1
|
Moon DO. MetAP2 as a Therapeutic Target for Obesity and Type 2 Diabetes: Structural Insights, Mechanistic Roles, and Inhibitor Development. Biomolecules 2024; 14:1572. [PMID: 39766279 PMCID: PMC11673396 DOI: 10.3390/biom14121572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/07/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Type 2 Diabetes Mellitus (T2DM) and obesity are globally prevalent metabolic disorders characterized by insulin resistance, impaired glucose metabolism, and excessive adiposity. Methionine aminopeptidase 2 (MetAP2), an intracellular metalloprotease, has emerged as a promising therapeutic target due to its critical role in regulating lipid metabolism, energy balance, and protein synthesis. This review provides a comprehensive analysis of MetAP2, including its structural characteristics, catalytic mechanism, and functional roles in the pathophysiology of T2DM and obesity. The unique architecture of MetAP2's active site and its interactions with substrates are examined to elucidate its enzymatic function. The review also explores the development of MetAP2 inhibitors, focusing on their mechanisms of action, preclinical and clinical findings, and therapeutic potential. Special emphasis is placed on docking studies to analyze the binding interactions of six key inhibitors (fumagillin, TNP-470, beloranib, ZGN-1061, indazole, and pyrazolo[4,3-b]indole) with MetAP2, revealing their structural determinants for efficacy and specificity. These findings underscore the potential of MetAP2 as a therapeutic target and provide valuable insights for the rational design of next-generation inhibitors to address obesity and T2DM.
Collapse
Affiliation(s)
- Dong Oh Moon
- Department of Biology Education, Daegu University, 201, Daegudae-ro, Gyeongsan-si 38453, Gyeongsangbuk-do, Republic of Korea
| |
Collapse
|
2
|
Han JC, Rasmussen MC, Forte AR, Schrage SB, Zafar SK, Haqq AM. Management of Monogenic and Syndromic Obesity. Gastroenterol Clin North Am 2023; 52:733-750. [PMID: 37919024 DOI: 10.1016/j.gtc.2023.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Similar to the general population, lifestyle interventions focused on nutrition and physical activity form the foundation for treating obesity caused by rare genetic disorders. Additional therapies, including metreleptin and setmelanotide, that target defects within the leptin signaling pathway can effectively synergize with lifestyle efforts to treat monogenic disorders of leptin, leptin receptor, proopiomelanocortin (POMC), and proprotein convertase subtilisin/kexin type 1 (PCSK1) and syndromic conditions, such as the ciliopathies Bardet-Biedl and Alström syndromes, whose pathophysiological mechanisms also converge on the leptin pathway. Investigational treatments for Prader-Willi syndrome target specific defects caused by reduced expression of paternally derived genes within the chromosome 15q region.
Collapse
Affiliation(s)
- Joan C Han
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Marcus C Rasmussen
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison R Forte
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stephanie B Schrage
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah K Zafar
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea M Haqq
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
3
|
Lee HJ, Jin BY, Park MR, Kim NH, Seo KS, Jeong YT, Wada T, Lee JS, Choi SH, Kim DH. Inhibition of adipose tissue angiogenesis prevents rebound weight gain after caloric restriction in mice fed a high-fat diet. Life Sci 2023; 332:122101. [PMID: 37730110 DOI: 10.1016/j.lfs.2023.122101] [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: 01/05/2023] [Revised: 08/30/2023] [Accepted: 09/15/2023] [Indexed: 09/22/2023]
Abstract
AIMS We investigated whether modulation of white adipose tissue (WAT) vasculature regulates rebound weight gain (RWG) after caloric restriction (CR) in mice fed a high-fat diet (HFD). MAIN METHODS We compared changes in energy balance, hypothalamic neuropeptide gene expression, and characteristics of WAT by RT-qPCR, ELISA, immunohistochemistry, and adipose-derived stromal vascular fraction spheroid sprouting assay in obese mice fed a HFD ad libitum (HFD-AL), mice under 40 % CR for 3 or 4 weeks, mice fed HFD-AL for 3 days after CR (CRAL), and CRAL mice treated with TNP-470, an angiogenic inhibitor. KEY FINDINGS WAT angiogenic genes were expressed at low levels, but WAT vascular density was maintained in the CR group compared to that in the HFD-AL group. The CRAL group showed RWG, fat regain, and hyperphagia with higher expression of angiogenic genes and reduced pericyte coverage of the endothelium in WAT on day 3 after CR compared to the CR group, indicating rapidly increased angiogenic activity after CR. Administration of TNP-470 suppressed RWG, fat regain, and hyperphagia only after CR compared to the CRAL group. Changes in circulating leptin levels and hypothalamic neuropeptide gene expression were correlated with changes in weight and fat mass, suggesting that TNP-470 suppressed hyperphagia independently of the hypothalamic melanocortin system. Additionally, TNP-470 increased gene expression related to thermogenesis, fuel utilization, and browning in brown adipose tissue (BAT) and WAT, indicating TNP-470-induced increase in thermogenesis. SIGNIFICANCE Modulation of the WAT vasculature attenuates RWG after CR by suppressing hyperphagia and increasing BAT thermogenesis and WAT browning.
Collapse
Affiliation(s)
- Hye-Jin Lee
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Republic of Korea; BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Bo-Yeong Jin
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Republic of Korea; BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Mi-Rae Park
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Republic of Korea; BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Nam Hoon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Kwan Sik Seo
- Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Yong Taek Jeong
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Tsutomu Wada
- Department of Clinical Pharmacology, University of Toyama, Toyama 930-0194, Japan
| | - Jun-Seok Lee
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Sang-Hyun Choi
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Dong-Hoon Kim
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Republic of Korea; BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea.
| |
Collapse
|
4
|
Yoysungnoen B, Srisawat U, Piyabhan P, Duansak N, Sookprasert N, Mathuradavong N, Poomipark N, Munkong N, Tingpej P, Changtam C. Short term effect of tetrahydrocurcumin on adipose angiogenesis in very high-fat diet-induced obesity mouse model. Front Nutr 2023; 10:1221935. [PMID: 37876615 PMCID: PMC10591188 DOI: 10.3389/fnut.2023.1221935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 09/26/2023] [Indexed: 10/26/2023] Open
Abstract
Tetrahydrocurcumin (THC) has been shown to possess anti-angiogenic activities. This study aims to investigate the effects of THC on adipose angiogenesis and expression of angiogenic factors that occurs in 60% high-fat diet-induced obese mice. Male ICR mice were randomly divided into 3 groups: mice fed with a low-fat diet (LFD group); mice fed with very high fat diet (VHFD group), and mice fed with VHFD supplemented with THC (300 mg/kg/day orally) (VHFD+THC treated group) for 6 weeks. Body weight (BW), food intake, fasting blood sugar (FBS), lipid profiles and visceral fats weight (VF) were measured. The microvascular density (MVD), TNF-α, VEGF, MMP-2, and MMP-9 expressions were evaluated. The VHFD group had significantly increased total cholesterol, triglyceride, food intake, BW, VF, VF/BW ratio, adipocyte size and the number of crown-liked structures as compared to LFD group. THC supplementation markedly reduced these parameters and adipocyte hypertrophy and inflammation in white adipose tissues. MVD, TNF-α, VEGF, MMP-2, and MMP-9 were over-expressed in the VHFD group. However, THC supplementation decreased MVD and reduced expression of TNF-α, VEGF, MMP-2, and MMP-9. In conclusion, THC suppressed angiogenesis in adipose tissue by the downregulation of TNF-α, VEGF, MMP-2, and MMP-9. With its effects on lipid metabolism as well as on food consumption, THC could contribute to lower visceral fat and body weight. Overall, our study demonstrated the potential benefit of THC in mitigating obesity and associated metabolic disorders along with elucidated the suppression of adipose angiogenesis as one of its underlying mechanisms.
Collapse
Affiliation(s)
- Bhornprom Yoysungnoen
- Division of Physiology, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Umarat Srisawat
- Division of Physiology, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Pritsana Piyabhan
- Division of Physiology, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Naphatsanan Duansak
- Division of Physiology, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Nattapon Sookprasert
- Division of Physiology, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Nakorn Mathuradavong
- Division of Physiology, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Natwadee Poomipark
- Division of Biochemistry, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Narongsuk Munkong
- Department of Pathology, School of Medicine, University of Phayao, Phayao, Thailand
| | - Pholawat Tingpej
- Division of Microbiology and Immunology, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Chatchawan Changtam
- Division of Physical Science, Faculty of Science and Technology, Huachiew Chalermprakiet University, Samutprakarn, Thailand
| |
Collapse
|
5
|
Wang B, Du M. Increasing adipocyte number and reducing adipocyte size: the role of retinoids in adipose tissue development and metabolism. Crit Rev Food Sci Nutr 2023; 64:10608-10625. [PMID: 37427553 PMCID: PMC10776826 DOI: 10.1080/10408398.2023.2227258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
The rising prevalence of obesity is a grave public health threat. In response to excessive energy intake, adipocyte hypertrophy impairs cellular function and leads to metabolic dysfunctions while de novo adipogenesis leads to healthy adipose tissue expansion. Through burning fatty acids and glucose, the thermogenic activity of brown/beige adipocytes can effectively reduce the size of adipocytes. Recent studies show that retinoids, especially retinoic acid (RA), promote adipose vascular development which in turn increases the number of adipose progenitors surrounding the vascular vessels. RA also promotes preadipocyte commitment. In addition, RA promotes white adipocyte browning and stimulates the thermogenic activity of brown/beige adipocytes. Thus, vitamin A is a promising anti-obesity micronutrient.
Collapse
Affiliation(s)
- Bo Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, PR China
| | - Min Du
- Laboratory of Nutrigenomics and Growth Biology, Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
6
|
Liao ZZ, Ran L, Qi XY, Wang YD, Wang YY, Yang J, Liu JH, Xiao XH. Adipose endothelial cells mastering adipose tissues metabolic fate. Adipocyte 2022; 11:108-119. [PMID: 35067158 PMCID: PMC8786343 DOI: 10.1080/21623945.2022.2028372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 11/16/2022] Open
Abstract
Dynamic communication within adipose tissue depends on highly vascularized structural characteristics to maintain systemic metabolic homoeostasis. Recently, it has been noted that adipose endothelial cells (AdECs) act as essential bridges for biological information transmission between adipose-resident cells. Hence, paracrine regulators that mediate crosstalk between AdECs and adipose stromal cells were summarized. We also highlight the importance of AdECs to maintain adipocytes metabolic homoeostasis by regulating insulin sensitivity, lipid turnover and plasticity. The differential regulation of AdECs in adipose plasticity often depends on vascular density and metabolic states. Although choosing pro-angiogenic or anti-angiogenic therapies for obesity is still a matter of debate in clinical settings, the growing numbers of drugs have been confirmed to play an anti-obesity effect by affecting vascularization. Pharmacologic angiogenesis intervention has great potential as therapeutic strategies for obesity.
Collapse
Affiliation(s)
- Zhe-Zhen Liao
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Li Ran
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiao-Yan Qi
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ya-Di Wang
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuan-Yuan Wang
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jing Yang
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiang-Hua Liu
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xin-Hua Xiao
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
7
|
Abstract
While most tissues exhibit their greatest growth during development, adipose tissue is capable of additional massive expansion in adults. Adipose tissue expandability is advantageous when temporarily storing fuel for use during fasting, but becomes pathological upon continuous food intake, leading to obesity and its many comorbidities. The dense vasculature of adipose tissue provides necessary oxygen and nutrients, and supports delivery of fuel to and from adipocytes under fed or fasting conditions. Moreover, the vasculature of adipose tissue comprises a major niche for multipotent progenitor cells, which give rise to new adipocytes and are necessary for tissue repair. Given the multiple, pivotal roles of the adipose tissue vasculature, impairments in angiogenic capacity may underlie obesity-associated diseases such as diabetes and cardiometabolic disease. Exciting new studies on the single-cell and single-nuclei composition of adipose tissues in mouse and humans are providing new insights into mechanisms of adipose tissue angiogenesis. Moreover, new modes of intercellular communication involving micro vesicle and exosome transfer of proteins, nucleic acids and organelles are also being recognized to play key roles. This review focuses on new insights on the cellular and signaling mechanisms underlying adipose tissue angiogenesis, and on their impact on obesity and its pathophysiological consequences.
Collapse
|
8
|
A Feed Additive Containing Encapsulated 6-Phytase within Recombinant Yarrowia lipolytica Cells Produced by Cultivation on Fat-Containing Waste. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12063094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Feed phytases are purchased as a dry culture medium of secreting producers, mostly micellar fungi. These preparations are required to withstand heating up to 75–80 °C because they are intended for mixing with feed components with subsequent granulation by spray drying. For this reason, many phytases that have a high specific activity at 37 °C and correspond to the optimal pH of intestinal chyme are not used in practice. A novel expression system allowing accumulation of the phytase from Obesumbacterium proteus within yeast Yarrowia lipolytica was proposed. Encapsulation increases thermal stability of the enzyme from 55 °C up to 70 °C. The obtained preparation exhibited a high impact on the daily weight gain of a weaned mouse model fed a phosphorus-deficient diet at a dosage 165 phytase activity units (FYT)/kg, whereas a commercial phytase preparation—Ladozyme Proxi derived from Aspergillus ficuum—did not improve the daily weight gain even at the dosage of 15,000 FYT/kg.
Collapse
|
9
|
Dimitri P. Treatment of Acquired Hypothalamic Obesity: Now and the Future. Front Endocrinol (Lausanne) 2022; 13:846880. [PMID: 35464063 PMCID: PMC9019363 DOI: 10.3389/fendo.2022.846880] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
The hypothalamus is the centre of neuroendocrine regulation of energy homeostasis and appetite. Maldevelopment of, or damage to, the key hypothalamic nuclei disrupts the coordinated balance between energy intake and expenditure leading, to rapid and excessive weight gain. Hypothalamic obesity is compounded by a disruption of the hypothalamic-pituitary axis, sleep disruption, visual compromise, and neurological and vascular sequalae. Amongst suprasellar tumors, craniopharyngioma is the most common cause of acquired hypothalamic obesity, either directly or following surgical or radiotherapeutic intervention. At present, therapy is limited to strategies to manage obesity but with a modest and variable impact. Current approaches include optimizing pituitary hormone replacement, calorie restriction, increased energy expenditure through physical activity, behavioral interventions, pharmacotherapy and bariatric surgery. Current pharmacotherapeutic approaches include stimulants that increase energy consumption, anti-diabetic agents, hypothalamic-pituitary substitution therapy, octreotide, and methionine aminopeptidase 2 (MetAP2) inhibitors. Some pharmacological studies of hypothalamic obesity report weight loss or stabilization but reported intervention periods are short, and others report no effect. The impact of bariatric surgery on weight loss in hypothalamic obesity again is variable. Novel or combined approaches to manage hypothalamic obesity are thus required to achieve credible and sustained weight loss. Identifying etiological factors contributing hypothalamic obesity may lead to multi-faceted interventions targeting hyperphagia, insulin resistance, decreased energy expenditure, sleep disturbance, hypopituitarism and psychosocial morbidity. Placebo-controlled trials using current single, or combination therapies are required to determine the impact of therapeutic agents. A well-defined approach to defining the location of hypothalamic damage may support the use of future targeted therapies. Intranasal oxytocin is currently being investigated as an anorexogenic agent. Novel agents including those targeting pro-opimelanocortin-C and AgRP/NPY expressing neurons and the MC4 receptor may result in better outcomes. This article discusses the current challenges in the management of hypothalamic obesity in children and young people and future therapeutic approaches to increasing weight loss and quality of life in these patients.
Collapse
Affiliation(s)
- Paul Dimitri
- The Department of Paediatric Endocrinology, Sheffield Children’s NHS Foundation Trust, Sheffield, United Kingdom
- College of Health, Wellbeing and Life Sciences, Sheffield Hallam University, Sheffield, United Kingdom
- *Correspondence: Paul Dimitri,
| |
Collapse
|
10
|
Craig SL, Irwin N, Gault VA. Xenin and Related Peptides: Potential Therapeutic Role in Diabetes and Related Metabolic Disorders. CLINICAL MEDICINE INSIGHTS-ENDOCRINOLOGY AND DIABETES 2021; 14:11795514211043868. [PMID: 34588834 PMCID: PMC8474313 DOI: 10.1177/11795514211043868] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/09/2021] [Indexed: 11/16/2022]
Abstract
Xenin bioactivity and its role in normal physiology has been investigated by several research groups since its discovery in 1992. The 25 amino acid peptide hormone is secreted from the same enteroendocrine K-cells as the incretin hormone glucose-dependent insulinotropic polypeptide (GIP), with early studies highlighting the biological significance of xenin in the gastrointestinal tract, along with effects on satiety. Recently there has been more focus directed towards the role of xenin in insulin secretion and potential for diabetes therapies, especially through its ability to potentiate the insulinotropic actions of GIP as well as utilisation in dual/triple acting gut hormone therapeutic approaches. Currently, there is a lack of clinically approved therapies aimed at restoring GIP bioactivity in type 2 diabetes mellitus, thus xenin could hold real promise as a diabetes therapy. The biological actions of xenin, including its ability to augment insulin secretion, induce satiety effects, as well as restoring GIP sensitivity, earmark this peptide as an attractive antidiabetic candidate. This minireview will focus on the multiple biological actions of xenin, together with its proposed mechanism of action and potential benefits for the treatment of metabolic diseases such as diabetes.
Collapse
Affiliation(s)
- Sarah L Craig
- Faculty of Life and Health Sciences, School of Biomedical Sciences, Ulster University, UK
| | - Nigel Irwin
- Faculty of Life and Health Sciences, School of Biomedical Sciences, Ulster University, UK
| | - Victor A Gault
- Faculty of Life and Health Sciences, School of Biomedical Sciences, Ulster University, UK
| |
Collapse
|
11
|
The methionine aminopeptidase 2 inhibitor, TNP-470, enhances the antidiabetic properties of sitagliptin in mice by upregulating xenin. Biochem Pharmacol 2020; 183:114355. [PMID: 33279496 DOI: 10.1016/j.bcp.2020.114355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/12/2020] [Accepted: 12/01/2020] [Indexed: 12/24/2022]
Abstract
The therapeutic mechanism of action of methionine aminopeptidase 2 (MetAP2) inhibitors for obesity-diabetes has not yet been fully defined. Xenin, a K-cell derived peptide hormone, possesses an N-terminal Met amino acid residue. Thus, elevated xenin levels could represent a potential pharmacological mechanism of MetAP2 inhibitors, since long-acting xenin analogues have been shown to improve obesity-diabetes. The present study has assessed the ability of the MetAP2 inhibitor, TNP-470, to augment the antidiabetic utility of the incretin-enhancer drug, sitagliptin, in high fat fed (HFF) mice. TNP-470 (1 mg/kg) and sitagliptin (25 mg/kg) were administered once-daily alone, or in combination, to diabetic HFF mice (n = 10) for 18 days. Individual therapy with TNP-470 or sitagliptin resulted in numerous metabolic benefits including reduced blood glucose, increased circulating and pancreatic insulin and improved glucose tolerance, insulin sensitivity, pyruvate tolerance and overall pancreatic islet architecture. Further assessment of metabolic rate revealed that all treatments reduced respiratory exchange ratio and increased locomotor activity. All sitagliptin treated mice also exhibited increased energy expenditure. In addition, treatment with TNP-470 alone, or in combination with sitagliptin, reduced food intake and body weight, as well as elevating plasma and intestinal xenin. Importantly, combined sitagliptin and TNP-470 therapy was associated with further significant benefits beyond that observed by either treatment alone. This included more rapid restoration of normoglycaemia, superior glucose tolerance, increased circulating GIP concentrations and an enhanced pancreatic beta:alpha cell ratio. In conclusion, these data demonstrate that TNP-470 increases plasma and intestinal xenin levels, and augments the antidiabetic advantages of sitagliptin.
Collapse
|
12
|
A multitarget angiogenesis inhibitor, CTT peptide-endostatin mimic-kringle 5, prevents diet-induced obesity. J Mol Med (Berl) 2020; 98:1753-1765. [PMID: 33141247 DOI: 10.1007/s00109-020-01993-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 09/28/2020] [Accepted: 10/13/2020] [Indexed: 12/14/2022]
Abstract
Adipose tissue vasculature has been considered an attractive target for prevention and treatment of obesity. AARP (CTT peptide-endostatin mimic-kringle 5) is a novel multitarget fusion protein against tumor angiogenesis. This study aimed to examine the effects of AARP on diet-induced obesity and its possible molecular mechanism. Treatment with AARP markedly prevented weight gains, improved metabolic disturbances, and decreased adipose tissue angiogenesis in diet-induced obese mice without noticeable toxicities. In addition to its potent antiangiogenic and MMP-2/9 inhibitory activities, AARP administration also significantly increased energy expenditure, influenced the metabolic and angiogenic gene expression profiles, and attenuated obesity-induced inflammation, demonstrating its systemic beneficial effects. Importantly, AARP exhibited no effect on mice fed with standard normal mouse diet. Furthermore, the AARP-treated HFD-fed mice experienced a significant increase in lifespan during the posttreatment observation period, compared with untreated HFD-fed mice. Our results suggest that AARP might be pharmacologically useful for treatment of obesity or obesity-related metabolic disorders in humans. KEY MESSAGES: What is already known • More effective and safe therapies for obesity are in urgent need. • AARP is a novel multitarget fusion protein against tumor angiogenesis. What this study adds • AARP prevents obesity, improves metabolic disorders in mice fed high-fat diet. • AARP increases energy expenditure, decreases adipose tissue angiogenesis, and increases lifespan. • AARP is well tolerated and exhibits no observable toxicity. Clinical significance • AARP may be a promising therapeutic agent against obesity or obesity-related metabolic disturbances.
Collapse
|
13
|
Shin SS, Yoon M. Regulation of Obesity by Antiangiogenic Herbal Medicines. Molecules 2020; 25:molecules25194549. [PMID: 33020443 PMCID: PMC7582783 DOI: 10.3390/molecules25194549] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 12/11/2022] Open
Abstract
Obesity is the result of an energy imbalance caused by an increased ratio of caloric intake to energy expenditure. In conjunction with obesity, related metabolic disorders, such as dyslipidemia, atherosclerosis, and type 2 diabetes, have become global health problems. Obesity progression is thought to be associated with angiogenesis and extracellular matrix (ECM) remodeling. Angiogenesis occurs in growing adult adipose tissues, which are similar to neoplastic tissues. Adipose tissue is highly vascularized, and each adipocyte is nourished by an extensive capillary network. Adipocytes produce proangiogenic factors, such as vascular endothelial growth factor A and fibroblast growth factor 2, which promote neovascularization within the adipose tissue. Furthermore, matrix metalloproteinases (MMPs), including MMP-2 and MMP-9, play important roles in adipose tissue development and microvessel maturation by modifying the ECM. Thus, modulation of angiogenesis and MMP activity provides a promising therapeutic approach for controlling human obesity and its related disorders. Over the past decade, there has been a great increase in the use of alternative treatments, such as herbal remedies, for these diseases. This review will focus on the role of angiogenesis in adipose tissue growth and the regulation of obesity by antiangiogenic herbal medicines.
Collapse
Affiliation(s)
- Soon Shik Shin
- Department of Formula Sciences, College of Oriental Medicine, Dongeui University, Busan 47340, Korea;
| | - Michung Yoon
- Department of Biomedical Engineering, Mokwon University, Daejeon 35349, Korea;
- Correspondence: ; Tel.: +8242-829-7581; Fax: 8242-829-7580
| |
Collapse
|
14
|
Siddik MAB, Das BC, Weiss L, Dhurandhar NV, Hegde V. A MetAP2 inhibitor blocks adipogenesis, yet improves glucose uptake in cells. Adipocyte 2019; 8:240-253. [PMID: 31264515 PMCID: PMC6768232 DOI: 10.1080/21623945.2019.1636627] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue expansion involves angiogenesis to remodel its capillary network. The enzymemethionine aminopeptidase 2(MetAP2) promotes angiogenesis.MetAP2 inhibitors suppress angiogenesis and have potential anti-obesity effect. However, impairment in adipose tissue expansion is also linked with impaired glycemic control.This study investigated the effect of BL6, a MetAP2 inhibitor, on adipogenesis and glucose disposal.To test effect on angiogenesis, Human Umbilical Vein Endothelial Cells(HUVECs) were treated with BL6 for 24h to determine tube formation. Further, to test effect on adipogenesis and glucose disposal,3T3-L1 pre-adipocytes were treated with BL6(0 µM, 20µM, 50 µM or 100µM) during differentiation. Differentiated cells were stained with Oil Red O for determining lipid accumulation, and glucose uptake assay. Protein levels and RNA expression for key genes involved in the adipogenic cascade were determined.BL6 treatment of HUVECs dose dependently blocked angiogenesis. During differentiation of pre-adipocytes, 50μM and 100µM BL6 significantly reduced lipid accumulation. Treatment with 100µM BL6 significantly decreased expression of adipogenic genes. Interestingly, BL6 treatment dose dependently increased glucose uptake by 3T3-L1 cells.MetAP2 inhibitor blocks angiogenesis, attenuates adipogenesis, yet increases cellular glucose uptake. Collectively this proof of concept study supports a possible role for MetAP2 inhibitor BL6, as a putative anti-obesity therapeutic agent.
Collapse
Affiliation(s)
| | - Bhaskar C. Das
- The Icahn School of Medicine, Department of Medicine, New York, NY, USA
| | - Louis Weiss
- Department of Pathology, The Albert Einstein College of Medicine, New York, NY, USA
| | | | - Vijay Hegde
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| |
Collapse
|
15
|
Farrell PJ, Zopf CJ, Huang HJ, Balakrishna D, Holub C, Bilakovics J, Fanjul A, Matuszkiewicz J, Plonowski A, Rolzin P, Banerjee U, Ermolieff J, Cheruvallath ZS, McBride C, Bartkowski D, Mazur C, Pachori A, Larson CJ. Using Target Engagement Biomarkers to Predict Clinical Efficacy of MetAP2 Inhibitors. J Pharmacol Exp Ther 2019; 371:299-308. [PMID: 31537613 DOI: 10.1124/jpet.119.259028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 09/04/2019] [Indexed: 01/09/2023] Open
Abstract
Target-engagement pharmacodynamic (PD) biomarkers are valuable tools in the prioritization of drug candidates, especially for novel, first-in-class mechanisms whose robustness to alter disease outcome is unknown. Methionine aminopeptidase 2 (MetAP2) is a cytosolic metalloenzyme that cleaves the N-terminal methionine from nascent proteins. Inhibition of MetAP2 leads to weight loss in obese rodents, dogs and humans. However, there is a need to develop efficacious compounds that specifically inhibit MetAP2 with an improved safety profile. The objective of this study was to identify a PD biomarker for selecting potent, efficacious compounds and for predicting clinical efficacy that would result from inhibition of MetAP2. Here we report the use of NMet14-3-3γ for this purpose. Treatment of primary human cells with MetAP2 inhibitors resulted in an approx. 10-fold increase in NMet14-3-3γ levels. Furthermore, treatment of diet-induced obese mice with these compounds reduced body weight (approx. 20%) and increased NMet14-3-3γ (approx. 15-fold) in adipose tissues. The effects on target engagement and body weight increased over time and were dependent on dose and administration frequency of compound. The relationship between compound concentration in plasma, NMet14-3-3γ in tissue, and reduction of body weight in obese mice was used to generate a pharmacokinetic-pharmacodynamic-efficacy model for predicting efficacy of MetAP2 inhibitors in mice. We also developed a model for predicting weight loss in humans using a target engagement PD assay that measures inhibitor-bound MetAP2 in blood. In summary, MetAP2 target engagement biomarkers can be used to select efficacious compounds and predict weight loss in humans. SIGNIFICANCE STATEMENT: The application of target engagement pharmacodynamic biomarkers during drug development provides a means to determine the dose required to fully engage the intended target and an approach to connect the drug target to physiological effects. This work exemplifies the process of using target engagement biomarkers during preclinical research to select new drug candidates and predict clinical efficacy. We determine concentration of MetAP2 antiobesity compounds needed to produce pharmacological activity in primary human cells and in target tissues from an appropriate animal model and establish key relationships between pharmacokinetics, pharmacodynamics, and efficacy, including the duration of effects after drug administration. The biomarkers described here can aid decision-making in early clinical trials of MetAP2 inhibitors for the treatment of obesity.
Collapse
Affiliation(s)
- Pamela J Farrell
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| | - Christopher J Zopf
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| | - Huey-Jing Huang
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| | - Deepika Balakrishna
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| | - Corine Holub
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| | - James Bilakovics
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| | - Andrea Fanjul
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| | - Jennifer Matuszkiewicz
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| | - Artur Plonowski
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| | - Paul Rolzin
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| | - Urmi Banerjee
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| | - Jacques Ermolieff
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| | - Zacharia S Cheruvallath
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| | - Christopher McBride
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| | - Darian Bartkowski
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| | - Crystal Mazur
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| | - Alok Pachori
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| | - Christopher J Larson
- Biological Sciences (P.J.F., H.-J.H., De.B., C.H., J.B., A.F., J.M., A.Pl., P.R., U.B., J.E., C.J.L.), Chemistry (Z.S.C., C.Mc.), and Drug Metabolism and Pharmacokinetics (Da.B.), Takeda California, San Diego, California; Modeling and Simulation, Takeda Boston, Cambridge, Massachusetts (C.J.Z.); and Translational Research Institute for Metabolism and Diabetes, Florida Hospital Campus, Orlando, Florida (C.Ma., A.Pa.)
| |
Collapse
|
16
|
Di Stefano AB, Massihnia D, Grisafi F, Castiglia M, Toia F, Montesano L, Russo A, Moschella F, Cordova A. Adipose tissue, angiogenesis and angio-MIR under physiological and pathological conditions. Eur J Cell Biol 2019; 98:53-64. [DOI: 10.1016/j.ejcb.2018.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 01/06/2023] Open
|
17
|
Huang HJ, Holub C, Rolzin P, Bilakovics J, Fanjul A, Satomi Y, Plonowski A, Larson CJ, Farrell PJ. MetAP2 inhibition increases energy expenditure through direct action on brown adipocytes. J Biol Chem 2019; 294:9567-9575. [PMID: 31048375 DOI: 10.1074/jbc.ra118.007302] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 04/26/2019] [Indexed: 11/06/2022] Open
Abstract
Inhibitors of methionine aminopeptidase 2 (MetAP2) have been shown to reduce body weight in obese mice and humans. The target tissue and cellular mechanism of MetAP2 inhibitors, however, have not been extensively examined. Using compounds with diverse chemical scaffolds, we showed that MetAP2 inhibition decreases body weight and fat mass and increases lean mass in the obese mice but not in the lean mice. Obesity is associated with catecholamine resistance and blunted β-adrenergic receptor signaling activities, which could dampen lipolysis and energy expenditure resulting in weight gain. In the current study, we examined effect of MetAP2 inhibition on brown adipose tissue and brown adipocytes. Norepinephrine increases energy expenditure in brown adipose tissue by providing fatty acid substrate through lipolysis and by increasing expression of uncoupled protein-1 (UCP1). Metabolomic analysis shows that in response to MetAP2 inhibitor treatment, fatty acid metabolites in brown adipose tissue increase transiently and subsequently decrease to basal or below basal levels, suggesting an effect on fatty acid metabolism in this tissue. Treatment of brown adipocytes with MetAP2 inhibitors enhances norepinephrine-induced lipolysis and energy expenditure, and prolongs the activity of norepinephrine to increase ucp1 gene expression and energy expenditure in norepinephrine-desensitized brown adipocytes. In summary, we showed that the anti-obesity activity of MetAP2 inhibitors can be mediated, at least in part, through direct action on brown adipocytes by enhancing β-adrenergic-signaling-stimulated activities.
Collapse
Affiliation(s)
| | - Corine Holub
- From Takeda California, San Diego, California 92121
| | - Paul Rolzin
- From Takeda California, San Diego, California 92121
| | | | | | - Yoshinori Satomi
- Takeda Pharmaceutical Company Limited, Fujisawa 251-0012 Japan, and
| | | | - Christopher J Larson
- From Takeda California, San Diego, California 92121.,Sanford Burham Prebys Medical Discovery Institute, San Diego, California 92037
| | | |
Collapse
|
18
|
Wang J, He L, Huwatibieke B, Liu L, Lan H, Zhao J, Li Y, Zhang W. Ghrelin Stimulates Endothelial Cells Angiogenesis through Extracellular Regulated Protein Kinases (ERK) Signaling Pathway. Int J Mol Sci 2018; 19:ijms19092530. [PMID: 30149681 PMCID: PMC6164813 DOI: 10.3390/ijms19092530] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/16/2018] [Accepted: 08/19/2018] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue is hyper-vascularized. Vessels in adipose tissue not only supply nutrients and oxygen to nourish adipocytes, but also provide cytokines that regulate mass and function of adipose tissue. Understanding the fundamental mechanisms how vessels modulate adipocyte functions would provide new therapeutic options for treatment of metabolic disease and obesity. In recent years, researches about ghrelin are focused on glucose and lipid metabolism, but its effect on vascular function remains uncharacterized. In the present study, ghrelin receptor gene deletion mice (Ghsr-/- mice) were used to study ghrelin-regulated vascular metabolism in white adipose tissue. Ghsr-/- mice demonstrated lower food intake, lower body weight, and resistance to high-fat diet-induced obesity. The number of vessels in white adipose tissue was decreased in Ghsr-/- mice when compared with wild type mice fed with high-fat diet. To further define ghrelin effects in vitro, we used endothelial progenitor cells from wild type and Ghsr-/- mice as well as human umbilical vein endothelial cells in our experiments. We found that ghrelin stimulated endothelial cells angiogenesis and migration through the MEK-ERK signaling pathway. [d-Lys3]-GHRP-6 and PD98059 could reverse the effects of ghrelin on endothelial cells. Our study indicates that ghrelin activates its receptor on endothelial cells to promote angiogenesis and migration via a mechanism involving the extracellular regulated protein kinases (ERK) signaling pathway.
Collapse
Affiliation(s)
- Jun Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| | - Lin He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing 100191, China.
| | - Bahetiyaer Huwatibieke
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| | - Lingchao Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| | - He Lan
- Department of Clinical Laboratory, Capital Medical University, Beijing 100053, China.
| | - Jing Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| | - Yin Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| |
Collapse
|
19
|
McCandless SE, Yanovski JA, Miller J, Fu C, Bird LM, Salehi P, Chan CL, Stafford D, Abuzzahab MJ, Viskochil D, Barlow SE, Angulo M, Myers SE, Whitman BY, Styne D, Roof E, Dykens EM, Scheimann AO, Malloy J, Zhuang D, Taylor K, Hughes TE, Kim DD, Butler MG. Effects of MetAP2 inhibition on hyperphagia and body weight in Prader-Willi syndrome: A randomized, double-blind, placebo-controlled trial. Diabetes Obes Metab 2017; 19:1751-1761. [PMID: 28556449 PMCID: PMC5673540 DOI: 10.1111/dom.13021] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 01/01/2023]
Abstract
AIMS There are no treatments for the extreme hyperphagia and obesity in Prader-Willi syndrome (PWS). The bestPWS clinical trial assessed the efficacy, safety and tolerability of the methionine aminopeptidase 2 (MetAP2) inhibitor, beloranib. MATERIALS AND METHODS Participants with PWS (12-65 years old) were randomly assigned (1:1:1) to biweekly placebo, 1.8 mg beloranib or 2.4 mg beloranib injection for 26 weeks at 15 US sites. Co-primary endpoints were the changes in hyperphagia [measured by Hyperphagia Questionnaire for Clinical Trials (HQ-CT); possible score 0-36] and weight by intention-to-treat. ClinicalTrials.gov registration: NCT02179151. RESULTS One-hundred and seven participants were included in the intention-to-treat analysis: placebo (n = 34); 1.8 mg beloranib (n = 36); or 2.4 mg beloranib (n = 37). Improvement (reduction) in HQ-CT total score was greater in the 1.8 mg (mean difference -6.3, 95% CI -9.6 to -3.0; P = .0003) and 2.4 mg beloranib groups (-7.0, 95% CI -10.5 to -3.6; P = .0001) vs placebo. Compared with placebo, weight change was greater with 1.8 mg (mean difference - 8.2%, 95% CI -10.8 to -5.6; P < .0001) and 2.4 mg beloranib (-9.5%, 95% CI -12.1 to -6.8; P < .0001). Injection site bruising was the most frequent adverse event with beloranib. Dosing was stopped early due to an imbalance in venous thrombotic events in beloranib-treated participants (2 fatal events of pulmonary embolism and 2 events of deep vein thrombosis) compared with placebo. CONCLUSIONS MetAP2 inhibition with beloranib produced statistically significant and clinically meaningful improvements in hyperphagia-related behaviours and weight loss in participants with PWS. Although investigation of beloranib has ceased, inhibition of MetAP2 is a novel mechanism for treating hyperphagia and obesity.
Collapse
Affiliation(s)
- Shawn E McCandless
- UH Cleveland Medical Center and Case Western Reserve University, Cleveland, Ohio
| | - Jack A Yanovski
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | | | - Cary Fu
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lynne M Bird
- UCSD Rady Children's Hospital, San Diego, California
| | - Parisa Salehi
- Division of Endocrinology & Diabetes, Seattle Children's Hospital, Seattle, Washington
| | | | | | | | | | | | | | - Susan E Myers
- Saint Louis University School of Medicine, Saint Louis, Missouri
| | | | - Dennis Styne
- UC Davis Children's Hospital, UC Davis Medical Center, Davis, California
| | - Elizabeth Roof
- Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Ann O Scheimann
- Baylor College of Medicine and Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | | | | | | | | | - Merlin G Butler
- Departments of Psychiatry, Behavioral Sciences and Pediatrics, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
20
|
Zhang Y, He B, Liu K, Ning L, Luo D, Xu K, Zhu W, Wu Z, Huang J, Xu X. A novel peptide specifically binding to VEGF receptor suppresses angiogenesis in vitro and in vivo. Signal Transduct Target Ther 2017; 2:17010. [PMID: 29263914 PMCID: PMC5661615 DOI: 10.1038/sigtrans.2017.10] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 02/21/2017] [Accepted: 02/21/2017] [Indexed: 12/27/2022] Open
Abstract
Vascular endothelial growth factor (VEGF), one of the most important angiogenic factors, plays an essential role in both physiological and pathological angiogenesis through binding to VEGF receptors (VEGFRs). Here we report a novel peptide designated HRHTKQRHTALH (peptide HRH), which was isolated from the Ph.D. -12 phage display library using VEGFR-Fc fusion protein as the bait. This peptide was found to dose-dependently inhibit the proliferation of human umbilical vein endothelial cells stimulated by VEGF. The anti-angiogenesis effect of the HRH peptide was further confirmed in vivo using the chick chorioallantoic membrane assay, which was also dose-dependent. Besides, peptide HRH was proved to inhibit corneal neovascularization in an alkali-burnt rat corneal model and a suture-induced rat corneal model. Taken together, these findings suggest that the HRH peptide can inhibit angiogenesis both in vitro and in vivo. Consequently, the HRHTKQRHTALH peptide might be a promising lead peptide for the development of potential angiogenic inhibitors.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bifang He
- Key Laboratory for NeuroInformation of Ministry of Education, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Kun Liu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lin Ning
- Key Laboratory for NeuroInformation of Ministry of Education, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Delun Luo
- Chengdu Nuoen Biotechnologies, LTD, Chengdu, China
| | - Kai Xu
- Chengdu Nuoen Biotechnologies, LTD, Chengdu, China
| | - Wenli Zhu
- Chengdu Nuoen Biotechnologies, LTD, Chengdu, China
| | - Zhigang Wu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Chengdu Nuoen Biotechnologies, LTD, Chengdu, China
| | - Jian Huang
- Key Laboratory for NeuroInformation of Ministry of Education, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Xun Xu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Elfers CT, Roth CL. Robust Reductions of Excess Weight and Hyperphagia by Beloranib in Rat Models of Genetic and Hypothalamic Obesity. Endocrinology 2017; 158:41-55. [PMID: 27849360 DOI: 10.1210/en.2016-1665] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 11/15/2016] [Indexed: 02/08/2023]
Abstract
Hypothalamic lesions or deficient melanocortin (MC) signaling via MC4 receptor (MC4r) mutations often lead to hyperphagia and severe treatment-resistant obesity. We tested the methionine aminopeptidase 2-inhibitor beloranib (ZGN-440) in 2 male rat models of obesity, one modeling hypothalamic obesity with a combined medial hypothalamic lesion (CMHL) and the other modeling a monogenic form of obesity with MC4r mutations (MC4r knockout [MC4rKO]). In CMHL rats (age 3 months), postsurgery excess weight gain was significantly inhibited (ZGN-440, 0.2 ± 0.7 g/d; vehicle, 3.8 ± 0.6 g/d; P < 0.001) during 12 days of ZGN-440 treatment (0.1 mg/kg daily subcutaneously) together with a 30% reduction of daily food intake vs vehicle injection. In addition, ZGN-440 treatment improved glucose tolerance and reduced plasma insulin, and circulating levels of α-melanocyte stimulating hormone were increased. Serum lipid levels did not differ significantly in ZGN-440-treated vs vehicle-treated rats. Similar results were found in MC4rKO rats: ZGN-440 treatment (14-21 d) was associated with significant reductions of body weight gain (MC4rKO, -1.7 ± 0.6 vs 2.8 ± 0.4 g/d; lean wild-type controls, -0.7 ± 0.2 vs 1.7 ± 0.7 g/d; ZGN-440 vs vehicle, respectively), reduction of food intake (MC4rKO, -28%; lean controls, -7.5%), and insulin resistance, whereas circulating levels of interleukin-1β did not change. In both obesity models, body temperature and locomotor activity were not affected by ZGN-440 treatment. In conclusion, the robust reduction of body weight in response to ZGN-440 observed in rats with severe obesity is related to a strong reduction of food intake that is likely related to changes in the central regulation of feeding.
Collapse
Affiliation(s)
- Clinton T Elfers
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101; and
| | - Christian L Roth
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101; and
- Division of Endocrinology, Department of Pediatrics, University of Washington, Seattle, Washington 98105
| |
Collapse
|
22
|
Krishna S, Keralapurath MM, Lin Z, Wagner JJ, de La Serre CB, Harn DA, Filipov NM. Neurochemical and electrophysiological deficits in the ventral hippocampus and selective behavioral alterations caused by high-fat diet in female C57BL/6 mice. Neuroscience 2015; 297:170-81. [PMID: 25849614 DOI: 10.1016/j.neuroscience.2015.03.068] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 03/28/2015] [Accepted: 03/31/2015] [Indexed: 01/08/2023]
Abstract
Mounting experimental evidence, predominantly from male rodents, demonstrates that high-fat diet (HFD) consumption and ensuing obesity are detrimental to the brain. To shed additional light on the neurological consequences of HFD consumption in female rodents and to determine the relatively early impact of HFD in the likely continuum of neurological dysfunction in the context of chronic HFD intake, this study investigated effects of HFD feeding for up to 12weeks on selected behavioral, neurochemical, and electrophysiological parameters in adult female C57BL/6 mice; particular focus was placed on the ventral hippocampus (vHIP). Selected locomotor, emotional and cognitive functions were evaluated using behavioral tests after 5weeks on HFD or control (low-fat diet) diets. One week later, mice were sacrificed and brain regional neurochemical (monoamine) analysis was performed. Behaviorally naïve mice were maintained on their respective diets for an additional 5-6weeks at which time synaptic plasticity was determined in ex vivo slices from the vHIP. HFD-fed female mice exhibited increased: (i) locomotor activity in the open field testing, (ii) mean turn time on the pole test, (iii) swimming time in the forced swim test, and (iv) number of marbles buried in the marble burying test. In contrast, the novel object recognition memory was unaffected. Mice on HFD also had decreased norepinephrine and dopamine turnover, respectively, in the prefrontal cortex and the vHIP. HFD consumption for a total of 11-12weeks altered vHIP synaptic plasticity, evidenced by significant reductions in the paired-pulse ratio and long-term potentiation (LTP) magnitude. In summary, in female mice, HFD intake for several weeks induced multiple behavioral alterations of mainly anxiety-like nature and impaired monoamine pathways in a brain region-specific manner, suggesting that in the female, certain behavioral domains (anxiety) and associated brain regions, i.e., the vHIP, are preferentially targeted by HFD.
Collapse
Affiliation(s)
- S Krishna
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - M M Keralapurath
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Z Lin
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - J J Wagner
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - C B de La Serre
- Department of Foods and Nutrition, College of Family and Consumer Sciences, University of Georgia, Athens, GA 30602, USA
| | - D A Harn
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - N M Filipov
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
23
|
Ban JJ, Ruthenborg RJ, Cho KW, Kim JW. Regulation of obesity and insulin resistance by hypoxia-inducible factors. HYPOXIA 2014; 2:171-183. [PMID: 27774475 PMCID: PMC5045065 DOI: 10.2147/hp.s68771] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
In obesity, dysregulated metabolism and aberrant expansion of adipose tissue lead to the development of tissue hypoxia that plays an important role in contributing to obesity-associated metabolic disorders. Recent studies utilizing adipocyte-specific hypoxia-inducible factor-α (HIF-α) gain- or loss-of-function animal models highlight the pivotal involvement of hypoxic responses in the pathogenesis of obesity-associated inflammation and insulin resistance. HIF-1α, a master transcription factor of oxygen homeostasis, induces inflammation and insulin resistance in obesity, whereas its isoform, HIF-2α, exerts opposing functions in these obesity-associated metabolic phenotypes. In this review, recent evidence elucidating functional implications of adipocyte HIFs in obesity and, more importantly, how these regulate obesity-associated inflammation, fibrosis, and insulin resistance will be discussed. Further, we propose that modulation of HIF-1 could be a potential novel therapeutic strategy for antidiabetic treatment.
Collapse
Affiliation(s)
- Jae-Jun Ban
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Robin J Ruthenborg
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Kevin W Cho
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Jung-Whan Kim
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
24
|
Zhang S, Yu M, Wei Y. Do anti-angiogenic cancer therapies increase risk of significant weight loss? Expert Opin Drug Saf 2014; 13:473-82. [PMID: 24588304 DOI: 10.1517/14740338.2014.894506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Angiogenesis is important in many disease states such as cancer. Anti-angiogenic cancer drugs are in broad use for the treatment of cancers. However, currently most of these anticancer drugs result in some adverse effects in the patient. AREAS COVERED In this paper, we review evidence on the association between anti-angiogenic therapies and weight loss. We report on basic experiments and clinical trials that measure weight loss with anti-angiogenic cancer therapies. EXPERT OPINION Few strong associations are found between anti-angiogenic cancer therapies and weight loss, with the exception of some multikinase inhibitors in clinical trials. Anti-angiogenic cancer therapies appear safe in relation to weight loss, but the result needs to be established by further clinical trials.
Collapse
Affiliation(s)
- Shuang Zhang
- Sichuan University, West China Hospital, State Key Laboratory of Biotherapy and Cancer Center , Chengdu 610041 , China
| | | | | |
Collapse
|
25
|
White HM, Acton AJ, Kamocka MM, Considine RV. Hepatocyte growth factor regulates neovascularization in developing fat pads. Am J Physiol Endocrinol Metab 2014; 306:E189-96. [PMID: 24302003 PMCID: PMC3920009 DOI: 10.1152/ajpendo.00394.2013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In this study, we used lentiviral-delivered shRNA to generate a clonal line of 3T3-F442A preadipocytes with stable silencing of hepatocyte growth factor (HGF) expression and examined the long-term consequence of this modification on fat pad development. HGF mRNA expression was reduced 94%, and HGF secretion 79% (P < 0.01), compared with preadipocytes treated with nontargeting shRNA. Fat pads derived from HGF knockdown preadipocytes were significantly smaller (P < 0.01) than control pads beginning at 3 days postinjection (0.022 ± 0.003 vs. 0.037 ± 0.004 g), and further decreased in size at day 7 (0.015 ± 0.004 vs. 0.037 ± 0.003 g) and day 14 (0.008 ± 0.002 vs. 0.045 ± 0.007 g). Expression of the endothelial cell genes TIE1 and PECAM1 increased over time in control fat pads (1.6 ± 0.4 vs. 11.4 ± 1.7 relative units at day 3 and 14, respectively; P < 0.05) but not in HGF knockdown fat pads (1.1 ± 0.5 vs. 5.9 ± 2.2 relative units at day 3 and 14). Contiguous vascular structures were observed in control fat pads but were much less developed in HGF knockdown fat pads. Differentiation of preadipocytes to mature adipocytes was significantly attenuated in HGF knockdown fat pads. Fat pads derived from preadipocytes with knockdown of the HGF receptor c-MET were smaller than control pads at day 3 postinjection (0.034 ± 0.002 vs. 0.049 ± 0.004 g; P < 0.05), and remained the same size through day 14. c-MET knockdown fat pads developed a robust vasculature, and preadipocytes differentiated to mature adipocytes. Overall these data suggest that preadipocyte-secreted HGF is an important regulator of neovascularization in developing fat pads.
Collapse
Affiliation(s)
- Heather M White
- Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; and
| | | | | | | |
Collapse
|
26
|
Joharapurkar AA, Dhanesha NA, Jain MR. Inhibition of the methionine aminopeptidase 2 enzyme for the treatment of obesity. Diabetes Metab Syndr Obes 2014; 7:73-84. [PMID: 24611021 PMCID: PMC3944999 DOI: 10.2147/dmso.s56924] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Worldwide prevalence of obesity has nearly doubled since 1980. Obesity is the result of interactions among the environmental factors, genetic predisposition, and human behavior. Even modest weight reduction in obese patients provides beneficial health outcomes. For effective weight reduction, a drug should either increase energy expenditure or decrease energy intake without causing serious adverse effects. To overcome lack of efficacy and central nervous system related side effects, exploitation of the peripheral mechanism of anti-obesity action is needed. Inhibition of pathological angiogenesis in adipose tissue is one such peripheral mechanism that has attracted the attention of researchers in this area. Although originally developed as anti-cancer agents, methionine aminopeptidase (MetAP2) inhibitors induce significant and sustained weight reduction. Here, we review preclinical and clinical pharmacology of MetAP2 inhibitors. Beloranib is a prototype MetAP2 inhibitor, and currently in advanced clinical trials for the treatment of obesity. Clinical data of beloranib indicate that MetAP2 inhibitors could be a future treatment option for weight reduction without serious adverse effects. Further clinical data from Phase III trials will add to our growing knowledge of MetAP2 inhibitor potential for anti-obesity therapy.
Collapse
Affiliation(s)
- Amit A Joharapurkar
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Ahmedabad, India
- Correspondence: Amit A Joharapurkar, Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH 8A, Moraiya, Ahmedabad 382210, India, Tel + 91 271 766 5555, Fax + 91 271 766 5155, Email
| | - Nirav A Dhanesha
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Ahmedabad, India
| | - Mukul R Jain
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Ahmedabad, India
| |
Collapse
|
27
|
Cao Y. Angiogenesis and vascular functions in modulation of obesity, adipose metabolism, and insulin sensitivity. Cell Metab 2013; 18:478-89. [PMID: 24035587 DOI: 10.1016/j.cmet.2013.08.008] [Citation(s) in RCA: 243] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
White and brown adipose tissues are hypervascularized and the adipose vasculature displays phenotypic and functional plasticity to coordinate with metabolic demands of adipocytes. Blood vessels not only supply nutrients and oxygen to nourish adipocytes, they also serve as a cellular reservoir to provide adipose precursor and stem cells that control adipose tissue mass and function. Multiple signaling molecules modulate the complex interplay between the vascular system and the adipocytes. Understanding fundamental mechanisms by which angiogenesis and vasculatures modulate adipocyte functions may provide new therapeutic options for treatment of obesity and metabolic disorders by targeting the adipose vasculature.
Collapse
Affiliation(s)
- Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden; Department of Medicine and Health Sciences, Linköping University, 581 85 Linköping, Sweden.
| |
Collapse
|