1
|
Lisco G, De Tullio A, Iovino M, Disoteo O, Guastamacchia E, Giagulli VA, Triggiani V. Dopamine in the Regulation of Glucose Homeostasis, Pathogenesis of Type 2 Diabetes, and Chronic Conditions of Impaired Dopamine Activity/Metabolism: Implication for Pathophysiological and Therapeutic Purposes. Biomedicines 2023; 11:2993. [PMID: 38001993 PMCID: PMC10669051 DOI: 10.3390/biomedicines11112993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Dopamine regulates several functions, such as voluntary movements, spatial memory, motivation, sleep, arousal, feeding, immune function, maternal behaviors, and lactation. Less clear is the role of dopamine in the pathophysiology of type 2 diabetes mellitus (T2D) and chronic complications and conditions frequently associated with it. This review summarizes recent evidence on the role of dopamine in regulating insular metabolism and activity, the pathophysiology of traditional chronic complications associated with T2D, the pathophysiological interconnection between T2D and chronic neurological and psychiatric disorders characterized by impaired dopamine activity/metabolism, and therapeutic implications. Reinforcing dopamine signaling is therapeutic in T2D, especially in patients with dopamine-related disorders, such as Parkinson's and Huntington's diseases, addictions, and attention-deficit/hyperactivity disorder. On the other hand, although specific trials are probably needed, certain medications approved for T2D (e.g., metformin, pioglitazone, incretin-based therapy, and gliflozins) may have a therapeutic role in such dopamine-related disorders due to anti-inflammatory and anti-oxidative effects, improvement in insulin signaling, neuroinflammation, mitochondrial dysfunction, autophagy, and apoptosis, restoration of striatal dopamine synthesis, and modulation of dopamine signaling associated with reward and hedonic eating. Last, targeting dopamine metabolism could have the potential for diagnostic and therapeutic purposes in chronic diabetes-related complications, such as diabetic retinopathy.
Collapse
Affiliation(s)
- Giuseppe Lisco
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari, 70124 Bari, Italy; (G.L.); (A.D.T.); (M.I.); (E.G.); (V.A.G.)
| | - Anna De Tullio
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari, 70124 Bari, Italy; (G.L.); (A.D.T.); (M.I.); (E.G.); (V.A.G.)
| | - Michele Iovino
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari, 70124 Bari, Italy; (G.L.); (A.D.T.); (M.I.); (E.G.); (V.A.G.)
| | - Olga Disoteo
- Diabetology Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy;
| | - Edoardo Guastamacchia
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari, 70124 Bari, Italy; (G.L.); (A.D.T.); (M.I.); (E.G.); (V.A.G.)
| | - Vito Angelo Giagulli
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari, 70124 Bari, Italy; (G.L.); (A.D.T.); (M.I.); (E.G.); (V.A.G.)
| | - Vincenzo Triggiani
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari, 70124 Bari, Italy; (G.L.); (A.D.T.); (M.I.); (E.G.); (V.A.G.)
| |
Collapse
|
2
|
Genchi VA, Palma G, Sorice GP, D'Oria R, Caccioppoli C, Marrano N, Biondi G, Caruso I, Cignarelli A, Natalicchio A, Laviola L, Giorgino F, Perrini S. Pharmacological modulation of adaptive thermogenesis: new clues for obesity management? J Endocrinol Invest 2023; 46:2213-2236. [PMID: 37378828 PMCID: PMC10558388 DOI: 10.1007/s40618-023-02125-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023]
Abstract
BACKGROUND Adaptive thermogenesis represents the main mechanism through which the body generates heat in response to external stimuli, a phenomenon that includes shivering and non-shivering thermogenesis. The non-shivering thermogenesis is mainly exploited by adipose tissue characterized by a brown aspect, which specializes in energy dissipation. A decreased amount of brown adipose tissue has been observed in ageing and chronic illnesses such as obesity, a worldwide health problem characterized by dysfunctional adipose tissue expansion and associated cardiometabolic complications. In the last decades, the discovery of a trans-differentiation mechanism ("browning") within white adipose tissue depots, leading to the generation of brown-like cells, allowed to explore new natural and synthetic compounds able to favour this process and thus enhance thermogenesis with the aim of counteracting obesity. Based on recent findings, brown adipose tissue-activating agents could represent another option in addition to appetite inhibitors and inhibitors of nutrient absorption for obesity treatment. PURPOSE This review investigates the main molecules involved in the physiological (e.g. incretin hormones) and pharmacological (e.g. β3-adrenergic receptors agonists, thyroid receptor agonists, farnesoid X receptor agonists, glucagon-like peptide-1, and glucagon receptor agonists) modulation of adaptive thermogenesis and the signalling mechanisms involved.
Collapse
Affiliation(s)
- V A Genchi
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - G Palma
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - G P Sorice
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - R D'Oria
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - C Caccioppoli
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - N Marrano
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - G Biondi
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - I Caruso
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Cignarelli
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Natalicchio
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - L Laviola
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - F Giorgino
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy.
| | - S Perrini
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| |
Collapse
|
3
|
Zeng L, Herdman DS, Lee SM, Tao A, Das M, Bertin S, Eckmann L, Mahata SK, Wu P, Hara M, Byun JW, Devulapalli S, Patel HH, Molina AJ, Osborn O, Corr M, Raz E, Webster NJ. Loss of cAMP Signaling in CD11c Immune Cells Protects Against Diet-Induced Obesity. Diabetes 2023; 72:1235-1250. [PMID: 37257047 PMCID: PMC10451016 DOI: 10.2337/db22-1035] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 05/21/2023] [Indexed: 06/02/2023]
Abstract
In obesity, CD11c+ innate immune cells are recruited to adipose tissue and create an inflammatory state that causes both insulin and catecholamine resistance. We found that ablation of Gnas, the gene that encodes Gαs, in CD11c expressing cells protects mice from obesity, glucose intolerance, and insulin resistance. Transplantation studies showed that the lean phenotype was conferred by bone marrow-derived cells and did not require adaptive immunity. Loss of cAMP signaling was associated with increased adipose tissue norepinephrine and cAMP signaling, and prevention of catecholamine resistance. The adipose tissue had reduced expression of catecholamine transport and degradation enzymes, suggesting that the elevated norepinephrine resulted from decreased catabolism. Collectively, our results identified an important role for cAMP signaling in CD11c+ innate immune cells in whole-body metabolism by controlling norepinephrine levels in white adipose tissue, modulating catecholamine-induced lipolysis and increasing thermogenesis, which, together, created a lean phenotype. ARTICLE HIGHLIGHTS We undertook this study to understand how immune cells communicate with adipocytes, specifically, whether cAMP signaling in the immune cell and the adipocyte are connected. We identified a reciprocal interaction between CD11c+ innate immune cells and adipocytes in which high cAMP signaling in the immune cell compartment induces low cAMP signaling in adipocytes and vice versa. This interaction regulates lipolysis in adipocytes and inflammation in immune cells, resulting in either a lean, obesity-resistant, and insulin-sensitive phenotype, or an obese, insulin-resistant phenotype.
Collapse
Affiliation(s)
- Liping Zeng
- The Second Affiliated Hospital of Guangzhou Medical University, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, China
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - D. Scott Herdman
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Sung Min Lee
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Ailin Tao
- The Second Affiliated Hospital of Guangzhou Medical University, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, China
| | - Manasi Das
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Samuel Bertin
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Lars Eckmann
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Sushil K. Mahata
- Department of Medicine, University of California San Diego, La Jolla, CA
- VA San Diego Healthcare System, San Diego, CA
| | - Panyisha Wu
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Miki Hara
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Ji-Won Byun
- Department of Dermatology, Inha University Hospital, Incheon, South Korea
| | - Shwetha Devulapalli
- Department of Anesthesiology, University of California San Diego, La Jolla, CA
| | - Hemal H. Patel
- VA San Diego Healthcare System, San Diego, CA
- Department of Anesthesiology, University of California San Diego, La Jolla, CA
| | | | - Olivia Osborn
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Maripat Corr
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Eyal Raz
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Nicholas J.G. Webster
- Department of Medicine, University of California San Diego, La Jolla, CA
- VA San Diego Healthcare System, San Diego, CA
- Moores Cancer Center, University of California San Diego, La Jolla CA
| |
Collapse
|
4
|
He F, Zhang P, Zhang Q, Qi G, Cai H, Li T, Li M, Lu J, Lin J, Ming J, Tian B. Dopaminergic Projection from Ventral Tegmental Area to Substantia Nigra Pars Reticulata Mediates Chronic Social Defeat Stress-Induced Hypolocomotion. Mol Neurobiol 2021; 58:5635-5648. [PMID: 34382160 DOI: 10.1007/s12035-021-02522-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/04/2021] [Indexed: 01/06/2023]
Abstract
Numerous human clinical studies have suggested that decreased locomotor activity is a common symptom of major depressive disorder (MDD), as well as other psychiatric diseases. In MDD, the midbrain ventral tegmental area (VTA) dopamine (DA) neurons are closely related to regulate the information processing of reward, motivation, cognition, and aversion. However, the neural circuit mechanism that underlie the relationship between VTA-DA neurons and MDD-related motor impairments, especially hypolocomotion, is still largely unknown. Herein, we investigate how the VTA-DA neurons contribute to the hypolocomotion performance in chronic social defeat stress (CSDS), a mouse model of depression-relevant neurobehavioral states. The results show that CSDS could affect the spontaneous locomotor activity of mice, but not the grip strength and forced locomotor ability. Chemogenetic activation of VTA-DA neurons alleviated CSDS-induced hypolocomotion. Subsequently, quantitative whole-brain mapping revealed decreased projections from VTA-DA neurons to substantia nigra pars reticulata (SNr) after CSDS treatment. Optogenetic activation of dopaminergic projection from VTA to SNr with the stimulation of phasic firing, but not tonic firing, could significantly increase the locomotor activity of mice. Moreover, chemogenetic activation of VTA-SNr dopaminergic circuit in CSDS mice could also rescued the decline of locomotor activity. Taken together, our data suggest that the VTA-SNr dopaminergic projection mediates CSDS-induced hypolocomotion, which provides a theoretical basis and potential therapeutic target for MDD.
Collapse
Affiliation(s)
- Feng He
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China
| | - Pei Zhang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China.,Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China.,Key Laboratory of Neurological Diseases, Ministry of Education, Wuhan, Hubei Province, 430030, People's Republic of China
| | - Qian Zhang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China
| | - Guangjian Qi
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China
| | - Hongwei Cai
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China
| | - Tongxia Li
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China
| | - Ming Li
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China
| | - Jiazhen Lu
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China
| | - Jiaen Lin
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China
| | - Jie Ming
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technolog, Wuhan, Hubei Province, 430022, People's Republic of China.
| | - Bo Tian
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China. .,Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China. .,Key Laboratory of Neurological Diseases, Ministry of Education, Wuhan, Hubei Province, 430030, People's Republic of China.
| |
Collapse
|
5
|
Hollstein T, Basolo A, Ando T, Votruba SB, Krakoff J, Piaggi P. Urinary Norepinephrine Is a Metabolic Determinant of 24-Hour Energy Expenditure and Sleeping Metabolic Rate in Adult Humans. J Clin Endocrinol Metab 2020; 105:5718326. [PMID: 32002540 PMCID: PMC7055738 DOI: 10.1210/clinem/dgaa047] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 01/28/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Interindividual variability in 24-hour energy expenditure (24EE) during energy-balance conditions is mainly determined by differences in body composition and demographic factors. Previous studies suggested that 24EE might also be influenced by sympathetic nervous system activity via catecholamine (norepinephrine, epinephrine) secretion. Therefore, we analyzed the association between catecholamines and energy expenditure in 202 individuals from a heterogeneous population of mixed ethnicities. METHODS Participants (n = 202, 33% female, 14% black, 32% white, 41% Native American, 11% Hispanic, age: 36.9 ± 10.3 y [mean ± SD], percentage body fat: 30.3 ± 9.4) resided in a whole-room calorimeter over 24 hours during carefully controlled energy-balance conditions to measure 24EE and its components: sleeping metabolic rate (SMR), awake-fed thermogenesis (AFT), and spontaneous physical activity (SPA). Urine samples were collected, and 24-h urinary epinephrine and norepinephrine excretion rates were assessed by high-performance liquid chromatography. RESULTS Both catecholamines were associated with 24EE and SMR (norepinephrine: +27 and +19 kcal/d per 10 μg/24h; epinephrine: +18 and +10 kcal/d per 1 μg/24h) in separate analyses after adjustment for age, sex, ethnicity, fat mass, fat-free mass, calorimeter room, temperature, and physical activity. In a multivariate model including both norepinephrine and epinephrine, only norepinephrine was independently associated with both 24EE and SMR (both P < .008), whereas epinephrine became insignificant. Neither epinephrine nor norepinephrine were associated with adjusted AFT (both P = .37) but epinephrine was associated with adjusted SPA (+0.5% per 1 μg/24h). CONCLUSIONS Our data provide compelling evidence that sympathetic nervous system activity, mediated via norepinephrine, is a determinant of human energy expenditure during nonstressed, eucaloric conditions.
Collapse
Affiliation(s)
- Tim Hollstein
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, US
| | - Alessio Basolo
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, US
| | - Takafumi Ando
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, US
| | - Susanne B Votruba
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, US
| | - Jonathan Krakoff
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, US
| | - Paolo Piaggi
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, US
- Department of Information Engineering, University of Pisa, Pisa, Italy
- Correspondence and Reprint Requests: Paolo Piaggi, PhD, Obesity and Diabetes Clinical Research Section, NIDDK, NIH, 4212 North 16th Street, Phoenix, AZ 85016. E-mail: ;
| |
Collapse
|
6
|
Mercader J, Sabater AG, Le Gonidec S, Decaunes P, Chaplin A, Gómez-Zorita S, Milagro FI, Carpéné C. Oral Phenelzine Treatment Mitigates Metabolic Disturbances in Mice Fed a High-Fat Diet. J Pharmacol Exp Ther 2019; 371:555-566. [PMID: 31270215 DOI: 10.1124/jpet.119.259895] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/01/2019] [Indexed: 12/15/2022] Open
Abstract
Novel mechanisms and health benefits have been recently suggested for the antidepressant drug phenelzine (PHE), known as a nonselective monoamine oxidase inhibitor. They include an antilipogenic action that could have an impact on excessive fat accumulation and obesity-related metabolic alterations. We evaluated the metabolic effects of an oral PHE treatment on mice fed a high-fat diet (HFD). Eleven-week-old male C57BL/6 mice were fed a HFD and either a 0.028% PHE solution (HFD + PHE) or water to drink for 11 weeks. PHE attenuated the increase in body weight and adiposity without affecting food consumption. Energy efficiency was lower in HFD + PHE mice. Lipid content was reduced in subcutaneous fat pads, liver, and skeletal muscle. In white adipose tissue (WAT), PHE reduced sterol regulatory element-binding protein-1c and phosphoenolpyruvate carboxykinase mRNA levels, inhibited amine-induced lipogenesis, and did not increase lipolysis. Moreover, HFD + PHE mice presented diminished levels of hydrogen peroxide release in subcutaneous WAT and reduced expression of leukocyte transmigration markers and proinflammatory cytokines in visceral WAT and liver. PHE reduced the circulating levels of glycerol, triacylglycerols, high-density lipoprotein cholesterol, and insulin. Insulin resistance was reduced, without affecting glucose levels and glucose tolerance. In contrast, PHE increased rectal temperature and slightly increased energy expenditure. The mitigation of HFD-induced metabolic disturbances points toward a promising role for PHE in obesity treatment and encourages further research on its mechanisms of action. SIGNIFICANCE STATEMENT: Phenelzine reduces body fat, markers of oxidative stress, inflammation, and insulin resistance in high-fat diet mice. Semicarbazide-sensitive amine oxidase, monoamine oxidase, phosphoenolpyruvate carboxykinase, and sterol regulatory element-binding protein-1c are involved in the metabolic effects of phenelzine. Phenelzine could be potentially used for the treatment of obesity-related complications.
Collapse
Affiliation(s)
- Josep Mercader
- Balearic Islands Health Research Institute, Palma de Mallorca, Spain (J.M.); Department of Fundamental Biology and Health Sciences, University of Balearic Islands (UIB), Palma de Mallorca, Spain (J.M.); Alimentómica, S.L., Spin-off from UIB, Palma de Mallorca, Spain (A.G.S.); Institute of Metabolic and Cardiovascular Diseases, INSERM, UMR1048, Teams 1 & 3, Toulouse, France (C.C., S.L.G., P.D.); I2MC, University of Toulouse, UMR1048, Paul Sabatier University, Toulouse Cedex 4, France (C.C., S.L.G., P.D.); Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio (A.C.); Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country and Lucio Lascaray Research Institute, Vitoria, Spain (S.G.-Z.); CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain (S.G.-Z., F.I.M.); Department of Nutrition, Food Science, and Physiology, University of Navarra, Pamplona, Spain (F.I.M.); and Centre for Nutrition Research, University of Navarra, Pamplona, Spain (F.I.M.)
| | - Agustín G Sabater
- Balearic Islands Health Research Institute, Palma de Mallorca, Spain (J.M.); Department of Fundamental Biology and Health Sciences, University of Balearic Islands (UIB), Palma de Mallorca, Spain (J.M.); Alimentómica, S.L., Spin-off from UIB, Palma de Mallorca, Spain (A.G.S.); Institute of Metabolic and Cardiovascular Diseases, INSERM, UMR1048, Teams 1 & 3, Toulouse, France (C.C., S.L.G., P.D.); I2MC, University of Toulouse, UMR1048, Paul Sabatier University, Toulouse Cedex 4, France (C.C., S.L.G., P.D.); Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio (A.C.); Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country and Lucio Lascaray Research Institute, Vitoria, Spain (S.G.-Z.); CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain (S.G.-Z., F.I.M.); Department of Nutrition, Food Science, and Physiology, University of Navarra, Pamplona, Spain (F.I.M.); and Centre for Nutrition Research, University of Navarra, Pamplona, Spain (F.I.M.)
| | - Sophie Le Gonidec
- Balearic Islands Health Research Institute, Palma de Mallorca, Spain (J.M.); Department of Fundamental Biology and Health Sciences, University of Balearic Islands (UIB), Palma de Mallorca, Spain (J.M.); Alimentómica, S.L., Spin-off from UIB, Palma de Mallorca, Spain (A.G.S.); Institute of Metabolic and Cardiovascular Diseases, INSERM, UMR1048, Teams 1 & 3, Toulouse, France (C.C., S.L.G., P.D.); I2MC, University of Toulouse, UMR1048, Paul Sabatier University, Toulouse Cedex 4, France (C.C., S.L.G., P.D.); Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio (A.C.); Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country and Lucio Lascaray Research Institute, Vitoria, Spain (S.G.-Z.); CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain (S.G.-Z., F.I.M.); Department of Nutrition, Food Science, and Physiology, University of Navarra, Pamplona, Spain (F.I.M.); and Centre for Nutrition Research, University of Navarra, Pamplona, Spain (F.I.M.)
| | - Pauline Decaunes
- Balearic Islands Health Research Institute, Palma de Mallorca, Spain (J.M.); Department of Fundamental Biology and Health Sciences, University of Balearic Islands (UIB), Palma de Mallorca, Spain (J.M.); Alimentómica, S.L., Spin-off from UIB, Palma de Mallorca, Spain (A.G.S.); Institute of Metabolic and Cardiovascular Diseases, INSERM, UMR1048, Teams 1 & 3, Toulouse, France (C.C., S.L.G., P.D.); I2MC, University of Toulouse, UMR1048, Paul Sabatier University, Toulouse Cedex 4, France (C.C., S.L.G., P.D.); Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio (A.C.); Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country and Lucio Lascaray Research Institute, Vitoria, Spain (S.G.-Z.); CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain (S.G.-Z., F.I.M.); Department of Nutrition, Food Science, and Physiology, University of Navarra, Pamplona, Spain (F.I.M.); and Centre for Nutrition Research, University of Navarra, Pamplona, Spain (F.I.M.)
| | - Alice Chaplin
- Balearic Islands Health Research Institute, Palma de Mallorca, Spain (J.M.); Department of Fundamental Biology and Health Sciences, University of Balearic Islands (UIB), Palma de Mallorca, Spain (J.M.); Alimentómica, S.L., Spin-off from UIB, Palma de Mallorca, Spain (A.G.S.); Institute of Metabolic and Cardiovascular Diseases, INSERM, UMR1048, Teams 1 & 3, Toulouse, France (C.C., S.L.G., P.D.); I2MC, University of Toulouse, UMR1048, Paul Sabatier University, Toulouse Cedex 4, France (C.C., S.L.G., P.D.); Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio (A.C.); Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country and Lucio Lascaray Research Institute, Vitoria, Spain (S.G.-Z.); CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain (S.G.-Z., F.I.M.); Department of Nutrition, Food Science, and Physiology, University of Navarra, Pamplona, Spain (F.I.M.); and Centre for Nutrition Research, University of Navarra, Pamplona, Spain (F.I.M.)
| | - Saioa Gómez-Zorita
- Balearic Islands Health Research Institute, Palma de Mallorca, Spain (J.M.); Department of Fundamental Biology and Health Sciences, University of Balearic Islands (UIB), Palma de Mallorca, Spain (J.M.); Alimentómica, S.L., Spin-off from UIB, Palma de Mallorca, Spain (A.G.S.); Institute of Metabolic and Cardiovascular Diseases, INSERM, UMR1048, Teams 1 & 3, Toulouse, France (C.C., S.L.G., P.D.); I2MC, University of Toulouse, UMR1048, Paul Sabatier University, Toulouse Cedex 4, France (C.C., S.L.G., P.D.); Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio (A.C.); Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country and Lucio Lascaray Research Institute, Vitoria, Spain (S.G.-Z.); CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain (S.G.-Z., F.I.M.); Department of Nutrition, Food Science, and Physiology, University of Navarra, Pamplona, Spain (F.I.M.); and Centre for Nutrition Research, University of Navarra, Pamplona, Spain (F.I.M.)
| | - Fermín I Milagro
- Balearic Islands Health Research Institute, Palma de Mallorca, Spain (J.M.); Department of Fundamental Biology and Health Sciences, University of Balearic Islands (UIB), Palma de Mallorca, Spain (J.M.); Alimentómica, S.L., Spin-off from UIB, Palma de Mallorca, Spain (A.G.S.); Institute of Metabolic and Cardiovascular Diseases, INSERM, UMR1048, Teams 1 & 3, Toulouse, France (C.C., S.L.G., P.D.); I2MC, University of Toulouse, UMR1048, Paul Sabatier University, Toulouse Cedex 4, France (C.C., S.L.G., P.D.); Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio (A.C.); Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country and Lucio Lascaray Research Institute, Vitoria, Spain (S.G.-Z.); CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain (S.G.-Z., F.I.M.); Department of Nutrition, Food Science, and Physiology, University of Navarra, Pamplona, Spain (F.I.M.); and Centre for Nutrition Research, University of Navarra, Pamplona, Spain (F.I.M.)
| | - Christian Carpéné
- Balearic Islands Health Research Institute, Palma de Mallorca, Spain (J.M.); Department of Fundamental Biology and Health Sciences, University of Balearic Islands (UIB), Palma de Mallorca, Spain (J.M.); Alimentómica, S.L., Spin-off from UIB, Palma de Mallorca, Spain (A.G.S.); Institute of Metabolic and Cardiovascular Diseases, INSERM, UMR1048, Teams 1 & 3, Toulouse, France (C.C., S.L.G., P.D.); I2MC, University of Toulouse, UMR1048, Paul Sabatier University, Toulouse Cedex 4, France (C.C., S.L.G., P.D.); Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio (A.C.); Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country and Lucio Lascaray Research Institute, Vitoria, Spain (S.G.-Z.); CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain (S.G.-Z., F.I.M.); Department of Nutrition, Food Science, and Physiology, University of Navarra, Pamplona, Spain (F.I.M.); and Centre for Nutrition Research, University of Navarra, Pamplona, Spain (F.I.M.)
| |
Collapse
|
7
|
Navarro M, Luhn KL, Kampov-Polevoy AB, Garbutt JC, Thiele TE. Bupropion, Alone and in Combination with Naltrexone, Blunts Binge-Like Ethanol Drinking and Intake Following Chronic Intermittent Access to Ethanol in Male C57BL/6J Mice. Alcohol Clin Exp Res 2019; 43:783-790. [PMID: 30817015 DOI: 10.1111/acer.13992] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 02/21/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Regular binge drinking is associated with numerous adverse consequences, yet the U.S. Food and Drug Administration (FDA) has approved only 4 medications for the treatment of alcohol use disorders, and none have been specifically targeted for treating binge drinking. Here, we assessed the effectiveness of the dopamine/norepinephrine re-uptake inhibitor, bupropion (BUP), alone and in combination with naltrexone (NAL), to reduce binge-like and chronic ethanol (EtOH) intake in mice. While BUP is an FDA-approved drug that is currently used to treat depression and nicotine dependence, there has been only limited investigation to assess the ability of BUP to reduce EtOH intake. METHODS Male C57BL/6J mice were tested with 20% (v/v) EtOH using "drinking in the dark" (DID) procedures to model binge-like EtOH intake and following intermittent access to EtOH (IAE). In Experiment 1, mice were given intraperitoneal (i.p.) injection of 0, 20, or 40 mg/kg BUP 30 minutes before DID testing; in Experiment 2, mice were given i.p. injection of vehicle, BUP (20 mg/kg), NAL (3 mg/kg), or BUP + NAL (20 and 3 mg/kg, respectively) 30 minutes before DID testing; and in Experiment 3, mice were given i.p. injection of 0, 20, 40, or 60 mg/kg BUP 30 minutes before EtOH access after mice had 16 weeks of IAE. RESULTS BUP dose dependently blunted EtOH intake with DID procedures and after 16 weeks of IAE. Administration of subthreshold doses of BUP + NAL also reduced binge-like EtOH intake. Finally, BUP failed to reduce consumption of a 3% (w/v) sucrose solution. CONCLUSIONS BUP, alone and in combination with NAL, may represent a novel approach to treating binge EtOH intake. We are currently assessing the efficacy of BUP to curb binge drinking in a phase II clinical trial experiment.
Collapse
Affiliation(s)
- Montserrat Navarro
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,The Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kendall L Luhn
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Alexey B Kampov-Polevoy
- The Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - James C Garbutt
- The Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Todd E Thiele
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,The Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
8
|
Simonds SE, Pryor JT, Cowley MA. Repeated weight cycling in obese mice causes increased appetite and glucose intolerance. Physiol Behav 2018; 194:184-190. [DOI: 10.1016/j.physbeh.2018.05.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 05/21/2018] [Accepted: 05/21/2018] [Indexed: 11/25/2022]
|
9
|
Stefanidis A, Man Lee CC, Brown WA, Oldfield BJ. Improving efficacy of the adjustable gastric band: studies of the use of adjuvant approaches in a rodent model. Surg Obes Relat Dis 2016; 13:291-304. [PMID: 27988274 DOI: 10.1016/j.soard.2016.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 08/30/2016] [Accepted: 09/03/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND The laparoscopic adjustable gastric band (AGB) has been effective in reducing excess weight by approximately 50% for at least 16 years. However, as with all weight loss approaches, reduction in weight resulting from bariatric surgery is associated with a compensatory reduction in energy expenditure, which may confound and limit weight loss. Adjuvant therapies that reduce food intake and increase energy expenditure may be used to improve weight loss outcomes by ameliorating, or even reversing, this reduction in energy expenditure. METHODS Rats were either fitted with an AGB or were sham operated and received one of 2 adjunctive pharmacologic treatments, (1) thyroxine or (2) bupropion/naltrexone (Contrave), at a range of doses and matched with vehicle controls (n = 6-8/group) over a 4-week period of combined treatments. Metabolic parameters including food intake, weight, fat mass, and energy expenditure in brown adipose tissue (BAT), whole body calorimetry, and physical activity were assessed. RESULTS Inflation of the AGB caused a reduction in weight gain that was further enhanced by cotreatment with either thyroxine or Contrave (P<.05). Thyroxine completely ameliorated the reduction in AGB-induced BAT thermogenesis and significantly improved weight loss, particularly in fat mass. Contrave also augmented the loss of weight and fat mass associated with the AGB and increased BAT thermogenesis in banded rats even at doses below that required to change food intake. CONCLUSION Adjuvant therapies can improve the efficacy of the AGB, at least in part by negating the compensatory reduction in energy expenditure, but also via a combined effect on food intake.
Collapse
Affiliation(s)
- Aneta Stefanidis
- Department of Physiology, Monash University, Melbourne, Australia
| | | | - Wendy A Brown
- Monash University Department of Surgery, Alfred Hospital, Melbourne, Australia; Centre for Obesity Research and Education, Monash University School of Public Health and Preventive Science, Melbourne, Australia
| | - Brian J Oldfield
- Department of Physiology, Monash University, Melbourne, Australia.
| |
Collapse
|
10
|
FoxO1 in dopaminergic neurons regulates energy homeostasis and targets tyrosine hydroxylase. Nat Commun 2016; 7:12733. [PMID: 27681312 PMCID: PMC5056402 DOI: 10.1038/ncomms12733] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/28/2016] [Indexed: 01/05/2023] Open
Abstract
Dopaminergic (DA) neurons are involved in the integration of neuronal and hormonal signals to regulate food consumption and energy balance. Forkhead transcriptional factor O1 (FoxO1) in the hypothalamus plays a crucial role in mediation of leptin and insulin function. However, the homoeostatic role of FoxO1 in DA system has not been investigated. Here we report that FoxO1 is highly expressed in DA neurons and mice lacking FoxO1 specifically in the DA neurons (FoxO1 KODAT) show markedly increased energy expenditure and interscapular brown adipose tissue (iBAT) thermogenesis accompanied by reduced fat mass and improved glucose/insulin homoeostasis. Moreover, FoxO1 KODAT mice exhibit an increased sucrose preference in concomitance with higher dopamine and norepinephrine levels. Finally, we found that FoxO1 directly targets and negatively regulates tyrosine hydroxylase (TH) expression, the rate-limiting enzyme of the catecholamine synthesis, delineating a mechanism for the KO phenotypes. Collectively, these results suggest that FoxO1 in DA neurons is an important transcriptional factor that directs the coordinated control of energy balance, thermogenesis and glucose homoeostasis. Dopaminergic neurons are important for regulating energy homeostasis. Here, the authors show the transcription factor FoxO1 negatively regulates tyrosine hydroxylase expression in midbrain dopaminergic neurons, and plays an important role in regulation of glucose homeostasis, energy expenditure, and resistance to diet-induced obesity.
Collapse
|
11
|
Billes SK, Sinnayah P, Cowley MA. Naltrexone/bupropion for obesity: an investigational combination pharmacotherapy for weight loss. Pharmacol Res 2014; 84:1-11. [PMID: 24754973 DOI: 10.1016/j.phrs.2014.04.004] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 04/08/2014] [Accepted: 04/09/2014] [Indexed: 01/01/2023]
Abstract
The mechanism of action of the combination therapy, naltrexone/bupropion (NB), for obesity has not been fully described to date. Weight loss attempts rarely result in long-term success. This is likely a result of complex interactions among multiple peripheral and CNS systems that defend against weight loss, and may explain the overwhelming lack of effective obesity treatments. NB is an investigational combination therapy for obesity that was developed based on evidence that obesity involves alterations in the hypothalamic melanocortin system as well as brain reward systems that influence food craving and mood. Naltrexone and bupropion both have actions in these brain regions that may cause them to influence food intake, food craving, and other aspects of eating behavior that affect body weight. We review the individual actions of naltrexone and bupropion in brain hypothalamic and reward systems, and describe the current in vitro, in vivo, and clinical evidence for how NB influences food intake and produces weight loss.
Collapse
Affiliation(s)
| | - Puspha Sinnayah
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Michael A Cowley
- Department of Physiology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
12
|
Ahmed M, El-Bakly WM, Zaki AM, abd Alzez LF, El serafi O. Bupropion effects on high-fat diet-induced steatohepatitis and endothelial dysfunction in rats: role of tumour necrosis factor-alpha. J Pharm Pharmacol 2014; 66:793-801. [PMID: 24471744 DOI: 10.1111/jphp.12213] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Accepted: 12/07/2013] [Indexed: 12/16/2022]
Abstract
OBJECTIVES This study aimed to elucidate the effect of bupropion (BUP) on high-fat diet (HFD)-treated rats that is to say the action of BUP on diabetes and hyperlipidemia with its consequences on liver and endothelial function. METHODS Male Wistar rats were fed HFD or normal chow for 15 weeks then given either BUP (50 mg/kg) or distilled water by gavage for 4 weeks. The effect of BUP on diabetes, hyperlipidemia, hepatic and vascular functions as well as tumour necrosis factor-alpha (TNF)-α were assessed. The intima-media thickness of the aorta was evaluated. KEY FINDINGS BUP significantly decreased serum lipid, liver enzyme, homeostasis model assessment for insulin resistance (HOMA-IR), serum TNF-α and the impaired glucose tolerance. Liver from rats with non-alcoholic steatohepatitis (NASH) demonstrated significant higher TNF-α level, inflammatory cell infiltration, ballooning and steatosis which significantly ameliorated by BUP treatment. Neither intima/media ratio nor vascular reactivity to acetylcholine is improved by BUP treatment. CONCLUSIONS NASH induced by a HFD was associated with hyperlipidemia, insulin resistance, endothelial dysfunction and increase in liver TNF-α. All of these may contribute to the pathogenesis of NASH. BUP has potential role in improving metabolic and hepatic function with negative vascular effect. Since BUP is a well-known antidepressant, it will be a candidate drug in treatment of depression in hepatic diseased or metabolic disturbed patients.
Collapse
Affiliation(s)
- Mai Ahmed
- Department of Pharmacology, Ain Shams University, Cairo, Egypt; Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | | | | |
Collapse
|
13
|
Janhunen SK, la Fleur SE, Adan RAH. Blocking alpha2A adrenoceptors, but not dopamine receptors, augments bupropion-induced hypophagia in rats. Obesity (Silver Spring) 2013; 21:E700-8. [PMID: 23894096 DOI: 10.1002/oby.20581] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 07/02/2013] [Accepted: 07/18/2013] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Anti-obesity drugs have adverse effects which limit their use, creating a need for novel anti-obesity compounds. We studied effects of dopamine (DA) and norepinephrine (NE) reuptake inhibitor bupropion (BUP), alone and after blocking α1- or α2-adrenoceptors (AR), D1/5, D2/3, or D4 receptors, to determine which receptors act downstream of BUP. DESIGN AND METHODS Effects on caloric intake, meal patterning and locomotion were assessed, using an automated weighing system and telemetry in male rats with 18-h access to Western Human style diet. RESULTS BUP (30 mg/kg) induced hypophagia by reducing meal size and postponing meal initiation. WB4101 (α1-AR; 2 mg/kg) and imiloxan (α2B-AR; 5 mg/kg) attenuated BUP's effect on meal size, while WB4101 and BRL 44408 (α2A/D-AR; 2 mg/kg) counteracted effect on meal initiation. Atipamezole (α2-AR; 1 mg/kg) and imiloxan further postponed initiation of meals. SKF 83566 (D1/5; 0.3 mg/kg), raclopride (D2/3; 0.5 mg/kg) and to a lesser extent FAUC 213 (D4; 0.5 mg/kg), attenuated BUP-induced hypophagia. BUP stimulated locomotion, which was blocked by all antagonists, except FAUC 213 or BRL 44408. CONCLUSIONS Alpha1-, α2A/D- and α2B-ARs, and DA receptors underlie BUP's effects on size and initiation of meals, while blocking pre-synaptic α2-ARs enhanced BUP-induced hypophagia. An inverse agonist of (pre-synaptic) α2A-ARs could enhance BUP-induced anorexia and treat eating disorders and obesity.
Collapse
Affiliation(s)
- Sanna K Janhunen
- Rudolf Magnus Institute of Neuroscience, Department of Neuroscience and Pharmacology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | |
Collapse
|
14
|
Acute behavioural effects of bupropion and naltrexone, alone and in combination, in non-deprived male rats presented with palatable mash. Psychopharmacology (Berl) 2013; 228:291-307. [PMID: 23455599 DOI: 10.1007/s00213-013-3036-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 02/11/2013] [Indexed: 01/28/2023]
Abstract
RATIONALE In appetite research, drugs frequently progress to clinical trials on the basis of outcome (reduced food intake/body weight gain) with insufficient attention to process (behavioural analysis). Although bupropion and naltrexone (alone and in combination) reduce food consumption in rodents and humans, their effects on behaviour during feeding tests have not been thoroughly investigated. OBJECTIVES This study aimed to assess the behavioural specificity of anorectic responses to bupropion, naltrexone and their combination. METHODS Video analysis was employed to characterise the behavioural effects of acute systemic treatment with bupropion (10.0-40.0 mg/kg), naltrexone (0.1-3.0 mg/kg) and combined bupropion (20 mg/kg) plus naltrexone (0.1-1.0 mg/kg) in non-deprived male rats exposed for 1 h to palatable mash. Particular attention was paid to the behavioural satiety sequence (BSS). RESULTS In experiment 1, the anorectic response to 40 mg/kg bupropion was associated with significant psychomotor stimulation and a complete disruption of the BSS. In experiment 2, the anorectic response to 3 mg/kg naltrexone was associated with an accelerated but otherwise normal BSS. In experiment 3, the co-administration of 20 mg/kg bupropion and naltrexone (0.1 and 1.0 mg/kg) not only produced an additive anorectic profile (including a reduced rate of eating), but the addition of the opioid receptor antagonist also concurrently attenuated the psychomotor stimulant response to the atypical antidepressant. CONCLUSIONS Low-dose co-treatment with naltrexone and bupropion produces a stronger suppression of appetite than that seen with either agent alone and has the additional advantage of reducing some of the unwanted effects of bupropion.
Collapse
|
15
|
McElroy SL, Guerdjikova AI, Kim DD, Burns C, Harris-Collazo R, Landbloom R, Dunayevich E. Naltrexone/Bupropion combination therapy in overweight or obese patients with major depressive disorder: results of a pilot study. Prim Care Companion CNS Disord 2013; 15:12m01494. [PMID: 24171147 PMCID: PMC3795584 DOI: 10.4088/pcc.12m01494] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 02/18/2013] [Indexed: 10/26/2022] Open
Abstract
OBJECTIVE To evaluate the effect of 32-mg/d naltrexone sustained release and 360-mg/d bupropion sustained release (NB32) in overweight and obese patients with major depressive disorder (MDD). METHOD Twenty-five female patients with a DSM-IV diagnosis of MDD, an Inventory of Depressive Symptomatology-Self-Report score > 26, and a body mass index ≥ 27 and ≤ 43 kg/m(2) received up to 24 weeks of open-label treatment with NB32 with dietary and behavioral counseling (data collection: March 2008-July 2009). The primary endpoint was change from baseline in the Montgomery-Asberg Depression Rating Scale (MADRS) total score at 12 weeks; secondary endpoints included MADRS total score at week 24, change in weight, and Clinical Global Impressions-Improvement scale responder status (CGI-I score ≤ 2) at weeks 12 and 24 (modified intent-to-treat [mITT]: patients with ≥ 1 postbaseline MADRS total score on study drug; N = 23). RESULTS MADRS scores showed significant reductions at weeks 12 and 24 (mITT-last observation carried forward [LOCF]: -13.1 ± 7.1 and -15.3 ± 8.1, respectively, P < .001 vs baseline for all). Mean ± SD weight loss was -4.0% ± 4.6% (mITT-LOCF) and -6.1% ± 4.7% (observed cases) at week 12 and -5.3% ± 6.5% (mITT-LOCF) and -9.2% ± 6.2% (observed cases) at week 24 (P < .001 vs baseline for all). By week 24, 95% of patients (mITT-LOCF) were responders (CGI-I score ≤ 2) and 70% were in remission (CGI-I score = 1). The safety/tolerability profile of NB32 was consistent with its individual components; the most common adverse events were nausea, constipation, headache, and insomnia, with no serious adverse events attributed to NB32. CONCLUSION Twenty-four weeks of open-label NB32 therapy with dietary and behavioral counseling was associated with improvement in depressive symptoms and reduced body weight in overweight/obese women with MDD. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT00624858.
Collapse
Affiliation(s)
- Susan L McElroy
- Research Institute, Lindner Center of HOPE, Mason, Ohio (Dr McElroy); Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio (Drs McElroy and Guerdjikova); and Orexigen Therapeutics, Inc, La Jolla, California (Drs Burns, Kim, Harris-Collazo, Landbloom, and Dunayevich). Dr Kim is currently employed by Zafgen. Dr Harris-Collazo is currently employed by Santarus. Dr Landbloom is currently employed by Merck. Dr Dunayevich is currently employed by Amgen
| | | | | | | | | | | | | |
Collapse
|
16
|
Carrasco MC, Vidal J, Redolat R. Bupropion induced changes in exploratory and anxiety-like behaviour in NMRI male mice depends on the age. Behav Processes 2013; 98:117-24. [PMID: 23727544 DOI: 10.1016/j.beproc.2013.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Revised: 03/08/2013] [Accepted: 05/09/2013] [Indexed: 12/15/2022]
Abstract
The aim of this study was to assess the effects of the antidepressant bupropion on anxiety and novelty-seeking in adolescent mice of different ages and adults. Behavioural differences between early adolescent, late adolescent and adult NMRI mice were measured both in the elevated plus-maze and the hole-board tasks following acute administration of bupropion (5, 10, 15, 20mg/kg) or saline. In the plus maze test, early and late adolescent mice treated with bupropion (10, 15mg/kg, respectively) had lower percentages of entries in the open-arms compared to their vehicle controls. Adult mice treated with bupropion did not differ from their vehicle controls. These results suggest that the effect of this drug on anxiety-like behaviour in mice depends on the age, showing adolescents an anxiogenic-like profile. In the hole-board, adolescents showed more elevated levels of novelty-seeking than adults, exhibiting shorter latency to the first head-dip (HD) and a higher number of HD's. Bupropion increases the latency to the first HD and decreases the number of HD's in all age-groups, indicating a decline in exploratory tendency. Findings reveal that the age can modulate the behaviour displayed by mice in both animal models, and that adolescents are more sensitive to bupropion's anxiogenic effects.
Collapse
Affiliation(s)
- M Carmen Carrasco
- Departamento Psicobiología, Facultad de Psicología, Universitat de València, Blasco Ibañez, 21, Valencia 46010, Spain.
| | | | | |
Collapse
|
17
|
Arch JRS, Trayhurn P. Detection of thermogenesis in rodents in response to anti-obesity drugs and genetic modification. Front Physiol 2013; 4:64. [PMID: 23580228 PMCID: PMC3619105 DOI: 10.3389/fphys.2013.00064] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 03/16/2013] [Indexed: 11/22/2022] Open
Abstract
Many compounds and genetic manipulations are claimed to confer resistance to obesity in rodents by raising energy expenditure. Examples taken from recent and older literature, demonstrate that such claims are often based on measurements of energy expenditure after body composition has changed, and depend on comparisons of energy expenditure divided by body weight. This is misleading because white adipose tissue has less influence than lean tissue on energy expenditure. Application of this approach to human data would suggest that human obesity is usually due to a low metabolic rate, which is not an accepted view. Increased energy expenditure per animal is a surer way of demonstrating thermogenesis, but even then it is important to know whether this is due to altered body composition (repartitioning), or increased locomotor activity rather than thermogenesis per se. Regression analysis offers other approaches. The thermogenic response to some compounds has a rapid onset and so cannot be due to altered body composition. These compounds usually mimic or activate the sympathetic nervous system. Thermogenesis occurs in, but may not be confined to, brown adipose tissue. It should not be assumed that weight loss in response to these treatments is due to thermogenesis unless there is a sustained increase in 24-h energy expenditure. Thyroid hormones and fibroblast growth factor 21 also raise energy expenditure before they affect body composition. Some treatments and genetic modifications alter the diurnal rhythm of energy expenditure. It is important to establish whether this is due to altered locomotor activity or efficiency of locomotion. There are no good examples of compounds that do not affect short-term energy expenditure but have a delayed effect. How and under what conditions a genetic modification or compound increases energy expenditure influences the decision on whether to seek drugs for the target or take a candidate drug into clinical studies.
Collapse
|
18
|
Bello NT, Walters AL, Verpeut JL, Cunha PP. High-fat diet-induced alterations in the feeding suppression of low-dose nisoxetine, a selective norepinephrine reuptake inhibitor. J Obes 2013; 2013:457047. [PMID: 23431425 PMCID: PMC3570928 DOI: 10.1155/2013/457047] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 12/17/2012] [Indexed: 11/17/2022] Open
Abstract
Central noradrenergic pathways are involved in feeding and cardiovascular control, physiological processes altered by obesity. The present studies determined how high-fat feeding and body weight gain alter the sensitivity to the feeding suppression and neural activation to a selective norepinephrine reuptake inhibitor, nisoxetine. Acute administration of nisoxetine (saline: 0, 3, 10, and 30 mg/kg; i.p.) resulted in a dose-dependent reduction in the 24 h refeeding response in male Sprague Dawley rats maintained on standard chow. In a similar fashion, nisoxetine resulted in reductions in blood pressure and a compensatory increase in heart rate. From these studies, the 3 mg/kg dose was subthreshold. In a separate experiment, however, 10 wk exposure to a high-fat diet (60% fat) resulted in weight gain and significant feeding suppression following administration of nisoxetine (3 mg/kg) compared with animals fed a control diet (10% fat). Nisoxetine (3 mg/kg) also resulted in greater neural activation, as measured by c-Fos immunohistochemistry, in the arcuate nucleus of the hypothalamus in animals exposed to the high-fat diet. Such data indicate acute nisoxetine doses that suppress food intake can impact cardiovascular measures. It also suggests that the feeding suppression to a low-dose nisoxetine is enhanced as a result of high-fat diet and weight gain.
Collapse
Affiliation(s)
- Nicholas T Bello
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, 84 Lipman Drive, New Brunswick, NJ 08901, USA.
| | | | | | | |
Collapse
|
19
|
Clapper JR, Athanacio J, Wittmer C, Griffin PS, D'Souza L, Parkes DG, Roth JD. Effects of amylin and bupropion/naltrexone on food intake and body weight are interactive in rodent models. Eur J Pharmacol 2013. [DOI: 10.1016/j.ejphar.2012.11.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
20
|
Role of nociceptive arcuate nucleus neurons in chloroquine-induced pruritic behaviors in mice. ACTA ACUST UNITED AC 2012; 32:919-922. [DOI: 10.1007/s11596-012-1058-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Indexed: 10/27/2022]
|
21
|
Abstract
INTRODUCTION The ability of nicotine, the primary psychoactive substance in tobacco smoke, to regulate appetite and body weight is one of the factors cited by smokers that prevents them from quitting and is the primary reason for smoking initiation in teenage girls. The regulation of feeding and metabolism by nicotine is complex, and recent studies have begun to identify nicotinic acetylcholine receptor (nAChR) subtypes and circuits or cell types involved in this regulation. DISCUSSION We will briefly describe the primary anatomical and functional features of the input, output, and central integration structures of the neuroendocrine systems that regulate energy homeostasis. Then, we will describe the nAChR subtypes expressed in these structures in mammals to identify the possible molecular targets for nicotine. Finally, we will review the effects of nicotine and its withdrawal on feeding and energy metabolism and attribute them to potential central and peripheral cellular targets.
Collapse
Affiliation(s)
- Michele Zoli
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy.
| | | |
Collapse
|
22
|
Beeler JA, Frazier CRM, Zhuang X. Putting desire on a budget: dopamine and energy expenditure, reconciling reward and resources. Front Integr Neurosci 2012; 6:49. [PMID: 22833718 PMCID: PMC3400936 DOI: 10.3389/fnint.2012.00049] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 07/02/2012] [Indexed: 01/09/2023] Open
Abstract
Accumulating evidence indicates integration of dopamine function with metabolic signals, highlighting a potential role for dopamine in energy balance, frequently construed as modulating reward in response to homeostatic state. Though its precise role remains controversial, the reward perspective of dopamine has dominated investigation of motivational disorders, including obesity. In the hypothesis outlined here, we suggest instead that the primary role of dopamine in behavior is to modulate activity to adapt behavioral energy expenditure to the prevailing environmental energy conditions, with the role of dopamine in reward and motivated behaviors derived from its primary role in energy balance. Dopamine has long been known to modulate activity, exemplified by psychostimulants that act via dopamine. More recently, there has been nascent investigation into the role of dopamine in modulating voluntary activity, with some investigators suggesting that dopamine may serve as a final common pathway that couples energy sensing to regulated voluntary energy expenditure. We suggest that interposed between input from both the internal and external world, dopamine modulates behavioral energy expenditure along two axes: a conserve-expend axis that regulates generalized activity and an explore-exploit axes that regulates the degree to which reward value biases the distribution of activity. In this view, increased dopamine does not promote consumption of tasty food. Instead increased dopamine promotes energy expenditure and exploration while decreased dopamine favors energy conservation and exploitation. This hypothesis provides a mechanistic interpretation to an apparent paradox: the well-established role of dopamine in food seeking and the findings that low dopaminergic functions are associated with obesity. Our hypothesis provides an alternative perspective on the role of dopamine in obesity and reinterprets the "reward deficiency hypothesis" as a perceived energy deficit. We propose that dopamine, by facilitating energy expenditure, should be protective against obesity. We suggest the apparent failure of this protective mechanism in Western societies with high prevalence of obesity arises as a consequence of sedentary lifestyles that thwart energy expenditure.
Collapse
Affiliation(s)
- Jeff A. Beeler
- Department of Neurobiology, The University of ChicagoChicago, IL, USA
| | | | - Xiaoxi Zhuang
- Department of Neurobiology, The University of ChicagoChicago, IL, USA
- Committee on Neurobiology, The University of ChicagoChicago, IL, USA
| |
Collapse
|
23
|
Calandra C, Russo RG, Luca M. Bupropion versus sertraline in the treatment of depressive patients with binge eating disorder: retrospective cohort study. Psychiatr Q 2012; 83:177-85. [PMID: 21927936 DOI: 10.1007/s11126-011-9192-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This study sought to compare Bupropion versus Sertraline in the treatment of depressed patients with Binge Eating Disorder (BED) prescribed off-label. Medical records of outpatients with diagnosis of BED and Depression (DSM-IV-TR criteria) were selected: 15 patients were treated with bupropion 150 mg/per day, and 15 with sertraline 200 mg/per day. During the screening and control visits (2°-6°-14°-24° week), the selected patients were first weighed and then evaluated using the following questionnaires: Binge Eating Disorder-Clinical Interview (BEDCI), Beck Depression Inventory (BDI), State-Trait Anxiety Inventory X (STAI-X) and Arizona Sexual Experience Scale (ASEX). Both drugs reduced anxious-depressive symptoms and binge frequency: Bupropion showed a better effectiveness in reducing weight and improving sexual performances; weight loss related to it was proportional to the body mass index. Bupropion may be associated with more weight loss in BED, depressed patients than sertraline.
Collapse
Affiliation(s)
- Carmela Calandra
- Department of Medical and Surgery Specialties, Psychiatry Unit of the University Hospital Policlinico-Vittorio Emanuele of Catania, University of Catania, Via S. Sofia 78, 95100, Catania (Sicily), Italy.
| | | | | |
Collapse
|
24
|
van de Giessen E, de Bruin K, la Fleur SE, van den Brink W, Booij J. Triple monoamine inhibitor tesofensine decreases food intake, body weight, and striatal dopamine D2/D3 receptor availability in diet-induced obese rats. Eur Neuropsychopharmacol 2012; 22:290-9. [PMID: 21889317 DOI: 10.1016/j.euroneuro.2011.07.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 07/18/2011] [Accepted: 07/22/2011] [Indexed: 11/25/2022]
Abstract
The novel triple monoamine inhibitor tesofensine blocks dopamine, serotonin and norepinephrine re-uptake and is a promising candidate for the treatment of obesity. Obesity is associated with lower striatal dopamine D2 receptor availability, which may be related to disturbed regulation of food intake. This study assesses the effects of chronic tesofensine treatment on food intake and body weight in association with changes in striatal dopamine D2/D3 receptor (D2/3R) availability of diet-induced obese (DIO) rats. Four groups of 15 DIO rats were randomized to one of the following treatments for 28 days: 1. tesofensine (2.0 mg/kg), 2. vehicle, 3. vehicle+restricted diet isocaloric to caloric intake of group 1, and 4. tesofensine (2.0 mg/kg)+ a treatment-free period of 28 days. Caloric intake and weight gain decreased significantly more in the tesofensine-treated rats compared to vehicle-treated rats, which confirms previous findings. After treatment discontinuation, caloric intake and body weight gain gradually increased again. Tesofensine-treated rats showed significantly lower D2/3R availability in nucleus accumbens and dorsal striatum than both vehicle-treated rats and vehicle-treated rats on restricted isocaloric diet. No correlations were observed between food intake or body weight and D2/3R availability. Thus, chronic tesofensine treatment leads to decreased food intake and weight gain. However, this appears not to be directly related to the decreased striatal D2/3R availability, which is mainly a pharmacological effect.
Collapse
Affiliation(s)
- Elsmarieke van de Giessen
- Department of Nuclear Medicine, Academic Medical Center University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
25
|
Umezu T. Unusual effects of nicotine as a psychostimulant on ambulatory activity in mice. ISRN PHARMACOLOGY 2012; 2012:170981. [PMID: 22530136 PMCID: PMC3317018 DOI: 10.5402/2012/170981] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 12/26/2011] [Indexed: 01/08/2023]
Abstract
The present study examined the effect of nicotine, alone and in combination with various drugs that act on the CNS, on ambulatory activity, a behavioral index for locomotion, in ICR (CD-1) strain mice. Nicotine at 0.25–2 mg/kg acutely reduced ambulatory activity of ICR mice. The effect of nicotine was similar to that of haloperidol and fluphenazine but distinct from that of bupropion and methylphenidate. ICR mice developed tolerance against the inhibitory effect of nicotine on ambulatory activity when nicotine was repeatedly administered. This effect was also distinct from bupropion and methylphenidate as they produced augmentation of their ambulation-stimulating effects in ICR mice. Nicotine reduced the ambulation-stimulating effects of bupropion and methylphenidate as well as haloperidol and fluphenazine. Taken together, nicotine exhibited unusual effects as a psychostimulant on ambulatory activity in ICR mice.
Collapse
Affiliation(s)
- Toyoshi Umezu
- Biological Imaging and Analysis Section, Center for Environmental Measurement and Analysis, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba 305-8506, Japan
| |
Collapse
|
26
|
Ueno T, Tomita J, Kume S, Kume K. Dopamine modulates metabolic rate and temperature sensitivity in Drosophila melanogaster. PLoS One 2012; 7:e31513. [PMID: 22347491 PMCID: PMC3274542 DOI: 10.1371/journal.pone.0031513] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 01/11/2012] [Indexed: 01/06/2023] Open
Abstract
Homeothermal animals, such as mammals, maintain their body temperature by heat generation and heat dissipation, while poikilothermal animals, such as insects, accomplish it by relocating to an environment of their favored temperature. Catecholamines are known to regulate thermogenesis and metabolic rate in mammals, but their roles in other animals are poorly understood. The fruit fly, Drosophila melanogaster, has been used as a model system for the genetic studies of temperature preference behavior. Here, we demonstrate that metabolic rate and temperature sensitivity of some temperature sensitive behaviors are regulated by dopamine in Drosophila. Temperature-sensitive molecules like dTrpA1 and shits induce temperature-dependent behavioral changes, and the temperature at which the changes are induced were lowered in the dopamine transporter-defective mutant, fumin. The mutant also displays a preference for lower temperatures. This thermophobic phenotype was rescued by the genetic recovery of the dopamine transporter in dopamine neurons. Flies fed with a dopamine biosynthesis inhibitor (3-iodo-L-tyrosine), which diminishes dopamine signaling, exhibited preference for a higher temperature. Furthermore, we found that the metabolic rate is up-regulated in the fumin mutant. Taken together, dopamine has functions in the temperature sensitivity of behavioral changes and metabolic rate regulation in Drosophila, as well as its previously reported functions in arousal/sleep regulation.
Collapse
Affiliation(s)
- Taro Ueno
- Department of Stem Cell Biology, Institute of Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Jun Tomita
- Department of Stem Cell Biology, Institute of Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Shoen Kume
- Department of Stem Cell Biology, Institute of Embryology and Genetics, Kumamoto University, Kumamoto, Japan
- Global COE program, Kumamoto University, Kumamoto, Japan
| | - Kazuhiko Kume
- Department of Stem Cell Biology, Institute of Embryology and Genetics, Kumamoto University, Kumamoto, Japan
- * E-mail:
| |
Collapse
|
27
|
Leptin action in the dorsomedial hypothalamus increases sympathetic tone to brown adipose tissue in spite of systemic leptin resistance. J Neurosci 2011; 31:12189-97. [PMID: 21865462 DOI: 10.1523/jneurosci.2336-11.2011] [Citation(s) in RCA: 231] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Leptin regulates body weight in mice by decreasing appetite and increasing sympathetic nerve activity (SNA), which increases energy expenditure in interscapular brown adipose tissue (iBAT). Diet-induced obese mice (DIO) are resistant to the anorectic actions of leptin. We evaluated whether leptin still stimulated sympathetic outflow in DIO mice. We measured iBAT temperature as a marker of SNA. We found that obese hyperleptinemic mice have higher iBAT temperature than mice on regular diet. Conversely, obese leptin-deficient ob/ob mice have lower iBAT temperature. Additionally, leptin increased SNA in obese (DIO and ob/ob) and control mice, despite DIO mice being resistant to anorectic action of leptin. We demonstrated that neurons in the dorsomedial hypothalamus (DMH) of DIO mice mediate the thermogenic responses to hyperleptinemia in obese mammals because blockade of leptin receptors in the DMH prevented the thermogenic effects of leptin. Peripheral Melotan II (MTII) injection increased iBAT temperature, but it was blunted by blockade of DMH melanocortin receptors (MC4Rs) by injecting agouti-related peptide (AgRP) directly into the DMH, suggesting a physiological role of the DMH on temperature regulation in animals with normal body weight. Nevertheless, obese mice without a functional melanocortin system (MC4R KO mice) have an increased sympathetic outflow to iBAT compared with their littermates, suggesting that higher leptin levels drive sympathoexcitation to iBAT by a melanocortin-independent pathway. Because the sympathetic nervous system contributes in regulating blood pressure, heart rate, and hepatic glucose production, selective leptin resistance may be a crucial mechanism linking adiposity and metabolic syndrome.
Collapse
|
28
|
Clapham JC. Central control of thermogenesis. Neuropharmacology 2011; 63:111-23. [PMID: 22063719 DOI: 10.1016/j.neuropharm.2011.10.014] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 10/18/2011] [Accepted: 10/24/2011] [Indexed: 01/26/2023]
Abstract
In mammals and birds, conservation of body heat at around 37 °C is vital to life. Thermogenesis is the production of this heat which can be obligatory, as in basal metabolic rate, or it can be facultative such as the response to cold. A complex regulatory system has evolved which senses environmental or core temperature and integrates this information in hypothalamic regions such as the preoptic area and dorsomedial hypothalamus. These areas then send the appropriate signals to generate and conserve heat (or dissipate it). In this review, the importance of the sympathetic nervous system is discussed in relation to its role in basal metabolic rate and adaptive thermogenesis with a particular emphasis to human obesity. The efferent sympathetic pathway does not uniformly act on all tissues; different tissues can receive different levels of sympathetic drive at the same time. This is an important concept in the discussion of the pharmacotherapy of obesity. Despite decades of work the medicine chest contains only one pill for the long term treatment of obesity, orlistat, a lipase inhibitor that prevents the absorption of lipid from the gut and is itself not systemically absorbed. The central controlling system for thermogenesis has many potential intervention points. Several drugs, previously marketed, awaiting approval or in the earlier stages of development may have a thermogenic effect via activation of the sympathetic nervous system at some point in the thermoregulatory circuit and are discussed in this review. If the balance is weighted to the "wrong" side there is the burden of increased cardiovascular risk while a shift to the "right" side, if possible, will afford a thermogenic benefit that is conducive to weight loss maintenance. This article is part of a Special Issue entitled 'Central Control Food Intake'
Collapse
Affiliation(s)
- John C Clapham
- AstraZeneca R&D, Alderley Park, Macclesfield, SK10 4TG, UK.
| |
Collapse
|
29
|
Targeting thermogenesis and related pathways in anti-obesity drug discovery. Pharmacol Ther 2011; 131:295-308. [PMID: 21514319 DOI: 10.1016/j.pharmthera.2011.04.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2011] [Accepted: 03/29/2011] [Indexed: 01/12/2023]
Abstract
The health consequences of the obesity epidemic are a huge burden on patients and society. Yet it remains an unmet therapeutic need. Lifestyle or behaviour modification, although desirable, seems to benefit only a few and bariatric surgery is not an option for all and not without risks. Nevertheless, bariatric surgery is currently the gold standard in terms of weight loss therapy and any weight loss agent will be in combination with management of lifestyle modification. Sadly, there is a poor history for the pharmacological treatment of obesity and repeated safety concerns have attracted intense regulatory scrutiny. Indeed, recent market withdrawals leave us with just one agent approved for the long term treatment of obesity and that is only mildly efficacious in terms of weight loss, although it is beneficial in terms of metabolic health. There are two broad pharmacological approaches that can be applied in obesity drug discovery: reduce intake (or absorption) or increase expenditure (thermogenesis) of calories. In this review we will look at the latter approach. We will cover regulatory requirements and the rationale for this approach. We believe that post-obese subjects display abnormal metabolic responses to weight loss that almost inevitably leads to weight regain. We will then explore a number of approaches that potentially increase thermogenesis in humans. The challenge we have is in accumulating enough human data to validate this approach using drugs.
Collapse
|
30
|
Hobgood DK. Personality and illness: genetic connections? Med Hypotheses 2010; 76:89-93. [PMID: 20826063 DOI: 10.1016/j.mehy.2010.08.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Revised: 07/30/2010] [Accepted: 08/07/2010] [Indexed: 10/19/2022]
Abstract
Illness is strongly influenced by genetics. Personality traits are influenced by genetics and have linkages with, at least, affective illness. Because genetics influences both personality and illness, we studied by literature review the genes that illness and personality traits share and, by office assessment, our patient population's illnesses and personality traits. This led to the following hypothesis: illness is related to more Submissive than Dominant personality trait and thus to lower norepinephrine activity. Patient affect and personality are often noted to be subdued in chronic illness, but whether from cause or effect is difficult to determine. After reviewing the literature on wellness, personality and genes, we endeavored to explore and better delineate our impressions of illness and personality relationships and to set the stage for a genetic hypothesis by using an online personality test in an ob-gyn practice to determine which traits could be used as proxies for personality trait genes. We thus confirmed our clinical impression that women with psychiatric and/or medical illness have more Submissive personality. The medical literature on gene markers shared by Submissive personality and illness in our population suggests the hypothesis that this relationship could be mediated by genetically lower neurotransmitter activity. Both norepinephrine activity and monoamine oxidase A, the enzyme controlling its catabolism, are in concert to blunt neuronal activity in such diverse states as depression, asthma, obesity, autoimmune illness, and in smokers. Conversely, norepinephrine activity is enhanced in breastfeeders and in other settings known to be related to good health. A point for possible divergence from the hypothesis is that African-Americans have enhanced norepinephrine activity yet poorer health outcomes although the effect of environmental risk factors in African-Americans is considered prominent by researchers. The possible importance of this hypothesis is that it changes emphasis slightly from one of splitting diagnoses to one of grouping them to achieve fresh insight. If we assess what is found in common in diverse illnesses of mind and body, we have yet another tool to assess causation. Although this hypothesis is offered somewhat in a boldness of spirit to take a new look at our many perplexing clinical problems, it is also offered with humility in light of the complexity with which we deal in patient care.
Collapse
Affiliation(s)
- Donna K Hobgood
- Women's Institute for Specialized Health, University of Tennessee at Chattanooga Medical Units, Chattanooga, TN 37403, USA.
| |
Collapse
|
31
|
Kennett GA, Clifton PG. New approaches to the pharmacological treatment of obesity: can they break through the efficacy barrier? Pharmacol Biochem Behav 2010; 97:63-83. [PMID: 20688100 DOI: 10.1016/j.pbb.2010.07.020] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Revised: 07/22/2010] [Accepted: 07/27/2010] [Indexed: 01/31/2023]
Abstract
In this review we assess the range of centrally active anorectics that are either in human clinical trials, or are likely to be so in the near future. We describe their weight loss efficacy, mode of action at both pharmacological and behavioural levels, where understood, together with the range of side effects that might be expected in clinical use. We have however evaluated these compounds against the considerably more rigorous criteria that are now being used by the Federal Drugs Agency and European Medicines Agency to decide approvals and market withdrawals. Several trends are evident. Recent advances in the understanding of energy balance control have resulted in the exploitation of a number of new targets, some of which have yielded promising data in clinical trials for weight loss. A second major trend is derived from the hypothesis that improved weight loss efficacy over current therapy is most likely to emerge from treatments targeting multiple mechanisms of energy balance control. This reasoning has led to the development of a number of new treatments for obesity where multiple mechanisms are targeted, either by a single molecule, such as tesofensine, or through drug combinations such as qnexa, contrave, empatic, and pramlintide+metreleptin. Many of these approaches also utilise advances in formulation technology to widen safety margins. Finally, the practicality of peptide therapies for obesity has become better validated in recent studies and this may allow more rapid exploitation of novel targets, rather than awaiting the development of orally available small molecules. We conclude that novel, more efficacious and better tolerated treatments for obesity may become available in the near future.
Collapse
Affiliation(s)
- G A Kennett
- Saretius Limited, Science and Technology Centre, Earley Gate, University of Reading, Reading, Berkshire, UK.
| | | |
Collapse
|
32
|
Tesofensine, a novel triple monoamine reuptake inhibitor, induces appetite suppression by indirect stimulation of alpha1 adrenoceptor and dopamine D1 receptor pathways in the diet-induced obese rat. Neuropsychopharmacology 2010; 35:1464-76. [PMID: 20200509 PMCID: PMC3055463 DOI: 10.1038/npp.2010.16] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Tesofensine is a novel monoamine reuptake inhibitor that inhibits both norepinephrine, 5-HT, and dopamine (DA) reuptake function. Tesofensine is currently in clinical development for the treatment of obesity, however, the pharmacological basis for its strong effect in obesity management is not clarified. Using a rat model of diet-induced obesity (DIO), we characterized the pharmacological mechanisms underlying the appetite suppressive effect of tesofensine. DIO rats treated with tesofensine (2.0 mg/kg, s.c.) for 16 days showed significantly lower body weights than vehicle-treated DIO rats, being reflected by a marked hypophagic response. Using an automatized food intake monitoring system during a 12 h nocturnal test period, tesofensine-induced hypophagia was investigated further by studying the acute interaction of a variety of monoamine receptor antagonists with tesofensine-induced hypophagia in the DIO rat. Tesofensine (0.5-3.0 mg/kg, s.c.) induced a dose-dependent and marked decline in food intake with an ED(50) of 1.3 mg/kg. The hypophagic response of tesofensine (1.5 mg/kg, s.c.) was almost completely reversed by co-administration of prazosin (1.0 mg/kg, alpha(1) adrenoceptor antagonist) and partially antagonized by co-administration of SCH23390 (0.03 mg/kg, DA D(1) receptor antagonist). In contrast, tesofensine-induced hypophagia was not affected by RX821002 (0.3 mg/kg, alpha(2) adrenoceptor antagonist), haloperidol (0.03 mg/kg, D(2) receptor antagonist), NGB2904 (0.1 mg/kg, D(3) receptor antagonist), or ritanserin (0.03 mg/kg, 5-HT(2A/C) receptor antagonist). Hence, the mechanism underlying the suppression of feeding by tesofensine in the obese rat is dependent on the drug's ability to indirectly stimulate alpha(1) adrenoceptor and DA D(1) receptor function.
Collapse
|
33
|
Roitman MF, Wescott S, Cone JJ, McLane MP, Wolfe HR. MSI-1436 reduces acute food intake without affecting dopamine transporter activity. Pharmacol Biochem Behav 2010; 97:138-43. [PMID: 20478327 DOI: 10.1016/j.pbb.2010.05.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 04/15/2010] [Accepted: 05/10/2010] [Indexed: 01/16/2023]
Abstract
Many therapies designed to reduce food intake and body weight act, in part, by blocking the dopamine transporter (DAT) - a protein responsible for clearing extracellular dopamine (DA) after release thereby terminating its action. Here, we found that a single injection of the drug trodusquemine (MSI-1436) decreased food intake in rats. To assess the effects of MSI-1436 on DAT function, fast-scan cyclic voltammetry was used to measure DA concentration changes in the ventral striatum. DA release was evoked by electrical stimulation of the ventral tegmental area every 5 min. After 3 baseline measurements, rats were injected with MSI-1436 (10 mg/kg), the known DAT blocker bupropion (80 mg/kg) or saline and evoked DA release and reuptake were monitored for an additional hour. Neither saline nor MSI-1436 caused a significant change in the magnitude of evoked release from baseline values whereas bupropion caused a significant increase. In addition, neither saline nor MSI-1436 significantly increased DA decay rates while such an increase was observed with bupropion. Thus, over a time course when MSI-1436 suppresses food intake it does not affect DAT function. The results support MSI-1436 as an anti-obesity treatment which spares DAT.
Collapse
Affiliation(s)
- Mitchell F Roitman
- Department of Psychology, University of Illinois, Chicago, IL 60607, USA.
| | | | | | | | | |
Collapse
|
34
|
Hansen HH, Hansen G, Tang-Christensen M, Larsen PJ, Axel AMD, Raben A, Mikkelsen JD. The novel triple monoamine reuptake inhibitor tesofensine induces sustained weight loss and improves glycemic control in the diet-induced obese rat: comparison to sibutramine and rimonabant. Eur J Pharmacol 2010; 636:88-95. [PMID: 20385125 DOI: 10.1016/j.ejphar.2010.03.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 02/22/2010] [Accepted: 03/14/2010] [Indexed: 11/19/2022]
Abstract
Tesofensine, a novel triple monoamine reuptake inhibitor, produces a significant weight loss in humans. The present study aimed at characterizing the weight-reducing effects of tesofensine in a rat model of diet-induced obesity. Sibutramine and rimonabant were used as reference comparators. Compared to baseline, long-term treatment with tesofensine (28 days, 1.0 or 2.5mg/kg, p.o.) resulted in a significant, dose-dependent and sustained weight loss of 5.7 and 9.9%, respectively. Sibutramine (7.5mg/kg, p.o.) treatment caused a sustained weight loss of 7.6%, whereas the employed dose of rimonabant (10mg/kg, p.o.) only produced a transient weight reduction. While all compounds exhibited a significant inhibitory effect on food intake which gradually wore off, the hypophagic effect of tesofensine was longer lasting than sibutramine and rimonabant. In contrast to tesofensine, the body weight of pair-fed rats returned to baseline at the end of the study, which may indicate that tesofensine stimulated energy expenditure. The differential efficacy on weight reduction was also reflected in lowered body fat depots, as tesofensine and sibutramine most efficiently reduced abdominal and subcutaneous fat mass which was paralleled by reduced plasma lipid levels. In an oral glucose tolerance test, only tesofensine significantly suppressed the plasma insulin response below the level that could be obtained by paired feeding, indicating that tesofensine further improved glycemic control. In conclusion, the robust weight loss with long-term tesofensine treatment is likely due to a combined synergistic effect of appetite suppression and increased energy expenditure.
Collapse
Affiliation(s)
- Henrik H Hansen
- NeuroSearch A/S, Pederstrupvej 93, DK-2750 Ballerup, Copenhagen, Denmark.
| | | | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Lai CH. Duloxetine related binge eating behaviors: a case report. Prog Neuropsychopharmacol Biol Psychiatry 2009; 33:1581-2. [PMID: 19778574 DOI: 10.1016/j.pnpbp.2009.09.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 09/11/2009] [Accepted: 09/15/2009] [Indexed: 11/18/2022]
|
37
|
|
38
|
Rajput SK, Krishnamoorthy S, Pawar C, Kaur N, Monga V, Meena CL, Jain R, Sharma SS. Antiepileptic potential and behavioral profile of L-pGlu-(2-propyl)-L-His-L-ProNH2, a newer thyrotropin-releasing hormone analog. Epilepsy Behav 2009; 14:48-53. [PMID: 18952198 DOI: 10.1016/j.yebeh.2008.10.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 09/25/2008] [Accepted: 10/05/2008] [Indexed: 11/27/2022]
Abstract
Thyrotropin-releasing hormone (TRH) and its analogs have a number of neurobiological functions and therapeutic uses in disorders of the central nervous system. In this study, the newly synthesized TRH analogs were evaluated for central nervous system activity in pentobarbital-induced sleeping in mice. The most potent TRH analog (L-pGlu-(2-propyl)-L-His-L-ProNH(2) coded as NP-647) was evaluated for its antiepileptic potential in various seizure models in mice in comparison with TRH. Intravenous pretreatment with NP-647 (10 and 20 micromol/kg body wt) significantly delayed the onset and reduced the frequency of convulsions in the pentylenetetrazole model, but not in the maximum electroshock seizure model. Also, it was found to be protective against picrotoxin- and kainic acid-induced seizures. However, NP-647 did not significantly affect theophylline-induced seizures. Further study of the effect of NP-647 on locomotor activity and a functional observational battery revealed that it did not significantly exhibit any undesirable effects as compared with vehicle and TRH. NP-647 did not significantly affect cerebral blood flow, whereas the native peptide TRH markedly increased cerebral blood flow. Furthermore, NP-647 exerted antiepileptic activity without significantly altering plasma thyroid-stimulating hormone levels and mean arterial blood pressure. This suggests that NP-647 is more selective for central nervous system activity and devoid of hormonal and cerebrovascular system effects. In contrast, TRH exhibited cardiac and endocrine effects as marked by significant elevation in mean arterial blood pressure and plasma thyroid-stimulating hormone levels. This study demonstrates that NP-647 has potential antiepileptic activity devoid of undesirable effects and, thus, can be exploited for the prevention and treatment of epilepsy.
Collapse
Affiliation(s)
- Satyendra Kumar Rajput
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Greenway FL, Whitehouse MJ, Guttadauria M, Anderson JW, Atkinson RL, Fujioka K, Gadde KM, Gupta AK, O'Neil P, Schumacher D, Smith D, Dunayevich E, Tollefson GD, Weber E, Cowley MA. Rational design of a combination medication for the treatment of obesity. Obesity (Silver Spring) 2009; 17:30-9. [PMID: 18997675 DOI: 10.1038/oby.2008.461] [Citation(s) in RCA: 208] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Existing obesity therapies are limited by safety concerns and modest efficacy reflecting a weight loss plateau. Here, we explore combination therapy with bupropion (BUP), a putative stimulator of melanocortin pathways, and an opioid antagonist, naltrexone (NAL), to antagonize an inhibitory feedback loop that limits sustained weight reduction. In vitro electrophysiologic experiments were conducted to determine the extent to which BUP+NAL stimulated hypothalamic pro-opiomelanocortin (POMC) neurons in mouse brain. A subsequent study further characterized the effect of combination BUP+NAL treatment on food intake in lean and obese mice. Finally, a randomized, blinded, placebo-controlled trial in obese adult subjects was conducted. Randomization included: BUP (300 mg) + NAL (50 mg), BUP (300 mg) + placebo (P), NAL (50 mg) + P or P+P for up to 24 weeks. BUP+NAL stimulated murine POMC neurons in vitro and caused a greater reduction in acute food intake than either monotherapy, an effect consistent with synergism. Combined BUP+NAL provided sustained weight loss without evidence of an efficacy plateau through 24 weeks of treatment. BUP+NAL completers diverged from NAL+P (P < 0.01) and P+P (P < 0.001) at week 16 and from BUP+P by week 24 (P < 0.05). The combination was also well tolerated. Translational studies indicated that BUP+NAL therapy produced synergistic weight loss which exceeded either BUP or NAL alone. These results supported the hypothesis that NAL, through blockade of beta-endorphin mediated POMC autoinhibition, prevents the classic weight loss plateau observed with monotherapies such as BUP. This novel treatment approach (BUP+NAL) holds promise for the treatment of obesity.\
Collapse
Affiliation(s)
- Frank L Greenway
- 1Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Tyrosine hydroxylase deficit in the chemoafferent and the sympathoadrenergic pathways of the Mecp2 deficient mouse. Neurosci Lett 2008; 447:82-6. [DOI: 10.1016/j.neulet.2008.09.045] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Revised: 09/12/2008] [Accepted: 09/18/2008] [Indexed: 11/18/2022]
|