1
|
Abstract
Leptin for over 25 years has been a central theme in the study of appetite, obesity, and starvation. As the major site of leptin production is peripheral, and the site of action of greatest interest is the hypothalamus, how leptin accesses the central nervous system (CNS) and crosses the blood-brain barrier (BBB) has been of great interest. We review here the ongoing research that addresses fundamental questions such as the sites of leptin resistances in obesity and other conditions, the causes of resistances and their relations to one another, the three barrier sites of entry into the CNS, why recent studies using suprapharmacological doses cannot address these questions but give insight into nonsaturable entry of leptin into the CNS, and how that might be useful in using leptin therapeutically. The current status of the controversy of whether the short form of the leptin receptor acts as the BBB leptin transporter and how obesity may transform leptin transport is reviewed. Review of these and other topics summarizes in a new appreciation of what leptin may have actually evolved to do and what physiological role leptin resistance may play. © 2021 American Physiological Society. Compr Physiol 11:1-19, 2021.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
2
|
Brown WE, Bradford BJ. Invited review: Mechanisms of hypophagia during disease. J Dairy Sci 2021; 104:9418-9436. [PMID: 34099296 DOI: 10.3168/jds.2021-20217] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/24/2021] [Indexed: 12/26/2022]
Abstract
Suppression of appetite, or hypophagia, is among the most recognizable effects of disease in livestock, with the potential to impair growth, reproduction, and lactation. The continued evolution of the field of immunology has led to a greater understanding of the immune and endocrine signaling networks underlying this conserved response to disease. Inflammatory mediators, especially including the cytokines tumor necrosis factor-α and interleukin-1β, are likely pivotal to disease-induced hypophagia, based on findings in both rodents and cattle. However, the specific mechanisms linking a cytokine surge to decreased feeding behavior are more difficult to pin down and likely include direct effects on appetite centers in the brain, alteration of gastric motility, and modulation of other endocrine factors that influence appetite and satiety. These insights into the mechanisms for disease-induced hypophagia have great relevance for management of neonatal calves, mature cows transitioning to lactation, and cows experiencing mastitis; however, it is not necessarily the case that increasing feed intake by any means possible will improve health outcomes for diseased cattle. We explore conflicting effects of hypophagia on immune responses, which may be impaired by the lack of specific substrates, versus apparent benefits for controlling the growth of some pathogens. Anti-inflammatory strategies have shown promise for promoting recovery of feed intake following some conditions but not others. Finally, we explore the potential for early disease detection through automated monitoring of feeding behavior and consider which strategies may be implemented to respond to early hypophagia.
Collapse
Affiliation(s)
- W E Brown
- Department of Animal Sciences & Industry, Kansas State University, Manhattan 66506
| | - B J Bradford
- Department of Animal Science, Michigan State University, East Lansing 48824.
| |
Collapse
|
3
|
Peek V, Neumann E, Inoue T, Koenig S, Pflieger FJ, Gerstberger R, Roth J, Matsumura K, Rummel C. Age-Dependent Changes of Adipokine and Cytokine Secretion From Rat Adipose Tissue by Endogenous and Exogenous Toll-Like Receptor Agonists. Front Immunol 2020; 11:1800. [PMID: 32973755 PMCID: PMC7466552 DOI: 10.3389/fimmu.2020.01800] [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] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/06/2020] [Indexed: 01/04/2023] Open
Abstract
White adipose tissue but recently also brown adipose tissue have emerged as endocrine organs. Age-associated obesity is accompanied by prolonged and elevated lipopolysaccharide (LPS)-induced sickness symptoms and increased cytokine and adipokine levels in the circulation partially originating from adipose tissue. In the present study, ex vivo fat explants were used to investigate how the exogenous pathogen-associated molecular pattern (PAMP) LPS or the endogenous danger-associated molecular patterns (DAMPs) high mobility group box-1 protein (HMGB1) and biglycan modulate the release of cytokines and adipokines/batokines and, thus, could influence systemic and/or local inflammation. The response of adipose tissue (epididymal, retroperitoneal, subcutaneous, and brown) was compared between young lean and old obese rats (2 vs. 24 months old). LPS induced a strong interleukin (IL)-6 and tumor necrosis factor (TNF) alpha release into the supernatant of all adipose tissue types investigated. HMGB1 (subcutaneous) and biglycan (retroperitoneal) led to an increased release of IL-6 and TNFalpha (HMGB1) and decreased visfatin and adiponectin (biglycan) secretion from epididymal adipose tissue (young rats). Visfatin was also decreased by HMGB1 in retroperitoneal adipose tissue of old rats. We found significantly higher leptin (all fat pads) and adiponectin (subcutaneous) levels in supernatants of adipose tissue from old compared to young rats, whereas visfatin secretion showed the opposite. The expression of the biglycan receptor Toll-like receptor (TLR) 2 as well as the LPS and HMGB1 receptors TLR4 and receptor for advanced glycation end products (RAGE) were reduced with age (TLR4/RAGE) and by stimulation with their ligands (subcutaneous). Overall, we revealed that adipokines/adipose-tissue released cytokines show some modulation of their release caused by mediators of septic (batokines) and sterile inflammation with potential implication for acute and chronic disease. Moreover, aging may increase or decrease the release of fat-derived mediators. These data show that DAMPS and LPS locally modulate cytokine secretion while only DAMPS but not LPS can locally alter adipokine secretion during inflammation.
Collapse
Affiliation(s)
- Verena Peek
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Elena Neumann
- Department of Rheumatology and Clinical Immunology, Campus Kerckhoff, Justus Liebig University Gießen, Bad Nauheim, Germany
| | - Tomohiro Inoue
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka, Japan
| | - Sandy Koenig
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Fabian Johannes Pflieger
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Rüdiger Gerstberger
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Joachim Roth
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany.,Joachim Roth and Christoph Rummel, Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany
| | - Kiyoshi Matsumura
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka, Japan
| | - Christoph Rummel
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany.,Joachim Roth and Christoph Rummel, Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany
| |
Collapse
|
4
|
Gregg BA, Parker PA, Waller KM, Schneider LG, Garcia M, Bradford B, Daniel JA, Whitlock BK. Effects of central and peripheral administration of an acute-phase protein, α-1-acid-glycoprotein, on feed intake and rectal temperature in sheep. J Anim Sci 2019; 97:4783-4791. [PMID: 31679022 PMCID: PMC6915238 DOI: 10.1093/jas/skz336] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 10/29/2019] [Indexed: 12/21/2022] Open
Abstract
In rodents, an acute-phase protein, α-1-acid-glycoprotein (AGP), was shown to provide a link between inflammation and suppression of feed intake by acting as a leptin receptor agonist. The objective of this study was to determine the effects of AGP on feed intake and rectal temperature in sheep. Ewes were ovariectomized, implanted with a cannula into a lateral ventricle of the brain, and kept indoors in individual pens. Feed intake and rectal temperature were determined for sheep in all experiments. In the first experiment, ewes (n = 4) received 1 of 4 treatments [0 (control), 0.012 (low), 0.06 (medium), or 0.30 (high) mg/kg BW AGP] into the lateral ventricle (ICV). All sheep received all treatments in a Latin square design balanced for carryover effects with 10 d between treatments. In the second experiment, ewes (n = 10) received 1 of 2 treatments (0 and 3 mg/kg BW of AGP) intravenously (IV) in a completely randomized design. In the third experiment, ewes (n = 19) received peripheral treatments (IV) of an antipyretic [0 (control) or 2.2 mg/kg BW flunixin meglumine (FLU)] 30 min before receiving central AGP [0 (control) or 0.3 mg/kg BW of AGP] in a completely randomized design. All data were analyzed using a mixed model analysis of variance and tested for effects of treatment, time, and the interaction of treatment and time. Cumulative 48-h feed intake after administration of treatments was also determined. In the first experiment, there was no effect of ICV treatment (P = 0.37) on feed intake rate or on cumulative feed intake (P = 0.31). There was an effect of ICV treatment (P = 0.002) on rectal temperatures, which were greater (P < 0.05) after the high dose of centrally administered AGP. In the second experiment, there was no effect of AGP administration IV on feed intake rate (P = 0.98), on cumulative feed intake (P = 0.41) or on rectal temperature (P = 0.71). In the third experiment, there was an effect of central AGP treatment (P < 0.0001) and an interaction of central AGP and time (P < 0.0001) on rectal temperature, whereas FLU had no effect (P = 0.93), demonstrating that AGP increased rectal temperatures regardless of antipyretic treatment. These results indicate that central AGP increases rectal temperature in sheep by pathways that do not involve prostaglandins. Further research is needed to determine whether AGP may be an important integrator of energy balance and inflammation.
Collapse
Affiliation(s)
- Brittany A Gregg
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN
- Department of Animal Science, University of Tennessee, Knoxville, TN
| | - Paxton A Parker
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN
| | - Kathryn M Waller
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN
| | | | - Miriam Garcia
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS
| | - Barry Bradford
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS
| | - Joseph A Daniel
- School of Mathematical and Natural Sciences, Berry College, Mount Berry, GA
| | - Brian K Whitlock
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN
- Department of Animal Science, University of Tennessee, Knoxville, TN
| |
Collapse
|
5
|
Tachibana T, Kodama T, Yamane S, Makino R, Khan SI, Cline MA. Possible role of central interleukins on the anorexigenic effect of lipopolysaccharide in chicks. Br Poult Sci 2017; 58:305-311. [DOI: 10.1080/00071668.2017.1280774] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- T. Tachibana
- Department of Agrobiological Science, Faculty of Agriculture, Ehime University, Matsuyama, Japan
| | - T. Kodama
- Department of Agrobiological Science, Faculty of Agriculture, Ehime University, Matsuyama, Japan
| | - S. Yamane
- Department of Agrobiological Science, Faculty of Agriculture, Ehime University, Matsuyama, Japan
| | - R. Makino
- Department of Agrobiological Science, Faculty of Agriculture, Ehime University, Matsuyama, Japan
| | - S. I. Khan
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon 791-0212, Ehime, Japan
| | - M. A. Cline
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| |
Collapse
|
6
|
Kurosawa N, Shimizu K, Seki K. The development of depression-like behavior is consolidated by IL-6-induced activation of locus coeruleus neurons and IL-1β-induced elevated leptin levels in mice. Psychopharmacology (Berl) 2016; 233:1725-37. [PMID: 26385227 DOI: 10.1007/s00213-015-4084-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 09/10/2015] [Indexed: 12/17/2022]
Abstract
RATIONALE Many studies have supported the cytokine hypothesis as the underlying pathophysiology of depressive disorder. OBJECTIVES We previously reported that lipopolysaccharide (LPS)-induced depression-like behavior is abrogated by the α1-adrenoceptor antagonist prazosin. Since cytokines are involved in LPS effects on the brain, we investigated the effects of cytokines on noradrenergic neurons in the locus coeruleus (LC) and whether central α1-adrenoceptors can cause the development of depression-like behavior. METHODS Adult male CD1 mice were treated with LPS (1 mg/kg, i.p.) or saline and sacrificed 2 h later for immunofluorescence studies of c-fos and tyrosine hydroxylase (TH) expression in LC neurons. Serum cytokines were measured using enzyme-linked immunosorbent assay (ELISA). Another group of mice were implanted with intracerebroventricular (i.c.v.) cannulae and given artificial cerebrospinal fluid (CSF) (control), interleukin (IL)-1β (0.5 μg), IL-6 (1 μg), or tumor necrosis factor (TNF)-α (1 μg), and sacrificed 2 h later for c-fos and TH immunofluorescence analysis. Serum samples were analyzed for leptin levels. In addition, tail suspension test (TST), forced swimming test (FST), and sucrose preference (SP) test were conducted in a separate group of mice treated i.c.v. with cytokines, recombinant mouse leptin (5 μg) or phenylephrine (40 μg). These effects were countered by i.c.v. administration of prazosin and a leptin antagonist. RESULTS LPS increased c-fos expression in TH-positive neurons. Central administration of IL-6 and IL-1β increased c-fos immunoreactivity and serum leptin levels. Phenylephrine, an α1-adrenoceptor agonist, given i.c.v., increased the immobility time during FST and decreased SP, but had no effect on TST. Central leptin administration increased immobility time during FST but did not affect TST or SP. The combination of phenylephrine and leptin increased immobility time during FST and TST, and decreased SP. Induction of depression-like behavior by co-administration of IL-1β and IL-6 was prevented by pretreatment with prazosin alone. CONCLUSION These results suggest that IL-6-dependent LC neuronal activation induced depression-like behavior and IL-1β-induced increase in leptin levels enhanced α1-adrenoceptor-mediated depression-like behavior.
Collapse
Affiliation(s)
- Natsuki Kurosawa
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima, 963-8611, Japan
| | - Koh Shimizu
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima, 963-8611, Japan
| | - Kenjiro Seki
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima, 963-8611, Japan.
| |
Collapse
|
7
|
Aguilar-Valles A, Inoue W, Rummel C, Luheshi GN. Obesity, adipokines and neuroinflammation. Neuropharmacology 2015; 96:124-34. [PMID: 25582291 DOI: 10.1016/j.neuropharm.2014.12.023] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/16/2014] [Accepted: 12/19/2014] [Indexed: 12/14/2022]
Abstract
Global levels of obesity are reaching epidemic proportions, leading to a dramatic increase in incidence of secondary diseases and the significant economic burden associated with their treatment. These comorbidities include diabetes, cardiovascular disease, and some psychopathologies, which have been linked to a low-grade inflammatory state. Obese individuals exhibit an increase in circulating inflammatory mediators implicated as the underlying cause of these comorbidities. A number of these molecules are also manufactured and released by white adipose tissue (WAT), in direct proportion to tissue mass and are collectively known as adipokines. In the current review we focused on the role of two of the better-studied members of this family namely, leptin and adiponectin, with particular emphasis on their role in neuro-immune interactions, neuroinflammation and subsequent brain diseases. This article is part of a Special Issue entitled 'Neuroimmunology and Synaptic Function'.
Collapse
Affiliation(s)
- Argel Aguilar-Valles
- Department of Neuroscience, Université de Montréal and Goodman Cancer Centre, Department of Biochemistry, McGill University, Montréal, Canada
| | - Wataru Inoue
- Robarts Research Institute, Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Christoph Rummel
- Department of Veterinary-Physiology and -Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - Giamal N Luheshi
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Quebec H4H 1R3, Canada.
| |
Collapse
|
8
|
Duan K, Yu W, Lin Z, Tan S, Bai X, Gao T, Xi F, Li N. Endotoxemia-induced muscle wasting is associated with the change of hypothalamic neuropeptides in rats. Neuropeptides 2014; 48:379-86. [PMID: 25459520 DOI: 10.1016/j.npep.2014.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 10/08/2014] [Accepted: 10/09/2014] [Indexed: 11/21/2022]
Abstract
In critical patients, sepsis-induced muscle wasting is considered to be an important contributor to complications and mortality. Previous work mainly focuses on the peripheral molecular mechanism of muscle degradation, however little evidence exists for the role of central nervous system in the process. In the present study, we, for the first time, characterized the relationship between muscle wasting and central neuropeptide changes in a septic model. Thirty-six adult male Sprague-Dawley rats were intraperitoneally injected with lipopolysaccharide (LPS) or saline. Twelve, 24 and 48 hrs after injection, skeletal muscle and hypothalamus tissues were harvested. Muscle wasting was measured by the mRNA expression of two E3 ubiquitin ligases, muscle ring finger 1 (MuRF-1) and muscle atrophy F-box (MAFbx), as well as 3-methyl-histidine (3-MH) and tyrosine release. Hypothalamic neuropeptides and inflammatory marker expressions were also measured in three time points. LPS injection caused an increase expression of MuRF-1 and MAFbx, and a significant higher release of 3-MH and tyrosine. Hypothalamic neuropeptides, proopiomelanocortin (POMC), cocaine- and amphetamine-regulated transcript (CART), agouti-related protein (AgRP) and neuropeptide Y (NPY) presented a dynamic change after LPS injection. Also, hypothalamic inflammatory markers, interleukin-1 β (IL-1β) and tumor necrosis factor α (TNF-α) increased substantially after LPS administration. Importantly, the expressions of POMC, AgRP and CART were well correlated with muscle atrophy gene, MuRF-1 expression. These findings suggest hypothalamic peptides and inflammation may participate in the sepsis-induced muscle wasting, but the exact mechanism needs further study.
Collapse
Affiliation(s)
- Kaipeng Duan
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Wenkui Yu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Zhiliang Lin
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Shanjun Tan
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Xiaowu Bai
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Tao Gao
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Fengchan Xi
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Ning Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| |
Collapse
|
9
|
Quinson N, Vitton V, Bouvier M, Grimaud JC, Abysique A. Effects of tumor necrosis factor α on leptin-sensitive intestinal vagal mechanoreceptors in the cat. Can J Physiol Pharmacol 2013; 91:941-50. [PMID: 24117262 DOI: 10.1139/cjpp-2013-0025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The involvement of tumour necrosis factor α (TNF-α) in inflammatory bowel disease (IBD) has been established, and anti-TNF-α has been suggested as a therapeutic approach for the treatment of these pathologies. We studied the effects of TNF-α on leptin-sensitive intestinal vagal units to determine whether TNF-α exerts its effects through the intestinal vagal mechanoreceptors and to investigate its interactions with substances regulating food intake. The activity of intestinal vagal mechanoreceptors was recorded via microelectrodes implanted into the nodose ganglion in anesthetized cats. TNF-α (1 μg, i.a.) increased the discharge frequency of leptin-activated units (type 1 units; P < 0.05) and had no effect on the discharge frequency of leptin-inhibited units (type 2 units). When TNF-α was administered 20 min after sulfated cholecystokinin-8 (CCK), its excitatory effects on type 1 units were significantly enhanced (P < 0.0001) and type 2 units were significantly (P < 0.05) activated. Pre-treatment with Il-1ra (250 μg, i.a.) blocked the excitatory effects of TNF-α on type 1 units whereas the excitatory effects of TNF-α administration after CCK treatment on type 2 units were not modified. The activation of leptin-sensitive units by TNF-α may explain, at least in part, the weight loss observed in IBD.
Collapse
Affiliation(s)
- Nathalie Quinson
- a Aix Marseille Université, Physiologie et Physiopathologie du Système Nerveux Somatomoteur et Neurovégétatif (PPSN, EA4674), Avenue Escadrille Normandie Niemen, 13397 Marseille Cedex 20, France
| | | | | | | | | |
Collapse
|
10
|
Gómez-SanMiguel AB, Martín AI, Nieto-Bona MP, Fernández-Galaz C, López-Menduiña M, Villanúa MÁ, López-Calderón A. Systemic α-melanocyte-stimulating hormone administration decreases arthritis-induced anorexia and muscle wasting. Am J Physiol Regul Integr Comp Physiol 2013; 304:R877-86. [PMID: 23515620 DOI: 10.1152/ajpregu.00447.2012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Rheumatoid cachexia is associated with rheumatoid arthritis and it increases mortality and morbidity. Adjuvant-induced arthritis is an experimental model of rheumatoid arthritis that causes anorexia and muscle wasting. α-Melanocyte-stimulating hormone (α-MSH) has anti-inflammatory actions, and it is able to decrease inflammation in several inflammatory diseases including experimental arthritis. In this study we tested whether systemic α-MSH treatment is able to ameliorate cachexia in arthritic rats. On day 8 after adjuvant injection control and arthritic rats were treated with α-MSH (50 μg/rat ip) twice a day, until day 16 when all rats were euthanized. Arthritis decreased food intake, but it increased hypothalamic expression of neuropeptide Y (NPY) and Agouti-related peptides (AgRP) as well as interleukin-1β (IL-1β) and cyclooxygenase-2 (COX-2) mRNA. In arthritic rats, α-MSH decreased the external signs of arthritis and increased food intake (P < 0.01). In addition, α-MSH decreased hypothalamic expression of IL-1β, COX-2, proopiomelanocortin, and prohormone-converting (PC) enzymes PC1/3 and PC2 mRNA in arthritic rats. In control rats, α-MSH did not modify food intake or hypothalamic expression of aforementioned mRNA. α-MSH prevented arthritis-induced increase in gastrocnemius COX-2, muscle-specific RING-finger protein-1 (MuRF1), and atrogin-1 expression, and it increased fast myofiber size. In conclusion our data show that in arthritic rats peripheral α-MSH treatment has an anti-cachectic action increasing food intake and decreasing muscle wasting.
Collapse
|
11
|
Sarsu SB, Ozokutan BH, Tarakcioglu M, Sarı I, Bağcı C. Effects of Leptin on Intestinal Ischemia-Reperfusion Injury. Indian J Surg 2013; 77:351-5. [PMID: 26730024 DOI: 10.1007/s12262-013-0836-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 01/16/2013] [Indexed: 10/27/2022] Open
Abstract
Many clinical conditions such as shock, sepsis, mesenteric thrombosis, necrotizing enterocolitis, and bowel transplantation can cause intestinal ischemia-reperfusion (IR) injury. This study was designed to determine the effects of leptin on intestinal IR injury. Thirty rats were divided into three groups, each containing ten rats: group A (IR group), group B (treatment group), and group C (sham group). After 1 h of intestinal ischemia, the clamp was removed in order to perform reperfusion. In group B, 100 mg/kg leptin was administered subcutaneously 30 min before reperfusion. In groups A and C, 0.1 ml physiologic saline was injected. In group A, serum and tissue nitric oxide (NO) levels were significantly decreased, and malondialdehyde levels were significantly increased compared to sham group (p < 0.05). Histopathologic injury was significantly lower in sham group compared to group A. In group B, serum and tissue malondialdehyde levels were significantly decreased (p < 0.05), but serum and tissue NO levels were significantly increased compared to group A (p < 0.05). Histopathologic injury was significantly lower in group B compared to group A (p < 0.05). The results of the present study demonstrated that leptin decreases intestinal IR injury by increasing NO production, rearranging mucosal blood flow, and inhibiting polymorphonuclear leukocyte infiltration.
Collapse
Affiliation(s)
- Sevgi Buyukbese Sarsu
- Department of Pediatric Surgery, Gaziantep Children's Hospital, 27060 Gaziantep, Turkey ; Ataturk mah. Adnan Inanıcı cad. 1107 nolu sok. Buyukbese Apt. Kat 4. Daire No.8 Sehitkamil, Gaziantep, Turkey
| | - Bulent Hayri Ozokutan
- Department of Pediatric Surgery, Faculty of Medicine, University of Gaziantep, 27310 Gaziantep, Turkey
| | - Mehmet Tarakcioglu
- Department of Biochemistry, Faculty of Medicine, University of Gaziantep, 27310 Gaziantep, Turkey
| | - Ibrahim Sarı
- Department of Pathology, Faculty of Medicine, University of Gaziantep, 27310 Gaziantep, Turkey
| | - Cahit Bağcı
- Department of Physiology, Faculty of Medicine, University of Gaziantep, 27310 Gaziantep, Turkey
| |
Collapse
|
12
|
Balasubramanian P, Jagannathan L, Subramanian M, Gilbreath ET, MohanKumar PS, MohanKumar SM. High fat diet affects reproductive functions in female diet-induced obese and dietary resistant rats. J Neuroendocrinol 2012; 24:748-55. [PMID: 22192683 PMCID: PMC3330165 DOI: 10.1111/j.1365-2826.2011.02276.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The incidence of ovulatory disorders is common in obese animal models. The mechanism behind this effect is unclear. We hypothesised that a high-fat (HF) diet induces alterations in neuroendocrine mechanisms resulting in anovulation in diet-induced obese (DIO) animals. Adult female DIO and diet-resistant (DR) rats were fed either chow or a HF diet (45% calories from fat) for 6 weeks. Oestrous cyclicity and body weight were monitored regularly. At the end of treatment, rats were implanted with a jugular catheter to monitor luteinising hormone (LH) levels on the day of pro-oestrous. Rats were sacrificed on the next pro-oestrous, and their brains and ovaries were collected. Plasma from trunk blood was analysed for oestradiol and leptin concentrations. Ovaries were fixed and sectioned for histological analysis. Brains were removed, frozen and sectioned, and norepinephrine (NE) concentrations in discrete hypothalamic areas were measured using high-performance liquid chromatography with electrochemical detection. A HF diet exposure affected oestrous cyclicity in both DIO and DR rats, with the effect being more pronounced in DIO animals. HF diet exposure increased leptin levels in both DIO and DR rats. Oestradiol levels were low in the DIO-HF group. NE levels in the hypothalamus were unaffected by HF diet or genotype. A normal LH surge was observed in DR-Chow rats and LH levels were low in the remaining groups. These results lead to the conclusion that DIO rats have an inherently reduced reproductive capacity and exposure to a HF diet decreases it further. A reduction in oestradiol and LH surge levels could contribute to this effect; however, the underlying mechanisms need to be investigated further.
Collapse
Affiliation(s)
- Priya Balasubramanian
- Neuroendocrine Research Laboratory, Department of Pharmacology & Toxicology, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - LakshmiKripa Jagannathan
- Neuroendocrine Research Laboratory, Department of Pathobiology & Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - Madhan Subramanian
- Neuroendocrine Research Laboratory, Department of Pathobiology & Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - Ebony T. Gilbreath
- Neuroendocrine Research Laboratory, Department of Pathobiology & Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - P. S. MohanKumar
- Neuroendocrine Research Laboratory, Department of Pathobiology & Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - Sheba M.J. MohanKumar
- Neuroendocrine Research Laboratory, Department of Pharmacology & Toxicology, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| |
Collapse
|
13
|
MacDonald L, Radler M, Paolini AG, Kent S. Calorie restriction attenuates LPS-induced sickness behavior and shifts hypothalamic signaling pathways to an anti-inflammatory bias. Am J Physiol Regul Integr Comp Physiol 2011; 301:R172-84. [PMID: 21525175 DOI: 10.1152/ajpregu.00057.2011] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Calorie restriction (CR) has been demonstrated to alter cytokine levels; however, its potential to modify sickness behavior (fever, anorexia, cachexia) has not. The effect of CR on sickness behavior was examined in male C57BL/6J mice fed ad libitum or restricted 25% (CR25%) or restricted 50% (CR50%) in food intake for 28 days and injected with 50 μg/kg of LPS on day 29. Changes in body temperature, locomotor activity, body weight, and food intake were determined. A separate cohort of mice were fed ad libitum or CR50% for 28 days, and hypothalamic mRNA expression of inhibitory factor κB-α (IκB-α), cyclooxygenase-2 (COX-2), prostaglandin E(2) (PGE(2)), suppressor of cytokine signaling 3 (SOCS3), IL-10, neuropeptide Y (NPY), leptin, proopiomelanocortin (POMC), and corticotrophin-releasing hormone (CRH) were determined at 0, 2, and 4 h post-LPS. CR50% mice did not develop fevers, whereas the CR25% mice displayed a fever shorter in duration but with the same peak as the controls. Both CR25% and CR50% mice showed no sign of anorexia and reduced cachexia after LPS administration. Hypothalamic mRNA expression of NPY and CRH were both increased by severalfold in CR50% animals preinjection compared with controls. The CR50% mice did not demonstrate the expected rise in hypothalamic mRNA expression of COX-2, microsomal prostaglandin E synthase-1, POMC, or CRH 2 h post-LPS, and leptin expression was decreased at this time point. Increases in SOCS3, IL-10, and IκB-α expression in CR50% animals were enhanced compared with ad libitum-fed controls at 4 h post-LPS. CR results in a suppression of sickness behavior in a dose-dependent manner, which may be due to CR attenuating proinflammatory pathways and enhancing anti-inflammatory pathways.
Collapse
Affiliation(s)
- Leah MacDonald
- School of Psychological Science, La Trobe University, Bundoora, VIC 3086, Australia
| | | | | | | |
Collapse
|
14
|
Abstract
Consumption of DHA has numerous beneficial effects, but little is known about these effects during the first few days of the DHA dietary intake. The main objectives of the present study were to determine the time course of DHA incorporation into phospholipids in different mouse tissues and the effects of DHA supplementation on adiponectin and leptin secretion. Mice were fed either a control diet or a DHA-rich diet, and some were killed on days 0, 4, 8, 16 and 32. Some mice were fed the DHA-rich diet for 16 d, and were then maintained on the control diet for sixteen more days (washout period). DHA supplementation increased plasma adiponectin secretion by 2·4-fold as early as 4 d after the initiation of the DHA-rich diet feeding. The adiponectin concentration remained 1·6-fold higher after the 16 d washout period. Plasma leptin levels were significantly lower after 4 d of feeding with DHA. These effects were associated with a significant increase in DHA incorporation in phosphatidylethanolamine and phosphatidylcholine of all analysed tissues (liver, heart and white adipose tissues). DHA mainly got incorporated at the expense of n-6 arachidonic acid. The present data show that DHA rapidly improved the profile of secreted adipokines, and that these protective effects were long lasting.
Collapse
|
15
|
Grinevich V, Kolleker A, Eliava M, Takada N, Takuma H, Fukazawa Y, Shigemoto R, Kuhl D, Waters J, Seeburg PH, Osten P. Fluorescent Arc/Arg3.1 indicator mice: a versatile tool to study brain activity changes in vitro and in vivo. J Neurosci Methods 2009; 184:25-36. [PMID: 19628007 DOI: 10.1016/j.jneumeth.2009.07.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Revised: 07/08/2009] [Accepted: 07/09/2009] [Indexed: 01/13/2023]
Abstract
The brain-specific immediate early gene Arc/Arg3.1 is induced in response to a variety of stimuli, including sensory and behavior-linked neural activity. Here we report the generation of transgenic mice, termed TgArc/Arg3.1-d4EGFP, expressing a 4-h half-life form of enhanced green fluorescent protein (d4EGFP) under the control of the Arc/Arg3.1 promoter. We show that d4EGFP-mediated fluorescence faithfully reports Arc/Arg3.1 induction in response to physiological, pathological and pharmacological stimuli, and that this fluorescence permits electrical recording from activated neurons in the live mouse. Moreover, the fluorescent Arc/Arg3.1 indicator revealed activity changes in circumscribed brain areas in distinct modes of stress and in a mouse model of Alzheimer's disease. These findings identify the TgArc/Arg3.1-d4EGFP mouse as a versatile tool to monitor Arc/Arg3.1 induction in neural circuits, both in vitro and in vivo.
Collapse
Affiliation(s)
- Valery Grinevich
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Molecular Neurobiology, Max-Planck-Institute for Medical Research, D-69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Yeh SS, Blackwood K, Schuster MW. The cytokine basis of cachexia and its treatment: are they ready for prime time? J Am Med Dir Assoc 2008; 9:219-36. [PMID: 18457797 DOI: 10.1016/j.jamda.2008.01.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Accepted: 01/04/2008] [Indexed: 01/14/2023]
Abstract
Cachexia is a hypercatabolic condition that is often associated with the terminal stages of many diseases, in which the patient's resting metabolic rate is high and loss of muscle and fat tissue mass occur at an alarming rate. The patient also usually has concurrent anorexia, amplifying the wasting syndrome that is cachexia. The greater the extent of cachexia (regardless of underlying disease), the worse the prognosis. Efforts to treat cachexia over the years have fallen short of satisfactorily reversing the wasting syndrome. This article reviews the pathophysiology of cachexia, enumerating the different pro-inflammatory cytokines that contribute to the syndrome and attempting to illustrate their interwoven pathways. We also review the different treatments that have been explored, as well as the recent literature addressing the use of anti-cytokine therapy to treat cachexia.
Collapse
|
17
|
Becskei C, Riediger T, Hernádfalvy N, Arsenijevic D, Lutz TA, Langhans W. Inhibitory effects of lipopolysaccharide on hypothalamic nuclei implicated in the control of food intake. Brain Behav Immun 2008; 22:56-64. [PMID: 17624718 DOI: 10.1016/j.bbi.2007.06.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Revised: 06/01/2007] [Accepted: 06/02/2007] [Indexed: 10/23/2022] Open
Abstract
The arcuate nucleus (Arc) and the lateral hypothalamic area (LHA), two key hypothalamic nuclei regulating feeding behavior, express c-Fos, a marker of neuronal activation in fasted animals. This is reversed by refeeding. In the present study we tested whether an anorectic dose of lipopolysaccharide (LPS), the cell wall component of Gram-negative bacteria, also inhibits fasting-induced c-Fos expression in these hypothalamic nuclei. This would suggest that they are involved in anorexia during bacterial infections as well. We also studied whether LPS modulates the activity of orexin-A positive (OX+) LHA neurons. Food deprived BALB/c mice were injected with LPS or saline and were sacrificed 4 or 6h later. Four hours after injection, LPS reduced the number of c-Fos positive cells in the Arc and in the LHA, but had no effect on c-Fos in OX+ neurons. Six hours after injection, LPS reduced c-Fos expression in the LHA, both in the OX- and OX+ neurons, but not in the Arc. These results show that LPS modulates neuronal activity in the Arc and LHA similar to feeding-related stimuli, suggesting that the observed effects might contribute to the anorectic effect of LPS. Thus, physiological satiety signals released during refeeding and anorexia during bacterial infection seem to engage similar neuronal substrates.
Collapse
Affiliation(s)
- Csilla Becskei
- Institute of Veterinary Physiology and Zurich Centre of Human Integrative Physiology, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland.
| | | | | | | | | | | |
Collapse
|
18
|
Harada N, Kusuyama A, Morishima M, Okada K, Takahashi A, Nakaya Y. Bezafibrate improves bacterial lipopolysaccharide-induced dyslipidemia and anorexia in rats. Metabolism 2007; 56:517-22. [PMID: 17379010 DOI: 10.1016/j.metabol.2006.11.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2006] [Accepted: 11/06/2006] [Indexed: 11/24/2022]
Abstract
Bacterial endotoxin/lipopolysaccharide (LPS)-induced cachexia is characterized by weight loss, anorexia, and a disturbance in lipid metabolism, namely, hypertriacylglycerolemia. The aim of this study in rats with acute endotoxicity induced by an injection of LPS was to investigate whether bezafibrate, a ligand for peroxisome proliferator-activated receptor alpha and a lipoprotein lipase (LPL) activator, improved cachectic conditions, including impaired lipid metabolism. Short-term administration of LPS in the rats resulted in impairment of triacylglycerol clearance in plasma after the intake of fresh cream. In addition, LPS increased whole-body energy expenditure, reduced fasting body weight and caused anorexia in the rats. Bezafibrate treatment resulted in significant improvements in LPS-induced dyslipidemia and anorexia, but had no effect on energy expenditure, respiratory quotient, or fasting body weight in the endotoxic rats. Administration of LPS was also associated with a decrease in the level of messenger RNA (mRNA) expression for LPL in white adipose tissue and skeletal muscle and an increase in the mRNA levels for uncoupling protein 3 in skeletal muscle. Bezafibrate treatment reversed the decline in LPL mRNA levels in white adipose tissue but not in the skeletal muscle tissue of the rats. The enhanced uncoupling protein 3 mRNA level in the endotoxic rats was not affected by bezafibrate treatment. Plasma concentration of leptin was increased by short-term LPS treatment. Bezafibrate decreased the level of plasma leptin significantly without affecting the level of leptin mRNA expression. These results suggest that bezafibrate may be an effective drug not only for impaired triacylglycerol metabolism, but also for anorexia in cachectic states induced by bacterial infections.
Collapse
Affiliation(s)
- Nagakatsu Harada
- Department of Nutrition and Metabolism, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima City, 770-8503, Japan.
| | | | | | | | | | | |
Collapse
|
19
|
Kim YW, Kim KH, Ahn DK, Kim HS, Kim JY, Lee DC, Park SY. Time-Course Changes of Hormones and Cytokines by Lipopolysaccharide and Its Relation with Anorexia. J Physiol Sci 2007; 57:159-65. [PMID: 17481364 DOI: 10.2170/physiolsci.rp003407] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Accepted: 05/05/2007] [Indexed: 11/05/2022]
Abstract
We assessed the time course effects of lipopolysaccaride (LPS) on food intake, cytokines, and hormones in rats and evaluated the relation between LPS-induced anorexia and its possible causative factors. Food intake was reduced 2 h after LPS injection (500 microg/kg, intraperitoneally) and remained decreased for 24 h. Plasma TNF-alpha and IL-6 levels increased by LPS administration at 0.5 and 2 h, and at 2 and 4 h, respectively. Plasma leptin and glucose levels were elevated at 8 and 16 h, and insulin levels were elevated at 2, 4, 8, and 16 h in the LPS-injected group, as compared to the counterpart controls. IL-6 levels in the CSF were elevated at 2 and 4 h. Hypothalamic cytokines tended to increase as early as 0.5 h after LPS injection and remained increased until 16 h. LPS-induced anorexia was attenuated in insulin-deficient STZ rats and was abolished by insulin treatment. The hypothalamic expression of NPY, a target of insulin's anorexic effect, was decreased 2 h after LPS administration, and central NPY injection (3 nM) prevented LPS-induced anorexia. In conclusion, cytokines, insulin, and leptin levels evidence different time courses by LPS administration. In LPS-induced anorexia, insulin may constitute a newly found causative factor, whereas leptin appears to be uninvolved in an early period in rats.
Collapse
Affiliation(s)
- Yong-Woon Kim
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Korea
| | | | | | | | | | | | | |
Collapse
|
20
|
Steiner AA, Romanovsky AA. Leptin: at the crossroads of energy balance and systemic inflammation. Prog Lipid Res 2006; 46:89-107. [PMID: 17275915 PMCID: PMC1976277 DOI: 10.1016/j.plipres.2006.11.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2006] [Revised: 11/10/2006] [Accepted: 11/16/2006] [Indexed: 11/21/2022]
Abstract
In addition to playing a central role in energy homeostasis, leptin is also an important player in the inflammatory response. Systemic inflammation is accompanied by fever (less severe cases) or hypothermia (more severe cases). In leptin-irresponsive mutants, the hypothermia of systemic inflammation is exaggerated, presumably due to the enhanced production and cryogenic action of tumor necrosis factor (TNF)-alpha. Mechanisms that exaggerate hypothermia can also attenuate fever, particularly in a cool environment. Another common manifestation of systemic inflammation is behavioral depression. Along with the production of interleukin (IL)-1beta, this manifestation is exaggerated in leptin-irresponsive mutants. The enhanced production of TNF-alpha and IL-1beta may be due, at least in part, to insufficient activation of the anti-inflammatory hypothalamo-pituitary-adrenal axis by immune stimuli in the absence of leptin signaling. In experimental animals and humans that are responsive to leptin, suppression of leptin production under conditions of negative energy balance (e.g., fasting) can exaggerate both hypothermia and behavioral depression. Since these manifestations aid energy conservation, exaggeration of these manifestations under conditions of negative energy balance is likely to be beneficial.
Collapse
Affiliation(s)
- Alexandre A Steiner
- Systemic Inflammation Laboratory, Trauma Research, St Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | | |
Collapse
|
21
|
Vélez ML, Costamagna E, Kimura ET, Fozzatti L, Pellizas CG, Montesinos MM, Lucero AM, Coleoni AH, Santisteban P, Masini-Repiso AM. Bacterial lipopolysaccharide stimulates the thyrotropin-dependent thyroglobulin gene expression at the transcriptional level by involving the transcription factors thyroid transcription factor-1 and paired box domain transcription factor 8. Endocrinology 2006; 147:3260-75. [PMID: 16627577 DOI: 10.1210/en.2005-0789] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The bacterial lipopolysaccharide (LPS) is a biological activator that induces expression of multiple genes in several cell types. LPS has been proposed as an etiopathogenic agent in autoimmune diseases. However, whether LPS affects the expression of autoantigens has not been explored. Thyroglobulin (TG) is a key protein in thyroid hormonogenesis and one of the major thyroid autoantigens. This study aimed to analyze the action of LPS on TG gene expression in Fisher rat thyroid cell line FRTL-5 thyroid cells. We demonstrate that LPS increases the TSH-induced TG protein and mRNA level. Evidence that the effect of LPS is exerted at the transcriptional level was obtained by transfecting the minimal TG promoter. The C element of the TG promoter, which contains sequences for paired box domain transcription factor 8 (Pax8) and thyroid transcription factor (TTF)-1 binding, is essential for full TG promoter expression under TSH stimulation. The transcriptional activity of a construct containing five tandem repeats of the C site is increased by LPS, indicating a possible involvement of the C site in the LPS-induced TG gene transcription. We demonstrate that the TG promoter mutated at the Pax8 or TTF-1 binding element in the C site does not respond to LPS. In band shift assays, binding of Pax8 and TTF-1 to the C site is increased by LPS. The Pax8 and TTF-1 mRNA and protein levels are augmented by LPS. The half-lives of TG, Pax8, and TTF-1 are increased in endotoxin-treated cells. Our results reveal the ability of LPS to stimulate the expression of TG, a finding of potential pathophysiological implication.
Collapse
Affiliation(s)
- María L Vélez
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Córdoba, 5000 Córdoba, Argentina
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Ozer C, Gülen S, Dileköz E, Babül A, Ercan ZS. The effect of systemic leptin administration on aorta smooth muscle responses in diabetic rats. Mol Cell Biochem 2006; 282:187-91. [PMID: 16317526 DOI: 10.1007/s11010-006-1927-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Accepted: 08/09/2005] [Indexed: 10/25/2022]
Abstract
Leptin produces effects in central nervous system and peripheral tissues via its specific receptors. Leptin also stimulates nitric oxide release in a concentration-dependent manner. In this study, our aim was to test the hypothesis that whether leptin has a modulatory role on endothelium or smooth muscle function in streptozotocin (STZ)-induced diabetic rats. Wistar-Albino rats were divided into four groups: 1 -- Control, 2 -- Diabetic, 3 -- Control + leptin and 4 -- Diabetic + leptin. Experimental diabetes was produced by intraperitoneal injection of a single dose of STZ (55 mg/kg). Diabetes was determined by increased fasting blood glucose level on the 7th day of the experiment. Leptin (0.1 mg/kg/day) was administered intraperitoneally for 5 days. At the end of the 5th day, thoracic aortas were isolated and phenylephrine (Phe)-induced contractions and acetylcholine (ACh)-induced relaxations of each group were estimated. In diabetic rats, Phe-induced contractility was increased (p < 0.05). Leptin pre-treatment increased the Phe-induced contractility significantly in aortic rings obtained from diabetic rats (p < 0.05). In normal rats, leptin administration produced only a slight and non-significant increase in Phe-induced contractions. Although the relaxant responses were decreased in diabetic rats, leptin administration enhanced the ACh-induced relaxation in both normal and diabetic animals significantly. As a conclusion; chronic leptin pre-treatment caused a significant increase both in Phe-induced contractions and ACh-induced Endothelial-Derived Relaxing Factor (EDRF)/Nitric oxide-mediated relaxations in the aortic rings isolated from streptozotocin-induced diabetic rats. This peptide hormone caused a significant increase in the relaxations obtained by ACh while not inducing a significant alteration in the contractile effect of Phe in control rats.
Collapse
MESH Headings
- Acetylcholine/pharmacology
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/physiopathology
- Diabetes Mellitus, Experimental/physiopathology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/physiopathology
- Endothelium-Dependent Relaxing Factors/metabolism
- In Vitro Techniques
- Leptin/administration & dosage
- Leptin/pharmacology
- Male
- Muscle Contraction/drug effects
- Muscle Contraction/physiology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiopathology
- Nitric Oxide/metabolism
- Phenylephrine/pharmacology
- Rats
- Rats, Wistar
- Vasoconstrictor Agents/pharmacology
- Vasodilator Agents/pharmacology
Collapse
Affiliation(s)
- Ciğdem Ozer
- Department of Physiology, Faculty of Medicine, Gazi University, 06510 Beşevler, Ankara, Turkey
| | | | | | | | | |
Collapse
|
23
|
Abstract
Cachexia, usually defined as the loss of >5% of an individual's baseline bodyweight over 2-6 months, occurs with a number of diseases that includes not only AIDS and advanced cancer but also chronic heart failure, rheumatoid arthritis, chronic obstructive pulmonary disease, Crohn disease, and renal failure. Anorexia is considered a key component of the anorexia-cachexia syndrome. Progestogens, particularly megestrol acetate, are commonly used to treat anorexia-cachexia. The mechanism of action of megestrol is believed to involve stimulation of appetite by both direct and indirect pathways and antagonism of the metabolic effects of the principal catabolic cytokines. Because the bioavailability of megestrol acetate directly affects its efficacy and safety, the formulation was refined to enhance its pharmacokinetics. Such efforts yielded megestrol acetate in a tablet form, followed by a concentrated oral suspension form, and an oral suspension form developed using nanocrystal technology. Nanocrystal technology was designed specifically to optimize drug delivery and enhance the bioavailability of drugs that have poor solubility in water. Megestrol acetate nanocrystal oral suspension is currently under review by the US FDA for the treatment of cachexia in patients with AIDS. Preclinical pharmacokinetic data suggest that the new megestrol acetate formulation has the potential to significantly shorten the time to clinical response and thus may improve outcomes in patients with anorexia-cachexia.
Collapse
Affiliation(s)
- Robert A Femia
- Scientific and Regulatory Affairs, Par Pharmaceutical, Inc., Spring Valley, New York 10977, USA.
| | | |
Collapse
|
24
|
Lugarini F, Hrupka BJ, Schwartz GJ, Plata-Salaman CR, Langhans W. Acute and chronic administration of immunomodulators induces anorexia in Zucker rats. Physiol Behav 2004; 84:165-73. [PMID: 15642620 DOI: 10.1016/j.physbeh.2004.11.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2004] [Revised: 11/03/2004] [Accepted: 11/08/2004] [Indexed: 11/20/2022]
Abstract
To investigate the possible involvement of leptin signaling in lipopolysaccharide (LPS) anorexia, we compared the anorectic effect of LPS in genetically obese (fa/fa) Zucker rats and in their lean (Fa/?) counterparts. The effects of interleukin-1beta (IL-1beta) and muramyl dipeptide (MDP) were also tested. LPS [100 microg/kg body weight (BW)], IL-1beta (2 microg/kg BW) and MDP (2.2 mg/kg BW) injected intraperitoneally (i.p.) at lights out reduced food intake similarly in obese and lean rats. LPS injection at 500 or 1000 microg/kg BW (i.p.) also reduced food intake and BW similarly in obese and lean rats, but obese regained BW faster than lean rats. LPS (2.45 microg or 9.8 microg/h/rat) administered chronically with i.p. implanted osmotic pumps reduced food intake similarly on experimental day 1, regardless of the genotype. After day 3, the lean rats' anorectic response and recovery were dose-dependent, whereas the anorectic response in obese rats was minimally affected by dose (significant dose effect only on day 3). Again, obese rats regained lost BW faster than lean rats. These results do not support a role for leptin as the sole mediator of anorexia induced by bacterial products (LPS and MDP) and IL-1beta.
Collapse
Affiliation(s)
- F Lugarini
- Institute of Animal Sciences, Physiology and Animal Husbandry, Swiss Federal Institute of Technology, Schorenstrasse 16, Postfach, 8603 Schwerzenbach, Switzerland
| | | | | | | | | |
Collapse
|
25
|
Lin J, Yan GT, Wang LH, Hao XH, Zhang K, Xue H. Leptin fluctuates in intestinal ischemia-reperfusion injury as inflammatory cytokine. Peptides 2004; 25:2187-93. [PMID: 15572209 DOI: 10.1016/j.peptides.2004.08.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2004] [Revised: 08/07/2004] [Accepted: 08/09/2004] [Indexed: 01/02/2023]
Abstract
As leptin is an active mediator mainly secreted by adipose tissue and is closely related with energy metabolism, we evaluate both the changes of leptin levels in serum and adipose tissue with a concise radioimmunoassay and the changes of leptin mRNA expression in adipose tissue with RT-PCR, during the severe metabolic impediment in rat intestinal ischemia-reperfusion (I/R) injury. Results show that not only leptin levels in serum and adipose tissue but also its mRNA expression in adipose tissue undergo a fluctuation according to different injury times. Therefore, we conclude that leptin has a time-dependent response to acute inflammatory stimuli and acts as an anti-inflammatory cytokine.
Collapse
Affiliation(s)
- Ji Lin
- Research Laboratory of Biochemistry, Basic Medical Institute, General Hospital of PLA, 28 Fuxing Road, Beijing 100853, PR China
| | | | | | | | | | | |
Collapse
|
26
|
García-Suárez C, Crespo J, Fernández-Gil PL, Amado JA, García-Unzueta MT, Pons Romero F. [Plasma leptin levels in patients with primary biliary cirrhosis and their relationship with degree of fibrosis]. GASTROENTEROLOGIA Y HEPATOLOGIA 2004; 27:47-50. [PMID: 14733878 DOI: 10.1016/s0210-5705(03)79085-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVES a) To analyze plasma leptin levels in patients with primary biliary cirrhosis (PBC) and b) to investigate the relationship between leptin levels and liver fibrosis stage in a cohort of patients with PBC. PATIENTS AND METHODS Serum leptin levels were evaluated through radioimmunoassay in 30 patients with PBC (mean age: 37.2 +/- 11.0 years; range:19-75) and in 29 controls matched for age and weight. Venous blood obtained after a 12-hour fast was centrifuged in EDTA tubes. Weight, height and body mass index (BMI) were measured using standard methods. Hepatitis C virus RNA was determined using qualitative and quantitative polymerase chain reaction. In all patients liver biopsies were performed and the degree of fibrosis and extent of inflammatory infiltrate were evaluated. RESULTS Plasma leptin levels in patients with PBC were lower than those obtained in control subjects (p<0.0001). No significant differences were found between the two groups in age, weight, height, BMI or body fat index. There was a clear increase in serum leptin levels according to histological stage of PBC (stage I: 2.1 ng/ml; stage II: 4.3 ng/ml; stage III: 5.3 ng/ml; stage IV: 12.1 ng/ml; p<0.01) CONCLUSIONS The present study demonstrates the correlation between leptin and stage of liver fibrosis in a cohort of patients with PBC, providing further evidence of the involvement of leptin in the process of liver fibrosis.
Collapse
Affiliation(s)
- C García-Suárez
- Servicio de Aparato Digestivo, Hospital Universitario Valdecilla, Universidad de Cantabria, Santander, España
| | | | | | | | | | | |
Collapse
|
27
|
Rummel C, Hübschle T, Gerstberger R, Roth J. Nuclear translocation of the transcription factor STAT3 in the guinea pig brain during systemic or localized inflammation. J Physiol 2004; 557:671-87. [PMID: 14966301 PMCID: PMC1665088 DOI: 10.1113/jphysiol.2003.058834] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The purpose of the present study was to investigate a possible lipopolysaccharide (LPS)-induced activation of brain cells that is mediated by the pleiotropic cytokine interleukin-6 (IL-6) and its transcription factor STAT3 during systemic or localized inflammation. In guinea pigs, intra-arterial (i.a., 10 microg x kg(-1)) or intraperitoneal (i.p., 30 microg x kg(-1)) injections of bacterial LPS cause a systemic inflammatory response which is accompanied by a robust fever. A febrile response can also be induced by administration of LPS into artificial subcutaneously implanted Teflon chambers (s.c. 100 or 10 microg x kg(-1)), which reflects an experimental model that mimics local tissue inflammation. Baseline plasma levels of bioactive IL-6 determined 60 min prior to injections of LPS or vehicle amounted to 35-80 international units (i.u.) ml(-1). Within 90 min of LPS injection, plasma IL-6 rose about 1000-fold in the groups injected i.a. or i.p., about 50-fold in the group injected s.c. with 100 microg x kg(-1) LPS, and only 5-fold in guinea pigs injected with the lower dose of LPS (10 microg x kg(-1)). At this time point, a distinct nuclear translocation pattern of the transcription factor STAT3 became evident in several brain structures. Amongst those, the sensory circumventricular organs known to lack a tight blood-brain barrier such as the area postrema, the vascular organ of the lamina terminalis and the subfornical organ, as well as the hypothalamic supraoptic nucleus showed intense nuclear STAT3 signals in the i.a. or i.p. injected groups. In contrast a moderate (s.c. group, 100 microg x kg(-1)), or even no (s.c. group, 10 microg x kg(-1)), nuclear STAT3 translocation occurred in response to s.c. injections of LPS. These results suggest that STAT3-mediated genomic activation of target gene transcription in brain cells occurred only in those cases in which sufficiently high concentrations of circulating IL-6 were formed during systemic (i.a. and i.p. groups) or localized (s.c. group, 100 microg x kg(-1)) inflammation.
Collapse
Affiliation(s)
- Christoph Rummel
- Institut für Veterinär-Physiologie, Justus-Liebig-Universität Giessen, Frankfurter Strasse 100, 35392 Giessen, Germany
| | | | | | | |
Collapse
|
28
|
Gaigé S, Abou E, Abysique A, Bouvier M. Effects of interactions between interleukin-1 beta and leptin on cat intestinal vagal mechanoreceptors. J Physiol 2003; 555:297-310. [PMID: 14645453 PMCID: PMC1664812 DOI: 10.1113/jphysiol.2003.054379] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In a previous study, we established that leptin acts on chemosensitive intestinal vagal mechanoreceptors and that its excitatory effects are blocked by the endogenous interleukin-1beta receptor antagonist (Il-1ra). To determine how interleukin-1beta (Il-1beta) is involved in the action of leptin, we studied the effects of this drug on the single vagal afferent activities of intestinal mechanoreceptors in anaesthetized cats. For this purpose, the activity of 34 intestinal vagal mechanoreceptors was recorded via glass microelectrodes implanted in the nodose ganglion. Il-1beta (1 microg) administered into the artery irrigating the upper part of the intestine activated both the 16 leptin-activated units (type 1 units; P < 0.01) and the 12 leptin-inhibited units (type 2 units; P < 0.001), but had no effect on the six leptin-insensitive units. Cholecystokinin (CCK, 10 microg) induced an activatory response only in the two types of Il-1beta-sensitive units. When Il-1beta was administered after CCK, its excitatory effects on type 1 units were enhanced, whereas the excitatory effects on type 2 units were abolished. Pre-treatment with Il-1ra (250 microg) blocked all the effects of Il-1beta and the excitatory effects of leptin on type 1 units, whereas it enhanced the inhibitory effects of leptin on type 2 units. It can therefore be concluded that (i) leptin acts on intestinal vagal mechanoreceptors via Il-1beta in the case of the type 1 units and independently of Il-1beta in the case of the type 2 units, and (ii) type 1 and type 2 units belong to two different populations of vagal afferents that transmit different information about ingestion or inflammation to the CNS, depending on the chemical environment.
Collapse
Affiliation(s)
- Stéphanie Gaigé
- Laboratoire de Physiologie Neurovégétative (UMR CNRS 6153, UMR INRA 1147), Faculté des Sciences et Techniques Saint-Jérôme, Université Aix-Marseille 3, Cases postales 351-352, Avenue Escadrille Normandie Niemen, 13397 Marseille Cedex 20, France
| | | | | | | |
Collapse
|
29
|
Xiao E, Xia-Zhang L, Vulliémoz NR, Ferin M, Wardlaw SL. Leptin modulates inflammatory cytokine and neuroendocrine responses to endotoxin in the primate. Endocrinology 2003; 144:4350-3. [PMID: 12959996 DOI: 10.1210/en.2003-0532] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Leptin, which plays a crucial role in regulating energy balance, can also modulate the inflammatory response. Although leptin-deficient rodents are more sensitive to the toxic effects of bacterial endotoxin, it is unknown if leptin can modulate inflammatory cytokine or neuroendocrine responses to inflammation in a primate model. We have therefore studied the effects of leptin on plasma cytokine and hypothalamic-pituitary-adrenal responses to endotoxin (5 microg iv) in nine ovariectomized rhesus monkeys. Human leptin (50 microg/h) or saline was infused iv for 16 h before and 4 h after endotoxin injection; mean plasma leptin increased from 3.6 +/- 1.0 ng/ml to 18 +/- 1.7 ng/ml (P < 0.001). Leptin infusion had no effect on baseline plasma cytokine and hormone levels before endotoxin injection. As expected, endotoxin stimulated TNF-alpha, IL-6, IL-1 receptor antagonist (IL-1ra), ACTH, and cortisol in the saline-infused animals (P < 0.001). There was a significant attenuation of the IL-6 (P < 0.005) and cortisol (P < 0.001) responses (repeated measures ANOVA) to endotoxin in the leptin-infused animals. There was a significant reduction (by paired analysis) in the responses of the leptin compared with saline-treated animals: 47% for TNF-alpha, 48% for IL-6, 30% for IL1ra, 42% for ACTH, and 22% for cortisol (P < 0.05). We conclude that an increase in circulating leptin, within the physiological range of our monkey colony, can blunt the inflammatory cytokine and hypothalamic-pituitary-adrenal responses to an inflammatory challenge. These results, coupled with our recent finding that endotoxin stimulates leptin release in the monkey, demonstrate that leptin can be both released in response to inflammatory cytokines and act to attenuate the responses to these cytokines.
Collapse
Affiliation(s)
- Ennian Xiao
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York 10032, USA
| | | | | | | | | |
Collapse
|
30
|
Landman RE, Puder JJ, Xiao E, Freda PU, Ferin M, Wardlaw SL. Endotoxin stimulates leptin in the human and nonhuman primate. J Clin Endocrinol Metab 2003; 88:1285-91. [PMID: 12629120 DOI: 10.1210/jc.2002-021393] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Leptin, which plays a key role in regulating energy homeostasis, may also modulate the inflammatory response. An inflammatory challenge with endotoxin has been shown to stimulate leptin release in the rodent. This finding has not been reproduced in humans or in nonhuman primates, although leptin levels have been reported to increase in septic patients. We have therefore examined the effects of endotoxin injection on plasma leptin levels in nine ovariectomized monkeys and four postmenopausal women. In an initial study in five monkeys, mean leptin levels did not increase during the first 5 h after endotoxin treatment, but did increase significantly from 6.4 +/- 2.1 ng/ml at baseline to 12.3 +/- 4.4 ng/ml at 24 h (P = 0.043). In a second study, a significant increase in leptin over time was noted after endotoxin treatment (P < 0.001); leptin release during the 16- to 24-h period after endotoxin injection was 48% higher than during the control period (P = 0.043). A similar stimulatory effect of endotoxin on leptin was observed when monkeys received estradiol replacement. In a third study, repeated injections of endotoxin over a 3-d period stimulated IL-6, ACTH, cortisol, and leptin release (P < 0.001). Leptin increased during the first day of treatment in all animals, but only monkeys with baseline plasma leptin levels greater than 10 ng/ml exhibited a sustained increase in leptin throughout the 3-d period. There was a significant correlation (r = 0.81; P = 0.008) between the mean baseline leptin level and the percent increase in leptin over baseline on the last day of treatment. In the human subjects, plasma leptin concentrations did not change significantly during the 7-h period after endotoxin injection. However, leptin increased in all four women from a mean baseline of 8.34 +/- 3.1 to 13.1 +/- 4.3 ng/ml 24 h after endotoxin (P = 0.038). In summary, endotoxin stimulates the release of leptin into peripheral blood in the human and nonhuman primate, but the time course is different from that reported in the rodent. These results are consistent with previous reports of increased blood leptin levels in patients with sepsis. The significance of these findings and the potential role of leptin in modulating the response to inflammation in the human require further study.
Collapse
Affiliation(s)
- Rita E Landman
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | | | | | | | | | | |
Collapse
|
31
|
Caldefie-Chézet F, Guillot J, Vasson MP. La leptine : hormone et cytokine impliquée dans la réponse à l’agression. NUTR CLIN METAB 2003. [DOI: 10.1016/s0985-0562(03)00007-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
32
|
Abstract
In a previous study, we established that leptin controls food intake and immune responses by acting on intestinal vagal chemosensitive mechanoreceptors via a functional link with interleukin-1 beta (Il-1 beta). Since the control of intestinal motility is one of the main roles of the vagal afferent fibres, we investigated the effects of leptin on intestinal electromyographic (EMG) activity which reflects intestinal motility. For this purpose, the effects of locally injected leptin on small intestine spontaneous EMG activity were studied in 23 anaesthetised cats. The EMG activity was recorded using bipolar electrodes implanted in the proximal small intestine. Leptin and Il-1 beta (0.1, 1 and 10 microg), administered through the artery irrigating the upper part of the intestine 20 min after cholecystokinin (CCK, 10 microg, I.A.), had significant (P < 0.001) excitatory effects on intestinal EMG activity. The effects of both substances were blocked by the endogenous interleukin-1 beta receptor antagonist (Il-1ra, 250 microg, I.A.), by atropine (250 microg, I.A.) and by vagotomy. In the absence of CCK, leptin and Il-1 beta had no effect on intestinal electrical activity. It can therefore be concluded that: (1) leptin is effective only after the previous intervention of CCK, (2) the enhancement of the electrical activity induced by leptin involves Il-1 beta receptors and the cholinergic excitatory pathway, (3) the modes whereby the leptin-induced enhancement of EMG activity occurs strongly suggest that these effects are due to a long-loop reflex involving intestinal vagal afferent fibres and the parasympathetic nervous system.
Collapse
Affiliation(s)
- Stéphanie Gaigé
- Laboratoire de Physiologie Neurovégétative, UMR CNRS 6153, UMR INRA, Faculté des Sciences et Techniques Saint-Jérôme, Université Aix-Marseille 3, Case postale 351-352, Avenue Escadrille Normandie Niemen, 13397 Marseille Cedex 20, France
| | | | | |
Collapse
|
33
|
Coyne CP, Howell T, Baravik J, Baravik E, Willetto C, Fenwick BW. Biochemical entities that influence membrane-associated TNF RII (80-kDa) and IL-1 RI (80-kDa) complex expression and receptor fragment production in adherent vascular endothelium. PATHOPHYSIOLOGY 2003; 9:115-125. [PMID: 14567944 DOI: 10.1016/s0928-4680(02)00056-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The research aim of the present investigation was to identify leukocyte enzyme-proteases that have the capacity to biochemically recruit the passive participation of vascular endothelium in cytokine receptor 'shedding' phenomenon involving membrane-associated TNF RII (80-kDa) and IL-1 RI (80-kDa) complexes. Achieving this research objective involved the design of a laboratory approach that delineated to what extent enzyme-proteases released by activated macrophages directly interact with, and liberate soluble fragments of membrane-associated cytokine receptor complexes. Results from this segment of the investigation revealed that cathepsin-D, a leukocyte carboxyl/aspartate protease, altered the integrity and generated soluble fragments of TNF RII (80-kDa) and IL-1 RI (80-kDa) receptor complexes expressed by vascular endothelium. Furthermore, laboratory findings also suggested that cathepsin-D possessed the ability to variably deplete biologically functional membrane-associated TNF RII (80-kDa) and IL-1 RI (80-kDa) complexes. Complementary investigations isolated a carboxyl/aspartate protease from activated macrophages utilizing pepstatin-A affinity chromatography. Exposure of vascular endothelium to pepstatin-A binding proteins resulted in a detectable depletion of membrane-associated TNF RII (80-kDa) and IL-1 RI (80-kDa) in addition to the generation of soluble receptor fragments. Analysis of macrophage pepstatin-A binding proteins by SDS-PAGE identified a primary fraction with a molecular mass of 47-52-kDa that closely correlated with the known molecular mass of leukocyte cathepsin-D. Evaluation of macrophage pepstatin-A binding-protein fractions by non-denaturing Hb-PAGE detected a lucent proteolytic band at 47-52-kDa compatible with the known molecular mass of leukocyte cathepsin-D. Macrophage pepstatin-A binding proteins also hydrolyzed a synthetic enzyme-specific substrate that selectively recognizes cathepsin-D biochemical activity. In conclusion, the leukocyte carboxyl/aspartate protease, cathepsin-D can biochemically alter the integrity and generate soluble fragments of membrane-associated TNF RII (80-kDa) and IL-1 RI (80-kDa) receptor complexes expressed by vascular endothelium. The relevance of this concept is in part based on investigations that have discovered that genetic 'knock-out' mice incapable of expressing IL-1 RI (80-kDa) or TNF RI (55-kDa) receptor complexes are highly resistant to developing the pathophysiological alterations classically associated with conditions of endotoxic-shock.
Collapse
Affiliation(s)
- C P. Coyne
- The Veterinary Pharmacology Research Laboratory, Department of Basic Science, Veterinary Research Program, College of Veterinary Medicine, Wise Center Drawer V, Mississippi State University, 39762, Mississippi, USA
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
Vagal afferent nerve fibres are involved in the transmission to the central nervous system of information relating to food intake and immune reactions. Leptin is involved in the control of food intake and has specific receptors in afferent vagal neurones. To investigate the role of these receptors, we studied the effects of leptin on single vagal afferent activities from intestinal mechanoreceptors in anaesthetized cats. The activity of 35 intestinal vagal mechanoreceptors was recorded by means of glass microelectrodes implanted in the nodose ganglion. Leptin (10 microg), administered into the artery irrigating the upper part of the intestine, induced activation in 17 units (P < 0.001), inhibition in 8 units (P < 0.05), and had no effect on 10 units. The excitatory effects of leptin were blocked by the endogenous interleukine-1beta receptor antagonist, (Il-1ra, 250 microg, I.A.). Cholecystokinin (CCK, 10 microg, I.A.) induced an activatory response only in the two types of units which were responsive to leptin alone. When leptin was administered after CCK, its excitatory effects were enhanced and its inhibitory effects were blocked, whereas it had no effect on the units which were not affected by leptin alone. The interactions between leptin and CCK are specific ones, since other substances (phenylbiguanide, a serotoninergic agonist; substance P) known to activate the mechanoreceptors did not modify the effects of leptin. These results indicate that leptin appears to play a role in the control of immune responses and food intake via intestinal vagal afferent nerve fibres and that there is a functional link between leptin and Il-1beta.
Collapse
Affiliation(s)
- Stéphanie Gaigé
- Laboratoire de Physiologie Neurovégétative, (UMR CNRS 6153, UMR INRA), Faculté des Sciences et Techniques Saint-Jérôme, Université Aix-Marseille 3, Case postale 351-352, 13397 Marseille Cedex 20, France
| | | | | |
Collapse
|
35
|
Crespo J, Rivero M, Fábrega E, Cayón A, Amado JA, García-Unzeta MT, Pons-Romero F. Plasma leptin and TNF-alpha levels in chronic hepatitis C patients and their relationship to hepatic fibrosis. Dig Dis Sci 2002; 47:1604-10. [PMID: 12141823 DOI: 10.1023/a:1015835606718] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The aim of this study was to examine the possible relationship between the plasma levels of leptin and tumor necrosis factor (TNF)-alpha and the stage of hepatic fibrosis in a cohort of patients with chronic hepatitis C. Leptin and TNF levels were measured by RIA in 135 patients and in 75 age- and sex-matched controls. Liver disease was evaluated by the stage of fibrosis and the extent of inflammatory infiltrate in the liver biopsy. Leptin levels correlated with BMI values in healthy controls and in patients with chronic hepatitis C (men, r = 0.61, P = 0.0001; women, r = 0.68, P = 0.003). Leptin levels increased as hepatic fibrosis stage progressed both in male and in female patients (P < 0.001); also, TNF levels were higher in patients with an advanced stage of fibrosis (P = 0.006). In these patients, levels of leptin increased according to the progression of the stage of fibrosis; these data suggest that leptin may play a role in the regulation of hepatic fibrosis.
Collapse
Affiliation(s)
- Javier Crespo
- Gastroenterology and Hepatology Unit, University Hospital Marqués de Valdecilla, University of Cantabria, Santander, Spain
| | | | | | | | | | | | | |
Collapse
|
36
|
Faggioni R, Feingold KR, Grunfeld C. Leptin regulation of the immune response and the immunodeficiency of malnutrition. FASEB J 2001; 15:2565-71. [PMID: 11726531 DOI: 10.1096/fj.01-0431rev] [Citation(s) in RCA: 320] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Leptin is a 16 kDa protein mainly produced by adipose tissue in proportion to adipose tissue mass. Originally thought to be a satiety factor, leptin is a pleiotropic molecule. In addition to playing a role in energy regulation, leptin also regulates endocrine and immune functions. Both the structure of leptin and that of its receptor suggest that leptin might be classified as a cytokine. The secondary structure of leptin has similarities to the long-chain helical cytokines family, which includes interleukin 6 (IL-6), IL-11, CNTF, and LIF, and the leptin receptor is homologous to the gp-130 signal-transducing subunit of the IL-6-type cytokine receptors. Leptin plays a role in innate and acquired immunity. Leptin levels increase acutely during infection and inflammation, and may represent a protective component of the host response to inflammation. More important, leptin deficiency increases susceptibility to infectious and inflammatory stimuli and is associated with dysregulation of cytokine production. Leptin deficiency also causes a defect in hematopoiesis. Leptin regulates T cells responses, polarizing Th cells toward a Th1 phenotype. Low leptin levels occurring during starvation mediate the neuroendocrine and immune dysfunction of starvation.
Collapse
Affiliation(s)
- R Faggioni
- Department of Veterans Affairs Medical Center, University of California, San Francisco, San Francisco, California, 94121, USA.
| | | | | |
Collapse
|
37
|
Ingvartsen KL, Boisclair YR. Leptin and the regulation of food intake, energy homeostasis and immunity with special focus on periparturient ruminants. Domest Anim Endocrinol 2001; 21:215-50. [PMID: 11872319 DOI: 10.1016/s0739-7240(02)00119-4] [Citation(s) in RCA: 119] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The biology of leptin has been studied most extensively in rodents and in humans. Leptin is involved in the regulation of food intake, energy homeostasis and immunity. Leptin is primarily produced in white adipose tissue and acts via a family of membrane bound receptors, including an isoform with a long intracellular domain (OB-Rb), and many isoforms with short intracellular domains (Ob-Rs). OB-Rb is predominantly expressed in the hypothalamic regions involved in the regulation of food intake and energy homeostasis. The other isoforms are distributed ubiquitously and are found in most peripheral tissues in far greater abundance than OB-Rb. The effects of leptin on food intake and energy homeostasis are central and are mediated via a network of orexigenic neuropeptides (neuropeptide Y, galanin, galanin-like peptide, melanin-concentrating hormone, orexins, agouti-related peptide) and anorexigenic neuropeptides (corticotropin-releasing hormone, pro-opiomelanocortin, alpha-melanocyte stimulating hormone and cocaine- and amphetamine-regulated transcript). In addition, leptin acts directly on immune cells to stimulate hematopoesis, T-cell immunity, phagocytosis, cytokine production, and to attenuate susceptibility to infectious insults. Emerging data in ruminants suggest that leptin is dynamically regulated by many factors and physiological states. Thus, leptin is secreted in a pulsatile fashion, but without a marked diurnal rhythm. A positive relationship between adiposity and plasma leptin concentration exists in growing and lactating ruminants. The concentration of plasma leptin increases during pregnancy, starts to decline 1--2 wk before parturition, and reaches a nadir in early lactation. The reduction of plasma leptin at parturition is likely to promote centrally mediated adaptations required in periods of energy deficit, but could have negative effects on immune cell function. Future research is needed in ruminants to address the roles played by leptin and the central nervous system in orchestrating metabolism during the periparturient period and during infectious diseases.
Collapse
Affiliation(s)
- K L Ingvartsen
- Danish Institute of Agricultural Sciences, Department of Animal Health and Welfare, Research Centre Foulum, DK-8830, Tjele, Denmark.
| | | |
Collapse
|
38
|
Coyne C, Baravick J, Howell T, Baravick E, Willetto C, Fenwick BW. Biochemical mechanisms that interact with membrane-associated IL-1 RII (60-kDa decoy) receptors in populations of adherent macrophages and vascular endothelium. Cell Signal 2001; 13:765-76. [PMID: 11602187 DOI: 10.1016/s0898-6568(01)00196-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The aim of this investigation was to identify the potential biochemical mechanisms that alter the integrity of membrane-associated IL-1 RII (decoy) receptor complexes expressed by populations of adherent macrophages and vascular endothelium. The initial research strategy utilized to achieve this objective involved delineating the ability of macrophage activation or exposure of macrophages and vascular endothelium to a spectrum of enzyme proteases to influence the expression of membrane-associated IL-1 RII (decoy) or generate soluble fragments of this receptor complex. Results from these investigations revealed that stimulated macrophages displayed proportional increases in both the expression of membrane-associated IL-1 RII (decoy) and release of soluble receptor fragments. Exposure of macrophages and vascular endothelium to the reference proteases discovered the ability of cathepsin-D to biochemically deplete membrane-associated IL-1 RII (decoy) in addition to generating soluble fragments of this receptor complex. Complementary investigations isolated a carboxyl/aspartate protease from activated macrophages utilizing pepstatin-A affinity chromatography. Exposure of vascular endothelium to pepstatin-A binding proteins resulted in a detectable depletion of membrane-associated IL-1 RII (decoy) and generation of soluble receptor fragments. Evaluation of pepstatin-A binding proteins by SDS-PAGE identified a primary protein fraction with a molecular mass of 47-52 kDa that closely correlates with the known molecular size of leukocyte cathepsin-D fractions. Macrophage pepstatin-A binding protein fractions evaluated by nondenaturing haemoglobin-substrate PAGE (Hb-PAGE) analysis detected a lucent proteolytic band at 47-52 kDa. Macrophage pepstatin-A binding proteins also hydrolyzed a synthetic enzyme-specific substrate that selectively recognizes cathepsin-D biochemical activity. In conclusion, the leukocyte carboxyl/aspartate protease cathepsin-D can biochemically alter the integrity and generate soluble fragments of membrane-associated IL-1 RII (60-kDa decoy) receptor complexes expressed by macrophages and vascular endothelium.
Collapse
Affiliation(s)
- C Coyne
- Veterinary Pharmacology Research Laboratory, Veterinary Research Programme, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS 39762, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Parhami F, Tintut Y, Ballard A, Fogelman AM, Demer LL. Leptin enhances the calcification of vascular cells: artery wall as a target of leptin. Circ Res 2001; 88:954-60. [PMID: 11349006 DOI: 10.1161/hh0901.090975] [Citation(s) in RCA: 225] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Leptin, the product of the ob gene, regulates food intake, energy expenditure, and other physiological functions of the peripheral tissues. Leptin receptors have been identified in the hypothalamus and in extrahypothalamic tissues. Increased circulating leptin levels have been correlated with cardiovascular disease, obesity, aging, infection with bacterial lipopolysaccharide, and high-fat diets. All these conditions have also been correlated with increased vascular calcification, a hallmark of atherosclerotic and age-related vascular disease. In addition, the differentiation of marrow osteoprogenitor cells is regulated by leptin. Thus, we hypothesized that leptin may regulate the calcification of vascular cells. In this report, we tested the effects of leptin on a previously characterized subpopulation of vascular cells that undergo osteoblastic differentiation and calcification in vitro. When treated with leptin, these calcifying vascular cells had a significant 5- to 10-fold increase in alkaline phosphatase activity, a marker of osteogenic differentiation of osteoblastic cells. Prolonged treatment with leptin enhanced the calcification of these cells, further supporting the pro-osteogenic differentiation effects of leptin. Furthermore, the presence of the leptin receptor on calcifying vascular cells was demonstrated using reverse transcriptase polymerase chain reaction, immunocytochemistry, and Western blot analysis. We also identified the presence of leptin receptor in the mouse artery wall, localized to subpopulations of medial and adventitial cells, and the expression of leptin by artery wall cells and atherosclerotic lesions in mice. Taken together, these results suggest that leptin regulates the osteoblastic differentiation and calcification of vascular cells and that the artery wall may be an important peripheral tissue target of leptin action.
Collapse
MESH Headings
- Alkaline Phosphatase/drug effects
- Alkaline Phosphatase/metabolism
- Animals
- Arteries/drug effects
- Arteries/metabolism
- Arteries/pathology
- Calcinosis/chemically induced
- Calcium/metabolism
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cattle
- Cells, Cultured
- Female
- Gene Expression Regulation/drug effects
- Immunohistochemistry
- Leptin/metabolism
- Leptin/pharmacology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- RNA/drug effects
- RNA/genetics
- RNA/metabolism
- Receptors, Cell Surface
- Receptors, Leptin
- Reverse Transcriptase Polymerase Chain Reaction
- Vascular Diseases/chemically induced
- Vascular Diseases/metabolism
- Vascular Diseases/pathology
Collapse
Affiliation(s)
- F Parhami
- Departments of Medicine, University of California, Los Angeles, USA.
| | | | | | | | | |
Collapse
|
40
|
Sergeyev V, Broberger C, Hökfelt T. Effect of LPS administration on the expression of POMC, NPY, galanin, CART and MCH mRNAs in the rat hypothalamus. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2001; 90:93-100. [PMID: 11406287 DOI: 10.1016/s0169-328x(01)00088-2] [Citation(s) in RCA: 112] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Anorexia and weight loss are manifestations of inflammation seen both in patients and in experimental animal models such as the lipopolysaccharide (LPS)-treated rat. Using in situ hybridization, the levels of mRNAs encoding proopiomelanocortin (POMC), neuropeptide Y (NPY), galanin, melanin-concentrating hormone (MCH) and cocaine- and amphetamine-regulated transcript (CART) were investigated in the rat hypothalamus after a single intraperitoneal dose (125 microg/kg) of LPS. Four hours after LPS injection the food intake was significantly decreased. POMC and CART mRNA levels were increased in the arcuate nucleus, and MCH, CART and galanin mRNAs were all decreased in the lateral hypothalamic area in LPS-treated rats. Levels of mRNAs for NPY and galanin in the arcuate nucleus, and for MCH and CART in the zona incerta did not change significantly after LPS treatment. These findings support the hypothesis that LPS-induced factors mediate signalling to the POMC/CART neurons in the arcuate nucleus which could lead to reduced food intake by decreasing MCH, CART and galanin synthesis in target lateral hypothalamic neurons.
Collapse
Affiliation(s)
- V Sergeyev
- Department of Neuroscience, Karolinska Institutet, S-171 77, Stockholm, Sweden
| | | | | |
Collapse
|
41
|
Francis J, MohanKumar SM, MohanKumar PS. Correlations of norepinephrine release in the paraventricular nucleus with plasma corticosterone and leptin after systemic lipopolysaccharide: blockade by soluble IL-1 receptor. Brain Res 2000; 867:180-7. [PMID: 10837812 DOI: 10.1016/s0006-8993(00)02311-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The purpose of the study was to investigate the effects of systemic lipopolysaccharide (LPS) on norepinephrine (NE) release in the paraventricular nucleus (PVN) and on plasma concentrations of corticosterone and leptin. Soluble IL-1 receptor (sIL-1R) was used to determine the role of interleukin-1 (IL-1) in these effects. Adult male rats were implanted with a push-pull cannula in the PVN and a jugular catheter to facilitate blood sampling. On the day of the experiment, after the collection of a pretreatment blood and perfusate sample, rats were injected (i.p.) with the vehicle for LPS (saline), 2.5 or 10 microg/kg BW LPS. Other groups of animals were treated i.p. with 25 microg of sIL-1R, or a combination of 10 microg/kg BW of LPS and 25 microg of sIL-1R, 5 min before and 90 min after LPS. Blood and perfusate samples were collected at 30-min intervals for 6 h. NE concentrations in the perfusate were measured using HPLC-EC and corticosterone and leptin levels in the plasma were measured using radioimmunoassay. NE release in the PVN was dose dependent and increased significantly within 90 min in response to the high dose of LPS and reached maximum levels around 180 min before declining gradually to pretreatment levels at 330 min. The corticosterone profile in LPS-treated animals was similar to the NE release profile in the PVN. In contrast, the LPS-induced increase in leptin levels reached a maximum at 210 min and remained elevated even at the end of the observation period. Treatment with sIL-1R completely blocked the LPS-induced effects. It is concluded that LPS stimulates NE release in the PVN and increases plasma concentrations of corticosterone and leptin and that these effects are mediated at least in part by IL-1.
Collapse
Affiliation(s)
- J Francis
- Neuroendocrine Research Laboratory, Department of Diagnostic Medicine and Pathobiology, 1800 Denison Avenue, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | | | | |
Collapse
|
42
|
|