1
|
Zor E, Alpaydin S, Bingol H. Quantitative determination of leptin hormone using gold nanoparticle-based lateral flow assay. Mikrochim Acta 2025; 192:63. [PMID: 39789177 PMCID: PMC11717784 DOI: 10.1007/s00604-024-06945-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
A lateral flow assay (LFA) has been developed that can be used in point-of-care (PoC) use for the sensitive determination of leptin hormone. The limit of detection value was 0.158 ng/mL and the limit of quantification value was 0.479 ng/mL for leptin hormone. The stability studies showed that the assay response was equal to the initial value even after 6 months. LFA accuracy tests performed in commercial artificial serum samples were compared with the enzyme-linked immunosorbent assay (ELISA). The obtained recovery values between 95 and 110% show that the developed LFA can be used for quantitative determination of leptin hormone. As the result of the study, a fast and disposable assay was developed for the determination of leptin hormone, which can be performed with a small amount of sample in less than 15 min without the need for long-term analysis stages, qualified personnel, expensive equipment and devices.
Collapse
Affiliation(s)
- Erhan Zor
- Science and Technology Research and Application Center (BITAM), Necmettin Erbakan University, Konya, Türkiye.
- Department of Science Education, A.K. Education Faculty, Necmettin Erbakan University, Konya, Türkiye.
| | - Sabri Alpaydin
- Department of Chemistry Education, A.K. Education Faculty, Necmettin Erbakan University, Konya, Türkiye
| | - Haluk Bingol
- Science and Technology Research and Application Center (BITAM), Necmettin Erbakan University, Konya, Türkiye
- Department of Basic Sciences, Faculty of Engineering, Necmettin Erbakan University, Konya, Türkiye
| |
Collapse
|
2
|
Lee DH, Song J. Impaired olfactory system in metabolic imbalance-related neuropathology. Life Sci 2024; 355:122967. [PMID: 39142504 DOI: 10.1016/j.lfs.2024.122967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/25/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
Olfactory dysfunction, influenced by factors such as aging and environmental stress, is linked to various neurological disorders. The olfactory bulb's connections to brain areas like the hypothalamus, piriform cortex, entorhinal cortex, and limbic system make olfactory dysfunction a contributor to a range of neuropathological conditions. Recent research has underscored that olfactory deficits are prevalent in individuals with both metabolic syndrome and dementia. These systemic metabolic alterations correlate with olfactory impairments, potentially affecting brain regions associated with the olfactory bulb. In cases of metabolic syndrome, phenomena such as insulin resistance and disrupted glucose metabolism may result in compromised olfactory function, leading to multiple neurological issues. This review synthesizes key findings on the interplay between metabolic-induced olfactory dysfunction and neuropathology. It emphasizes the critical role of olfactory assessment in diagnosing and managing neurological diseases related to metabolic syndrome.
Collapse
Affiliation(s)
- Dong Hoon Lee
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School & Hwasun Hospital, Hwasun 58128, Republic of Korea.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Republic of Korea.
| |
Collapse
|
3
|
Iyer DR, Venkatraman J, Tanguy E, Vitale N, Mahapatra NR. Chromogranin A and its derived peptides: potential regulators of cholesterol homeostasis. Cell Mol Life Sci 2023; 80:271. [PMID: 37642733 PMCID: PMC11072126 DOI: 10.1007/s00018-023-04908-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/31/2023]
Abstract
Chromogranin A (CHGA), a member of the granin family of proteins, has been an attractive therapeutic target and candidate biomarker for several cardiovascular, neurological, and inflammatory disorders. The prominence of CHGA stems from the pleiotropic roles of several bioactive peptides (e.g., catestatin, pancreastatin, vasostatins) generated by its proteolytic cleavage and by their wide anatomical distribution. These peptides are emerging as novel modulators of cardiometabolic diseases that are often linked to high blood cholesterol levels. However, their impact on cholesterol homeostasis is poorly understood. The dynamic nature of cholesterol and its multitudinous roles in almost every aspect of normal body function makes it an integral component of metabolic physiology. A tightly regulated coordination of cholesterol homeostasis is imperative for proper functioning of cellular and metabolic processes. The deregulation of cholesterol levels can result in several pathophysiological states. Although studies till date suggest regulatory roles for CHGA and its derived peptides on cholesterol levels, the mechanisms by which this is achieved still remain unclear. This review aims to aggregate and consolidate the available evidence linking CHGA with cholesterol homeostasis in health and disease. In addition, we also look at common molecular regulatory factors (viz., transcription factors and microRNAs) which could govern the expression of CHGA and genes involved in cholesterol homeostasis under basal and pathological conditions. In order to gain further insights into the pathways mediating cholesterol regulation by CHGA/its derived peptides, a few prospective signaling pathways are explored, which could act as primers for future studies.
Collapse
Affiliation(s)
- Dhanya R Iyer
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Janani Venkatraman
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Emeline Tanguy
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212 and Université de Strasbourg, 5 Rue Blaise Pascal, 67000, Strasbourg, France
| | - Nicolas Vitale
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212 and Université de Strasbourg, 5 Rue Blaise Pascal, 67000, Strasbourg, France.
| | - Nitish R Mahapatra
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India.
| |
Collapse
|
4
|
Effects of Long-Term Low-Protein Diets Supplemented with Sodium Dichloroacetate and Glucose on Metabolic Biomarkers and Intestinal Microbiota of Finishing Pigs. Animals (Basel) 2022; 12:ani12192522. [PMID: 36230260 PMCID: PMC9558518 DOI: 10.3390/ani12192522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/09/2022] [Accepted: 09/19/2022] [Indexed: 11/25/2022] Open
Abstract
The objective of this study was to evaluate the effects of low-protein (LP) diets supplemented with sodium dichloroacetate (DCA) and glucose (GLUC) on metabolic markers and intestinal microbiota of finishing pigs. A total of 80 crossbred growing barrows were allocated randomly to one of the five treatments, including the normal protein level diet (CON), the LP diets, LP with 120 mg/kg DCA (LP + DCA) or 1.8% glucose (LP + GLUC), and LP with 120 mg/kg DCA and 1.8% glucose (LP + DCA + GLUC). The LP diet increased the plasma HDL, triglyceride, and cholesterol concentrations and reduced the bile acid, urea nitrogen, albumin, and total protein concentrations compared to the CON diet (p < 0.05). The LP + DCA + GLUC diet reduced the plasma VLDL, triglyceride, and cholesterol concentrations and increased the bile acid concentration compared with the LP diet (p < 0.05). Pigs fed the LP + DCA and LP + GLUC diets showed reduced 3-Hydroxy-3-Methylglutaryl-CoA Reductase content and increased Cytochrome P450 Family 7 Subfamily A Member 1 activity of liver compared that of the CON diet (p < 0.05). Moreover, the LP diets with or without DCA and GLUC supplementation increased the relative abundance of colonic microbiota related to carbohydrate fermentation in finishing pigs. In conclusion, 120 mg/kg DCA or 1.8% GLUC supplementation in an LP diet modulated the hepatic lipid metabolism of pigs, while the DCA along with GLUC supplementation likely improved the lipid metabolism by stimulating bile acid secretion.
Collapse
|
5
|
The Roles and Associated Mechanisms of Adipokines in Development of Metabolic Syndrome. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27020334. [PMID: 35056647 PMCID: PMC8781412 DOI: 10.3390/molecules27020334] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/30/2021] [Accepted: 01/03/2022] [Indexed: 12/12/2022]
Abstract
Metabolic syndrome is a cluster of metabolic indicators that increase the risk of diabetes and cardiovascular diseases. Visceral obesity and factors derived from altered adipose tissue, adipokines, play critical roles in the development of metabolic syndrome. Although the adipokines leptin and adiponectin improve insulin sensitivity, others contribute to the development of glucose intolerance, including visfatin, fetuin-A, resistin, and plasminogen activator inhibitor-1 (PAI-1). Leptin and adiponectin increase fatty acid oxidation, prevent foam cell formation, and improve lipid metabolism, while visfatin, fetuin-A, PAI-1, and resistin have pro-atherogenic properties. In this review, we briefly summarize the role of various adipokines in the development of metabolic syndrome, focusing on glucose homeostasis and lipid metabolism.
Collapse
|
6
|
Akinci G, Celik M, Akinci B. Complications of lipodystrophy syndromes. Presse Med 2021; 50:104085. [PMID: 34728268 DOI: 10.1016/j.lpm.2021.104085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 10/09/2021] [Accepted: 10/20/2021] [Indexed: 10/19/2022] Open
Abstract
Lipodystrophy syndromes are rare complex multisystem disorders caused by generalized or partial lack of adipose tissue. Adipose tissue dysfunction in lipodystrophy is associated with leptin deficiency. Lipodystrophy leads to severe metabolic problems. These abnormalities include, but are not limited to, insulin-resistant diabetes, severe hypertriglyceridemia, and lipid accumulation in ectopic organs such as the liver, and are associated with end-organ complications. Metabolic abnormalities can be present at the time of diagnosis or may develop over time as the disease progresses. In addition to metabolic abnormalities, subtype-specific presentations due to underlying molecular etiology in genetic forms and autoimmunity in acquired forms contribute to severe morbidity in lipodystrophy.
Collapse
Affiliation(s)
- Gulcin Akinci
- Division of Pediatric Neurology, Dr. Behcet Uz Children's Hospital, Izmir, Turkey
| | - Merve Celik
- Division of Endocrinology and Metabolism, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Baris Akinci
- Division of Endocrinology and Metabolism, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey.
| |
Collapse
|
7
|
O'Brien CJO, Haberman ER, Domingos AI. A Tale of Three Systems: Toward a Neuroimmunoendocrine Model of Obesity. Annu Rev Cell Dev Biol 2021; 37:549-573. [PMID: 34613819 PMCID: PMC7614880 DOI: 10.1146/annurev-cellbio-120319-114106] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The prevalence of obesity is on the rise. What was once considered a simple disease of energy imbalance is now recognized as a complex condition perpetuated by neuro- and immunopathologies. In this review, we summarize the current knowledge of the neuroimmunoendocrine mechanisms underlying obesity. We examine the pleiotropic effects of leptin action in addition to its established role in the modulation of appetite, and we discuss the neural circuitry mediating leptin action and how this is altered with obesity, both centrally (leptin resistance) and in adipose tissues (sympathetic neuropathy). Finally, we dissect the numerous causal and consequential roles of adipose tissue macrophages in obesity and highlight recent key studies demonstrating their direct role in organismal energy homeostasis.
Collapse
Affiliation(s)
- Conan J O O'Brien
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom;
| | - Emma R Haberman
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom;
| | - Ana I Domingos
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom;
| |
Collapse
|
8
|
Sevrin T, Sirvins C, David A, Aguesse A, Gandon A, Castellano B, Darmaun D, Boquien CY, Alexandre-Gouabau MC. Dietary Arginine Supplementation during Gestation and Lactation Increases Milk Yield and Mammary Lipogenesis in Rats. J Nutr 2021; 151:2188-2198. [PMID: 34091672 DOI: 10.1093/jn/nxab152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/29/2021] [Accepted: 04/26/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Arginine, an essential amino acid during the reproductive period, has been shown to enhance lactation performances in livestock. Whether it could help mothers with breastfeeding difficulties is not known. OBJECTIVES This study aimed to determine whether dietary arginine supplementation would enhance milk production in rat dams nursing large 12-pup litters and, if so, what mechanisms are involved. METHODS In 3 series of experiments, differing in dam killing timing, 59 primiparous, pregnant Sprague-Dawley rats (mean ± SD weight: 254 ± 24.7 g) were randomly assigned to receive either 1) an AIN-93G diet supplemented with l-arginine at 2.0% (ARG diet), through lactation and gestation (AGL group); 2) a control AIN-93G diet including at 3.5% an isonitrogenous mix of amino acids that are not essential for lactation (MA diet), during gestation and lactation (MA group); or 3) the MA diet during gestation and the ARG diet during lactation (AL group). Milk flow was measured using deuterated water enrichment between days 11 and 18. Plasma hormones and mammary expression of genes involved in lactation were measured using ELISA and qRT-PCR, respectively, at lactation days 12, 18, or 21 in the 3 experiments. Data were analyzed by ANOVA. RESULTS Dam food intake, pup weight gain, milk flow normalized to dam weight, and milk fat concentration were 17%, 9%, 20%, and 20% greater in the AGL group than in the MA group, respectively (P < 0.05). Genes involved in lipogenesis and lipid regulation were overexpressed ≤2.76-fold in the mammary gland of AGL dams compared with MA dams (P < 0.05) and plasma leptin concentration was 39% higher (P = 0.008). Milk flow and composition and mammary gene expression of the AL group did not differ from those of the MA group, whereas milk fat concentration and flow were 26% and 37% lower than in the AGL group, respectively. CONCLUSIONS Arginine supplementation during gestation and lactation enhances milk flow and mammary lipogenesis in rats nursing large litters.
Collapse
Affiliation(s)
- Thomas Sevrin
- Laboratoire FRANCE Bébé Nutrition, Laval, France
- UMR 1280-Pathophysiology of Nutritional Adaptations (PhAN), Nantes University, INRAE, IMAD, CRNH-Ouest, Nantes, France
| | - Charlène Sirvins
- UMR 1280-Pathophysiology of Nutritional Adaptations (PhAN), Nantes University, INRAE, IMAD, CRNH-Ouest, Nantes, France
| | - Agnès David
- UMR 1280-Pathophysiology of Nutritional Adaptations (PhAN), Nantes University, INRAE, IMAD, CRNH-Ouest, Nantes, France
| | - Audrey Aguesse
- UMR 1280-Pathophysiology of Nutritional Adaptations (PhAN), Nantes University, INRAE, IMAD, CRNH-Ouest, Nantes, France
| | - Alexis Gandon
- UMR 1280-Pathophysiology of Nutritional Adaptations (PhAN), Nantes University, INRAE, IMAD, CRNH-Ouest, Nantes, France
| | - Blandine Castellano
- UMR 1280-Pathophysiology of Nutritional Adaptations (PhAN), Nantes University, INRAE, IMAD, CRNH-Ouest, Nantes, France
| | - Dominique Darmaun
- UMR 1280-Pathophysiology of Nutritional Adaptations (PhAN), Nantes University, INRAE, IMAD, CRNH-Ouest, Nantes, France
- University Hospital of Nantes, Nantes, France
| | - Clair-Yves Boquien
- UMR 1280-Pathophysiology of Nutritional Adaptations (PhAN), Nantes University, INRAE, IMAD, CRNH-Ouest, Nantes, France
| | | |
Collapse
|
9
|
Abstract
Severe insulin resistance syndromes are a heterogeneous group of rare disorders characterized by profound insulin resistance, substantial metabolic abnormalities, and a variety of clinical manifestations and complications. The etiology of these syndromes may be hereditary or acquired, due to defects in insulin potency and action, cellular responsiveness to insulin, and/or aberrations in adipose tissue function or development. Over the past decades, advances in medical technology, particularly in genomic technologies and genetic analyses, have provided insights into the underlying pathophysiological pathways and facilitated the more precise identification of several of these conditions. However, the exact cellular and molecular mechanisms of insulin resistance have not yet been fully elucidated for all syndromes. Moreover, in clinical practice, many of the syndromes are often misdiagnosed or underdiagnosed. The majority of these disorders associate with an increased risk of severe complications and mortality; thus, early identification and personalized clinical management are of the essence. This Review aims to categorize severe insulin resistance syndromes by disease process, including insulin receptor defects, signaling defects, and lipodystrophies. We also highlight several complex syndromes and emphasize the need to identify patients, investigate underlying disease mechanisms, and develop specific treatment regimens.
Collapse
Affiliation(s)
- Angeliki M. Angelidi
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Andreas Filippaios
- Department of Medicine, Lowell General Hospital, Lowell, Massachusetts, USA
| | - Christos S. Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Section of Endocrinology, Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Pedicelli S, de Palma L, Pelosini C, Cappa M. Metreleptin for the treatment of progressive encephalopathy with/without lipodystrophy (PELD) in a child with progressive myoclonic epilepsy: a case report. Ital J Pediatr 2020; 46:158. [PMID: 33099310 PMCID: PMC7585287 DOI: 10.1186/s13052-020-00916-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 10/05/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND A number of genetic syndromes associated with variants in the BSCL2/seipin gene have been identified. Variants that cause skipping of exon 7 are associated with progressive encephalopathy with/without lipodystrophy (PELD), which is characterized by the development of progressive myoclonic epilepsy at a young age, severe progressive neurological impairment, and early death, often in childhood. Because the genetic basis of PELD is similar to that of congenital lipodystrophy type 2, we hypothesized that a patient with PELD may respond to treatments approved for other congenital lipodystrophic syndromes. CASE PRESENTATION We describe a 5-year-old boy with an extremely rare phenotype involving severe progressive myoclonic epilepsy who received metreleptin (a recombinant analogue of leptin) to control metabolic abnormalities. At the age of two, he had no subcutaneous adipose tissue, with hypertriglyceridemia, hypertransaminasemia and hepatic steatosis. He also had a moderate psychomotor delay and generalized tonic seizures. At 4 years, he had insulin resistance, hypercholesterolemia, hypertriglyceridemia, mild hepatosplenomegaly and mild hepatic steatosis; he began a hypolipidemic diet. Severe psychomotor delay and myoclonic/myoclonic atonic seizures with absences was evident. At 5 years of age, metreleptin 0.06 mg/kg/day was initiated; after 2 months, the patient's lipid profile improved and insulin resistance resolved. After 1 year of treatment, hepatic steatosis improved and abdominal ultrasound showed only mild hepatomegaly. Seizure frequency decreased but was not eliminated during metreleptin therapy. CONCLUSIONS Metreleptin may be used to control metabolic disturbances and may lead to better seizure control in children with PELD.
Collapse
Affiliation(s)
| | - Luca de Palma
- Rare and Complex Epilepsy, Department of Neuroscience, Bambino Gesù Children Hospital, Rome, Italy
| | - Caterina Pelosini
- Chemistry and Endocrinology Laboratory, University Hospital of Pisa, Pisa, Italy
| | - Marco Cappa
- Unit of Endocrinology, Bambino Gesù Children's Hospital, Rome, Italy
| |
Collapse
|
11
|
Female mice exposed to low doses of dioxin during pregnancy and lactation have increased susceptibility to diet-induced obesity and diabetes. Mol Metab 2020; 42:101104. [PMID: 33075544 PMCID: PMC7683344 DOI: 10.1016/j.molmet.2020.101104] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/05/2020] [Accepted: 10/13/2020] [Indexed: 12/30/2022] Open
Abstract
Objective Exposure to persistent organic pollutants is consistently associated with increased diabetes risk in humans. We investigated the short- and long-term impact of transient low-dose dioxin (2,3,7,8-tetrachlorodibenzo-p-dioxin, TCDD) exposure during pregnancy and lactation on glucose homeostasis and beta cell function in female mice, including their response to a metabolic stressor later in life. Methods Female mice were injected with either corn oil (CO; vehicle control) or 20 ng/kg/d TCDD 2x/week throughout mating, pregnancy and lactation, and then tracked for 6–10 weeks after chemical exposure stopped. A subset of CO- and TCDD-exposed dams was then transferred to a 45% high-fat diet (HFD) or remained on a standard chow diet for an additional 11 weeks to assess the long-term effects of TCDD on adaptability to a metabolic stressor. To summarize, female mice were transiently exposed to TCDD and then subsequently tracked beyond when TCDD had been excreted to identify lasting metabolic effects of TCDD exposure. Results TCDD-exposed dams were hypoglycemic at birth but otherwise had normal glucose homeostasis during and post-TCDD exposure. However, TCDD-exposed dams on a chow diet were modestly heavier than controls starting 5 weeks after the last TCDD injection, and their weight gain accelerated after transitioning to a HFD. TCDD-exposed dams also had an accelerated onset of hyperglycemia, impaired glucose-induced plasma insulin levels, reduced islet size, increased MAFA-ve beta cells, and increased proinsulin accumulation following HFD feeding compared to controls. Overall, our study demonstrates that low-dose TCDD exposure during pregnancy has minimal effects on metabolism during the period of active exposure, but has detrimental long-term effects on metabolic adaptability to HFD feeding. Conclusions Our study suggests that transient low-dose TCDD exposure in female mice impairs metabolic adaptability to HFD feeding, demonstrating that dioxin exposure may be a contributing factor to obesity and diabetes pathogenesis in females. Female mice exposed to TCDD during pregnancy are hypoglycemic at birth. TCDD exposure promotes weight gain long after exposure ceases. TCDD-exposed dams fed a high-fat diet have accelerated onset of glucose intolerance. TCDDHFD dams have defects in islet morphology and beta cell function.
Collapse
|
12
|
Lawler K, Huang-Doran I, Sonoyama T, Collet TH, Keogh JM, Henning E, O’Rahilly S, Bottolo L, Farooqi IS. Leptin-Mediated Changes in the Human Metabolome. J Clin Endocrinol Metab 2020; 105:dgaa251. [PMID: 32392278 PMCID: PMC7282709 DOI: 10.1210/clinem/dgaa251] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/06/2020] [Indexed: 02/08/2023]
Abstract
CONTEXT While severe obesity due to congenital leptin deficiency is rare, studies in patients before and after treatment with leptin can provide unique insights into the role that leptin plays in metabolic and endocrine function. OBJECTIVE The aim of this study was to characterize changes in peripheral metabolism in people with congenital leptin deficiency undergoing leptin replacement therapy, and to investigate the extent to which these changes are explained by reduced caloric intake. DESIGN Ultrahigh performance liquid chromatography-tandem mass spectroscopy (UPLC-MS/MS) was used to measure 661 metabolites in 6 severely obese people with congenital leptin deficiency before, and within 1 month after, treatment with recombinant leptin. Data were analyzed using unsupervised and hypothesis-driven computational approaches and compared with data from a study of acute caloric restriction in healthy volunteers. RESULTS Leptin replacement was associated with class-wide increased levels of fatty acids and acylcarnitines and decreased phospholipids, consistent with enhanced lipolysis and fatty acid oxidation. Primary and secondary bile acids increased after leptin treatment. Comparable changes were observed after acute caloric restriction. Branched-chain amino acids and steroid metabolites decreased after leptin, but not after acute caloric restriction. Individuals with severe obesity due to leptin deficiency and other genetic obesity syndromes shared a metabolomic signature associated with increased BMI. CONCLUSION Leptin replacement was associated with changes in lipolysis and substrate utilization that were consistent with negative energy balance. However, leptin's effects on branched-chain amino acids and steroid metabolites were independent of reduced caloric intake and require further exploration.
Collapse
Affiliation(s)
- Katherine Lawler
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - Isabel Huang-Doran
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - Takuhiro Sonoyama
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - Tinh-Hai Collet
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
- Service of Endocrinology, Diabetes and Metabolism, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Julia M Keogh
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - Elana Henning
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - Stephen O’Rahilly
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - Leonardo Bottolo
- University Department of Medical Genetics, Addenbrooke’s Hospital, Cambridge, UK
- The Alan Turing Institute, London, UK
- MRC Biostatistics Unit, University of Cambridge, Robinson Way, Cambridge, UK
| | - I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| |
Collapse
|
13
|
DHA reduces hypothalamic inflammation and improves central leptin signaling in mice. Life Sci 2020; 257:118036. [PMID: 32622949 DOI: 10.1016/j.lfs.2020.118036] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/17/2020] [Accepted: 06/29/2020] [Indexed: 01/09/2023]
Abstract
AIMS Anti-obesity effects and improved leptin sensitivity from n-3 polyunsaturated fatty acids (n-3 PUFAs) have been reported in diet-induced obese animals. This study sought to determine the beneficial central effects and mechanism of docosahexaenoic acid (DHA, 22:6 n-3) in high-fat (HF) diet fed mice. MAIN METHODS Male C57BL/6J mice were given HF diet with or without intracerebroventricular (icv) injection of docosahexaenoic acid (DHA, 22:6 n-3) for two days. Central leptin sensitivity, hypothalamic inflammation, leptin signaling molecules and tyrosine hydroxylase (TH) were examined by central leptin sensitivity test and Western blot. Furthermore, the expression of hepatic genes involved in lipid metabolism was examined by RT-PCR. KEY FINDINGS We found that icv administration of DHA not only reduced energy intake and body weight gain but also corrected the HF diet-induced hypothalamic inflammation. DHA decreased leptin signaling inhibitor SOCS3 and improved the leptin JAK2-Akt signaling pathways in the hypothalamus. Furthermore, icv administration of DHA improved the effects of leptin in the regulation of mRNA expression of enzymes related to lipogenesis, fatty acid β-oxidation, and cholesterol synthesis in the liver. DHA increased leptin-induced activation of TH in the hypothalamus. SIGNIFICANCE Therefore, increasing central DHA concentration may prevent the deficit of hypothalamic regulation, which is associated with disorders of energy homeostasis in the liver as a result of a high-fat diet.
Collapse
|
14
|
Shahraki MR, Badini F, Shahraki E, Shahraki AR, Dashipour A. Effects of Capparis decidua Hydroalcoholic Extracts on Blood Glucose, Lipid Profile and Leptin of Wistar Male Rats with High Cholesterol Diets. NUTRITION AND FOOD SCIENCES RESEARCH 2020. [DOI: 10.29252/nfsr.7.1.25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
15
|
Kinzer AB, Shamburek RD, Lightbourne M, Muniyappa R, Brown RJ. Advanced Lipoprotein Analysis Shows Atherogenic Lipid Profile That Improves After Metreleptin in Patients with Lipodystrophy. J Endocr Soc 2019; 3:1503-1517. [PMID: 31620670 DOI: 10.1210/js.2019-00103] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/04/2019] [Indexed: 01/12/2023] Open
Abstract
Context Patients with lipodystrophy have dyslipidemia and insulin resistance. Leptin treatment with metreleptin in lipodystrophy decreases insulin resistance and lowers triglycerides without changing high-density lipoprotein. Detailed measurement of lipoprotein particles with nuclear magnetic resonance (NMR) spectroscopy can offer insights into cardiovascular disease (CVD) risk and lipid metabolism beyond a standard lipid panel. We hypothesized that patients with lipodystrophy would have a more atherogenic lipid profile than controls at baseline, which would be ameliorated with metreleptin treatment. Objective To characterize the lipoprotein profile in patients with lipodystrophy compared with controls and to evaluate effects of metreleptin treatment. Design Setting Patients and Intervention Patients with lipodystrophy (N = 17) were studied before and after metreleptin for 2 weeks and 6 months and compared with 51 insulin-sensitive sex-matched controls. Main Outcome Measures Lipoprotein profiles were measured by NMR with the LP4 deconvolution algorithm, which reports triglyceride-rich lipoprotein particles (TRLPs), high-density lipoprotein particles (HDLPs), and low-density lipoprotein particles (LDLPs). Results Patients with lipodystrophy had elevated large TRLPs and smaller HDLPs and LDLPs compared with controls. Five patients with lipodystrophy had chylomicrons, compared with zero controls. Metreleptin decreased the size and concentration of TRLPs, eliminated chylomicrons in all but one patient, decreased LDLPs, and increased LDLP size. Metreleptin treatment did not have major effects on HDLPs. Conclusions Patients with lipodystrophy had an atherogenic lipoprotein profile at baseline consistent with elevated CVD risk, which improved after metreleptin treatment. The presence of fasting chylomicrons in a subset of patients with lipodystrophy suggests saturation of chylomicron clearance by lipoprotein lipase.
Collapse
Affiliation(s)
- Alexandra B Kinzer
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | | | - Marissa Lightbourne
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland
| | - Ranganath Muniyappa
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Rebecca J Brown
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| |
Collapse
|
16
|
Lausten-Thomsen U, Lund MAV, Frithioff-Bøjsøe C, Hedley PL, Pedersen O, Hansen T, Christiansen M, Holm JC. Reference values for leptin/adiponectin ratio in healthy children and adolescents. Clin Chim Acta 2019; 493:123-128. [DOI: 10.1016/j.cca.2019.03.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 03/05/2019] [Indexed: 12/17/2022]
|
17
|
Brown RJ, Valencia A, Startzell M, Cochran E, Walter PJ, Garraffo HM, Cai H, Gharib AM, Ouwerkerk R, Courville AB, Bernstein S, Brychta RJ, Chen KY, Walter M, Auh S, Gorden P. Metreleptin-mediated improvements in insulin sensitivity are independent of food intake in humans with lipodystrophy. J Clin Invest 2018; 128:3504-3516. [PMID: 29723161 DOI: 10.1172/jci95476] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 05/01/2018] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Recombinant leptin (metreleptin) ameliorates hyperphagia and metabolic abnormalities in leptin-deficient humans with lipodystrophy. We aimed to determine whether metreleptin improves glucose and lipid metabolism in humans when food intake is held constant. METHODS Patients with lipodystrophy were hospitalized for 19 days, with food intake held constant by a controlled diet in an inpatient metabolic ward. In a nonrandomized, crossover design, patients previously treated with metreleptin (n = 8) were continued on metreleptin for 5 days and then taken off metreleptin for the next 14 days (withdrawal cohort). This order was reversed in metreleptin-naive patients (n = 14), who were reevaluated after 6 months of metreleptin treatment on an ad libitum diet (initiation cohort). Outcome measurements included insulin sensitivity by hyperinsulinemic-euglycemic clamp, fasting glucose and triglyceride levels, lipolysis measured using isotopic tracers, and liver fat by magnetic resonance spectroscopy. RESULTS With food intake constant, peripheral insulin sensitivity decreased by 41% after stopping metreleptin for 14 days (withdrawal cohort) and increased by 32% after treatment with metreleptin for 14 days (initiation cohort). In the initiation cohort only, metreleptin decreased fasting glucose by 11% and triglycerides by 41% and increased hepatic insulin sensitivity. Liver fat decreased from 21.8% to 18.7%. In the initiation cohort, changes in lipolysis were not independent of food intake, but after 6 months of metreleptin treatment on an ad libitum diet, lipolysis decreased by 30% (palmitate turnover) to 35% (glycerol turnover). CONCLUSION Using lipodystrophy as a human model of leptin deficiency and replacement, we show that metreleptin improves insulin sensitivity and decreases hepatic and circulating triglycerides and that these improvements are independent of its effects on food intake. TRIAL REGISTRATION ClinicalTrials.gov NCT01778556FUNDING. This research was supported by the intramural research program of the NIDDK.
Collapse
Affiliation(s)
- Rebecca J Brown
- Diabetes, Endocrinology, and Obesity Branch (DEOB), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Areli Valencia
- Diabetes, Endocrinology, and Obesity Branch (DEOB), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Megan Startzell
- Diabetes, Endocrinology, and Obesity Branch (DEOB), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Elaine Cochran
- Diabetes, Endocrinology, and Obesity Branch (DEOB), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | | | | | | | - Ahmed M Gharib
- Biomedical and Metabolic Imaging Branch, NIDDK, NIH, Bethesda, Maryland, USA
| | - Ronald Ouwerkerk
- Biomedical and Metabolic Imaging Branch, NIDDK, NIH, Bethesda, Maryland, USA
| | | | - Shanna Bernstein
- Nutrition Department, Clinical Center, NIH, Bethesda, Maryland, USA
| | - Robert J Brychta
- Diabetes, Endocrinology, and Obesity Branch (DEOB), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Kong Y Chen
- Diabetes, Endocrinology, and Obesity Branch (DEOB), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | | | - Sungyoung Auh
- Office of the Clinical Director, NIDDK, NIH, Bethesda, Maryland, USA
| | - Phillip Gorden
- Diabetes, Endocrinology, and Obesity Branch (DEOB), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| |
Collapse
|
18
|
Ochiai M, Misaki K, Takeuchi T, Narumi R, Azuma Y, Matsuo T. Egg White Hydrolysate Can Be a Low-Allergenic Food Material to Suppress Ectopic Fat Accumulation in Rats Fed an Equicaloric Diet. J Nutr Sci Vitaminol (Tokyo) 2018; 63:111-119. [PMID: 28552875 DOI: 10.3177/jnsv.63.111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Egg white (EW) is known as a nutritional protein but can induce allergic reactions in humans. We investigated the dietary effects of EW and its hydrolysate (EWH), which contains less allergen, on body fat accumulation in Wistar rats fed an equicaloric high-fat and high-sucrose diet for 8 wk (Exp A). The pair-feeding of EW and equicaloric-feeding of EWH increased fecal fat excretion and suppressed lipid accumulation in the liver and muscles but not in the abdominal adipose tissues, carcass, or total body. Dietary EWH also suppressed the serum glucose level and alkaline phosphatase activity. Further, we showed a higher dispersibility of EW and EWH in physicochemical assay (Exp B). Next, we investigated the suppressive effects of a single administration of EW and EWH on lipid-induced hypertriglyceridemia and small intestinal meal transit in ddY mice (Exp C). However, a single administration of EW or EWH did not suppress the lipid-induced hypertriglyceridemia nor did it delay the rate of small intestinal transit. These findings indicated that dietary EW and EWH reduce hepatic and muscular (ectopic) fat accumulation mainly by suppressing fat absorption and supplying fat to the liver and muscles. Therefore, the low-allergenic EWH can be effective for the prevention of high-fat-diet-induced obesity.
Collapse
Affiliation(s)
- Masaru Ochiai
- School of Veterinary Medicine, Kitasato University.,Faculty of Agriculture, Kagawa University
| | | | | | - Ryoyo Narumi
- School of Veterinary Medicine, Kitasato University
| | | | | |
Collapse
|
19
|
Vatier C, Arnaud L, Prieur X, Guyomarch B, Le May C, Bigot E, Pichelin M, Daguenel A, Vantyghem MC, Gautier JF, Vigouroux C, Cariou B. One-year metreleptin therapy decreases PCSK9 serum levels in diabetic patients with monogenic lipodystrophy syndromes. DIABETES & METABOLISM 2017; 43:275-279. [DOI: 10.1016/j.diabet.2016.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 08/25/2016] [Accepted: 08/29/2016] [Indexed: 12/23/2022]
|
20
|
Effects of clozapine on adipokine secretions/productions and lipid droplets in 3T3-L1 adipocytes. J Pharmacol Sci 2017; 133:79-87. [DOI: 10.1016/j.jphs.2017.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/29/2016] [Accepted: 01/13/2017] [Indexed: 12/14/2022] Open
|
21
|
Chelminski Y, Magnan C, Luquet SH, Everard A, Meunier N, Gurden H, Martin C. Odor-Induced Neuronal Rhythms in the Olfactory Bulb Are Profoundly Modified in ob/ob Obese Mice. Front Physiol 2017; 8:2. [PMID: 28154537 PMCID: PMC5244437 DOI: 10.3389/fphys.2017.00002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/03/2017] [Indexed: 01/03/2023] Open
Abstract
Leptin, the product of the Ob(Lep) gene, is a peptide hormone that plays a major role in maintaining the balance between food intake and energy expenditure. In the brain, leptin receptors are expressed by hypothalamic cells but also in the olfactory bulb, the first central structure coding for odors, suggesting a precise function of this hormone in odor-evoked activities. Although olfaction plays a key role in feeding behavior, the ability of the olfactory bulb to integrate the energy-related signal leptin is still missing. Therefore, we studied the fate of odor-induced activity in the olfactory bulb in the genetic context of leptin deficiency using the obese ob/ob mice. By means of an odor discrimination task with concomitant local field potential recordings, we showed that ob/ob mice perform better than wild-type (WT) mice in the early stage of the task. This behavioral gain of function was associated in parallel with profound changes in neuronal oscillations in the olfactory bulb. The distribution of the peaks in the gamma frequency range was shifted toward higher frequencies in ob/ob mice compared to WT mice before learning. More notably, beta oscillatory activity, which has been shown previously to be correlated with olfactory discrimination learning, was longer and stronger in expert ob/ob mice after learning. Since oscillations in the olfactory bulb emerge from mitral to granule cell interactions, our results suggest that cellular dynamics in the olfactory bulb are deeply modified in ob/ob mice in the context of olfactory learning.
Collapse
Affiliation(s)
- Yan Chelminski
- UMR 8165 Centre National de la Recherche Scientifique, IMNC, Paris Sud University, Paris Diderot University Orsay, France
| | - Christophe Magnan
- UMR 8251 Centre National de la Recherche Scientifique, BFA, Paris Diderot University, Sorbonne Paris Cité University Paris, France
| | - Serge H Luquet
- UMR 8251 Centre National de la Recherche Scientifique, BFA, Paris Diderot University, Sorbonne Paris Cité University Paris, France
| | - Amandine Everard
- UMR 8251 Centre National de la Recherche Scientifique, BFA, Paris Diderot University, Sorbonne Paris Cité University Paris, France
| | - Nicolas Meunier
- INRA, UR1197 NeuroBiologie de l'OlfactionJouy-en-Josas, France; Université de Versailles St-Quentin en YvelinesVersailles, France
| | - Hirac Gurden
- UMR 8165 Centre National de la Recherche Scientifique, IMNC, Paris Sud University, Paris Diderot UniversityOrsay, France; UMR 8251 Centre National de la Recherche Scientifique, BFA, Paris Diderot University, Sorbonne Paris Cité UniversityParis, France
| | - Claire Martin
- UMR 8165 Centre National de la Recherche Scientifique, IMNC, Paris Sud University, Paris Diderot UniversityOrsay, France; UMR 8251 Centre National de la Recherche Scientifique, BFA, Paris Diderot University, Sorbonne Paris Cité UniversityParis, France
| |
Collapse
|
22
|
Lausten-Thomsen U, Christiansen M, Louise Hedley P, Esmann Fonvig C, Stjernholm T, Pedersen O, Hansen T, Holm JC. Reference values for serum leptin in healthy non-obese children and adolescents. Scandinavian Journal of Clinical and Laboratory Investigation 2016; 76:561-567. [DOI: 10.1080/00365513.2016.1210226] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Ulrik Lausten-Thomsen
- Department of Pediatrics, The Children’s Obesity Clinic, Copenhagen University Hospital Holbæk, Holbæk, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Michael Christiansen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Paula Louise Hedley
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Cilius Esmann Fonvig
- Department of Pediatrics, The Children’s Obesity Clinic, Copenhagen University Hospital Holbæk, Holbæk, Denmark
- The Faculty of Health and Medical Sciences, Section of Metabolic Genetics, The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Theresa Stjernholm
- Department of Pediatrics, The Children’s Obesity Clinic, Copenhagen University Hospital Holbæk, Holbæk, Denmark
| | - Oluf Pedersen
- The Faculty of Health and Medical Sciences, Section of Metabolic Genetics, The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- The Faculty of Health and Medical Sciences, Section of Metabolic Genetics, The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Jens-Christian Holm
- Department of Pediatrics, The Children’s Obesity Clinic, Copenhagen University Hospital Holbæk, Holbæk, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- The Faculty of Health and Medical Sciences, Section of Metabolic Genetics, The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Ehrhardt RA, Foskolos A, Giesy SL, Wesolowski SR, Krumm CS, Butler WR, Quirk SM, Waldron MR, Boisclair YR. Increased plasma leptin attenuates adaptive metabolism in early lactating dairy cows. J Endocrinol 2016; 229:145-57. [PMID: 26957637 DOI: 10.1530/joe-16-0031] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 03/03/2016] [Indexed: 12/31/2022]
Abstract
Mammals meet the increased nutritional demands of lactation through a combination of increased feed intake and a collection of adaptations known as adaptive metabolism (e.g., glucose sparing via insulin resistance, mobilization of endogenous reserves, and increased metabolic efficiency via reduced thyroid hormones). In the modern dairy cow, adaptive metabolism predominates over increased feed intake at the onset of lactation and develops concurrently with a reduction in plasma leptin. To address the role of leptin in the adaptive metabolism of early lactation, we asked which adaptations could be countered by a constant 96-h intravenous infusion of human leptin (hLeptin) starting on day 8 of lactation. Compared to saline infusion (Control), hLeptin did not alter energy intake or milk energy output but caused a modest increase in body weight loss. hLeptin reduced plasma glucose by 9% and hepatic glycogen content by 73%, and these effects were associated with a 17% increase in glucose disposal during an insulin tolerance test. hLeptin attenuated the accumulation of triglyceride in the liver by 28% in the absence of effects on plasma levels of the anti-lipolytic hormone insulin or plasma levels of free fatty acids, a marker of lipid mobilization from adipose tissue. Finally, hLeptin increased the plasma concentrations of T4 and T3 by nearly 50% without affecting other neurally regulated hormones (i.e., cortisol and luteinizing hormone (LH)). Overall these data implicate the periparturient reduction in plasma leptin as one of the signals promoting conservation of glucose and energy at the onset of lactation in the energy-deficient dairy cow.
Collapse
Affiliation(s)
- Richard A Ehrhardt
- Departments of Animal Science and Large Animal Clinical SciencesMichigan State University, East Lansing, Michigan, USA
| | - Andreas Foskolos
- Institute of Biological, Environmental and Rural SciencesAberystwyth University, Aberystwyth, UK
| | - Sarah L Giesy
- Department of Animal ScienceCornell University, Ithaca, New York, USA
| | | | | | - W Ronald Butler
- Department of Animal ScienceCornell University, Ithaca, New York, USA
| | - Susan M Quirk
- Department of Animal ScienceCornell University, Ithaca, New York, USA
| | - Matthew R Waldron
- Department of Animal ScienceCornell University, Ithaca, New York, USA
| | - Yves R Boisclair
- Department of Animal ScienceCornell University, Ithaca, New York, USA
| |
Collapse
|
24
|
Richards L, Li M, van Esch B, Garssen J, Folkerts G. The effects of short-chain fatty acids on the cardiovascular system. PHARMANUTRITION 2016. [DOI: 10.1016/j.phanu.2016.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
25
|
Seroussi E, Cinnamon Y, Yosefi S, Genin O, Smith JG, Rafati N, Bornelöv S, Andersson L, Friedman-Einat M. Identification of the Long-Sought Leptin in Chicken and Duck: Expression Pattern of the Highly GC-Rich Avian leptin Fits an Autocrine/Paracrine Rather Than Endocrine Function. Endocrinology 2016; 157:737-51. [PMID: 26587783 DOI: 10.1210/en.2015-1634] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
More than 20 years after characterization of the key regulator of mammalian energy balance, leptin, we identified the leptin (LEP) genes of chicken (Gallus gallus) and duck (Anas platyrhynchos). The extreme guanine-cytosine content (∼70%), the location in a genomic region with low-complexity repetitive and palindromic sequence elements, the relatively low sequence conservation, and low level of expression have hampered the identification of these genes until now. In vitro-expressed chicken and duck leptins specifically activated signaling through the chicken leptin receptor in cell culture. In situ hybridization demonstrated expression of LEP mRNA in granular and Purkinje cells of the cerebellum, anterior pituitary, and in embryonic limb buds, somites, and branchial arches, suggesting roles in adult brain control of energy balance and during embryonic development. The expression patterns of LEP and the leptin receptor (LEPR) were explored in chicken, duck, and quail (Coturnix japonica) using RNA-sequencing experiments available in the Short Read Archive and by quantitative RT-PCR. In adipose tissue, LEP and LEPR were scarcely transcribed, and the expression level was not correlated to adiposity. Our identification of the leptin genes in chicken and duck genomes resolves a long lasting controversy regarding the existence of leptin genes in these species. This identification was confirmed by sequence and structural similarity, conserved exon-intron boundaries, detection in numerous genomic, and transcriptomic datasets and characterization by PCR, quantitative RT-PCR, in situ hybridization, and bioassays. Our results point to an autocrine/paracrine mode of action for bird leptin instead of being a circulating hormone as in mammals.
Collapse
Affiliation(s)
- Eyal Seroussi
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| | - Yuval Cinnamon
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| | - Sara Yosefi
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| | - Olga Genin
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| | - Julia Gage Smith
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| | - Nima Rafati
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| | - Susanne Bornelöv
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| | - Leif Andersson
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| | - Miriam Friedman-Einat
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| |
Collapse
|
26
|
Abstract
The adipokine leptin realizes signal transduction via four different leptin receptor (OB-R) isoforms. The amount of functionally active OB-R, however, is affected by constitutive shedding of the extracellular domain. The product of the cleavage process, the so-called soluble leptin receptor (sOB-R), is the main binding protein for leptin in human blood and modulates its bioavailability. Concentrations of sOB-R are differentially regulated in metabolic disorders, such as type 1 diabetes mellitus or obesity, and can, therefore, enhance or reduce leptin sensitivity. Lipotoxicity and apoptosis increase OB-R cleavage via ADAM10-dependent mechanisms. In contrast, although increased sOB-R concentrations seem to directly inhibit leptin effects, reduced amounts of sOB-R may reflect decreased membrane expression of OB-R. These findings, in part, explain alterations of leptin sensitivity that are associated with changes in serum sOB-R concentrations seen in metabolic disorders.
Collapse
Affiliation(s)
- Michael Schaab
- Institute of Laboratory Medicine Clinical Chemistry and Molecular Diagnostics, Leipzig, Germany.
| | - Juergen Kratzsch
- Institute of Laboratory Medicine Clinical Chemistry and Molecular Diagnostics, Leipzig, Germany
| |
Collapse
|
27
|
Cheng L, Yu Y, Zhang Q, Szabo A, Wang H, Huang XF. Arachidonic acid impairs hypothalamic leptin signaling and hepatic energy homeostasis in mice. Mol Cell Endocrinol 2015; 412:12-8. [PMID: 25986657 DOI: 10.1016/j.mce.2015.04.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 04/15/2015] [Accepted: 04/28/2015] [Indexed: 11/30/2022]
Abstract
Epidemiological evidence suggests that the consumption of a diet high in n-6 polyunsaturated fatty acids (PUFA) is associated with the development of leptin resistance and obesity. We aim to examine the central effect of n-6 PUFA, arachidonic acid (ARA) on leptin sensitivity and leptin-regulated hepatic glucose and lipid metabolism. We found that intracerebroventricular injection of ARA (25 nmol/day) for 2.5 days reversed the effect of central leptin on hypothalamic JAK2, pSTAT3, pAkt, and pFOXO1 protein levels, which was concomitant with a pro-inflammatory response in the hypothalamus. ARA also attenuated the effect of central leptin on hepatic glucose and lipid metabolism by reversing the mRNA expression of the genes involved in gluconeogenesis (G6Pase, PEPCK), glucose transportation (GLUT2), lipogenesis (FAS, SCD1), and cholesterol synthesis (HMG-CoA reductase). These results indicate that an increased exposure to central n-6 PUFA induces central cellular leptin resistance with concomitant defective JAK2-STAT3 and PI3K-Akt signaling.
Collapse
Affiliation(s)
- Licai Cheng
- School of Medicine, Illawarra Health and Medical Research Institute, University of Wollongong, NSW 2522, Australia
| | - Yinghua Yu
- School of Medicine, Illawarra Health and Medical Research Institute, University of Wollongong, NSW 2522, Australia; Schizophrenia Research Institute (SRI), 405 Liverpool St, Sydney, NSW 2010, Australia.
| | - Qingsheng Zhang
- School of Medicine, Illawarra Health and Medical Research Institute, University of Wollongong, NSW 2522, Australia; Schizophrenia Research Institute (SRI), 405 Liverpool St, Sydney, NSW 2010, Australia
| | - Alexander Szabo
- School of Medicine, Illawarra Health and Medical Research Institute, University of Wollongong, NSW 2522, Australia; ANSTO Life Sciences, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, NSW 2234, Australia
| | - Hongqin Wang
- School of Medicine, Illawarra Health and Medical Research Institute, University of Wollongong, NSW 2522, Australia
| | - Xu-Feng Huang
- School of Medicine, Illawarra Health and Medical Research Institute, University of Wollongong, NSW 2522, Australia; Schizophrenia Research Institute (SRI), 405 Liverpool St, Sydney, NSW 2010, Australia.
| |
Collapse
|
28
|
Munusamy S, do Carmo JM, Hosler JP, Hall JE. Obesity-induced changes in kidney mitochondria and endoplasmic reticulum in the presence or absence of leptin. Am J Physiol Renal Physiol 2015; 309:F731-43. [PMID: 26290368 DOI: 10.1152/ajprenal.00188.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 08/13/2015] [Indexed: 12/17/2022] Open
Abstract
We investigated obesity-induced changes in kidney lipid accumulation, mitochondrial function, and endoplasmic reticulum (ER) stress in the absence of hypertension, and the potential role of leptin in modulating these changes. We compared two normotensive genetic mouse models of obesity, leptin-deficient ob/ob mice and hyperleptinemic melanocortin-4 receptor-deficient mice (LoxTB MC4R-/-), with their respective lean controls. Compared with controls, ob/ob and LoxTB MC4R-/- mice exhibit significant albuminuria, increased creatinine clearance, and high renal triglyceride content. Renal ATP levels were decreased in both obesity models, and mitochondria isolated from both models showed alterations that would lower mitochondrial ATP production. Mitochondria from hyperleptinemic LoxTB MC4R-/- mice kidneys respired NADH-generating substrates (including palmitate) at lower rates due to an apparent decrease in complex I activity, and these mitochondria showed oxidative damage. Kidney mitochondria of leptin-deficient ob/ob mice showed normal rates of respiration with no evidence of oxidative damage, but electron transfer was partially uncoupled from ATP synthesis. A fourfold induction of C/EBP homologous protein (CHOP) expression indicated induction of ER stress in kidneys of hyperleptinemic LoxTB MC4R-/- mice. In contrast, ER stress was not induced in kidneys of leptin-deficient ob/ob mice. Our findings show that obesity, in the absence of hypertension, is associated with renal dysfunction in mice but not with major renal injury. Alterations to mitochondria that lower cellular ATP levels may be involved in obesity-induced renal injury. The type and severity of mitochondrial and ER dysfunction differs depending upon the presence or absence of leptin.
Collapse
Affiliation(s)
- Shankar Munusamy
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi; Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi; and College of Pharmacy, Qatar University, Doha, Qatar
| | - Jussara M do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi; Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi; and
| | - Jonathan P Hosler
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi
| | - John E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi; Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi; and
| |
Collapse
|
29
|
Rodríguez A, Moreno NR, Balaguer I, Méndez-Giménez L, Becerril S, Catalán V, Gómez-Ambrosi J, Portincasa P, Calamita G, Soveral G, Malagón MM, Frühbeck G. Leptin administration restores the altered adipose and hepatic expression of aquaglyceroporins improving the non-alcoholic fatty liver of ob/ob mice. Sci Rep 2015; 5:12067. [PMID: 26159457 PMCID: PMC4498231 DOI: 10.1038/srep12067] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 06/17/2015] [Indexed: 12/27/2022] Open
Abstract
Glycerol is an important metabolite for the control of lipid accumulation in white adipose tissue (WAT) and liver. We aimed to investigate whether exogenous administration of leptin improves features of non-alcoholic fatty liver disease (NAFLD) in leptin-deficient ob/ob mice via the regulation of AQP3 and AQP7 (glycerol channels mediating glycerol efflux in adipocytes) and AQP9 (aquaglyceroporin facilitating glycerol influx in hepatocytes). Twelve-week-old male wild type and ob/ob mice were divided in three groups as follows: control, leptin-treated (1 mg/kg/d) and pair-fed. Leptin deficiency was associated with obesity and NAFLD exhibiting an AQP3 and AQP7 increase in WAT, without changes in hepatic AQP9. Adipose Aqp3 and hepatic Aqp9 transcripts positively correlated with markers of adiposity and hepatic steatosis. Chronic leptin administration (4-weeks) was associated with improved body weight, whole-body adiposity, and hepatosteatosis of ob/ob mice and to a down-regulation of AQP3, AQP7 in WAT and an up-regulation of hepatic AQP9. Acute leptin stimulation in vitro (4-h) induced the mobilization of aquaglyceroporins towards lipid droplets (AQP3) and the plasma membrane (AQP7) in murine adipocytes. Our results show that leptin restores the coordinated regulation of fat-specific AQP7 and liver-specific AQP9, a step which might prevent lipid overaccumulation in WAT and liver in obesity.
Collapse
Affiliation(s)
- Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029, Madrid, Spain
- Obesity & Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Natalia R. Moreno
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigación Biomédica (IMIBIC)/Reina Sofia University Hospital/University of Córdoba, Córdoba, Spain
| | - Inmaculada Balaguer
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | - Leire Méndez-Giménez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029, Madrid, Spain
- Obesity & Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029, Madrid, Spain
- Obesity & Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029, Madrid, Spain
- Obesity & Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029, Madrid, Spain
- Obesity & Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology University of Bari Medical School, Policlinico Hospital, Bari, Italy
| | - Giuseppe Calamita
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari “Aldo Moro”, Bari, Italy
| | - Graça Soveral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - María M. Malagón
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigación Biomédica (IMIBIC)/Reina Sofia University Hospital/University of Córdoba, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029, Madrid, Spain
- Obesity & Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
30
|
Cheng L, Yu Y, Szabo A, Wu Y, Wang H, Camer D, Huang XF. Palmitic acid induces central leptin resistance and impairs hepatic glucose and lipid metabolism in male mice. J Nutr Biochem 2015; 26:541-8. [PMID: 25724108 DOI: 10.1016/j.jnutbio.2014.12.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/19/2014] [Accepted: 12/08/2014] [Indexed: 12/12/2022]
Abstract
The consumption of diets rich in saturated fat largely contributes to the development of obesity in modern societies. A diet high in saturated fats can induce inflammation and impair leptin signaling in the hypothalamus. However, the role of saturated fatty acids on hypothalamic leptin signaling, and hepatic glucose and lipid metabolism remains largely undiscovered. In this study, we investigated the effects of intracerebroventricular (icv) administration of a saturated fatty acid, palmitic acid (PA, C16:0), on central leptin sensitivity, hypothalamic leptin signaling, inflammatory molecules and hepatic energy metabolism in C57BL/6J male mice. We found that the icv administration of PA led to central leptin resistance, evidenced by the inhibition of central leptin's suppression of food intake. Central leptin resistance was concomitant with impaired hypothalamic leptin signaling (JAK2-STAT3, PKB/Akt-FOXO1) and a pro-inflammatory response (TNF-α, IL1-β, IL-6 and pIκBa) in the mediobasal hypothalamus and paraventricular hypothalamic nuclei. Furthermore, the pre-administration of icv PA blunted the effect of leptin-induced decreases in mRNA expression related to gluconeogenesis (G6Pase and PEPCK), glucose transportation (GLUT2) and lipogenesis (FAS and SCD1) in the liver of mice. Therefore, elevated central PA concentrations can induce pro-inflammatory responses and leptin resistance, which are associated with disorders of energy homeostasis in the liver as a result of diet-induced obesity.
Collapse
Affiliation(s)
- Licai Cheng
- School of Medicine, University of Wollongong and Illawarra Health and Medical Research Institute, NSW 2522, Australia
| | - Yinghua Yu
- School of Medicine, University of Wollongong and Illawarra Health and Medical Research Institute, NSW 2522, Australia; Schizophrenia Research Institute (SRI), 405 Liverpool St, Sydney, NSW 2010, Australia
| | - Alexander Szabo
- School of Medicine, University of Wollongong and Illawarra Health and Medical Research Institute, NSW 2522, Australia; ANSTO Life Sciences, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, NSW 2234, Australia
| | - Yizhen Wu
- School of Medicine, University of Wollongong and Illawarra Health and Medical Research Institute, NSW 2522, Australia
| | - Hongqin Wang
- School of Medicine, University of Wollongong and Illawarra Health and Medical Research Institute, NSW 2522, Australia
| | - Danielle Camer
- School of Medicine, University of Wollongong and Illawarra Health and Medical Research Institute, NSW 2522, Australia
| | - Xu-Feng Huang
- School of Medicine, University of Wollongong and Illawarra Health and Medical Research Institute, NSW 2522, Australia; Schizophrenia Research Institute (SRI), 405 Liverpool St, Sydney, NSW 2010, Australia.
| |
Collapse
|
31
|
Schleicher J, Tokarski C, Marbach E, Matz-Soja M, Zellmer S, Gebhardt R, Schuster S. Zonation of hepatic fatty acid metabolism - The diversity of its regulation and the benefit of modeling. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:641-56. [PMID: 25677822 DOI: 10.1016/j.bbalip.2015.02.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/26/2015] [Accepted: 02/03/2015] [Indexed: 02/07/2023]
Abstract
A pronounced heterogeneity between hepatocytes in subcellular structure and enzyme activities was discovered more than 50years ago and initiated the idea of metabolic zonation. In the last decades zonation patterns of liver metabolism were extensively investigated for carbohydrate, nitrogen and lipid metabolism. The present review focuses on zonation patterns of the latter. We review recent findings regarding the zonation of fatty acid uptake and oxidation, ketogenesis, triglyceride synthesis and secretion, de novo lipogenesis, as well as bile acid and cholesterol metabolism. In doing so, we expose knowledge gaps and discuss contradictory experimental results, for example on the zonation pattern of fatty acid oxidation and de novo lipogenesis. Thus, possible rewarding directions of further research are identified. Furthermore, recent findings about the regulation of metabolic zonation are summarized, especially regarding the role of hormones, nerve innervation, morphogens, gender differences and the influence of the circadian clock. In the last part of the review, a short collection of models considering hepatic lipid metabolism is provided. We conclude that modeling, despite its proven benefit for understanding of hepatic carbohydrate and ammonia metabolisms, has so far been largely disregarded in the study of lipid metabolism; therefore some possible fields of modeling interest are presented.
Collapse
Affiliation(s)
- J Schleicher
- Department of Bioinformatics, University of Jena, Jena, Germany.
| | - C Tokarski
- Department of Bioinformatics, University of Jena, Jena, Germany
| | - E Marbach
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - M Matz-Soja
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - S Zellmer
- Department of Chemicals and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - R Gebhardt
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - S Schuster
- Department of Bioinformatics, University of Jena, Jena, Germany
| |
Collapse
|
32
|
Tsoukas MA, Farr OM, Mantzoros CS. Leptin in congenital and HIV-associated lipodystrophy. Metabolism 2015; 64:47-59. [PMID: 25267014 DOI: 10.1016/j.metabol.2014.07.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/31/2014] [Accepted: 07/31/2014] [Indexed: 02/07/2023]
Abstract
Leptin is a hormone secreted by adipocytes that regulates energy metabolism via peripheral action on glucose synthesis and utilization as well as through central regulation of food intake. Patients with decreased amounts of fat in their adipose tissue (lipoatrophy) will have low leptin levels, and hypoleptinemic states have been associated with a variety of metabolic dysfunctions. Pronounced complications of insulin resistance, dyslipidemia and fatty liver are observed in patients suffering from congenital or acquired generalized lipodystrophy while somewhat less pronounced abnormalities are associated with human immunodeficiency virus (HIV) and the use of highly active antiretroviral therapy, the so-called HIV-associated lipodystrophy. Previous uncontrolled open-label studies have demonstrated that physiological doses of leptin repletion have corrected many of the metabolic derangements observed in subjects with rare fat maldistribution syndromes such as generalized lipodystrophy. In the much more commonly encountered HIV-associated lipodystrophy, leptin replacement has been shown to decrease central fat mass and to improve insulin sensitivity, dyslipidemia, and glucose levels. The United States Food and Drug Administration has recently granted approval for recombinant leptin therapy for congenital and acquired generalized lipodystrophy, however large, well-designed, placebo-controlled studies are needed to assess long-term efficacy, safety and adverse effects of leptin replacement. In this review, we present the role of leptin in the metabolic complications of congenital and acquired lipodystrophy and discuss current and emerging clinical therapeutic uses of leptin in humans with lipodystrophy.
Collapse
Affiliation(s)
- Michael A Tsoukas
- Section of Endocrinology, Boston VA Healthcare system and Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Olivia M Farr
- Section of Endocrinology, Boston VA Healthcare system and Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Christos S Mantzoros
- Section of Endocrinology, Boston VA Healthcare system and Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
33
|
Fang XL, Zhu XT, Chen SF, Zhang ZQ, Zeng QJ, Deng L, Peng JL, Yu JJ, Wang LN, Wang SB, Gao P, Jiang QY, Shu G. Differential gene expression pattern in hypothalamus of chickens during fasting-induced metabolic reprogramming: Functions of glucose and lipid metabolism in the feed intake of chickens. Poult Sci 2014; 93:2841-54. [DOI: 10.3382/ps.2014-04047] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
34
|
Ochiai M, Matsuo T. Effect of egg white and its hydrolysate on stearoyl-CoA desaturase index and fat accumulation in rat tissues. Int J Food Sci Nutr 2014; 65:948-52. [PMID: 25046367 DOI: 10.3109/09637486.2014.937800] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We investigated the dietary effects of egg white (EW) and its hydrolysate (EWH) on fat metabolism in rats. Wistar rats were divided into casein, EW and EWH dietary groups, and fed their respective diet for 8 weeks. Dietary EW and EWH decreased food intake, body weight gain and fat accumulation in the carcass, liver, muscles and adipose tissues, but muscle weight was increased. In addition, dietary EW and EWH decreased stearoyl-CoA desaturase (SCD) indices and glucose-6-phosphate dehydrogenase activity of the liver and gastrocnemius muscle. Dietary EW also increased the fecal excretion of triacylglycerol, cholesterol and total bile acids, and decreased the serum levels of triacylglycerol and leptin. The suppressive effects of dietary EW on food intake and body fat accumulation were weakened by dietary EWH. These findings indicate that EW and EWH, especially EW, are effective in reducing body fat accumulation by regulating hepatic and muscular SCD indices.
Collapse
Affiliation(s)
- Masaru Ochiai
- Faculty of Agriculture, Kagawa University , Kagawa , Japan
| | | |
Collapse
|
35
|
Valleau JC, Sullivan EL. The impact of leptin on perinatal development and psychopathology. J Chem Neuroanat 2014; 61-62:221-32. [PMID: 24862904 DOI: 10.1016/j.jchemneu.2014.05.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 04/18/2014] [Accepted: 05/11/2014] [Indexed: 01/29/2023]
Abstract
Leptin has long been associated with metabolism as it is a critical regulator of both food intake and energy expenditure, but recently, leptin dysregulation has been proposed as a mechanism of psychopathology. This review discusses the evidence supporting a role for leptin in mental health disorders and describes potential mechanisms that may underlie this association. Leptin plays a critical role in pregnancy and in fetal growth and development. Leptin's role and profile during development is examined in available human studies, and the validity of applying studies conducted in animal models to the human population are discussed. Rodents experience a postnatal leptin surge, which does not occur in humans or larger animal models. This suggests that further research using large mammal models, which have a leptin profile across pregnancy and development similar to humans, are of high importance. Maternal obesity and hyperleptinemia correlate with increased leptin levels in the umbilical cord, placenta, and fetus. Leptin levels are thought to impact fetal brain development; likely by activating proinflammatory cytokines that are known to impact many of the neurotransmitter systems that regulate behavior. Leptin is likely involved in behavioral regulation as leptin receptors are widely distributed in the brain, and leptin influences cortisol release, the mesoaccumbens dopamine pathway, serotonin synthesis, and hippocampal synaptic plasticity. In humans, both high and low levels of leptin are reported to be associated with psychopathology. This inconsistency is likely due to differences in the metabolic state of the study populations. Leptin resistance, which occurs in the obese state, may explain how both high and low levels of leptin are associated with psychopathology, as well as the comorbidity of obesity with numerous mental illnesses. Leptin resistance is likely to influence disorders such as depression and anxiety where high leptin levels have been correlated with symptomatology. Schizophrenia is also associated with both low and high leptin levels. However, as anti-psychotics pharmacotherapy induces weight gain, which elevates leptin levels, drug-naïve populations are needed for further studies. Elevated circulating leptin is consistently found in childhood neurodevelopmental disorders including autism spectrum disorders and Rhett disorder. Further, studies on the impact of leptin and leptin resistance on psychopathology and neurodevelopmental disorders are important directions for future research. Studies examining the mechanisms by which exposure to maternal obesity and hyperleptinemia during fetal development impact brain development and behavior are critical for the health of future generations.
Collapse
Affiliation(s)
- Jeanette C Valleau
- Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, 505 NW 185th Ave., Beaverton, OR, USA
| | - Elinor L Sullivan
- Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, 505 NW 185th Ave., Beaverton, OR, USA; Department of Biology, University of Portland, 5000 N Willamette Blvd., Portland, OR, USA.
| |
Collapse
|
36
|
Cornall LM, Mathai ML, Hryciw DH, McAinch AJ. The therapeutic potential of GPR43: a novel role in modulating metabolic health. Cell Mol Life Sci 2013; 70:4759-70. [PMID: 23852543 PMCID: PMC11113592 DOI: 10.1007/s00018-013-1419-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 06/09/2013] [Accepted: 06/28/2013] [Indexed: 02/07/2023]
Abstract
GPR43 is a receptor for short-chain fatty acids. Preliminary data suggest a putative role for GPR43 in regulating systemic health via processes including inflammation, carcinogenesis, gastrointestinal function, and adipogenesis. GPR43 is involved in secretion of gastrointestinal peptides, which regulate appetite and gastrointestinal motility. This suggests GPR43 may have a role in weight control. Moreover, GPR43 regulates plasma lipid profile and inflammatory processes, which further indicates that GPR43 could have the ability to modulate the etiology and pathogenesis of metabolic diseases such as obesity, type 2 diabetes mellitus, and cardiovascular disease. This review summarizes the current evidence regarding the ability of GPR43 to mediate both systemic and tissue specific functions and how GPR43 may be modulated in the treatment of metabolic disease.
Collapse
Affiliation(s)
- Lauren M Cornall
- Biomedical and Lifestyle Diseases Unit, College of Health and Biomedicine, Victoria University, PO Box 14428, Melbourne, VIC, 8001, Australia,
| | | | | | | |
Collapse
|
37
|
Wu T, Tang Q, Yu Z, Gao Z, Hu H, Chen W, Zheng X, Yu T. Inhibitory effects of sweet cherry anthocyanins on the obesity development in C57BL/6 mice. Int J Food Sci Nutr 2013; 65:351-9. [DOI: 10.3109/09637486.2013.854749] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
38
|
Carobbio S, Hagen RM, Lelliott CJ, Slawik M, Medina-Gomez G, Tan CY, Sicard A, Atherton HJ, Barbarroja N, Bjursell M, Bohlooly-Y M, Virtue S, Tuthill A, Lefai E, Laville M, Wu T, Considine RV, Vidal H, Langin D, Oresic M, Tinahones FJ, Fernandez-Real JM, Griffin JL, Sethi JK, López M, Vidal-Puig A. Adaptive changes of the Insig1/SREBP1/SCD1 set point help adipose tissue to cope with increased storage demands of obesity. Diabetes 2013; 62:3697-708. [PMID: 23919961 PMCID: PMC3806615 DOI: 10.2337/db12-1748] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The epidemic of obesity imposes unprecedented challenges on human adipose tissue (WAT) storage capacity that may benefit from adaptive mechanisms to maintain adipocyte functionality. Here, we demonstrate that changes in the regulatory feedback set point control of Insig1/SREBP1 represent an adaptive response that preserves WAT lipid homeostasis in obese and insulin-resistant states. In our experiments, we show that Insig1 mRNA expression decreases in WAT from mice with obesity-associated insulin resistance and from morbidly obese humans and in in vitro models of adipocyte insulin resistance. Insig1 downregulation is part of an adaptive response that promotes the maintenance of SREBP1 maturation and facilitates lipogenesis and availability of appropriate levels of fatty acid unsaturation, partially compensating the antilipogenic effect associated with insulin resistance. We describe for the first time the existence of this adaptive mechanism in WAT, which involves Insig1/SREBP1 and preserves the degree of lipid unsaturation under conditions of obesity-induced insulin resistance. These adaptive mechanisms contribute to maintain lipid desaturation through preferential SCD1 regulation and facilitate fat storage in WAT, despite on-going metabolic stress.
Collapse
Affiliation(s)
- Stefania Carobbio
- University of Cambridge, Metabolic Research Laboratories, Institute of Metabolic Science Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, U.K
| | - Rachel M. Hagen
- University of Cambridge, Metabolic Research Laboratories, Institute of Metabolic Science Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, U.K
| | | | - Marc Slawik
- University of Cambridge, Metabolic Research Laboratories, Institute of Metabolic Science Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, U.K
- Endocrine Research Unit, Medizinische Klinik-Innenstadt, Ludwig-Maximilians University, Munich, Germany
| | - Gema Medina-Gomez
- University of Cambridge, Metabolic Research Laboratories, Institute of Metabolic Science Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, U.K
- Departamento de Bioquímica, Fisiología y Genética Molecular, Universidad Rey Juan Carlos Facultad de Ciencias de la Salud Avda.de Atenas s/n28922 Alcorcón, Madrid, Spain
| | - Chong-Yew Tan
- University of Cambridge, Metabolic Research Laboratories, Institute of Metabolic Science Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, U.K
| | - Audrey Sicard
- INSERM, Paul Sabatier University, UMR1048, Institute of Metabolic and Cardiovascular Diseases (I2MC), Laboratory of Obesity, Toulouse, France
| | - Helen J. Atherton
- MRC Human Nutrition Research, Elsie Widdowson Laboratory & University of Cambridge, Department of Biochemistry, Cambridge, U.K
| | - Nuria Barbarroja
- University of Cambridge, Metabolic Research Laboratories, Institute of Metabolic Science Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, U.K
- Hospital Virgen de la Victoria, CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Malaga, Spain
| | - Mikael Bjursell
- Department of Biosciences, CVGI iMED, AstraZeneca Research and Development, Mölndal, Sweden
| | - Mohammad Bohlooly-Y
- Department of Biosciences, CVGI iMED, AstraZeneca Research and Development, Mölndal, Sweden
| | - Sam Virtue
- University of Cambridge, Metabolic Research Laboratories, Institute of Metabolic Science Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, U.K
| | - Antoinette Tuthill
- University of Cambridge, Metabolic Research Laboratories, Institute of Metabolic Science Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, U.K
| | - Etienne Lefai
- INSERM U-1060; INRA U-1235; Human Nutrition Research Center of Lyon CarMeN Laboratory, Lyon1 University, Lyon, France
| | - Martine Laville
- INSERM U-1060; INRA U-1235; Human Nutrition Research Center of Lyon CarMeN Laboratory, Lyon1 University, Lyon, France
| | - Tingting Wu
- Department of Biosciences, CVGI iMED, AstraZeneca Research and Development, Mölndal, Sweden
| | - Robert V. Considine
- Division of Endocrinology and Metabolism, Indiana University School of Medicine, Indianapolis, Indiana
| | - Hubert Vidal
- INSERM U-1060; INRA U-1235; Human Nutrition Research Center of Lyon CarMeN Laboratory, Lyon1 University, Lyon, France
| | - Dominique Langin
- INSERM, Paul Sabatier University, UMR1048, Institute of Metabolic and Cardiovascular Diseases (I2MC), Laboratory of Obesity, Toulouse, France
- Laboratory of Clinical Biochemistry, Toulouse, France
| | - Matej Oresic
- Department of Medicine, Division of Internal Medicine, and Department of Psychiatry, Obesity Research Unit, Helsinki University Central Hospital, Helsinki, Finland
| | - Francisco J. Tinahones
- Hospital Virgen de la Victoria, CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Malaga, Spain
| | - Jose Manuel Fernandez-Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomédica de Girona, CIBERobn Fisiopatología de la Obesidad y Nutrición CB06/03/010, Girona, Spain
| | - Julian L. Griffin
- MRC Human Nutrition Research, Elsie Widdowson Laboratory & University of Cambridge, Department of Biochemistry, Cambridge, U.K
| | - Jaswinder K. Sethi
- University of Cambridge, Metabolic Research Laboratories, Institute of Metabolic Science Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, U.K
| | - Miguel López
- University of Cambridge, Metabolic Research Laboratories, Institute of Metabolic Science Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, U.K
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Antonio Vidal-Puig
- University of Cambridge, Metabolic Research Laboratories, Institute of Metabolic Science Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, U.K
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, U.K
- Corresponding author: Antonio Vidal-Puig,
| |
Collapse
|
39
|
Wu T, Tang Q, Gao Z, Yu Z, Song H, Zheng X, Chen W. Blueberry and mulberry juice prevent obesity development in C57BL/6 mice. PLoS One 2013; 8:e77585. [PMID: 24143244 PMCID: PMC3797064 DOI: 10.1371/journal.pone.0077585] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Accepted: 09/03/2013] [Indexed: 12/16/2022] Open
Abstract
Objectives To establish whether blueberry (Vaccinium ashei) and mulberry (Morus australis Poir) juice, anthocyanin rich fruit juice, may help counteract obesity. Design And Methods: Four-week-old C57BL/6 mice were fed a high-fat diet (HFD) with or without blueberry and mulberry juice for 12 weeks. Body weight, serum and hepatic lipids, liver and adipose tissues morphology, insulin and leptin were assessed. Results Mice fed HFD exhibited increased body weight, insulin resistance, serum and hepatic lipids. In comparison, blueberry and mulberry juice inhibited body weight gain, decreased the serum cholesterol, reduced the resistance to insulin, attenuated lipid accumulation and decreased the leptin secretin. Conclusion These results indicate that blueberry and mulberry juice may help counteract obesity.
Collapse
Affiliation(s)
- Tao Wu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
- Fuli Institute of Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qiong Tang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zichun Gao
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhuoping Yu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haizhao Song
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaodong Zheng
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
- Fuli Institute of Food Science, Zhejiang University, Hangzhou, Zhejiang, China
- * E-mail: (XZ); (WC)
| | - Wei Chen
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
- Fuli Institute of Food Science, Zhejiang University, Hangzhou, Zhejiang, China
- * E-mail: (XZ); (WC)
| |
Collapse
|
40
|
Aizawa-Abe M, Ebihara K, Ebihara C, Mashimo T, Takizawa A, Tomita T, Kusakabe T, Yamamoto Y, Aotani D, Yamamoto-Kataoka S, Sakai T, Hosoda K, Serikawa T, Nakao K. Generation of leptin-deficient Lepmkyo/Lepmkyo rats and identification of leptin-responsive genes in the liver. Physiol Genomics 2013; 45:786-93. [DOI: 10.1152/physiolgenomics.00040.2013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Leptin is one of the key molecules in maintaining energy homeostasis. Although genetically leptin-deficient Lep ob /Lep ob mice have greatly contributed to elucidating leptin physiology, the use of more than one species can improve the accuracy of analysis results. Using the N-ethyl- N-nitrosourea mutagenesis method, we generated a leptin-deficient Lep mkyo /Lep mkyo rat that had a nonsense mutation (Q92X) in leptin gene. Lep mkyo /Lep mkyo rats showed obese phenotypes including severe fatty liver, which were comparable to Lep ob /Lep ob mice. To identify genes that respond to leptin in the liver, we performed microarray analysis with Lep mkyo /Lep mkyo rats and Lep ob /Lep ob mice. We sorted out genes whose expression levels in the liver of Lep mkyo /Lep mkyo rats were changed from wild-type (WT) rats and were reversed toward WT rats by leptin administration. In this analysis, livers were sampled for 6 h, a relatively short time after leptin administration to avoid the secondary effect of metabolic changes such as improvement of fatty liver. We did the same procedure in Lep ob /Lep ob mice and selected genes whose expression patterns were common in rat and mouse. We verified their gene expressions by real-time quantitative PCR. Finally, we identified eight genes that primarily respond to leptin in the liver commonly in rat and mouse. These genes might be important for the effect of leptin in the liver.
Collapse
Affiliation(s)
- Megumi Aizawa-Abe
- Department of Experimental Therapeutics, Translational Research Center, Kyoto University Hospital, Kyoto, Japan
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ken Ebihara
- Department of Experimental Therapeutics, Translational Research Center, Kyoto University Hospital, Kyoto, Japan
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Chihiro Ebihara
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomoji Mashimo
- Institute of Laboratory Animals, Kyoto University Graduate School of Medicine, Kyoto, Japan; and
| | - Akiko Takizawa
- Institute of Laboratory Animals, Kyoto University Graduate School of Medicine, Kyoto, Japan; and
| | - Tsutomu Tomita
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toru Kusakabe
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuji Yamamoto
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Daisuke Aotani
- Department of Experimental Therapeutics, Translational Research Center, Kyoto University Hospital, Kyoto, Japan
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sachiko Yamamoto-Kataoka
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeru Sakai
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kiminori Hosoda
- Department of Experimental Therapeutics, Translational Research Center, Kyoto University Hospital, Kyoto, Japan
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Health and Science, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tadao Serikawa
- Institute of Laboratory Animals, Kyoto University Graduate School of Medicine, Kyoto, Japan; and
| | - Kazuwa Nakao
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
41
|
Moon HS, Dalamaga M, Kim SY, Polyzos SA, Hamnvik OP, Magkos F, Paruthi J, Mantzoros CS. Leptin's role in lipodystrophic and nonlipodystrophic insulin-resistant and diabetic individuals. Endocr Rev 2013; 34:377-412. [PMID: 23475416 PMCID: PMC3660716 DOI: 10.1210/er.2012-1053] [Citation(s) in RCA: 190] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Leptin is an adipocyte-secreted hormone that has been proposed to regulate energy homeostasis as well as metabolic, reproductive, neuroendocrine, and immune functions. In the context of open-label uncontrolled studies, leptin administration has demonstrated insulin-sensitizing effects in patients with congenital lipodystrophy associated with relative leptin deficiency. Leptin administration has also been shown to decrease central fat mass and improve insulin sensitivity and fasting insulin and glucose levels in HIV-infected patients with highly active antiretroviral therapy (HAART)-induced lipodystrophy, insulin resistance, and leptin deficiency. On the contrary, the effects of leptin treatment in leptin-replete or hyperleptinemic obese individuals with glucose intolerance and diabetes mellitus have been minimal or null, presumably due to leptin tolerance or resistance that impairs leptin action. Similarly, experimental evidence suggests a null or a possibly adverse role of leptin treatment in nonlipodystrophic patients with nonalcoholic fatty liver disease. In this review, we present a description of leptin biology and signaling; we summarize leptin's contribution to glucose metabolism in animals and humans in vitro, ex vivo, and in vivo; and we provide insights into the emerging clinical applications and therapeutic uses of leptin in humans with lipodystrophy and/or diabetes.
Collapse
Affiliation(s)
- Hyun-Seuk Moon
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Bays HE, Toth PP, Kris-Etherton PM, Abate N, Aronne LJ, Brown WV, Gonzalez-Campoy JM, Jones SR, Kumar R, La Forge R, Samuel VT. Obesity, adiposity, and dyslipidemia: a consensus statement from the National Lipid Association. J Clin Lipidol 2013; 7:304-83. [PMID: 23890517 DOI: 10.1016/j.jacl.2013.04.001] [Citation(s) in RCA: 318] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 04/02/2013] [Accepted: 04/03/2013] [Indexed: 01/04/2023]
Abstract
The term "fat" may refer to lipids as well as the cells and tissue that store lipid (ie, adipocytes and adipose tissue). "Lipid" is derived from "lipos," which refers to animal fat or vegetable oil. Adiposity refers to body fat and is derived from "adipo," referring to fat. Adipocytes and adipose tissue store the greatest amount of body lipids, including triglycerides and free cholesterol. Adipocytes and adipose tissue are active from an endocrine and immune standpoint. Adipocyte hypertrophy and excessive adipose tissue accumulation can promote pathogenic adipocyte and adipose tissue effects (adiposopathy), resulting in abnormal levels of circulating lipids, with dyslipidemia being a major atherosclerotic coronary heart disease risk factor. It is therefore incumbent upon lipidologists to be among the most knowledgeable in the understanding of the relationship between excessive body fat and dyslipidemia. On September 16, 2012, the National Lipid Association held a Consensus Conference with the goal of better defining the effect of adiposity on lipoproteins, how the pathos of excessive body fat (adiposopathy) contributes to dyslipidemia, and how therapies such as appropriate nutrition, increased physical activity, weight-management drugs, and bariatric surgery might be expected to impact dyslipidemia. It is hoped that the information derived from these proceedings will promote a greater appreciation among clinicians of the impact of excess adiposity and its treatment on dyslipidemia and prompt more research on the effects of interventions for improving dyslipidemia and reducing cardiovascular disease risk in overweight and obese patients.
Collapse
Affiliation(s)
- Harold E Bays
- Louisville Metabolic and Atherosclerosis Research Center, 3288 Illinois Avenue, Louisville, KY 40213, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Dodson MV, Boudina S, Albrecht E, Bucci L, Culver MF, Wei S, Bergen WG, Amaral AJ, Moustaid-Moussa N, Poulos S, Hausman GJ. A long journey to effective obesity treatments: is there light at the end of the tunnel? Exp Biol Med (Maywood) 2013; 238:491-501. [PMID: 23856900 DOI: 10.1177/1535370213477603] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
As the obesity epidemic continues, more Americans are getting fatter, having more weight-related problems such as cardiovascular disease, and are experiencing new metabolic dysfunctions. For over 50 years, the adipose tissue (AT), commonly referred to as fat, has been of interest to academic and clinical scientists, public health officials and individuals interested in body composition and image including much of the average public, athletes, parents, etc. On one hand, efforts to alter body shape, weight and body fat percentage still include bizarre and scientifically unfounded methods. On the other hand, significant new scientific strides have been made in understanding the growth, function and regulation of anatomical and systemic AT. Markers of transition/conversion of precursor cells that mature to form lipid assimilating adipocytes have been identified. Molecular 'master' regulators such as peroxisome proliferator-activated receptor gamma and CCAAT-enhancer-binding proteins were uncovered and regulatory mechanisms behind variables of adiposity defined and refined. Interventions including pharmaceutical compounds, surgical, psychosocial interventions have also been tested. Has all of the preceding research helped alleviate the adverse physiologies of overweight and/or obese people? Does research to date point to new modalities that should be the focus of efforts to rid the world of obesity-related problems in the 21st century? This review provides a general overview of scientific efforts to date and a provocative view of the future for adiposity.
Collapse
Affiliation(s)
- Michael V Dodson
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Wu T, Yu Z, Tang Q, Song H, Gao Z, Chen W, Zheng X. Honeysuckle anthocyanin supplementation prevents diet-induced obesity in C57BL/6 mice. Food Funct 2013; 4:1654-61. [DOI: 10.1039/c3fo60251f] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
45
|
Burgos-Ramos E, Sackmann-Sala L, Baquedano E, Cruz-Topete D, Barrios V, Argente J, Kopchick JJ. Central leptin and insulin administration modulates serum cytokine- and lipoprotein-related markers. Metabolism 2012; 61:1646-57. [PMID: 22658937 DOI: 10.1016/j.metabol.2012.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 04/12/2012] [Accepted: 05/01/2012] [Indexed: 10/28/2022]
Abstract
UNLABELLED In most obese patients there is an inflammatory state characterized by lipid abnormalities, hyperleptinemia and hyperinsulinemia. OBJECTIVE The objective was to identify mechanisms involved in leptin's role in the attenuation of the response to insulin using a proteomic approach. MATERIAL/METHODS We studied the serum proteomic profile of rats treated by central leptin infusion followed by an injection of insulin. We analyzed the relationship between these proteins and serum cytokine and apolipoprotein levels. RESULTS Out of 81 protein spots, intensity differences were found in 11, corresponding to 5 proteins: three isoforms of α1 macroglobulin; three of haptoglobin and serum amyloid P component-precursor. All of these are acute-phase proteins involved in inflammation and are correlated with cytokine levels. Additionally, two apolipoprotein E and two apolipoprotein A1 isoforms were identified and were found to correlate with LDL and HDL. CONCLUSIONS Our results indicate that increased leptin and insulin levels change these circulating proteins, thus promoting systemic inflammation and changing lipid metabolism.
Collapse
Affiliation(s)
- Emma Burgos-Ramos
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto Investigación Sanitaria Princesa, Madrid, E-28009 Spain
| | | | | | | | | | | | | |
Collapse
|
46
|
Burgos-Ramos E, Canelles S, Perianes-Cachero A, Arilla-Ferreiro E, Argente J, Barrios V. Adipose tissue promotes a serum cytokine profile related to lower insulin sensitivity after chronic central leptin infusion. PLoS One 2012; 7:e46893. [PMID: 23056516 PMCID: PMC3462753 DOI: 10.1371/journal.pone.0046893] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 09/10/2012] [Indexed: 12/13/2022] Open
Abstract
Obesity is an inflammatory state characterized by an augment in circulating inflammatory factors. Leptin may modulate the synthesis of these factors by white adipose tissue decreasing insulin sensitivity. We have examined the effect of chronic central administration of leptin on circulating levels of cytokines and the possible relationship with cytokine expression and protein content as well as with leptin and insulin signaling in subcutaneous and visceral adipose tissues. In addition, we analyzed the possible correlation between circulating levels of cytokines and peripheral insulin resistance. We studied 18 male Wistar rats divided into controls (C), those treated icv for 14 days with a daily dose of 12 μg of leptin (L) and a pair-fed group (PF) that received the same food amount consumed by the leptin group. Serum leptin and insulin were measured by ELISA, mRNA levels of interferon-γ (IFN-γ), interleukin-2 (IL-2), IL-4, IL-6, IL-10 and tumor necrosis factor-α (TNF-α) by real time PCR and serum and adipose tissue levels of these cytokines by multiplexed bead immunoassay. Serum leptin, IL-2, IL-4, IFN-γ and HOMA-IR were increased in L and TNF-α was decreased in PF and L. Serum leptin and IL-2 levels correlate positively with HOMA-IR index and negatively with serum glucose levels during an ip insulin tolerance test. In L, an increase in mRNA levels of IL-2 was found in both adipose depots and IFN-γ only in visceral tissue. Activation of leptin signaling was increased and insulin signaling decreased in subcutaneous fat of L. In conclusion, leptin mediates the production of inflammatory cytokines by adipose tissue independent of its effects on food intake, decreasing insulin sensitivity.
Collapse
Affiliation(s)
- Emma Burgos-Ramos
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa and Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Canelles
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa and Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Arancha Perianes-Cachero
- Grupo de Neurobioquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Eduardo Arilla-Ferreiro
- Grupo de Neurobioquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa and Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa and Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- * E-mail:
| |
Collapse
|
47
|
Alves-Pereira JL, Colli S, Marques DS, Sampaio FJ, Ramos CF. Molecular and morphometric analysis of the rat ventral prostate injected with leptin. ACTA ACUST UNITED AC 2012; 176:6-12. [PMID: 22387703 DOI: 10.1016/j.regpep.2012.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 12/14/2011] [Accepted: 02/20/2012] [Indexed: 01/11/2023]
Abstract
The aim of this study was to evaluate the effect of leptin administration on the ventral prostate lobe of adult rat. Twenty adult male rats were divided into 2 groups: L-animals were daily injected with 50 μL of leptin (8 μg/100 g BW, subcutaneous) for four days and C-animals received the same volume of saline solution. Lipid profile and testosterone serum levels were evaluated. The prostate ventral lobe was processed for histomorphometric analysis. Gene expression of aromatase, androgen, leptin and estrogen receptors isoforms was evaluated by real-time PCR. Cell proliferation was evaluated by PCNA immunohistochemistry. Data were expressed as mean±standard error and analyzed by student's t-test. Serum levels of cholesterol (C=39.7±4.2;L=55.2±4.2, mg/dL; P<0.02) increased and testosterone (C=1.6±0.43;L=0.6±0.15, ng/dL; P<0.03) decreased in L group. The histomorphometric analysis showed a reduction in cell density (C=8868±242; L=8211±210, mm(2); P<0.04), in total (C=0.24±0.026; L=0.10±0.009, mm(2); P<0.001) and in the internal acini areas (C=0.16±0.009; L=0.08±0.006, mm(2); P<0.0002). On the other hand, there was an increase in the epithelial height (C=17.3±0.3; L=22.8±0.2, μm; P<0.0001) and in the number of acini (C=7.0±0.2; L=8.7±0.1, mm(2); P<0.0002). The histomorphometric analyses together with PCNA immunohistochemistry results suggest that leptin increases cell proliferation. In relation to the gene expression, leptin treatment increased the expression of all genes, but ER-α, in more than 200 times compared to the expression in C group. In conclusion, in this paper we showed that leptin has a direct effect on the prostate gland of adult rats leading to an increase in proliferation and in the gene expression of aromatase, androgen, leptin and estrogen receptors isoforms that are important for the physiology of the prostate gland.
Collapse
|
48
|
Berglund ED, Vianna CR, Donato J, Kim MH, Chuang JC, Lee CE, Lauzon DA, Lin P, Brule LJ, Scott MM, Coppari R, Elmquist JK. Direct leptin action on POMC neurons regulates glucose homeostasis and hepatic insulin sensitivity in mice. J Clin Invest 2012; 122:1000-9. [PMID: 22326958 DOI: 10.1172/jci59816] [Citation(s) in RCA: 255] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 01/04/2012] [Indexed: 01/01/2023] Open
Abstract
Leptin action on its receptor (LEPR) stimulates energy expenditure and reduces food intake, thereby lowering body weight. One leptin-sensitive target cell mediating these effects on energy balance is the proopiomelano-cortin (POMC) neuron. Recent evidence suggests that the action of leptin on POMC neurons regulates glucose homeostasis independently of its effects on energy balance. Here, we have dissected the physiological impact of direct leptin action on POMC neurons using a mouse model in which endogenous LEPR expression was prevented by a LoxP-flanked transcription blocker (loxTB), but could be reactivated by Cre recombinase. Mice homozygous for the Lepr(loxTB) allele were obese and exhibited defects characteristic of LEPR deficiency. Reexpression of LEPR only in POMC neurons in the arcuate nucleus of the hypothalamus did not reduce food intake, but partially normalized energy expenditure and modestly reduced body weight. Despite the moderate effects on energy balance and independent of changes in body weight, restoring LEPR in POMC neurons normalized blood glucose and ameliorated hepatic insulin resistance, hyperglucagonemia, and dyslipidemia. Collectively, these results demonstrate that direct leptin action on POMC neurons does not reduce food intake, but is sufficient to normalize glucose and glucagon levels in mice otherwise lacking LEPR.
Collapse
Affiliation(s)
- Eric D Berglund
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical School, Dallas, Texas 75390-9051, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Volpato AM, Schultz A, Magalhães-da-Costa E, Correia MLDG, Águila MB, Mandarim-de-Lacerda CA. Maternal high-fat diet programs for metabolic disturbances in offspring despite leptin sensitivity. Neuroendocrinology 2012; 96:272-84. [PMID: 22456428 DOI: 10.1159/000336377] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 12/30/2011] [Indexed: 02/04/2023]
Abstract
A fatty diet during pregnancy in mouse dams causes metabolic abnormalities (similar to metabolic syndrome in humans) in the rodents' offspring. We tested the hypothesis that the offspring of dams fed a high-fat diet during pregnancy and lactation develop metabolic abnormalities and leptin resistance. Pregnant C57BL/6 mice (n = 20) were fed either standard chow (SC; 19% fat) or a high-fat diet (HF; 49% fat). After weaning, male offspring were divided into four groups, according to the diet of dams and offspring: SC(dams)/SC(offspring), SC/HF, HF/SC and HF/HF (n = 30/group). For a metabolic analysis, we evaluated body mass, fat mass depots, blood plasma and adipocyte structure at 12 weeks of age. To analyse leptin sensitivity, each group was divided into two groups (vehicle or leptin) to identify the feeding response and pSTAT3 expression after acute intracerebroventricular (ICV) treatment. The offspring of mothers fed a high-fat diet presented increased body mass and visceral fat, adipocyte hypertrophy and insulin resistance. This phenotype was not associated with central leptin resistance. Thus, maternal programming by HF predisposes offspring to metabolic abnormalities despite leptin sensitivity.
Collapse
Affiliation(s)
- Ana Maria Volpato
- Laboratory of Morphometry, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | | |
Collapse
|
50
|
Warne JP, Alemi F, Reed AS, Varonin JM, Chan H, Piper ML, Mullin ME, Myers MG, Corvera CU, Xu AW. Impairment of central leptin-mediated PI3K signaling manifested as hepatic steatosis independent of hyperphagia and obesity. Cell Metab 2011; 14:791-803. [PMID: 22152304 PMCID: PMC3240844 DOI: 10.1016/j.cmet.2011.11.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 09/13/2011] [Accepted: 11/02/2011] [Indexed: 11/22/2022]
Abstract
Hepatic steatosis is generally thought to develop via peripheral mechanisms associated with obesity. We show that chronic central infusion of leptin suppresses hepatic lipogenic gene expression and reduces triglyceride content via stimulation of hepatic sympathetic activity. This leptin function is independent of feeding and body weight but requires phosphatidylinositol 3-kinase (PI3K) signaling. Attenuation of leptin-induced PI3K signaling, brought about by transgenic expression of phosphatase and tensin homolog (PTEN) in leptin receptor neurons, leads to decreased hepatic sympathetic tone and increased triglyceride levels without affecting adiposity or hepatic insulin signaling. Central leptin's effects on hepatic norepinephrine levels and triglyceride content are blunted in these mutant mice. Simultaneous downregulation of PI3K and signal transducer and activator of transcription-3 (Stat3) in leptin receptor neurons does not exacerbate obesity but causes more severe hepatic steatosis. Together, our results indicate that central cellular leptin resistance in PI3K signaling manifests as hepatic steatosis without causing obesity.
Collapse
Affiliation(s)
- James P. Warne
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Farzad Alemi
- General Surgery, San Francisco VA Medical Center, University of California, San Francisco, San Francisco, CA 94143
| | - Alison S. Reed
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jillian M. Varonin
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Helen Chan
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Merisa L. Piper
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mark E. Mullin
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Martin G. Myers
- Departments of Internal Medicine and Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Carlos U. Corvera
- General Surgery, San Francisco VA Medical Center, University of California, San Francisco, San Francisco, CA 94143
| | - Allison W. Xu
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|