mineral content and density were not detected among treatments groups following dietary intervention or between treatment groups and the baseline group. Similarly, indices of cancellous bone architecture (area, trabecular number, thickness, and separation) and bone turnover (mineralizing perimeter, mineral apposition rate, and bone formation rate) did not differ among groups following dietary intervention. Our findings suggest that neither weight gain nor increased serum leptin levels, over the range evaluated, influence bone metabolism in skeletally mature female rats.
Collapse
Affiliation(s)
- R.T. Turner
- Department of Nutrition and Exercise Sciences, Oregon State University, Corvallis, OR 97331
| | - U.T. Iwaniec
- Department of Nutrition and Exercise Sciences, Oregon State University, Corvallis, OR 97331
- Corresponding author: Urszula T. Iwaniec, Ph.D., Department of Nutrition and Exercise Sciences, 108 Milam Hall, Oregon State University, Corvallis, OR 97331, Tel: 541-737-9925, Fax: 541-737-6914,
| |
Collapse
|
27
|
Kalra SP. Central leptin gene therapy ameliorates diabetes type 1 and 2 through two independent hypothalamic relays; a benefit beyond weight and appetite regulation. Peptides 2009; 30:1957-63. [PMID: 19647774 PMCID: PMC2755606 DOI: 10.1016/j.peptides.2009.07.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Revised: 07/22/2009] [Accepted: 07/23/2009] [Indexed: 01/12/2023]
Abstract
Although its role in energy homeostasis is firmly established, the evidence accumulated over a decade linking the adipocyte leptin-hypothalamus axis in the pathogenesis of diabetes mellitus has received little attention in the contemporary thinking. In this context various lines of evidence are collated here to show that (1) under the direction of leptin two independent relays emanating from the hypothalamus restrain insulin secretion from the pancreas and mobilize peripheral organs--liver, skeletal muscle and brown adipose tissue--to upregulate glucose disposal, and (2), leptin insufficiency in the hypothalamus produced by either leptinopenia or restriction of leptin transport across the blood brain barrier due to hyperleptinemia of obesity and aging, initiate antecedent pathophysiological sequalae of diabetes type 1 and 2. Further, we document here the efficacy of leptin replenishment in vivo, especially by supplying it to the hypothalamus with the aid of gene therapy, in preventing the antecedent pathophysiological sequalae--hyperinsulinemia, insulin resistance and hyperglycemia--in various animal models and clinical paradigms of diabetes type 1 and 2 with or without attendant obesity. Overall, the new insights on the long-lasting antidiabetic potential of two independent hypothalamic relays engendered by central leptin gene therapy and the preclinical safety indicators in rodents warrant further validation in subhuman primates and humans.
Collapse
Affiliation(s)
- Satya P Kalra
- Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, PO Box 100244, Gainesville, FL 32610-0244, United States.
| |
Collapse
|
28
|
Kojima S, Asakawa A, Amitani H, Sakoguchi T, Ueno N, Inui A, Kalra SP. Central leptin gene therapy, a substitute for insulin therapy to ameliorate hyperglycemia and hyperphagia, and promote survival in insulin-deficient diabetic mice. Peptides 2009; 30:962-6. [PMID: 19428774 DOI: 10.1016/j.peptides.2009.01.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2008] [Revised: 01/08/2009] [Accepted: 01/09/2009] [Indexed: 12/22/2022]
Abstract
Long-term benefits of central leptin gene therapy in insulin-deficient diabetes are not known despite its therapeutic effects in obesity animal models such as ob/ob and diet-induced obese mice. Adult male mice were injected intraperitoneally with streptozotocin (STZ, 200mg/kg) to induce insulitis. A week later, only diabetic STZ-pretreated mice (blood glucose >350 mg/dl) received intracerebroventricularly (icv) an injection of recombinant adeno-associated virus vector (rAAV) encoding either green fluorescent protein (control), or leptin gene (rAAV-lep). Body weight (BW), food intake, blood glucose, insulin and survival rate responses were monitored post-icv injection at regular intervals for 52 weeks. The STZ pre-injected diabetic mice remained hyperphagic, gradually lost BW and died by week 6 after receiving control vector. In marked contrast, injection of rAAV-lep to raise hypothalamic leptin levels, rescued the STZ-pretreated mice from early mortality, gradually curbed hyperphagia to normalize intake by week 20, and maintained BW at significantly lower than the control range. Blood glucose levels in these mice started to recede dramatically by week 2-3 to normalize by week 8, and euglycemia was sustained during the remaining course of the experiment. rAAV-lep injected mice did not exhibit any discernible untoward gross behavioral changes and diabetic complications and showed a partial return of pancreatic beta-cell function. These results show for the first time that one time central leptin gene therapy is effective and durable in reinstating euglycemia and energy homeostasis for extended periods in the absence of insulin.
Collapse
Affiliation(s)
- Shinya Kojima
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | | | | | | | | | | | | |
Collapse
|
29
|
Kalra SP, Dube MG, Iwaniec UT. Leptin increases osteoblast-specific osteocalcin release through a hypothalamic relay. Peptides 2009; 30:967-73. [PMID: 19428775 PMCID: PMC2749976 DOI: 10.1016/j.peptides.2009.01.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Revised: 01/21/2009] [Accepted: 01/23/2009] [Indexed: 12/31/2022]
Abstract
Enhanced long-term expression of leptin by gene therapy selectively in the hypothalamus, without leakage to the systemic circulation, abrogated skeletal abnormalities and reinstated weight and insulin-glucose homeostasis in leptin-deficient ob/ob mice. Whether increases in osteocalcin, a hormone produced by osteoblasts and known to play a role in bone growth and recently in glucose-insulin homeostasis, may link these benefits of central leptin was assessed. The effects of a single intraventricular injection of non-immunogenic, non-pathogenic recombinant adeno-associated virus vector encoding leptin gene (rAAV-lep) or green fluorescent protein gene (rAAV-GFP, control) were studied in three genotypes, wild type (wt), obese diabetic, hyperinsulinemic ob/ob and non-obese, diabetic insulinopenic Akita mice. Selective hypothalamic leptin expression with central rAAV-lep treatment decreased weight, fat mass, food intake, suppressed insulin levels in ob/ob and wt mice, and conferred euglycemia by suppressing blood glucose in all three genotypes. Contemporaneously, rAAV-lep treatment also augmented blood osteocalcin levels. In wt mice, osteocalcin rose by 51% and, whereas, basal osteocalcin levels in ob/ob and Akita mice were significantly lower as compared to those in wt mice (26% and 55%, respectively), gene therapy reinstated levels to the control range in ob/ob mice, and raised 40% above the wt range even in the absence of insulin in Akita mice. These findings demonstrate that the central beneficial effects of leptin on bone growth involve increased hypothalamic relay of signals that augment osteocalcin efflux from osteoblasts into the general circulation, a response that, in turn, may also modulate glucose-insulin and weight homeostasis.
Collapse
Affiliation(s)
- Satya P Kalra
- Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610-0244, United States.
| | | | | |
Collapse
|
30
|
Iwaniec UT, Dube MG, Boghossian S, Song H, Helferich WG, Turner RT, Kalra SP. Body mass influences cortical bone mass independent of leptin signaling. Bone 2009; 44:404-12. [PMID: 19095090 PMCID: PMC3522417 DOI: 10.1016/j.bone.2008.10.058] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2008] [Revised: 10/01/2008] [Accepted: 10/31/2008] [Indexed: 11/19/2022]
Abstract
Obesity in humans is associated with increased bone mass. Leptin, a hormone produced by fat cells, functions as a sentinel of energy balance, and may mediate the putative positive effects of body mass on bone. We performed studies in male C57Bl/6 wild type (WT) and leptin-deficient ob/ob mice to determine whether body mass gain induced by high fat intake increases bone mass and, if so, whether this requires central leptin signaling. The relationship between body mass and bone mass and architecture was evaluated in 9-week-old and 24-week-old WT mice fed a regular mouse diet. Femora and lumbar vertebrae were analyzed by micro computed tomography. In subsequent studies, slowly and rapidly growing ob/ob mice were injected in the hypothalamus with a recombinant adeno-associated virus containing the leptin gene (rAAV-lep) or a control vector, rAAV-GFP (green fluorescent protein). The mice were maintained on a regular control diet for 5 or 7 weeks and then subdivided into groups and either continued on the control diet or fed a high fat diet (45% of kcal from fat) for 8 weeks. In the WT mice, femoral and vertebral bone mass was positively correlated with body mass (Pearson's r=0.65-0.88 depending on endpoint). rAAV-lep therapy dramatically decreased body mass (-61%) but increased femur length. However, in the distal femur and lumbar vertebra, rAAV-lep therapy reduced cancellous bone volume/tissue volume, trabecular number and trabecular thickness, and increased trabecular spacing. The high fat diet increased body mass, irrespective of vector treatment. Total femur bone volume, length, cross-sectional volume, and cortical volume and thickness were increased in mice with increased body mass, independent of rAAV treatment. In the distal femur, increased body mass had no effect on cancellous architecture and there were no vector x body mass interactions. In WT mice, increased body mass resulted in increased (+33%) vertebral cancellous bone volume/tissue volume. Increased body mass had minimal independent effect on cancellous vertebral bone mass in ob/ob mice. Taken together, these findings suggest that increased body mass has a positive effect on femur cortical bone mass that is independent of leptin signaling.
Collapse
Affiliation(s)
- U T Iwaniec
- Department of Nutrition and Exercise Sciences, Oregon State University, Corvallis, OR 97331, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Landersdorfer CB, DuBois DC, Almon RR, Jusko WJ. Mechanism-based modeling of nutritional and leptin influences on growth in normal and type 2 diabetic rats. J Pharmacol Exp Ther 2009; 328:644-51. [PMID: 18971370 DOI: 10.1124/jpet.108.144766] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
Influences of genetic and nutritional factors on body weight, fat mass, and leptin production and effects of leptin were assessed in normal [Wistar-Kyoto (WKY)] and diabetic [Goto-Kakizaki (GK)] rats by mechanism-based modeling. The study included 60 WKY and 60 GK rats; half received high-fat diet (HF), and the others received normal rat chow (N). Body weights and food consumption were measured twice weekly. Six rats per group were sacrificed at 4, 8, 12, 16, and 20 weeks. Abdominal fat was weighed, and plasma leptin was measured by enzyme-linked immunosorbent assay. All data were comodeled using NONMEM version VI level 1.1 (first-order conditional estimation with interaction) (Beal SL, Boeckmann AJ, Sheiner LB, and NONMEM Project Group, NONMEM Users Guides, University of California, San Francisco, CA, 2007). Weight gain was modeled as differences between energy intake and metabolic rate based on allometrically scaled lean body mass (LBM). The GK had higher metabolic rates (1.15 kcal/day/g LBM(0.75)) than WKY-N (0.92) and WKY-HF (1.02) rats and higher efficiency in transforming energy into body weight. Leptin effect was modeled as inhibition of food consumption. Total body fat was estimated from abdominal fat. Leptin production from fat was 4.7-fold higher for GK (3.03 ng/ml/day/g) than WKY (0.66 ng/ml/day/g). Leptin production rate from LBM was 0.53 ng/ml/day/g for all groups. The IC(50) for inhibition of food intake by leptin was approximately 3-fold higher in GK versus WKY, indicating leptin resistance for the effect on food consumption in GK. The GK had similar intake of kilocalories but lower body weights and fat mass than WKY, possibly because of higher metabolic rates. Our mechanism-based model explains intrinsic reasons for differences in growth, food intake, and leptin concentrations among these two strains of rats.
Collapse
Affiliation(s)
- Cornelia B Landersdorfer
- Department of Pharmaceutical Sciences, State University of New York, Buffalo, New York 14260, USA
| | | | | | | |
Collapse
|
32
|
Kalra SP. Disruption in the leptin-NPY link underlies the pandemic of diabetes and metabolic syndrome: new therapeutic approaches. Nutrition 2009; 24:820-6. [PMID: 18725078 DOI: 10.1016/j.nut.2008.06.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2008] [Accepted: 06/11/2008] [Indexed: 12/11/2022]
Abstract
Multidisciplinary research from my and my colleagues' laboratory has shown that disruption at various levels of leptin signaling to the interactive hypothalamic network of neuropeptide Y (NPY) and cohorts contributes to the antecedent pathophysiologic sequelae of the disease cluster of the metabolic syndrome. Disruptions in NPY signaling due to high or low abundance of NPY and cognate receptors dysregulate the homeostatic milieu to promote hyperinsulinemia, hyperglycemia, fat accrual, and overt diabetes. Hyperleptinemia induced by consumption of energy-rich diets inhibits leptin transport across the blood-brain barrier and thereby produces leptin insufficiency in the hypothalamus. Sustained leptin insufficiency results in loss of hypothalamic restraint on pancreatic insulin secretion and diminished glucose metabolism and energy expenditure. This chain of events culminates in hyperinsulinemia, hyperglycemia, and diabetes. Our recent studies have shown that increasing the supply of leptin centrally by gene therapy reinstates the restraint on hypothalamic NPY signaling and ameliorates diabetes and the attendant disease cluster of the metabolic syndrome. Thus, newer therapies that would enhance leptin transport across the blood-brain barrier in a timely manner or reinstate leptin restraint on NPY signaling through central leptin gene therapy or pharmacologically with leptin mimetics are likely to curtail the pathophysiologic sequelae of diabetes and related ailments of the metabolic syndrome.
Collapse
Affiliation(s)
- Satya P Kalra
- Department of Neuroscience, University of Florida, McKnight Brain Institute, Gainesville, Florida, USA.
| |
Collapse
|
33
|
Malhotra A, Vashistha H, Yadav VS, Dube MG, Kalra SP, Abdellatif M, Meggs LG. Inhibition of p66ShcA redox activity in cardiac muscle cells attenuates hyperglycemia-induced oxidative stress and apoptosis. Am J Physiol Heart Circ Physiol 2008; 296:H380-8. [PMID: 19060130 DOI: 10.1152/ajpheart.00225.2008] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Apoptotic myocyte cell death, diastolic dysfunction, and progressive deterioration in left ventricular pump function characterize the clinical course of diabetic cardiomyopathy. A key question concerns the mechanism(s) by which hyperglycemia (HG) transmits danger signals in cardiac muscle cells. The growth factor adapter protein p66ShcA is a genetic determinant of longevity, which controls mitochondrial metabolism and cellular responses to oxidative stress. Here we demonstrate that interventions which attenuate or prevent HG-induced phosphorylation at critical position 36 Ser residue (phospho-Ser36) inhibit the redox function of p66ShcA and promote the survival phenotype. Adult rat ventricular myocytes obtained by enzymatic dissociation were transduced with mutant-36 p66ShcA (mu-36) dominant-negative expression vector and plated in serum-free media containing 5 or 25 mM glucose. At HG, adult rat ventricular myocytes exhibit a marked increase in reactive oxygen species production, upregulation of phospho-Ser36, collapse of mitochondrial transmembrane potential, and increased formation of p66ShcA/cytochrome-c complexes. These indexes of oxidative stress were accompanied by a 40% increase in apoptosis and the upregulation of cleaved caspase-3 and the apoptosis-related proteins p53 and Bax. To test whether p66ShcA functions as a redox-sensitive molecular switch in vivo, we examined the hearts of male Akita diabetic nonobese (C57BL/6J) mice. Western blot analysis detected the upregulation of phospho-Ser36, the translocation of p66ShcA to mitochondria, and the formation of p66ShcA/cytochrome-c complexes. Conversely, the correction of HG by recombinant adeno-associated viral delivery of leptin reversed these alterations. We conclude that p66ShcA is a molecular switch whose redox function is turned on by phospho-Ser36 and turned off by interventions that prevent this modification.
Collapse
Affiliation(s)
- Ashwani Malhotra
- Division of Nephrology and Hypertension, Department of Medicine, UMDNJ-New Jersey Medical School, Newark, NJ 07103, USA.
| | | | | | | | | | | | | |
Collapse
|
34
|
Dube MG, Torto R, Kalra SP. Increased leptin expression selectively in the hypothalamus suppresses inflammatory markers CRP and IL-6 in leptin-deficient diabetic obese mice. Peptides 2008; 29:593-8. [PMID: 18325632 PMCID: PMC2291149 DOI: 10.1016/j.peptides.2008.01.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2007] [Revised: 01/03/2008] [Accepted: 01/04/2008] [Indexed: 01/30/2023]
Abstract
Low-grade systemic inflammation, as indicated by increased circulating levels of inflammatory markers CRP and IL-6, is linked to increased risks for cardiovascular diseases (CVD) and diabetes mellitus in obese subjects. Whereas hyperleptinemia in obesity are associated with increased CRP and IL-6 release, the hypothalamic versus peripheral site of leptin action has not been ascertained. The effects of increased leptin supply selectively in the hypothalamus by gene therapy on pro-inflammatory CRP and IL-6 levels and on markers of diabetes in the circulation of ob/ob mice displaying either age-related or dietary obesity were assessed. A recombinant adeno-associated viral vector encoding either green-fluorescent protein (control) or leptin gene was injected intracerebroventricularly. Five weeks later, one-half of each of the vector groups was switched to high-fat diet consumption and the other half continued to consume regular low-fat chow diet. Body weight and visceral white adipose tissue were drastically reduced and hyperinsulinemia and hyperglycemia were abrogated by leptin gene therapy, independent of the dietary fat content. The elevated plasma CRP and IL-6 levels seen in obese ob/ob mice receiving the control vector, regardless of the fat content of the diet, were markedly suppressed by increased hypothalamic leptin in both groups. The results show for the first time that leptin deficiency elevates and reinstatement of leptin selectively in the hypothalamus suppresses the release of pro-inflammatory biomarkers, a response likely to alleviate CVD associated with obesity.
Collapse
Affiliation(s)
- Michael G. Dube
- Department of Physiology and Functional Genomics, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL
| | - Rita Torto
- Department of Physiology and Functional Genomics, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL
| | - Satya P. Kalra
- Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL
| |
Collapse
|
35
|
Nogueiras R, Wilson H, Rohner-Jeanrenaud F, Tschöp MH. Central nervous system regulation of adipocyte metabolism. ACTA ACUST UNITED AC 2008; 149:26-31. [PMID: 18453013 DOI: 10.1016/j.regpep.2007.09.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2007] [Accepted: 09/14/2007] [Indexed: 01/23/2023]
Abstract
The white adipose tissue was initially largely known only as an energy storage tissue. It is now well recognized that white adipose tissue is a major endocrine and secretory organ, which releases a wide range of protein signals and factors termed adipokines. The regulation of adipocyte metabolism is an important factor for the understanding of obesity, and some mechanisms are still unknown. Many homeostatic processes, including appetite and food intake, are controlled by neuroendocrine circuits involving the central nervous system. There is substantial evidence demonstrating that the central nervous system also directly regulates adipocyte metabolism. In this review, we discuss the central actions of some peptides with an important role in energy balance regulation on adipocyte metabolism and the physiological relevance of these actions.
Collapse
Affiliation(s)
- Rubén Nogueiras
- Department of Psychiatry, Obesity Research Centre, University of Cincinnati, Cincinnati, OH 45226, USA.
| | | | | | | |
Collapse
|
36
|
Kiuru M, Crystal RG. Progress and prospects: gene therapy for performance and appearance enhancement. Gene Ther 2008; 15:329-37. [DOI: 10.1038/sj.gt.3303100] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
37
|
van den Hoek AM, Teusink B, Voshol PJ, Havekes LM, Romijn JA, Pijl H. Leptin deficiency per se dictates body composition and insulin action in ob/ob mice. J Neuroendocrinol 2008; 20:120-7. [PMID: 18081560 DOI: 10.1111/j.1365-2826.2007.01626.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Obese humans are often insulin- and leptin resistant. Since leptin can affect glucose metabolism, it is conceivable that a lack of leptin signal transduction contributes to insulin resistance. It remains unclear whether leptin affects glucose metabolism via peripheral and/or central mechanistic routes. In the present study, we aimed: (i) to determine the relative contributions of lack of leptin signal transduction and adiposity to insulin resistance and (ii) to establish the impact of central leptin action on glucose metabolism. To address the first point, ob/ob mice were subjected to severe calorie restriction, so that their body weight became similar to that of wild-type mice. Insulin sensitivity was measured in obese ob/ob, lean (food restricted) ob/ob and lean, weight-matched wild-type mice. To address the second point, leptin (or vehicle) was i.c.v. infused to the lateral cerebral ventricle of ob/ob mice and insulin sensitivity was determined. Hyperinsulinaemic euglyceamic clamps were used to quantify insulin sensitivity. Food restriction barely affected body composition, although it profoundly curtailed body weight. Insulin suppressed hepatic glucose production (HGP) to a greater extent in lean ob/ob than in obese ob/ob mice, but its impact remained considerably less than in wild-type mice (% suppression: 11.8 +/- 8.9 versus 1.3 +/- 1.1 versus 56.6 +/- 13.0%/nmol, for lean, obese ob/ob and wild-type mice, respectively; P < 0.05). The insulin-mediated glucose disposal (GD) of lean ob/ob mice was also in between that of obese ob/ob and wild-type mice (37.5 +/- 21.4 versus 25.1 +/- 14.6 versus 59.6 +/- 17.3 mumol/min/kg/nmol of insulin, respectively; P < 0.05 wild-type versus obese ob/ob mice). Leptin infusion acutely enhanced both hepatic insulin sensitivity (insulin-induced inhibition of HGP) and insulin-mediated GD (9.1 +/- 2.4 versus 5.0 +/- 2.7%/nmol of insulin, and 25.6 +/- 5.6 versus 13.6 +/- 4.8 mumol/min/kg/nmol of insulin, respectively; P < 0.05 for both comparisons) in ob/ob mice. Both a lack of leptin signals and adiposity may contribute to insulin resistance in obese individuals. Diminution of central leptin signalling can critically affect glucose metabolism in these individuals.
Collapse
Affiliation(s)
- A M van den Hoek
- TNO-Quality of Life, Gaubius Laboratory, Leiden, The Netherlands.
| | | | | | | | | | | |
Collapse
|
38
|
Central leptin insufficiency syndrome: an interactive etiology for obesity, metabolic and neural diseases and for designing new therapeutic interventions. Peptides 2007; 29:127-38. [PMID: 18053615 DOI: 10.1016/j.peptides.2007.10.017] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Revised: 10/17/2007] [Accepted: 10/18/2007] [Indexed: 01/10/2023]
Abstract
This review critically reappraises recent scientific evidence concerning central leptin insufficiency versus leptin resistance formulations to explain metabolic and neural disorders resulting from subnormal or defective leptin signaling in various sites in the brain. Research at various fronts to unravel the complexities of the neurobiology of leptin is surveyed to provide a comprehensive account of the neural and metabolic effects of environmentally imposed fluctuations in leptin availability at brain sites and the outcome of newer technology to restore leptin signaling in a site-specific manner. The cumulative new knowledge favors a unified central leptin insufficiency syndrome over the, in vogue, central resistance hypothesis to explain the global adverse impact of deficient leptin signaling in the brain. Furthermore, the leptin insufficiency syndrome delineates a novel role of leptin in the hypothalamus in restraining rhythmic pancreatic insulin secretion while concomitantly enhancing glucose metabolism and non-shivering thermogenic energy expenditure, sequelae that would otherwise promote fat accrual to store excess energy resulting from consumption of energy-enriched diets. A concerted effort should now focus on development of newer technologies for delivery of leptin or leptin mimetics to specifically target neural pathways for remediation of diverse ailments encompassing the central leptin insufficiency syndrome.
Collapse
|
39
|
Abstract
A identificação da leptina, hormônio secretado pelos adipócitos, cujo efeito sobre o sistema nervoso simpático e a função endócrina confere participação ativa no controle do dispêndio energético, bem como do apetite, acrescentou às funções do tecido adiposo no organismo humano o papel de órgão multifuncional, produtor e secretor de inúmeros peptídeos e proteínas bioativas, denominadas adipocitocinas. Alterações na quantidade de tecido adiposo, como ocorrem na obesidade, afetam a produção da maioria desses fatores secretados pelos adipócitos. Ainda que essas alterações estejam freqüentemente associadas às inúmeras disfunções metabólicas e ao aumento do risco de doenças cardiovasculares, permanece sob investigação o envolvimento do tecido adiposo no desenvolvimento dessas complicações, considerada a sua função endócrina. As concentrações de várias adipocitocinas elevam-se na obesidade e têm sido relacionadas à hipertensão (angiotensinogênio), ao prejuízo da fibrinólise (inibidor do ativador de plasminogênio-1) e à resistência à insulina (proteína estimuladora de acilação, fator de necrose tumoral-alfa, interleucina-6 e resistina). De outro modo, leptina e adiponectina têm efeitos sobre a sensibilidade à insulina. Na obesidade, a resistência insulínica também está relacionada à resistência à leptina e aos teores plasmáticos reduzidos de adiponectina. Leptina e adiponectina ainda exercem efeitos orgânicos adicionais distintos: frente à participação da leptina no controle da ingestão alimentar, a adiponectina apresenta potente ação anti-aterogênica. Algumas drogas utilizadas no controle do diabetes elevam a produção endógena de adiponectina, em roedores e humanos, indicando que o desenvolvimento de novos medicamentos com alvo nas adipocitocinas pode representar uma alternativa terapêutica de prevenção da resistência insulínica e da aterosclerose em indivíduos obesos.
Collapse
|
40
|
Iwaniec UT, Boghossian S, Lapke PD, Turner RT, Kalra SP. Central leptin gene therapy corrects skeletal abnormalities in leptin-deficient ob/ob mice. Peptides 2007; 28:1012-9. [PMID: 17346852 PMCID: PMC1986832 DOI: 10.1016/j.peptides.2007.02.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2006] [Revised: 01/29/2007] [Accepted: 02/01/2007] [Indexed: 10/23/2022]
Abstract
Skeletal growth is tightly coupled to energy balance via complex and incompletely understood mechanisms. Leptin-deficient ob/ob mice are obese and develop multiple pathologies associated with the metabolic syndrome. Additionally, ob/ob mice have skeletal abnormalities. The objective of this study was to evaluate the effects of leptin deficiency and long duration selective central leptin repletion via recombinant adeno-associated virus-leptin (rAAV-lep) gene therapy on bone in growing ob/ob mice. The ob/ob mice were injected in the hypothalamus with either rAAV-lep or rAAV-GFP (control vector). Treated ob/ob and untreated wild-type (WT) mice were then maintained on a normal diet for 15 weeks. In a second experiment, similarly treated mice along with a group of pair-fed mice were maintained for 30 weeks. Leptin was not detected in blood of either rAAV-lep- or rAAV-GFP-treated mice although rAAV-lep-treated mice displayed leptin transgene expression in the hypothalamus. As expected, rAAV-lep normalized body weight and food intake. Compared to WT mice, rAAV-GFP-treated ob/ob mice had decreased femoral length (by 1.6 mm or 10%, P<0.001), decreased total femur bone volume (by 3.3 mm(3) or 19%, P<0.001), but increased cancellous bone volume in the distal femur (by 0.04 mm(3) or 60%, P<0.09) and lumbar vertebrae (by 0.26 mm(3) or 118%, P<0.001). Treatment with rAAV-lep rescued the ob/ob skeletal phenotype by increasing femoral length and total bone volume, and decreasing femoral and vertebral cancellous bone volume, so that at 15 weeks post-rAAV-lep injection the ob/ob mice no longer differed from WT mice. No further skeletal changes in either the femur or lumbar vertebra were observed at 30 weeks post-rAAV-lep administration. The results suggest that hypothalamic leptin functions as an essential permissive factor for normal bone growth.
Collapse
Affiliation(s)
- Urszula T Iwaniec
- Department of Nutrition and Exercise Sciences, Oregon State University, Corvallis, OR 97331, USA.
| | | | | | | | | |
Collapse
|
41
|
Kalra SP, Kalra PS. To subjugate NPY is to improve the quality of life and live longer. Peptides 2007; 28:413-8. [PMID: 17215061 PMCID: PMC1839846 DOI: 10.1016/j.peptides.2006.08.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2006] [Accepted: 08/09/2006] [Indexed: 10/23/2022]
Abstract
The interactive network of neuropeptide Y (NPY) and cohorts is necessary for integrating the hypothalamic regulation of appetite and energy expenditure with the endocrine and neuroendocrine systems on a daily basis. Genetic and environmental factors that produce an insufficiency of leptin restraint on NPY and cognate receptors deregulate the homeostasis to engender various life-threatening risk factors. Recent studies from our laboratory show that neurotherapy consisting of a single central administration of recombinant adeno-associated virus vector encoding the leptin gene can repress the hypothalamic NPY system for the lifetime of rodents. A major benefit of this stable tonic restraint is deceleration of pathophysiologic sequalae that shorten life span. These include suppression of weight gain, fat accumulation, circulating adipokines, amelioration of major symptoms of metabolic syndrome, improved reproduction and bone health. Thus, sustained repression of NPY signaling in the hypothalamus by leptin transgene expression can improve the quality of life and extend longevity.
Collapse
Affiliation(s)
- Satya P Kalra
- Department of Neuroscience, McKnight Brain Institute, University of Florida, PO Box 100244, Gainesville, FL 32610-0244, USA.
| | | |
Collapse
|
42
|
Bibliography. Current world literature. Growth and development. Curr Opin Endocrinol Diabetes Obes 2007; 14:74-89. [PMID: 17940424 DOI: 10.1097/med.0b013e32802e6d87] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
43
|
Toyoshima M, Asakawa A, Fujimiya M, Inoue K, Inoue S, Kinboshi M, Koizumi A. Dimorphic gene expression patterns of anorexigenic and orexigenic peptides in hypothalamus account male and female hyperphagia in Akita type 1 diabetic mice. Biochem Biophys Res Commun 2007; 352:703-8. [PMID: 17141181 DOI: 10.1016/j.bbrc.2006.11.076] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2006] [Accepted: 11/14/2006] [Indexed: 12/28/2022]
Abstract
Progression of the diabetic trait is usually more rapid in males than females. The major aim of the present study is to explore sexual dimorphism in the hypothalamus in Akita mice. Akita male mice develop hyperphagia, reducing anorexigenic proopiomelanocortin (POMC), and increasing orexigenic neuropeptide Y (NPY) mRNA levels compared with wt males. Serum leptin level was suppressed in Akita males, though castration improved these levels leading to reductions of hyperphagia and blood glucose levels. While Akita female mice also developed hyperphagia, there was no difference in POMC, NPY and leptin levels between Akita and wt females. Anorexigenic Cocaine- and amphetamine-regulated transcript (CART) and corticotrophin-releasing factor (CRF) mRNA levels in Akita females were decreased against wt females. Gonadectomy did not have any effect on the regulation of these neuropeptides. These data suggested that there is sexual dimorphism of neuronal regulation in hyperphagia in Akita mice.
Collapse
Affiliation(s)
- Megumi Toyoshima
- Department of Health and Environmental Sciences, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | | | | | | | | | | | | |
Collapse
|
44
|
Boghossian S, Dube MG, Torto R, Kalra PS, Kalra SP. Hypothalamic clamp on insulin release by leptin-transgene expression. Peptides 2006; 27:3245-54. [PMID: 16962683 DOI: 10.1016/j.peptides.2006.07.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2006] [Revised: 07/24/2006] [Accepted: 07/26/2006] [Indexed: 12/15/2022]
Abstract
The effects of sustained leptin action locally in the hypothalamus on the functional link between fat accrual and insulin secretion after chronic high fat diet (HFD) consumption in leptin-deficient ob/ob mice, and on the post-prandial insulin response in rats consuming regular chow diet (RCD), was examined in this study. A single intracerebroventricular (icv) injection of recombinant adeno-associated virus vector encoding leptin gene (rAAV-lep) enhanced hypothalamic leptin-transgene expression in ob/ob mice consuming RCD and suppressed the time-related weight gain and fat accumulation concomitant with abrogation of hyperinsulinemia and enhanced glucose tolerance. This increased hypothalamic leptin-transgene expression continued to impose insulinopenia and increased glucose tolerance but was ineffective in suppressing weight gain and fat accumulation after these mice were switched to chronic HFD consumption. A similar icv rAAV-lep pretreatment in rats consuming RCD markedly attenuated the post-prandial rise in insulin release concomitant with suppressed weight and fat depots. These results show for the first time that a sustained hypothalamic leptin action can stably clamp pancreatic insulin secretion independent of the status of fat accrual engendered by diets of varying caloric enrichment. Thus, the efficacy of increased leptin afferent signaling in the hypothalamus to persistently restrain pancreatic insulin release and insulin resistance can be explored as an adjunct therapeutic modality to alleviate pathophysiological derrangements that confer type 2 diabetes.
Collapse
Affiliation(s)
- Stéphane Boghossian
- Department of Neuroscience, University of Florida, McKnight Brain Institute, PO Box 100244, Gainesville, FL 32610-0244, USA
| | | | | | | | | |
Collapse
|
45
|
Boghossian S, Ueno N, Dube MG, Kalra P, Kalra S. Leptin gene transfer in the hypothalamus enhances longevity in adult monogenic mutant mice in the absence of circulating leptin. Neurobiol Aging 2006; 28:1594-604. [PMID: 17011078 DOI: 10.1016/j.neurobiolaging.2006.08.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2006] [Revised: 08/17/2006] [Accepted: 08/24/2006] [Indexed: 12/18/2022]
Abstract
Leptin, a product of the ob gene, is a pleiotropic signal implicated in regulation of multiple physiological functions in the periphery and centrally, including hypothalamic integration of energy homeostasis. Recessive mutations of ob gene result in early onset of hyperphagia, morbid obesity, metabolic disorders, early mortality and shortened life-span. Intracerebroventricular injection of recombinant adeno-associated virus vector (rAAV) encoding the leptin gene in adult obese ob/ob mice enhanced leptin transgene expression only in the hypothalamus, normalized food intake, body weight and more than doubled the life-span as compared to control cohorts and extended it to near that of normal wild type mice. These life-extending benefits were associated with drastic reductions in visceral fat, and blood glucose and insulin levels, but elevated ghrelin levels, the anti-aging biomarkers. Thus, bioavailability of leptin transduced by ectopic gene in the hypothalamus alone is both necessary and sufficient to normalize life-span. Evidently, site-specific ectopic gene expression with rAAV is durable and safe for alleviating neural disorders that stem from missing or functional disruption of a single gene.
Collapse
Affiliation(s)
- Stéphane Boghossian
- Department of Neuroscience, University of Florida McKnight Brain Institute, Gainesville, FL, USA
| | | | | | | | | |
Collapse
|