1
|
Benita BA, Koss KM. Peptide discovery across the spectrum of neuroinflammation; microglia and astrocyte phenotypical targeting, mediation, and mechanistic understanding. Front Mol Neurosci 2024; 17:1443985. [PMID: 39634607 PMCID: PMC11616451 DOI: 10.3389/fnmol.2024.1443985] [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: 06/04/2024] [Accepted: 07/24/2024] [Indexed: 12/07/2024] Open
Abstract
Uncontrolled and chronic inflammatory states in the Central Nervous System (CNS) are the hallmark of neurodegenerative pathology and every injury or stroke-related insult. The key mediators of these neuroinflammatory states are glial cells known as microglia, the resident immune cell at the core of the inflammatory event, and astroglia, which encapsulate inflammatory insults in proteoglycan-rich scar tissue. Since the majority of neuroinflammation is exclusively based on the responses of said glia, their phenotypes have been identified to be on an inflammatory spectrum encompassing developmental, homeostatic, and reparative behaviors as opposed to their ability to affect devastating cell death cascades and scar tissue formation. Recently, research groups have focused on peptide discovery to identify these phenotypes, find novel mechanisms, and mediate or re-engineer their actions. Peptides retain the diverse function of proteins but significantly reduce the activity dependence on delicate 3D structures. Several peptides targeting unique phenotypes of microglia and astroglia have been identified, along with several capable of mediating deleterious behaviors or promoting beneficial outcomes in the context of neuroinflammation. A comprehensive review of the peptides unique to microglia and astroglia will be provided along with their primary discovery methodologies, including top-down approaches using known biomolecules and naïve strategies using peptide and phage libraries.
Collapse
Affiliation(s)
| | - Kyle M. Koss
- Department of Surgery, University of Arizona, Tucson, AZ, United States
- Department of Neurobiology, University of Texas Medical Branch (UTMB) at Galvestion, Galvestion, TX, United States
- Sealy Institute for Drug Discovery (SIDD), University of Texas Medical Branch (UTMB) at Galvestion, Galvestion, TX, United States
| |
Collapse
|
2
|
Keifi Bajestani A, Alavi MS, Etemad L, Roohbakhsh A. Role of orphan G-protein coupled receptors in tissue ischemia: A comprehensive review. Eur J Pharmacol 2024; 978:176762. [PMID: 38906238 DOI: 10.1016/j.ejphar.2024.176762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 06/23/2024]
Abstract
Ischemic events lead to many diseases and deaths worldwide. Ischemia/reperfusion (I/R) occurs due to reduced blood circulation in tissues followed by blood reflow. Reoxygenation of ischemic tissues is characterized by oxidative stress, inflammation, energy distress, and endoplasmic reticulum stress. There are still no adequate clinical protocols or pharmacological approaches to address the consequences of I/R damage. G protein-coupled receptors (GPCRs) are important therapeutic targets. They compose a large family of seven transmembrane-spanning proteins that are involved in many biological functions. Orphan GPCRs are a large subgroup of these receptors expressed in different organs. In the present review, we summarized the literature regarding the role of orphan GPCRs in I/R in different organs. We focused on the effect of these receptors on modulating cellular and molecular processes underlying ischemia including apoptosis, inflammation, and autophagy. The study showed that GPR3, GPR4, GPR17, GPR30, GPR31, GPR35, GPR37, GPR39, GPR55, GPR65, GPR68, GPR75, GPR81, and GPR91 are involved in ischemic events, mainly in the brain and heart. These receptors offer new possibilities for treating I/R injuries in the body.
Collapse
Affiliation(s)
- Alireza Keifi Bajestani
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohaddeseh Sadat Alavi
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Bora RR, Prasad R, Khatib MN. Cardio-Protective Role of a Gut Hormone Obestatin: A Narrative Review. Cureus 2023; 15:e37972. [PMID: 37223200 PMCID: PMC10202687 DOI: 10.7759/cureus.37972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 04/22/2023] [Indexed: 05/25/2023] Open
Abstract
Obestatin is a gut hormone composed of 23 amino acids that play a role in protecting the heart. It is synthesized from the same preproghrelin gut hormone gene as another gut hormone. The function and receptor of obestatin remain controversial, despite being present in various organs such as the liver, heart, mammary gland, pancreas, and more. The activity of obestatin is opposite to that of ghrelin, another hormone. The GPR-39 receptor is used by obestatin to exert its effects. Obestatin's cardioprotective role can be attributed to its ability to affect various factors, including adipose tissue, blood pressure regulation, heart, ischemia-reperfusion injury, endothelial cells, and diabetes. Because these factors are related to the cardiovascular system, modifying them via obestatin can provide cardioprotection. Furthermore, ghrelin, its antagonist hormone, regulates cardiovascular health. Diabetes mellitus, hypertension, and ischemia-reperfusion injury can all alter ghrelin/obestatin levels. Obestatin has also been shown to impact other organs, reducing weight and appetite, inhibiting food intake, and increasing adipogenesis. Obestatin has a brief half-life and is quickly degraded by proteases in the blood, liver, and kidneys after entering circulation. This article offers insights into the cardiac function of obestatin.
Collapse
Affiliation(s)
- Rajal R Bora
- Physiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Roshan Prasad
- Medicine and Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Mahalaqua Nazli Khatib
- Epidemiology and Public Health, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
4
|
Villarreal D, Pradhan G, Zhou Y, Xue B, Sun Y. Diverse and Complementary Effects of Ghrelin and Obestatin. Biomolecules 2022; 12:517. [PMID: 35454106 PMCID: PMC9028691 DOI: 10.3390/biom12040517] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/21/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023] Open
Abstract
Ghrelin and obestatin are two "sibling proteins" encoded by the same preproghrelin gene but possess an array of diverse and complex functions. While there are ample literature documenting ghrelin's functions, the roles of obestatin are less clear and controversial. Ghrelin and obestatin have been perceived to be antagonistic initially; however, recent studies challenge this dogma. While they have opposing effects in some systems, they function synergistically in other systems, with many functions remaining debatable. In this review, we discuss their functional relationship under three "C" categories, namely complex, complementary, and contradictory. Their functions in food intake, weight regulation, hydration, gastrointestinal motility, inflammation, and insulin secretion are complex. Their functions in pancreatic beta cells, cardiovascular, muscle, neuroprotection, cancer, and digestive system are complementary. Their functions in white adipose tissue, thermogenesis, and sleep regulation are contradictory. Overall, this review accumulates the multifaceted functions of ghrelin and obestatin under both physiological and pathological conditions, with the intent of contributing to a better understanding of these two important gut hormones.
Collapse
Affiliation(s)
- Daniel Villarreal
- Department of Nutrition, Texas A & M University, College Station, TX 77843, USA;
| | - Geetali Pradhan
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA;
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yu Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, China;
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA;
| | - Yuxiang Sun
- Department of Nutrition, Texas A & M University, College Station, TX 77843, USA;
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA;
| |
Collapse
|
5
|
Fernandez Rico C, Konate K, Josse E, Nargeot J, Barrère-Lemaire S, Boisguérin P. Therapeutic Peptides to Treat Myocardial Ischemia-Reperfusion Injury. Front Cardiovasc Med 2022; 9:792885. [PMID: 35252383 PMCID: PMC8891520 DOI: 10.3389/fcvm.2022.792885] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases (CVD) including acute myocardial infarction (AMI) rank first in worldwide mortality and according to the World Health Organization (WHO), they will stay at this rank until 2030. Prompt revascularization of the occluded artery to reperfuse the myocardium is the only recommended treatment (by angioplasty or thrombolysis) to decrease infarct size (IS). However, despite beneficial effects on ischemic lesions, reperfusion leads to ischemia-reperfusion (IR) injury related mainly to apoptosis. Improvement of revascularization techniques and patient care has decreased myocardial infarction (MI) mortality however heart failure (HF) morbidity is increasing, contributing to the cost-intense worldwide HF epidemic. Currently, there is no treatment for reperfusion injury despite promising results in animal models. There is now an obvious need to develop new cardioprotective strategies to decrease morbidity/mortality of CVD, which is increasing due to the aging of the population and the rising prevalence rates of diabetes and obesity. In this review, we will summarize the different therapeutic peptides developed or used focused on the treatment of myocardial IR injury (MIRI). Therapeutic peptides will be presented depending on their interacting mechanisms (apoptosis, necroptosis, and inflammation) reported as playing an important role in reperfusion injury following myocardial ischemia. The search and development of therapeutic peptides have become very active, with increasing numbers of candidates entering clinical trials. Their optimization and their potential application in the treatment of patients with AMI will be discussed.
Collapse
Affiliation(s)
- Carlota Fernandez Rico
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- Laboratory of Excellence Ion Channel Science and Therapeutics, Valbonne, France
| | - Karidia Konate
- PHYMEDEXP, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Emilie Josse
- PHYMEDEXP, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Joël Nargeot
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- Laboratory of Excellence Ion Channel Science and Therapeutics, Valbonne, France
| | - Stéphanie Barrère-Lemaire
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- Laboratory of Excellence Ion Channel Science and Therapeutics, Valbonne, France
| | - Prisca Boisguérin
- PHYMEDEXP, Université de Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
6
|
Comità S, Femmino S, Thairi C, Alloatti G, Boengler K, Pagliaro P, Penna C. Regulation of STAT3 and its role in cardioprotection by conditioning: focus on non-genomic roles targeting mitochondrial function. Basic Res Cardiol 2021; 116:56. [PMID: 34642818 PMCID: PMC8510947 DOI: 10.1007/s00395-021-00898-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 12/11/2022]
Abstract
Ischemia–reperfusion injury (IRI) is one of the biggest challenges for cardiovascular researchers given the huge death toll caused by myocardial ischemic disease. Cardioprotective conditioning strategies, namely pre- and post-conditioning maneuvers, represent the most important strategies for stimulating pro-survival pathways essential to preserve cardiac health. Conditioning maneuvers have proved to be fundamental for the knowledge of the molecular basis of both IRI and cardioprotection. Among this evidence, the importance of signal transducer and activator of transcription 3 (STAT3) emerged. STAT3 is not only a transcription factor but also exhibits non-genomic pro-survival functions preserving mitochondrial function from IRI. Indeed, STAT3 is emerging as an influencer of mitochondrial function to explain the cardioprotection phenomena. Studying cardioprotection, STAT3 proved to be crucial as an element of the survivor activating factor enhancement (SAFE) pathway, which converges on mitochondria and influences their function by cross-talking with other cardioprotective pathways. Clearly there are still some functional properties of STAT3 to be discovered. Therefore, in this review, we highlight the evidence that places STAT3 as a promoter of the metabolic network. In particular, we focus on the possible interactions of STAT3 with processes aimed at maintaining mitochondrial functions, including the regulation of the electron transport chain, the production of reactive oxygen species, the homeostasis of Ca2+ and the inhibition of opening of mitochondrial permeability transition pore. Then we consider the role of STAT3 and the parallels between STA3/STAT5 in cardioprotection by conditioning, giving emphasis to the human heart and confounders.
Collapse
Affiliation(s)
- Stefano Comità
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Torino, TO, Italy
| | - Saveria Femmino
- Department of Medical Sciences, University of Turin, Torino, Italy
| | - Cecilia Thairi
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Torino, TO, Italy
| | | | - Kerstin Boengler
- Institute of Physiology, University of Giessen, Giessen, Germany
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Torino, TO, Italy.
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Torino, TO, Italy.
| |
Collapse
|
7
|
Chen L, Shi D, Guo M. The roles of PKC-δ and PKC-ε in myocardial ischemia/reperfusion injury. Pharmacol Res 2021; 170:105716. [PMID: 34102229 DOI: 10.1016/j.phrs.2021.105716] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/01/2021] [Accepted: 06/03/2021] [Indexed: 01/14/2023]
Abstract
Ischemia and reperfusion (I/R) cause a reduction in arterial blood supply to tissues, followed by the restoration of perfusion and consequent reoxygenation. The reestablishment of blood flow triggers further damage to ischemic tissue through reactive oxygen species (ROS) accumulation, interference with cellular ion homeostasis, opening of mitochondrial permeability transition pores (mPTPs) and promotion of cell death (apoptosis or necrosis). PKC-δ and PKC-ε, belonging to a family of serine/threonine kinases, have been demonstrated to play important roles during I/R injury in cardiovascular diseases. However, the cardioprotective mechanisms of PKC-δ and PKC-ε in I/R injury have not been elaborated until now. This article discusses the roles of PKC-δ and PKC-ε during myocardial I/R in redox regulation (redox signaling and oxidative stress), cell death (apoptosis and necrosis), Ca2+ overload, and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Li Chen
- Peking University Traditional Chinese Medicine Clinical Medical School (Xi yuan), Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dazhuo Shi
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Ming Guo
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
8
|
Gupta S, Mitra A. Heal the heart through gut (hormone) ghrelin: a potential player to combat heart failure. Heart Fail Rev 2020; 26:417-435. [PMID: 33025414 DOI: 10.1007/s10741-020-10032-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/21/2020] [Indexed: 12/17/2022]
Abstract
Ghrelin, a small peptide hormone (28 aa), secreted mainly by X/A-like cells of gastric mucosa, is also locally produced in cardiomyocytes. Being an orexigenic factor (appetite stimulant), it promotes release of growth hormone (GH) and exerts diverse physiological functions, viz. regulation of energy balance, glucose, and/or fat metabolism for body weight maintenance. Interestingly, administration of exogenous ghrelin significantly improves cardiac functions in CVD patients as well as experimental animal models of heart failure. Ghrelin ameliorates pathophysiological condition of the heart in myocardial infarction, cardiac hypertrophy, fibrosis, cachexia, and ischemia reperfusion injury. This peptide also exerts significant impact at the level of vasculature leading to lowering high blood pressure and reversal of endothelial dysfunction and atherosclerosis. However, the molecular mechanism of actions elucidating the healing effects of ghrelin on the cardiovascular system is still a matter of conjecture. Some experimental data indicate its beneficial effects via complex cellular cross talks between autonomic nervous system and cardiovascular cells, some other suggest more direct receptor-mediated molecular actions via autophagy or ionotropic regulation and interfering with apoptotic and inflammatory pathways of cardiomyocytes and vascular endothelial cells. Here, in this review, we summarise available recent data to encourage more research to find the missing links of unknown ghrelin receptor-mediated pathways as we see ghrelin as a future novel therapy in cardiovascular protection.
Collapse
Affiliation(s)
- Shreyasi Gupta
- Department of Zoology, Triveni Devi Bhalotia College, Raniganj, Paschim Bardhaman, 713347, India
| | - Arkadeep Mitra
- Department of Zoology, City College , 102/1, Raja Rammohan Sarani, Kolkata, 700009, India.
| |
Collapse
|
9
|
Penna C, Alloatti G, Crisafulli A. Mechanisms Involved in Cardioprotection Induced by Physical Exercise. Antioxid Redox Signal 2020; 32:1115-1134. [PMID: 31892282 DOI: 10.1089/ars.2019.8009] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Significance: Regular exercise training can reduce myocardial damage caused by acute ischemia/reperfusion (I/R). Exercise can reproduce the phenomenon of ischemic preconditioning, due to the capacity of brief periods of ischemia to reduce myocardial damage caused by acute I/R. In addition, exercise may also activate the multiple kinase cascade responsible for cardioprotection even in the absence of ischemia. Recent Advances: Animal and human studies highlighted the fact that, besides to reduce risk factors related to cardiovascular disease, the beneficial effects of exercise are also due to its ability to induce conditioning of the heart. Exercise behaves as a physiological stress that triggers beneficial adaptive cellular responses, inducing a protective phenotype in the heart. The factors contributing to the exercise-induced heart preconditioning include stimulation of the anti-radical defense system and nitric oxide production, opioids, myokines, and adenosine-5'-triphosphate (ATP) dependent potassium channels. They appear to be also involved in the protective effect exerted by exercise against cardiotoxicity related to chemotherapy. Critical Issues and Future Directions: Although several experimental evidences on the protective effect of exercise have been obtained, the mechanisms underlying this phenomenon have not yet been fully clarified. Further studies are warranted to define precise exercise prescriptions in patients at risk of myocardial infarction or undergoing chemotherapy.
Collapse
Affiliation(s)
- Claudia Penna
- National Institute for Cardiovascular Research (INRC), Bologna, Italy.,Department of Clinical and Biological Sciences, University of Turin, Torino, Italy
| | | | - Antonio Crisafulli
- Department of Medical Sciences and Public Health, Sports Physiology Lab., University of Cagliari, Cagliari, Italy
| |
Collapse
|
10
|
Geale PF, Sheehy PA, Giles C, Thomson PC, Wynn PC. Efficacy of two adjuvant systems to promote humoral immunity to the pre-proghrelin peptide obestatin in pigs: consequences for the growth of piglets to weaning. ANIMAL PRODUCTION SCIENCE 2020. [DOI: 10.1071/an18404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The poor antigenicity of peptide antigens demands the selection of effective adjuvants to induce humoral immunity. The peptides obestatin and ghrelin from the pro-hormone pre-proghrelin were initially identified as antagonistic in regulating feeding behaviour, with obestatin being suppressive. The efficacy of two adjuvant systems, DEAE with the oil polysorbate emulsion of BP85:Span80 and the surfactant-oil system Montanide (ISA 50v) were therefore assessed with an obestatin-ovalbumin conjugate injected into late pregnant sows. This enabled the supply of antibodies directed against obestatin to newborn piglets through colostrum with the objective of promoting ghrelin secretion and therefore increasing feeding behaviour. Pregnant Landrace × Large White sows (n = 28) were immunised with 0.5 mg obestatin-ovalbumin in 2 mL DEAE:BP85:Span80 (DEAE; n = 14) or with 2 mL Montanide (ISA 50v: n = 14) as adjuvants at days 91 and 105 of gestation. After farrowing, piglets remained with their mothers during the lactation period and were weighed after weaning at Day 28. Antibody titres (unitless) in colostrum were assessed by ELISA as 5543 ± 2388 and 3139 ± 1151 for the DEAE and Montanide adjuvants respectively. These were associated with total IgG of 67.7 ± 3 and 82.3 ± 4.8 mg/mL respectively (P = 0.018). Piglet plasma titres were 5100 ± 1576 and 5762 ± 1688 for DEAE and Montanide respectively at Day 5 postpartum. These titres were still detectable through to Day 28 (titres of 1213 ± 389 and 665 ± 203 respectively (P = 0.176). However, sow colostral antibody titres were not related to piglet antibody concentrations on D5 (r = –0.225, P = 0.341). Sow plasma antibody titres were not related to titres at Day 28 in piglets across treatments (r = 0.198, P = 0.402). The concentration of ghrelin in colostrum was 672 ± 78 and 666 ± 39 pg/mL for the DEAE and Montanide groups, respectively, leading to piglet plasma concentrations on Day 5 of 1105 ± 164 and 530 ± 84 pg/mL (P = 0.002). Animals grew from birthweights of 1.7 ± 0.1 and 1.8 ± 0.1 (P = 0.993) to 7.7 ± 1.2 and 7.8 ± 1.0 kg (P = 0.295) at weaning, representing growth rates of 200.5 ± 52.9 and 225.5 ± 53.4 g/day (P = 0.181). There was a significant negative correlation between piglet D28 antibody titre and growth rate to weaning with the Montanide adjuvant (r = 0.116, P = 0.035) but not for the DEAE (r = –0.118, P = 0.411). Although both adjuvants were capable of generating high antibody titres, the DEAE dextran was likely to be the most effective adjuvant to induce a humoral immune response to develop further with a commercial vaccine.
Collapse
|
11
|
Foroughi K, Jahanbani S, Khaksari M, Shayannia A. Obestatin attenuated methamphetamine-induced PC12 cells neurotoxicity via inhibiting autophagy and apoptosis. Hum Exp Toxicol 2019; 39:301-310. [PMID: 31726888 DOI: 10.1177/0960327119886036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Methamphetamine (METH) is an illicit dopaminergic neurotoxin and is an extremely addictive psychostimulant drug that influences monoamine neurotransmitter system of the brain and is responsible for enhancing energy and satisfaction and feelings of alertness. Long-lasting exposure to METH causes psychosis and increases the risk of Parkinson's disease. Studies have revealed that obestatin (OB) is a novel endogenous ligand, which may have neuroprotective effects. Hence, we hypothesized that OB might appropriately limit METH-induced neurotoxicity via the control of apoptotis and autophagy. In the current study, PC12 cells were exposed to both METH (0.5, 1, 2, 3, 4, and 6 mmol/L) and pretreatment OB (1, 10, 100, and 200 nmol/L) in vitro for 24 h to determine appropriate dose, and then downstream pathways were measured to investigate apoptosis and autophagy. The results have shown that OB reduced the apoptotic response post-METH exposure in PC12 cells by developing cell viability and diminishing apoptotic rates. Furthermore, the study has exhibited OB decreased gene expression of Beclin-1 by real-time polymerase chain reaction and LC3-II by Western blotting in METH-induced PC12 cells, which demonstrated that autophagy is reduced. The study is proposed that OB is useful in reducing oxidative stress, which may also play an essential role in the regulation of METH-triggered apoptotic response. So these data indicate that OB could potentially alleviate METH-induced neurotoxicity via the reduction of apoptotic and autophagy responses.
Collapse
Affiliation(s)
- K Foroughi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - S Jahanbani
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - M Khaksari
- Addiction Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - A Shayannia
- Department of Medical Biotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
12
|
Landecho MF, Tuero C, Valentí V, Bilbao I, de la Higuera M, Frühbeck G. Relevance of Leptin and Other Adipokines in Obesity-Associated Cardiovascular Risk. Nutrients 2019; 11:nu11112664. [PMID: 31694146 PMCID: PMC6893824 DOI: 10.3390/nu11112664] [Citation(s) in RCA: 183] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/02/2019] [Accepted: 10/17/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity, which is a worldwide epidemic, confers increased risk for multiple serious conditions including type 2 diabetes, nonalcoholic fatty liver disease, and cardiovascular diseases. Adipose tissue is considered one of the largest endocrine organs in the body as well as an active tissue for cellular reactions and metabolic homeostasis rather than an inert tissue only for energy storage. The functional pleiotropism of adipose tissue relies on its ability to synthesize and release a large number of hormones, cytokines, extracellular matrix proteins, and growth and vasoactive factors, which are collectively called adipokines known to influence a variety of physiological and pathophysiological processes. In the obese state, excessive visceral fat accumulation causes adipose tissue dysfunctionality that strongly contributes to the onset of obesity-related comorbidities. The mechanisms underlying adipose tissue dysfunction include adipocyte hypertrophy and hyperplasia, increased inflammation, impaired extracellular matrix remodeling, and fibrosis together with an altered secretion of adipokines. This review describes the relevance of specific adipokines in the obesity-associated cardiovascular disease.
Collapse
Affiliation(s)
- Manuel F. Landecho
- Department of Internal Medicine, General Health Check-up Unit, Clínica Universidad de Navarra, Avenida Pío XII, 36, 31008 Pamplona, Navarra, Spain; (M.F.L.); (I.B.)
| | - Carlota Tuero
- Department of Surgery, Bariatric and Metabolic Surgery Unit, Clínica Universidad de Navarra, 31008 Pamplona, Navarra, Spain; (C.T.); (V.V.)
| | - Víctor Valentí
- Department of Surgery, Bariatric and Metabolic Surgery Unit, Clínica Universidad de Navarra, 31008 Pamplona, Navarra, Spain; (C.T.); (V.V.)
- Instituto de Salud Carlos III, CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 31008 Pamplona, Navarra, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Navarra, Spain
| | - Idoia Bilbao
- Department of Internal Medicine, General Health Check-up Unit, Clínica Universidad de Navarra, Avenida Pío XII, 36, 31008 Pamplona, Navarra, Spain; (M.F.L.); (I.B.)
| | - Magdalena de la Higuera
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, 28027 Madrid, Spain;
| | - Gema Frühbeck
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Navarra, Spain
- Metabolic Research Laboratory, Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Pamplona, Spain
- Correspondence: ; Tel.: +0034-948-255-400
| |
Collapse
|
13
|
Ghrelin Derangements in Idiopathic Dilated Cardiomyopathy: Impact of Myocardial Disease Duration and Left Ventricular Ejection Fraction. J Clin Med 2019; 8:jcm8081152. [PMID: 31375017 PMCID: PMC6723091 DOI: 10.3390/jcm8081152] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/29/2019] [Accepted: 07/31/2019] [Indexed: 11/30/2022] Open
Abstract
Background: Ghrelin may exert positive effects on cardiac structure and function in heart failure (HF) patients. Methods: We assessed ghrelin levels in 266 dilated cardiomyopathy (DCM) patients and in 200 age, gender and body mass index (BMI) matched controls. Further, we evaluated the expression of ghrelin and growth hormone secretagogue-receptor (GHSR) in the myocardium of 41 DCM patients and in 11 controls. Results: DCM patients had significantly lower levels of total, acylated and unacylated ghrelin when compared to controls (p < 0.05 for all). In controls, we observed a negative correlation of ghrelin with age, male gender and BMI. These correlations were lost in the DCM group, except for male gender. Total ghrelin was higher in patients with more recent diagnosis when compared to patients with longer duration of the DCM (p = 0.033). Further, total ghrelin was higher in patients with lower left ventricular systolic function (<40% LVEF, vs. 40% ≤ LVEF < 49% vs. LVEF ≥ 50%: 480.8, vs. 429.7, vs. 329.5 pg/mL, respectively, p = 0.05). Ghrelin prepropeptide was expressed more in DCM patients than in controls (p = 0.0293) while GHSR was expressed less in DCM patients (p < 0.001). Furthermore, ghrelin showed an inverse correlation with its receptor (ρ = −0.406, p = 0.009), and this receptor showed a significant inverse correlation with Interleukin-1β (ρ = −0.422, p = 0.0103). Conclusion: DCM duration and severity are accompanied by alterations in the ghrelin–GHSR system.
Collapse
|
14
|
Mirarab E, Hojati V, Vaezi G, Shiravi A, Khaksari M. Obestatin inhibits apoptosis and astrogliosis of hippocampal neurons following global cerebral ischemia reperfusion via antioxidant and anti-inflammatory mechanisms. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:617-622. [PMID: 31231488 PMCID: PMC6570752 DOI: 10.22038/ijbms.2019.34118.8110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Objective(s): Obestatin is a newly discovered peptide with antioxidant activities in different animal models. Recent studies have shown that Obestatin inhibits apoptosis following cardiac ischemia/reperfusion injury. Brain ischemia/reperfusion induces irreversible damage especially in the hippocampus area. This study aimed at examining the protective impact of Obestatin on apoptosis, protein expression and reactive astrogliosis level in hippocampal CA1 region of rat following transient global cerebral ischemia. Materials and Methods: Forty-eight male Wistar rats were randomly assigned into 4 groups (sham, ischemia/reperfusion, ischemia/reperfusion+ Obestatin 1, and 5 µg/kg, n=12). Ischemia induced occlusion of both common carotid arteries for 20 min. Obestatin 1 and 5 µg/kg were injected intraperitoneally at the beginning of reperfusion period and 24 and 48 hr after reperfusion. Assessment of the antioxidant enzymes and tumor necrosis factor alpha (TNF-α) was performed by ELISA method. Caspase-3 and glial fibrillary acidic protein (GFAP) proteins expression levels were evaluated by immunohistochemical staining 7 days after ischemia. Results: Based on the result of the current study, lower superoxide dismutase (SOD) and glutathione (GSH) (P<0.05) and higher malondialdehyde (MDA) and TNF-α levels were observed in the ischemia group than those of the sham group (P<0.01). Obestatin treatment could increase both SOD and GSH (P<0.05) and reduce MDA and TNF-α (P<0.05) versus the ischemia group. Moreover, obestatin could significantly decrease caspase-3 and GFAP positive cells in the CA1 region of hippocampus (P<0.01). Conclusion: Obestatin exerts protective effects against ischemia injury by inhibition of astrocytes activation and decreases neuronal cell apoptosis via its antioxidant properties.
Collapse
Affiliation(s)
- Elahe Mirarab
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Vida Hojati
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Golamhassan Vaezi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | | | - Mehdi Khaksari
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
15
|
Stempniewicz A, Ceranowicz P, Warzecha Z. Potential Therapeutic Effects of Gut Hormones, Ghrelin and Obestatin in Oral Mucositis. Int J Mol Sci 2019; 20:ijms20071534. [PMID: 30934722 PMCID: PMC6479885 DOI: 10.3390/ijms20071534] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/15/2019] [Accepted: 03/19/2019] [Indexed: 12/16/2022] Open
Abstract
Chemotherapy and/or head and neck radiotherapy are frequently associated with oral mucositis. Oral pain, odynophagia and dysphagia, opioid use, weight loss, dehydration, systemic infection, hospitalization and introduction of a feeding tube should be mentioned as the main determinated effect of oral mucositis. Oral mucositis leads to a decreased quality of life and an increase in treatment costs. Moreover, oral mucositis is a life-threatening disease. In addition to its own direct life-threatening consequences, it can also lead to a reduced survival due to the discontinuation or dose reduction of anti-neoplasm therapy. There are numerous strategies for the prevention or treatment of oral mucositis; however, their effectiveness is limited and does not correspond to expectations. This review is focused on the ghrelin and obestatin as potentially useful candidates for the prevention and treatment of chemo- or/and radiotherapy-induced oral mucositis.
Collapse
Affiliation(s)
- Agnieszka Stempniewicz
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Grzegórzecka 16 St., 31-531 Krakow, Poland.
| | - Piotr Ceranowicz
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Grzegórzecka 16 St., 31-531 Krakow, Poland.
| | - Zygmunt Warzecha
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Grzegórzecka 16 St., 31-531 Krakow, Poland.
| |
Collapse
|
16
|
Salman OF, El-Rayess HM, Abi Khalil C, Nemer G, Refaat MM. Inherited Cardiomyopathies and the Role of Mutations in Non-coding Regions of the Genome. Front Cardiovasc Med 2018; 5:77. [PMID: 29998127 PMCID: PMC6028572 DOI: 10.3389/fcvm.2018.00077] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/04/2018] [Indexed: 01/16/2023] Open
Abstract
Cardiomyopathies (CMs) are a group of cardiac pathologies caused by an intrinsic defect within the myocardium. The relative contribution of genetic mutations in the pathogenesis of certain CMs, such as hypertrophic cardiomyopathy (HCM), arrythmogenic right/left ventricular cardiomyopathy (ARVC) and left ventricular non-compacted cardiomyopathy (LVNC) has been established in comparison to dilated cardiomyopathy (DCM) and restrictive cardiomyopathy (RCM). The aim of this article is to review mutations in the non-coding parts of the genome, namely, microRNA, promoter elements, enhancer/silencer elements, 3′/5′UTRs and introns, that are involved in the pathogenesis CMs. Additionally, we will explore the role of some long non-coding RNAs in the pathogenesis of CMs.
Collapse
Affiliation(s)
- Oday F Salman
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hebah M El-Rayess
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Charbel Abi Khalil
- Department of Genetic Medicine, Weill Cornell Medical College, Doha, Qatar
| | - Georges Nemer
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Marwan M Refaat
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
17
|
Jovanović A. Cardioprotective signalling: Past, present and future. Eur J Pharmacol 2018; 833:314-319. [PMID: 29935170 DOI: 10.1016/j.ejphar.2018.06.029] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/08/2018] [Accepted: 06/19/2018] [Indexed: 11/29/2022]
Abstract
A few decades ago, cardiac muscle was discovered to possess signalling pathways that, when activated, protect the myocardium against the damage induced by ischaemia-reperfusion. The ability of cardiac muscle to protect itself against injury has been termed 'cardioprotection'. Many compounds and procedures can trigger cardioprotection including conditionings (exposure to brief episodes of ischaemia-reperfusion to protect against sustained ischaemia-reperfusion), hypoxia, adenosine, acetylcholine, adrenomedullin, angiotensin, bradykinin, catecholamines, endothelin, estrogens, phenylephrine, opioids, testosterone, and many more. These triggers activate many intracellular signalling factors including protein kinases, different enzymes, transcription factors and defined signalling pathways to target structures in mitochondria, sarcoplasmic reticulum, nucleus and sarcolemma to mediate cardioprotection. Although a lot of information about cardioprotection has been acquired, there are still two major outstanding issues to be addressed in the future 1) better understanding of spatio-temporal relationships between signalling elements, and; 2) devising therapeutic strategies against myocardial diseases based on cardioprotective signalling. Further research is required to paint integral picture of cardioprotective signalling and more clinical studies are required to properly test clinical efficacy and safety of potential cardioprotective strategies. Therapies against cardiac diseases based on cardioprotective strategies would be a perfect adjunct to current therapeutic strategies based on restitution of coronary blood flow and regulation of myocardial metabolic demands.
Collapse
Affiliation(s)
- Aleksandar Jovanović
- University of Nicosia Medical School, 21 Ilia Papakyriakou, 2414 Engomi, P.O. Box 24005, CY-1700 Nicosia, Cyprus.
| |
Collapse
|
18
|
Konarska K, Cieszkowski J, Warzecha Z, Ceranowicz P, Chmura A, Kuśnierz-Cabala B, Gałązka K, Kowalczyk P, Miskiewicz A, Konturek TJ, Pędziwiatr M, Dembiński A. Treatment with Obestatin-A Ghrelin Gene-Encoded Peptide-Reduces the Severity of Experimental Colitis Evoked by Trinitrobenzene Sulfonic Acid. Int J Mol Sci 2018; 19:ijms19061643. [PMID: 29865176 PMCID: PMC6032262 DOI: 10.3390/ijms19061643] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 05/25/2018] [Accepted: 05/28/2018] [Indexed: 02/06/2023] Open
Abstract
Obestatin is a 23-amino acid peptide derived from proghrelin, a common prohormone for ghrelin and obestatin. Previous studies showed that obestatin exhibited some protective and therapeutic effects in the gut. The aim of our presented study was to examine the effect of treatment with obestatin on trinitrobenzene sulfonic acid (TNBS)-induced colitis. In rats anesthetized with ketamine, colitis was induced through intrarectal administration of 25 mg of 2,4,6-trinitrobenzene sulfonic acid (TNBS). Obestatin was administered intraperitoneally at doses of 4, 8, or 16 nmol/kg, twice per day for four consecutive days. The first dose of obestatin was given one day before the induction of colitis, and the last one was given two days after administration of TNBS. Fourteen days after the induction of colitis, rats were anesthetized again with ketamine, and the severity of colitis was determined. The administration of obestatin had no effect on the parameters tested in rats without the induction of colitis. In rats with colitis, administration of obestatin at doses of 8 or 16 nmol/kg reduced the area of colonic damage, and improved mucosal blood flow in the colon. These effects were accompanied by a reduction in the colitis-evoked increase in the level of blood leukocytes, and mucosal concentration of pro-inflammatory interleukin-1β. Moreover, obestatin administered at doses of 8 or 16 nmol/kg reduced histological signs of colonic damage. The administration of obestatin at a dose of 4 nmol/kg failed to significantly affect the parameters tested. Overall, treatment with obestatin reduced the severity of TNBS-induced colitis in rats. This effect was associated with an improvement in mucosal blood flow in the colon, and a decrease in local and systemic inflammatory processes.
Collapse
Affiliation(s)
- Katarzyna Konarska
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland.
| | - Jakub Cieszkowski
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland.
| | - Zygmunt Warzecha
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland.
| | - Piotr Ceranowicz
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland.
| | - Anna Chmura
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland.
| | - Beata Kuśnierz-Cabala
- Department of Clinical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, 31-501 Cracow, Poland.
| | - Krystyna Gałązka
- Department of Pathomorphology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland.
| | - Paweł Kowalczyk
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jablonna, Poland.
| | - Andrzej Miskiewicz
- Department of Periodontology and Oral Diseases, Medical University of Warsaw, 00-246 Warsaw, Poland.
| | - Thomas Jan Konturek
- Department of Medicine, St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA 02135, USA.
| | - Michał Pędziwiatr
- Second Department of General Surgery, Faculty of Medicine, Jagiellonian University Medical College, 31-501 Cracow, Poland.
| | - Artur Dembiński
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland.
| |
Collapse
|
19
|
Beberashvili I, Katkov A, Sinuani I, Azar A, Shapiro G, Feldman L, Gorelik O, Stav K, Efrati S. Serum Obestatin: A Biomarker of Cardiovascular and All-Cause Mortality in Hemodialysis Patients. Am J Nephrol 2018; 47:254-265. [PMID: 29694945 DOI: 10.1159/000488285] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 03/07/2018] [Indexed: 01/10/2023]
Abstract
BACKGROUND Recent experimental studies have suggested that obestatin, a proposed anorexigenic gut hormone and a physiological opponent of acyl-ghrelin, has protective cardiovascular effects. We tested the hypothesis that obestatin is independent of inflammatory mediators and/or acyl-ghrelin in predicting outcomes of the maintenance hemodialysis (MHD) population. METHODS It was a 6-year cohort study on 261 MHD patients. Obestatin, acyl-ghrelin, adipokines (leptin and adiponectin), markers of inflammation and nutrition, prospective all-cause and cardiovascular mortality were studied. RESULTS During the follow-up, 160 patients died in total, with 74 deaths due to cardiovascular causes. For each ng/mL increase in baseline obestatin level in fully adjusted models (including malnutrition-inflammation score, Interleukin-6 [IL-6], adipokines and acyl-ghrelin), the hazard for death from all causes was 0.90 (95% CI 0.81-0.99) and for cardiovascular death 0.85 (95% CI 0.73-0.99). However, these associations were more robust in the subgroup of patients aged above 71 years: 0.85 (95% CI 0.73-0.98) for all-cause death and 0.66 (95% CI 0.52-0.85) for cardiovascular death. An interaction between high IL-6 (above median) and low obestatin (below median) levels for increased risk of all-cause mortality (synergy index [SI] 5.14, p = 0.001) and cardiovascular mortality (SI 4.81, p = 0.02) emerged in the development of multivariable adjusted models. Interactions were also observed between obestatin, Tumor necrosis factor-alpha, adipokines and acyl-ghrelin, which were associated with mortality risk. CONCLUSION Serum obestatin behaves as a biomarker for cardiovascular and all-cause mortality in MHD patients. The prognostic ability of obestatin in this regard is independent of inflammation, nutritional status, acyl-ghrelin's and adipokines' activity and is modified by age being very prominent in patients older than 71 years.
Collapse
Affiliation(s)
- Ilia Beberashvili
- Division of Nephrology, Assaf Harofeh Medical Center, Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Zerifin, Israel
| | - Anna Katkov
- Division of Nephrology, Assaf Harofeh Medical Center, Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Zerifin, Israel
| | - Inna Sinuani
- Department of Pathology, Assaf Harofeh Medical Center, Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Zerifin, Israel
| | - Ada Azar
- Department of Nutrition, Assaf Harofeh Medical Center, Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Zerifin, Israel
| | - Gregory Shapiro
- Division of Nephrology, Assaf Harofeh Medical Center, Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Zerifin, Israel
| | - Leonid Feldman
- Division of Nephrology, Assaf Harofeh Medical Center, Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Zerifin, Israel
| | - Oleg Gorelik
- Department of Internal Medicine F, Assaf Harofeh Medical Center, Affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Zerifin, Israel
| | - Kobi Stav
- Department of Urology, Assaf Harofeh Medical Center, Affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Zerifin, Israel
| | - Shai Efrati
- Division of Nephrology, Assaf Harofeh Medical Center, Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Zerifin, Israel
| |
Collapse
|
20
|
Green BD, Grieve DJ. Biochemical properties and biological actions of obestatin and its relevence in type 2 diabetes. Peptides 2018; 100:249-259. [PMID: 29412827 DOI: 10.1016/j.peptides.2017.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/05/2017] [Accepted: 12/06/2017] [Indexed: 12/15/2022]
Abstract
Obestatin was initially discovered in rat stomach extract, and although it is principally produced in the gastric mucosa, it can be found throughout the gastrointestinal tract. This 23-amino acid C-terminally amidated peptide is derived from preproghrelin and has been ascribed a wide range of metabolic effects relevant to type 2 diabetes. Obestatin reportedly inhibits gastrointestinal motility, reduces food intake and lowers body weight and improves lipid metabolism. Furthermore, it appears to exert actions on the pancreatic β-cell, most notably increasing β-cell mass and upregulating genes associated with insulin production and β-cell regeneration, with relevance to type 2 diabetes. It is becoming evident that obestatin also exerts pleiotropic effects on the cardiovascular system, possibly modulating blood pressure, endothelial function and triggering cardioprotective mechanisms, which may be important in determining cardiovascular outcomes in type 2 diabetes. Furthermore, it seems that like other gut peptides obestatin has neuroprotective properties. This review examines the biochemical properties of the obestatin peptide (its structure, sequence, stability and distribution) and the candidate receptors through which it may act. It provides a balanced examination of the reported pancreatic and extrapancreatic actions of obestatin and evaluates its potential relevance with respect to diabetes therapy, together with discussion of direct evidence linking alterations in obestatin signalling with obesity/diabetes and other diseases.
Collapse
Affiliation(s)
- Brian D Green
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, BT9 5BN, UK.
| | - David J Grieve
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7AE, UK
| |
Collapse
|
21
|
Liu Y, Xing YX, Gao XY, Kuang HY, Zhang J, Liu R. Obestatin prevents H 2O 2-induced damage through activation of TrkB in RGC-5 cells. Biomed Pharmacother 2017; 97:1061-1065. [PMID: 29136785 DOI: 10.1016/j.biopha.2017.11.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 11/03/2017] [Accepted: 11/03/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUD In the early stage of diabetic retinopathy, the damage of retinal ganglion cells already exists, promoting the development of the disease. The aim of this study was to investigate the protective role and the mechanisms of obestatin against H2O2-induced damage in RGC-5 cells. METHODS RGC-5 cells were incubated with various concentrations of obestatin for 24h before H2O2 added. The survival rates of RGC-5 were measured by MTT assay. The expression of apoptosis-related proteins and TrkB pathway-related proteins were detected by Western blot analysis. RESULTS Our data showed that H2O2 evidently decreased the survival rate of RGC-5 cells. However, obestatin pretreatment reversed the decreased activity. Moreover, obestatin effectively increased the expression of Bcl-2 and decreased the expression of Bax. In addition, obestatin potentially plays a role in protecting RGC-5 by activating of TrkB. Obestatin notablely increased the phosphorylation of TrkB, AKT and ERK1/2. All these effects of obestatin can be inhibited by GLP-1R antagonist exendin (9-39). CONCLUSIONS Obestatin prevents H2O2-induced damage in RGC-5 cells by activating TrkB pathway. Moreover, GLP-1R is closely related to the function of obestatin in RGC-5 cells.
Collapse
Affiliation(s)
- Yang Liu
- Department of Endocrinology, The First Clinical Hospital of Harbin Medical University, Harbin, 150001, China.
| | - Yue-Xian Xing
- Department of Endocrinology, The First Clinical Hospital of Harbin Medical University, Harbin, 150001, China.
| | - Xin-Yuan Gao
- Department of Endocrinology, The First Clinical Hospital of Harbin Medical University, Harbin, 150001, China.
| | - Hong-Yu Kuang
- Department of Endocrinology, The First Clinical Hospital of Harbin Medical University, Harbin, 150001, China.
| | - Jing Zhang
- Department of Endocrinology, The First Clinical Hospital of Harbin Medical University, Harbin, 150001, China.
| | - Rong Liu
- Department of Endocrinology, The First Clinical Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
22
|
Liu ZY, Hu S, Zhong QW, Tian CN, Ma HM, Yu JJ. N-Methyl-D-Aspartate Receptor-Driven Calcium Influx Potentiates the Adverse Effects of Myocardial Ischemia-Reperfusion Injury Ex Vivo. J Cardiovasc Pharmacol 2017; 70:329-338. [PMID: 28777252 PMCID: PMC5673305 DOI: 10.1097/fjc.0000000000000527] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 05/18/2017] [Indexed: 01/30/2023]
Abstract
BACKGROUND Despite the adverse effects of N-methyl-D-aspartate receptor (NMDAR) activity in cardiomyocytes, no study has yet examined the effects of NMDAR activity under ex vivo ischemic-reperfusion (I/R) conditions. Therefore, our aim was to comprehensively evaluate the effects of NMDAR activity through an ex vivo myocardial I/R rat model. METHODS Isolated rat hearts were randomly segregated into 6 groups (n = 20 in each group): (1) an untreated control group; (2) a NMDA-treated control group; (3) an untreated I/R group; (4) an I/R+NMDA group treated with NMDA; (5) an I/R+NMDA+MK-801 group treated with NMDA and the NMDAR inhibitor MK-801; and (6) an I/R+NMDA+[Ca]-free group treated with NMDA and [Ca]-free buffer. The 4 I/R groups underwent 30 minutes of ischemia followed by 50 minutes of reperfusion. Left ventricular pressure signals were analyzed to assess cardiac performance. Myocardial intracellular calcium levels ([Ca]i) were assessed in isolated ventricular cardiomyocytes. Creatine kinase, creatine kinase isoenzyme MB, lactate dehydrogenase, cardiac troponin I, and cardiac troponin T were assayed from coronary effluents. TTC and TUNEL staining were used to measure generalized myocardial necrosis and apoptosis levels, respectively. Western blotting was applied to assess the phosphorylation of PKC-δ, PKC-ε, Akt, and extracellular signal-regulated kinase. RESULTS Enhanced NMDAR activity under control conditions had no significant effects on the foregoing variables. In contrast, enhanced NMDAR activity under I/R conditions produced significant increases in [Ca]i levels (∼1.2% increase), significant losses in left ventricular function (∼5.4% decrease), significant multi-fold increases in creatine kinase, creatine kinase isoenzyme MB, lactate dehydrogenase, cardiac troponin I, and cardiac troponin T, significant increases in generalized myocardial necrosis (∼36% increase) and apoptosis (∼150% increase), and significant multi-fold increases in PKC-δ, PKC-ε, Akt, and extracellular signal-regulated kinase phosphorylation (all P < 0.05). These adverse effects were rescued by the NMDAR inhibitor MK-801 or [Ca]-free buffer (all P < 0.05). CONCLUSIONS NMDAR-driven calcium influx potentiates the adverse effects of myocardial I/R injury ex vivo.
Collapse
Affiliation(s)
- Zi-You Liu
- Department of Heart Center, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jianxi, China
| | | | | | | | | | | |
Collapse
|
23
|
Penna C, Tullio F, Femminò S, Rocca C, Angelone T, Cerra MC, Gallo MP, Gesmundo I, Fanciulli A, Brizzi MF, Pagliaro P, Alloatti G, Granata R. Obestatin regulates cardiovascular function and promotes cardioprotection through the nitric oxide pathway. J Cell Mol Med 2017; 21:3670-3678. [PMID: 28744974 PMCID: PMC5706590 DOI: 10.1111/jcmm.13277] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/15/2017] [Indexed: 02/06/2023] Open
Abstract
Patients with ischaemic heart disease or chronic heart failure show altered levels of obestatin, suggesting a role for this peptide in human heart function. We have previously demonstrated that GH secretagogues and the ghrelin gene-derived peptides, including obestatin, exert cardiovascular effects by modulating cardiac inotropism and vascular tone, and reducing cell death and contractile dysfunction in hearts subjected to ischaemia/reperfusion (I/R), through the Akt/nitric oxide (NO) pathway. However, the mechanisms underlying the cardiac actions of obestatin remain largely unknown. Thus, we suggested that obestatin-induced activation of PI3K/Akt/NO and PKG signalling is implicated in protection of the myocardium when challenged by adrenergic, endothelinergic or I/R stress. We show that obestatin exerts an inhibitory tone on the performance of rat papillary muscle in both basal conditions and under β-adrenergic overstimulation, through endothelial-dependent NO/cGMP/PKG signalling. This pathway was also involved in the vasodilator effect of the peptide, used both alone and under stress induced by endothelin-1. Moreover, when infused during early reperfusion, obestatin reduced infarct size in isolated I/R rat hearts, through an NO/PKG pathway, comprising ROS/PKC signalling, and converging on mitochondrial ATP-sensitive potassium [mitoK(ATP)] channels. Overall, our results suggest that obestatin regulates cardiovascular function in stress conditions and induces cardioprotection by mechanisms dependent on activation of an NO/soluble guanylate cyclase (sGC)/PKG pathway. In fact, obestatin counteracts exaggerated β-adrenergic and endothelin-1 activity, relevant factors in heart failure, suggesting multiple positive effects of the peptide, including the lowering of cardiac afterload, thus representing a potential candidate in pharmacological post-conditioning.
Collapse
Affiliation(s)
- Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy.,National Institute of Cardiovascular Research, Bologna, Italy
| | - Francesca Tullio
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Carmine Rocca
- National Institute of Cardiovascular Research, Bologna, Italy.,Department of Biology, Ecology and E.S., University of Calabria, Rende, CS, Italy
| | - Tommaso Angelone
- National Institute of Cardiovascular Research, Bologna, Italy.,Department of Biology, Ecology and E.S., University of Calabria, Rende, CS, Italy
| | - Maria C Cerra
- National Institute of Cardiovascular Research, Bologna, Italy.,Department of Biology, Ecology and E.S., University of Calabria, Rende, CS, Italy
| | - Maria Pia Gallo
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Iacopo Gesmundo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | | | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy.,National Institute of Cardiovascular Research, Bologna, Italy
| | - Giuseppe Alloatti
- National Institute of Cardiovascular Research, Bologna, Italy.,Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Riccarda Granata
- Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
24
|
Zhang Q, Dong XW, Xia JY, Xu KY, Xu ZR. Obestatin Plays Beneficial Role in Cardiomyocyte Injury Induced by Ischemia-Reperfusion In Vivo and In Vitro. Med Sci Monit 2017; 23:2127-2136. [PMID: 28472020 PMCID: PMC5426386 DOI: 10.12659/msm.901361] [Citation(s) in RCA: 11] [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: 09/01/2016] [Accepted: 10/13/2016] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Obestatin, primarily recognized as a peptide within the gastrointestinal system, has been shown to benefit the cardiovascular system. We designed this experiment to study the protective role and underlying mechanism of obestatin against ischemia-reperfusion(I/R) injury in myocardial cells. MATERIAL AND METHODS In an In vivo experiment, LAD was ligated for 0.5 h and then opened for reperfusion with obestatin for 24 h. Then, the infarction area was shown with TTC staining, and inflammation factors in serum were analyzed by qRT-PCR. In primary cultured cardiomyocytes, we measured the level of LDH, MDA, GSH, and SOD. Finally, we assessed cells apoptosis using flow cytometry and detected the concentrations of caspase-3, Bax, and Bcl-2 using Western blot analysis. RESULTS TTC staining showed that in the 3 obestatin groups, the infarct area became smaller with the increase of obestatin concentration. Obestatin also inhibited LDH expression in rat serum and decreased mRNA levels of TNF-α, IL-6, ICAM-1, and iNOS in rat cardiomyocytes after reperfusion. In primary cultured cardiomyocytes, obestatin decreased LDH content and increased GSH level after I/R injury. Obestatin was also found to antagonize the apoptosis of cardiomyocytes in a dose-dependent manner. Western blot analysis showed that obestatin downregulated the expression of caspase-3 and Bax and upregulated the expression of Bcl-2. CONCLUSIONS Obestatin can protect cardiomyocyte from I/R-induced injury in vitro and in vivo. This beneficial effect is closely related with its properties of anti-inflammation, anti-cytotoxicity, and anti-apoptosis. The protective effect of obestatin might be associated with activation of Bcl-2 and inhibition of caspase-3 and Bax.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Geriatrics, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Xin-wei Dong
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Jia-ying Xia
- Department of Geriatrics, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Ke-ying Xu
- Department of Geriatrics, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Zhe-rong Xu
- Department of Geriatrics, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
25
|
Pradhan G, Wu CS, Han Lee J, Kanikarla P, Guo S, Yechoor VK, Samson SL, Sun Y. Obestatin stimulates glucose-induced insulin secretion through ghrelin receptor GHS-R. Sci Rep 2017; 7:979. [PMID: 28428639 PMCID: PMC5430520 DOI: 10.1038/s41598-017-00888-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 03/07/2017] [Indexed: 11/19/2022] Open
Abstract
Orexigenic hormone ghrelin and anorexic hormone obestatin are encoded by the same preproghrelin gene. While it is known that ghrelin inhibits glucose-stimulated insulin secretion (GSIS), the effect of obestatin on GSIS is unclear. Ghrelin's effect is mediated by its receptor Growth Hormone Secretagogue Receptor (GHS-R), but the physiologically relevant receptor of obestatin remains debatable. Here we have investigated the effect of obestatin on GSIS in vitro, in vivo and ex vivo, and tested whether obestatin regulates insulin secretion through GHS-R. We found that under hyperglycemic condition, obestatin augments GSIS in rat insulinoma cells (INS-1) and in pancreatic islets from ghrelin -/- mice. Surprisingly, obestatin-induced GSIS was absent in β-cells in which GHS-R was suppressed. Obestatin-induced insulin secretion was abolished in the circulation of Ghsr -/- mice, and in pancreatic islets isolated from Ghsr -/- mice. We also found that obestatin-induced GSIS was attenuated in islets isolated from β-cell-specific Ghsr knockout MIP-Cre/ERT;Ghsrf/f mice. Our data collectively demonstrate that obestatin is a potent insulin secretagogue under hyperglycemic condition, and obestatin's effect on insulin secretion is mediated by GHS-R in pancreatic β-cells. Our findings reveal an intriguing insight that obestatin and ghrelin have opposing effects on insulin secretion, and both are mediated through ghrelin receptor GHS-R.
Collapse
Affiliation(s)
- Geetali Pradhan
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Chia-Shan Wu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA
| | - Jong Han Lee
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- College of Pharmacy, Gachon University, Incheon, 21936, Korea
| | - Preeti Kanikarla
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Shaodong Guo
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA
| | - Vijay K Yechoor
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Susan L Samson
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Yuxiang Sun
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
26
|
Wang S, Zhang F, Zhao G, Cheng Y, Wu T, Wu B, Zhang YE. Mitochondrial PKC-ε deficiency promotes I/R-mediated myocardial injury via GSK3β-dependent mitochondrial permeability transition pore opening. J Cell Mol Med 2017; 21:2009-2021. [PMID: 28266127 PMCID: PMC5571523 DOI: 10.1111/jcmm.13121] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 01/05/2017] [Indexed: 11/29/2022] Open
Abstract
Mitochondrial fission is critically involved in cardiomyocyte apoptosis, which has been considered as one of the leading causes of ischaemia/reperfusion (I/R)‐induced myocardial injury. In our previous works, we demonstrate that aldehyde dehydrogenase‐2 (ALDH2) deficiency aggravates cardiomyocyte apoptosis and cardiac dysfunction. The aim of this study was to elucidate whether ALDH2 deficiency promotes mitochondrial injury and cardiomyocyte death in response to I/R stress and the underlying mechanism. I/R injury was induced by aortic cross‐clamping for 45 min. followed by unclamping for 24 hrs in ALDH2 knockout (ALDH2−/−) and wild‐type (WT) mice. Then myocardial infarct size, cell apoptosis and cardiac function were examined. The protein kinase C (PKC) isoform expressions and their mitochondrial translocation, the activity of dynamin‐related protein 1 (Drp1), caspase9 and caspase3 were determined by Western blot. The effects of N‐acetylcysteine (NAC) or PKC‐δ shRNA treatment on glycogen synthase kinase‐3β (GSK‐3β) activity and mitochondrial permeability transition pore (mPTP) opening were also detected. The results showed that ALDH2−/− mice exhibited increased myocardial infarct size and cardiomyocyte apoptosis, enhanced levels of cleaved caspase9, caspase3 and phosphorylated Drp1. Mitochondrial PKC‐ε translocation was lower in ALDH2−/− mice than in WT mice, and PKC‐δ was the opposite. Further data showed that mitochondrial PKC isoform ratio was regulated by cellular reactive oxygen species (ROS) level, which could be reversed by NAC pre‐treatment under I/R injury. In addition, PKC‐ε inhibition caused activation of caspase9, caspase3 and Drp1Ser616 in response to I/R stress. Importantly, expression of phosphorylated GSK‐3β (inactive form) was lower in ALDH2−/− mice than in WT mice, and both were increased by NAC pre‐treatment. I/R‐induced mitochondrial translocation of GSK‐3β was inhibited by PKC‐δ shRNA or NAC pre‐treatment. In addition, mitochondrial membrane potential (∆Ψm) was reduced in ALDH2−/− mice after I/R, which was partly reversed by the GSK‐3β inhibitor (SB216763) or PKC‐δ shRNA. Collectively, our data provide the evidence that abnormal PKC‐ε/PKC‐δ ratio promotes the activation of Drp1 signalling, caspase cascades and GSK‐3β‐dependent mPTP opening, which results in mitochondrial injury‐triggered cardiomyocyte apoptosis and myocardial dysfuction in ALDH2−/− mice following I/R stress.
Collapse
Affiliation(s)
- Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Feng Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gang Zhao
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong Cheng
- Heart Centre of Zhengzhou Ninth People's Hospital, Zhengzhou, Henan, China
| | - Ting Wu
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Bing Wu
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - You-En Zhang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
27
|
Xing YX, Yang L, Kuang HY, Gao XY, Liu HL. Function of obestatin in the digestive system. Nutrition 2017; 34:21-28. [DOI: 10.1016/j.nut.2016.08.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 04/18/2016] [Accepted: 08/29/2016] [Indexed: 02/07/2023]
|
28
|
Li HQ, Wu YB, Yin CS, Chen L, Zhang Q, Hu LQ. Obestatin attenuated doxorubicin-induced cardiomyopathy via enhancing long noncoding Mhrt RNA expression. Biomed Pharmacother 2016; 81:474-481. [DOI: 10.1016/j.biopha.2016.04.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 04/06/2016] [Accepted: 04/08/2016] [Indexed: 11/26/2022] Open
|
29
|
Cowan E, Burch KJ, Green BD, Grieve DJ. Obestatin as a key regulator of metabolism and cardiovascular function with emerging therapeutic potential for diabetes. Br J Pharmacol 2016; 173:2165-81. [PMID: 27111465 DOI: 10.1111/bph.13502] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/05/2016] [Accepted: 04/15/2016] [Indexed: 01/01/2023] Open
Abstract
Obestatin is a 23-amino acid C-terminally amidated gastrointestinal peptide derived from preproghrelin and which forms an α helix. Although obestatin has a short biological half-life and is rapidly degraded, it is proposed to exert wide-ranging pathophysiological actions. Whilst the precise nature of many of its effects is unclear, accumulating evidence supports positive actions on both metabolism and cardiovascular function. For example, obestatin has been reported to inhibit food and water intake, body weight gain and gastrointestinal motility and also to mediate promotion of cell survival and prevention of apoptosis. Obestatin-induced increases in beta cell mass, enhanced adipogenesis and improved lipid metabolism have been noted along with up-regulation of genes associated with beta cell regeneration, insulin production and adipogenesis. Furthermore, human circulating obestatin levels generally demonstrate an inverse association with obesity and diabetes, whilst the peptide has been shown to confer protective metabolic effects in experimental diabetes, suggesting that it may hold therapeutic potential in this setting. Obestatin also appears to be involved in blood pressure regulation and to exert beneficial effects on endothelial function, with experimental studies indicating that it may also promote cardioprotective actions against, for example, ischaemia-reperfusion injury. This review will present a critical appraisal of the expanding obestatin research area and discuss the emerging therapeutic potential of this peptide for both metabolic and cardiovascular complications of diabetes.
Collapse
Affiliation(s)
- Elaine Cowan
- Queen's University Belfast, Institute for Global Food Security, School of Biological Sciences, Belfast, UK
| | - Kerry J Burch
- Queen's University Belfast, Wellcome-Wolfson Institute for Experimental Medicine, Belfast, UK
| | - Brian D Green
- Queen's University Belfast, Institute for Global Food Security, School of Biological Sciences, Belfast, UK
| | - David J Grieve
- Queen's University Belfast, Wellcome-Wolfson Institute for Experimental Medicine, Belfast, UK
| |
Collapse
|
30
|
Abstract
OBJECTIVE The aim of this study was to determine the impact of obestatin therapy on the course of cerulein-induced pancreatitis. METHODS Acute pancreatitis was induced by cerulein given intraperitoneally 5 times with 1 hour intervals at the dose of 50 μg/kg per dose. Obestatin was administered twice a day at the dose of 8 nmol/kg per dose, starting the first dose 24 hours after the last injection of cerulein. Severity of acute pancreatitis (AP) was examined at 0 hour or 1, 2, 3, 5, 7, and 10 days after the last injection of cerulein. RESULTS Administration of cerulein led to development of acute edematous pancreatitis in all rats, and maximal severity of this disease was observed 24 hours after induction of pancreatitis. Treatment with obestatin reduced morphological signs of pancreatic damage (pancreatic edema, leukocyte infiltration, vacuolization of acinar cells) and led to earlier regeneration of the pancreas. Biochemical indexes of severity of pancreatitis such as serum activity of pancreatic digestive enzymes were significantly reduced in animals treated with obestatin. These effects were accompanied by increase in pancreatic DNA synthesis and decrease in serum level of proinflammatory interleukin 1β. In addition, administration of obestatin improved pancreatic blood flow in rats with AP. CONCLUSIONS Treatment with exogenous obestatin reduces severity of AP and accelerates pancreatic recovery.
Collapse
|
31
|
Gargantini E, Lazzari L, Settanni F, Taliano M, Trovato L, Gesmundo I, Ghigo E, Granata R. Obestatin promotes proliferation and survival of adult hippocampal progenitors and reduces amyloid-β-induced toxicity. Mol Cell Endocrinol 2016; 422:18-30. [PMID: 26586206 DOI: 10.1016/j.mce.2015.11.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 11/05/2015] [Accepted: 11/07/2015] [Indexed: 12/31/2022]
Abstract
The ghrelin gene-derived peptide obestatin promotes survival in different cell types through a yet undefined receptor; however, its potential neuroprotective activities are still unknown. Here, obestatin effects were investigated on proliferation and survival of adult rat hippocampal progenitor cells (AHPs). Obestatin immunoreactivity was found in AHPs; moreover, obestatin binding to AHPs was displaced by the GLP-1R agonist Ex-4 and antagonist Ex-9. Furthermore, obestatin increased cell proliferation and survival in growth factor deprived medium and inhibited apoptosis; these effects were blocked by Ex-9. The underlying mechanisms involved Gαs/cAMP/PKA/CREB signaling, phosphorylation of ERK1/2 and PI3K/Akt, and the PI3K targets GSK-3β/β-catenin and mTOR. Obestatin also counteracted Aβ1-42-induced detrimental effects through inhibition of GSK-3β activity and Tau hyperphosphorylation, main hallmarks of neuronal death in Alzheimer's disease. These findings indicate a novel protective role for obestatin in AHPs and candidate this peptide as potential therapeutic target for increasing neurogenesis and for approaching neurodegenerative disorders.
Collapse
Affiliation(s)
- Eleonora Gargantini
- Laboratory of Molecular and Cellular Endocrinology, University of Torino, 10126 Torino, Italy; Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Laura Lazzari
- Laboratory of Molecular and Cellular Endocrinology, University of Torino, 10126 Torino, Italy; Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Fabio Settanni
- Laboratory of Molecular and Cellular Endocrinology, University of Torino, 10126 Torino, Italy; Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Marina Taliano
- Laboratory of Molecular and Cellular Endocrinology, University of Torino, 10126 Torino, Italy; Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Letizia Trovato
- Laboratory of Molecular and Cellular Endocrinology, University of Torino, 10126 Torino, Italy; Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Iacopo Gesmundo
- Laboratory of Molecular and Cellular Endocrinology, University of Torino, 10126 Torino, Italy; Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Ezio Ghigo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Riccarda Granata
- Laboratory of Molecular and Cellular Endocrinology, University of Torino, 10126 Torino, Italy; Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Torino, 10126 Torino, Italy.
| |
Collapse
|
32
|
Pamukcu O, Baykan A, Bayram LC, Narin F, Cetin N, Narin N, Argun M, Ozyurt A, Uzum K. Anti-inflammatory role of obestatin in autoimmune myocarditis. Clin Exp Pharmacol Physiol 2015; 43:47-55. [PMID: 26426263 DOI: 10.1111/1440-1681.12497] [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: 06/12/2015] [Revised: 09/23/2015] [Accepted: 09/24/2015] [Indexed: 11/28/2022]
Abstract
Obestatin is a popular endogeneous peptide, known to have an autoimmune regulatory effect on energy metabolism and the gastrointestinal system. Studies regarding the anti-inflammatory effects of obestatin are scarce. The aim of this study was to show the anti-inflammatory effect of obestatin in an experimental model of autoimmune myocarditis in rats. Experimental autoimmune myocarditis was induced in Lewis rats by immunization with subcutaneous administration of porcine cardiac myosin, twice at 7-day intervals. Intraperitoneal pretreatment with obestatin (50 μg/kg) was started before the induction of myocarditis and continued for 3 weeks. The severity of myocarditis was evidenced by clinical, echocardiographic and histological findings. In addition, by-products of neutrophil activation, lipid peroxidation, inflammatory and anti-inflammatory cytokines were measured in serum. Obestatin significantly ameliorated the clinical and histopathological severity of autoimmune myocarditis. Therapeutic effects of obestatin in myocarditis were associated with reduced lipid peroxidation, suppression of polymorphonuclear leukocyte infiltration and enhancement of glutathione synthesis, inhibition of serum inflammatory and activation of anti-inflammatory cytokines. Histopathologically, the left ventricle was significantly dilated, and its wall thickened, along with widespread lymphocytic and histocytic infiltration. The myocardium was severely infiltrated with relatively large mononuclear cells. These histopathological changes were observed in lesser degrees in obestatin-treated rats. This study demonstrated a novel anti-inflammatory effect of obestatin in an experimental model of autoimmune myocarditis. Consequently, obestatin administration may represent a promising therapeutic approach for myocarditis and dilated cardiomyopathy in the future.
Collapse
Affiliation(s)
- Ozge Pamukcu
- Division of Pediatric Cardiology, Erciyes University, Kayseri, Turkey
| | - Ali Baykan
- Division of Pediatric Cardiology, Erciyes University, Kayseri, Turkey
| | | | - Figen Narin
- Division of Biochemistry, School of Medicine, Erciyes University, Kayseri, Turkey
| | - Nazmi Cetin
- School of Veterinarian Physiology, Erciyes University, Kayseri, Turkey
| | - Nazmi Narin
- Division of Pediatric Cardiology, Erciyes University, Kayseri, Turkey
| | - Mustafa Argun
- Division of Pediatric Cardiology, Erciyes University, Kayseri, Turkey
| | - Abdullah Ozyurt
- Division of Pediatric Cardiology, Erciyes University, Kayseri, Turkey
| | - Kazim Uzum
- Division of Pediatric Cardiology, Erciyes University, Kayseri, Turkey
| |
Collapse
|
33
|
Şen LS, Karakoyun B, Yeğen C, Akkiprik M, Yüksel M, Ercan F, Özer A, Yeğen BÇ. Treatment with either obestatin or ghrelin attenuates mesenteric ischemia-reperfusion-induced oxidative injury of the ileum and the remote organ lung. Peptides 2015; 71:8-19. [PMID: 26032330 DOI: 10.1016/j.peptides.2015.04.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 04/06/2015] [Accepted: 04/14/2015] [Indexed: 12/14/2022]
Abstract
To evaluate the effects of exogenous ghrelin or obestatin on intestinal injury and accompanying pulmonary injury, intestinal ischemia-reperfusion (I/R) was induced in rats by obstructing the superior mesenteric artery for 60min, whereas laparotomy was performed in the sham group. At the beginning of the 90-min reperfusion period, the rats were injected with obestatin (100μg/kg), ghrelin (10ng/kg), or saline intravenously (iv). At the end of reperfusion, the blood, ileum, and lung samples were taken for the histological and biochemical assays. In the saline-treated I/R group, the increased serum interleukin (IL)-1β level, high damage scores, and elevated tissue malondialdehyde level and collagen content in both tissues were significantly reduced by obestatin or ghrelin. Increased ileal myeloperoxidase activity of the saline-treated I/R group was reduced by treatment with obestatin or ghrelin, whereas increased pulmonary myeloperoxidase activity was reduced with administration of obestatin. Increased DNA fragmentation in the ileum of the saline-treated I/R group was reduced by both peptides. Elevated luminol-lucigenin chemiluminescence levels and nuclear factor kappa B (NF-κB) messenger RNA (mRNA) expression in the ileum of the saline-treated-I/R group were significantly decreased by obestatin or ghrelin treatment. I/R-induced depletion of the antioxidant glutathione in both ileal and pulmonary tissues was prevented with either obestatin or ghrelin treatment. Administration of either obestatin or ghrelin exerts similar protective effects against I/R-induced ileal and pulmonary injury, thus warranting further investigation for their possible use against ischemic intestinal injury.
Collapse
Affiliation(s)
- Leyla Semiha Şen
- Marmara University School of Medicine, Department of General Surgery, Istanbul, Turkey
| | - Berna Karakoyun
- Marmara University Faculty of Health Sciences, Department of Basic Health Sciences, Istanbul, Turkey
| | - Cumhur Yeğen
- Marmara University School of Medicine, Department of General Surgery, Istanbul, Turkey
| | - Mustafa Akkiprik
- Marmara University School of Medicine, Department of Medical Biology, Istanbul, Turkey
| | - Meral Yüksel
- Marmara University Vocational School of Health Related Professions, Department of Medical Laboratory, Istanbul, Turkey
| | - Feriha Ercan
- Marmara University School of Medicine, Department of Histology and Embryology, Istanbul, Turkey
| | - Ayşe Özer
- Marmara University School of Medicine, Department of Medical Biology, Istanbul, Turkey
| | - Berrak Ç Yeğen
- Marmara University School of Medicine, Department of Physiology, Istanbul, Turkey.
| |
Collapse
|
34
|
Mason D, Chen YZ, Krishnan HV, Sant S. Cardiac gene therapy: Recent advances and future directions. J Control Release 2015; 215:101-11. [PMID: 26254712 DOI: 10.1016/j.jconrel.2015.08.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 07/31/2015] [Accepted: 08/01/2015] [Indexed: 11/15/2022]
Abstract
Gene therapy has the potential to serve as an adaptable platform technology for treating various diseases. Cardiovascular disease is a major cause of mortality in the developed world and genetic modification is steadily becoming a more plausible method to repair and regenerate heart tissue. Recently, new gene targets to treat cardiovascular disease have been identified and developed into therapies that have shown promise in animal models. Some of these therapies have advanced to clinical testing. Despite these recent successes, several barriers must be overcome for gene therapy to become a widely used treatment of cardiovascular diseases. In this review, we evaluate specific genetic targets that can be exploited to treat cardiovascular diseases, list the important delivery barriers for the gene carriers, assess the most promising methods of delivering the genetic information, and discuss the current status of clinical trials involving gene therapies targeted to the heart.
Collapse
Affiliation(s)
- Daniel Mason
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Yu-Zhe Chen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Harini Venkata Krishnan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
35
|
Ghrelin improves functional survival of engrafted adipose-derived mesenchymal stem cells in ischemic heart through PI3K/Akt signaling pathway. BIOMED RESEARCH INTERNATIONAL 2015; 2015:858349. [PMID: 25879037 PMCID: PMC4387976 DOI: 10.1155/2015/858349] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 01/26/2015] [Accepted: 01/27/2015] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) have been proposed as a promising cell population for cell therapy and regenerative medicine applications. However, the low retention and poor survival of engrafted cells hampered the therapeutic efficacy of engrafted MSCs. Ghrelin is a 28-amino-acid peptide hormone and is proved to exert a protective effect on the cardiovascular system. This study is designed to investigate the protective effects of ghrelin on engrafted adipose-derived mesenchymal stem cells (ADMSCs) and its beneficial effects with cellular therapy in mice myocardial infarction (MI). Results showed that intramyocardial injection of ADMSCs combining with ghrelin administration inhibited host cardiomyocyte apoptosis, reduced fibrosis, and improved cardiac function. To reveal possible mechanisms, ADMSCs were subjected to hypoxia/serum deprivation (H/SD) injury to simulate ischemic conditions in vivo. Ghrelin (10−8 M, 33712 pg/ml) improved ADMSCs survival under H/SD condition. Western blot assay revealed that ghrelin increased
AKT phosphorylation both in vivo and in vitro, decreased the proapoptotic protein Bax, and increased the antiapoptotic protein Bcl-2 in vitro, while these effects were abolished by PI3K inhibitor LY294002. These revealed that ghrelin may serve as a promising candidate for hormone-driven approaches to improve the efficacy of mesenchymal stem cell-based therapy for cardiac ischemic disease via PI3K/AKT pathway.
Collapse
|
36
|
Mondello P, Lacquaniti A, Mondello S, Bolignano D, Pitini V, Aloisi C, Buemi M. Emerging markers of cachexia predict survival in cancer patients. BMC Cancer 2014; 14:828. [PMID: 25400234 PMCID: PMC4239407 DOI: 10.1186/1471-2407-14-828] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 10/29/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Cachexia may occur in 40% of cancer patients, representing the major cause of death in more than 20% of them. The aim of this study was to investigate the role of leptin, ghrelin and obestatin as diagnostic and predictive markers of cachexia in oncologic patients. Their impact on patient survival was also evaluated. METHODS 140 adults with different cancer diagnoses were recruited. Thirty healthy volunteers served as control. Serum ghrelin, obestatin and leptin were tested at baseline and after a follow-up period of 18 months. RESULTS Ghrelin levels were significantly higher in cancer patients than in healthy subjects (573.31 ± 130 vs 320.20 ± 66.48 ng/ml, p < 0.0001), while obestatin (17.42 ± 7.12 vs 24.89 ± 5.54 ng/ml, p < 0.0001) and leptin (38.4 ± 21.2 vs 76.28 ± 17.48 ng/ml, p < 0.0001) values were lower. At ROC analyses the diagnostic profile of ghrelin (AUC 0.962; sensitivity 83%; specificity 98%), obestatin (AUC 0.798; sensitivity 74.5%; specificity 81.5%) and leptin (AUC 0.828; sensitivity 79%; specificity 73%) was superior to that of albumin (AUC 0.547; sensitivity 63%, specificity 69.4%) for detecting cachexia among cancer patients. On Cox multivariate analyses ghrelin (HR 1.02; 95% CI 1.01 - 1.03; p < 0.0001) and leptin (HR 0.94; 95% CI 0.92 - 0.96; p < 0.0001) were significant predictors of death even after correction for other known risk factors such as presence of metastasis and chronic kidney disease. CONCLUSION Ghrelin and leptin are promising biomarkers to diagnose cachexia and to predict survival in cancer patients.
Collapse
Affiliation(s)
- Patrizia Mondello
- />Department of Human Pathology, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Antonio Lacquaniti
- />Department of Internal Medicine, University of Messina, Messina, Italy
| | - Stefania Mondello
- />Department of Neurosciences, University of Messina, Messina, Italy
| | | | - Vincenzo Pitini
- />Department of Human Pathology, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Carmela Aloisi
- />Department of Internal Medicine, University of Messina, Messina, Italy
| | - Michele Buemi
- />Department of Internal Medicine, University of Messina, Messina, Italy
| |
Collapse
|
37
|
Koç M, Kumral ZNÖ, Özkan N, Memi G, Kaçar Ö, Bilsel S, Çetinel Ş, Yeğen BÇ. Obestatin improves ischemia/reperfusion-induced renal injury in rats via its antioxidant and anti-apoptotic effects: role of the nitric oxide. Peptides 2014; 60:23-31. [PMID: 25086266 DOI: 10.1016/j.peptides.2014.07.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 07/21/2014] [Accepted: 07/21/2014] [Indexed: 01/23/2023]
Abstract
Obestatin was shown to have anti-inflammatory effects in several inflammatory models. To elucidate the potential renoprotective effects of obestatin, renal I/R injury was induced in male Sprague Dawley rats by placing a clamp across left renal artery for 60min following a right nephrectomy. Clamp was released and the rats were injected with either saline or obestatin (10, 30, 100μg/kg). In some experiments, obestatin (10μg/kg) was administered with L-NAME (10mg/kg) or L-Nil (0.36mg/kg). Following a 24-h reperfusion, the rats were decapitated to measure serum creatinine and nitrite/nitrate levels, renal malondialdehyde (MDA), glutathione (GSH) levels and myeloperoxidase (MPO) activity and to assess cortical necrosis and apoptosis scores. Obestatin treatment reduced I/R-induced increase in creatinine levels, renal MPO activity and renal MDA levels, while renal GSH levels were significantly increased by obestatin. Histological analysis revealed that severe I/R injury and high apoptosis score in the kidney samples of saline-treated rats were significantly reduced and the cortical/medullary injury was ameliorated by obestatin. Expression of eNOS, which was increased by I/R injury, was further increased by obestatin, while serum NO levels were significantly decreased. iNOS inhibitor L-Nil reduced oxidative renal damage and improved the functional and histopathological parameters. I/R-induced elevation in eNOS expression, which was further increased by obestatin, was depressed by L-NAME and L-Nil treatments. The present data demonstrate that obestatin ameliorates renal I/R-injury by its possible anti-oxidative, anti-inflammatory and anti-apoptotic properties, which appear to involve the suppression of neutrophil accumulation and modulation of NO metabolism.
Collapse
Affiliation(s)
- Mehmet Koç
- Marmara University School of Medicine, Department of Internal Medicine, Division of Nephrology, Istanbul, Turkey; Marmara University School of Medicine, Department of Physiology, Istanbul, Turkey
| | | | - Naziye Özkan
- Marmara University School of Medicine, Department of Histology, Istanbul, Turkey
| | - Gülsün Memi
- Kırklareli University Health School, Kırklareli, Turkey
| | - Ömer Kaçar
- Marmara University School of Medicine, Department of Biochemistry, Istanbul, Turkey
| | - Serpil Bilsel
- Marmara University School of Medicine, Department of Biochemistry, Istanbul, Turkey
| | - Şule Çetinel
- Marmara University School of Medicine, Department of Histology, Istanbul, Turkey
| | - Berrak Ç Yeğen
- Marmara University School of Medicine, Department of Physiology, Istanbul, Turkey.
| |
Collapse
|
38
|
Prodam F, Filigheddu N. Ghrelin gene products in acute and chronic inflammation. Arch Immunol Ther Exp (Warsz) 2014; 62:369-84. [PMID: 24728531 DOI: 10.1007/s00005-014-0287-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 03/21/2014] [Indexed: 12/27/2022]
Abstract
Ghrelin gene products--the peptides ghrelin, unacylated ghrelin, and obestatin--have several actions on the immune system, opening new perspectives within neuroendocrinology, metabolism and inflammation. The aim of this review is to summarize the available evidence regarding the less known role of these peptides in the machinery of inflammation and autoimmunity, outlining some of their most promising therapeutic applications.
Collapse
Affiliation(s)
- Flavia Prodam
- Departmant of Health Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | | |
Collapse
|
39
|
Tang S, Dong X, Zhang W. Obestatin changes proliferation, differentiation and apoptosis of porcine preadipocytes. ANNALES D'ENDOCRINOLOGIE 2014; 75:1-9. [PMID: 24534601 DOI: 10.1016/j.ando.2013.10.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 10/25/2013] [Indexed: 01/04/2023]
Abstract
Obestatin, originally identified and purified from rat stomach extracts, was reported to bind to orphan G protein-coupled receptor, GPR39, and inhibit appetite and gastric motility. This study was conducted to investigate the effects of porcine obestatin on proliferation, differentiation and apoptosis of porcine preadipocytes isolated from subcutaneous fat of piglets. At indicated times of culture, morphology of preadipocytes and accumulated lipid droplets within the cells were identified by invert microscope. After treating with obestatin (0, 0.1, 1, 10 and 100nM), cell proliferation was measured by MTT method and protein expression of CCAAT/enhancer binding protein-α (C/EBPα), peroxisome proliferator-activated receptor-γ (PPARγ), Caspase-7 and Caspase-9 was determined by Western Blot, mRNA expression of GPR39 and Caspase-3 was analyzed by RT-PCR, and the activity of Caspase-3 was measured by spectrophotometric method. The results showed that obestatin had no effect on GPR39 expression, while promotes the optical density (OD) value of cells, enhanced protein expression of PPARγ and C/EBPa, decreased mRNA expression and activity of Caspase-3, and inhibited protein expression of Caspase-7 and Caspase-9 in a dose-dependent manner. These results suggested that obestatin enhances proliferation and differentiation of preadipocytes promoting PPARγ and C/EBPa expression, and inhibiting preadipocyte apoptosis by decreasing expression of Caspase-3, Caspase-7 and Caspase-9.
Collapse
Affiliation(s)
- Shengqiu Tang
- College of Yingdong agricultural science and engineering, Shaoguan university, Daxue road, Zhenjiang district, Shaoguan 512005, China
| | - Xiaoying Dong
- College of Yingdong agricultural science and engineering, Shaoguan university, Daxue road, Zhenjiang district, Shaoguan 512005, China.
| | - Wei Zhang
- Hubei Key laboratory of animal embryo and molecular breeding, Hubei academy of agricultural science, Wuhan 430064, China
| |
Collapse
|
40
|
Su XJ, Dong RX, Li YP, Yang SG, Li ZF. Obestatin and cardiovascular health. Peptides 2014; 52:58-60. [PMID: 24333655 DOI: 10.1016/j.peptides.2013.11.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 11/23/2013] [Accepted: 11/25/2013] [Indexed: 10/25/2022]
Abstract
Obestatin, encoded by the same gene as ghrelin, was first described as a physiological opponent of ghrelin through an interaction with the orphan receptor GPR39. However, the effects of obestatin were not totally contrary to the effects of ghrelin in cardiovascular regulations based on the recent studies. We summarize here the current evidences surrounding the cardiovascular actions of obestatin, and the possible implications of obestatin as a therapeutic agent in common conditions such as hypertension and heart failure.
Collapse
Affiliation(s)
- Xue-Jia Su
- Department of Thoracic Surgery, No. 88 Hospital of PLA, Tai'an City, Shandong Province 271000, China
| | - Rui-Xin Dong
- Department of Cardiology, No. 88 Hospital of PLA, Tai'an City, Shandong Province 271000, China
| | - Yan-Peng Li
- Department of Out-patient, No. 88 Hospital of PLA, Tai'an City, Shandong Province 271000, China
| | - Shu-Guang Yang
- Department of Cardiology, No. 88 Hospital of PLA, Tai'an City, Shandong Province 271000, China.
| | - Zhao-Feng Li
- Department of Cardiology, No. 88 Hospital of PLA, Tai'an City, Shandong Province 271000, China.
| |
Collapse
|
41
|
|
42
|
Gu PY, Kang DM, Wang WD, Chen Y, Zhao ZH, Zheng H, Ye SD. Relevance of plasma obestatin and early arteriosclerosis in patients with type 2 diabetes mellitus. J Diabetes Res 2013; 2013:563919. [PMID: 24350300 PMCID: PMC3855972 DOI: 10.1155/2013/563919] [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: 05/22/2013] [Revised: 09/04/2013] [Accepted: 09/10/2013] [Indexed: 11/18/2022] Open
Abstract
We investigated the correlation between obestatin and metabolic parameters and carotid intima-media thickness (IMT) in plasma of patients with type 2 diabetes mellitus (T2DM). We collected 103 patients aged from 60 to 83 years (69.26 ± 5.83 years) form January, 2007 to May, 2009. All patients were divided into normal glucose tolerance (NGT), impaired glucose tolerance (IGT), and T2DM according to the oral glucose tolerance test (OGTT). We found that higher levels of fasting insulin (Fins), fasting blood glucose, 2 h OGTT glucose, homeostasis model assessment of insulin resistance (HOMA-IR), low density lipoprotein cholesterol, glycated haemoglobin, and C-reactive protein (CRP), as well as lower obestatin level and higher intima-media thickness level (IMT), existed in T2DM group compared with NGT group and IGT group (P < 0.01). Also, obestatin level was independently associated with HOMA-IR and CRP, while IMT level was independently associated with HOMA-IR, triglyceride, Fins, and obestatin (P < 0.01), based on stepwise multiple regression analysis. Therefore, we deduced that the low level of plasma obestatin might be related to early arteriosclerosis in patients with T2DM via increasing IMT level, and elevated plasma obestatin levels might protect T2DM patients against carotid atherosclerosis to some extent.
Collapse
Affiliation(s)
- Peng-ying Gu
- Department of Geriatric Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
| | - Dong-mei Kang
- Department of Geriatric Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
| | - Wei-dong Wang
- Department of Geriatric Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
| | - Yan Chen
- Department of Endocrine Laboratory, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
| | - Zhi-hong Zhao
- Department of Ultrasonic Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
| | - Hui Zheng
- Department of Clinical Laboratory, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
| | - Shan-dong Ye
- Department of Endocrinology, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
| |
Collapse
|
43
|
Gesmundo I, Gallo D, Favaro E, Ghigo E, Granata R. Obestatin: a new metabolic player in the pancreas and white adipose tissue. IUBMB Life 2013; 65:976-82. [PMID: 24217898 DOI: 10.1002/iub.1226] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 10/10/2013] [Accepted: 10/11/2013] [Indexed: 01/14/2023]
Abstract
Obestatin is a 23 amino acid amidated peptide, member of the preproghrelin gene-derived peptides. Initially, obestatin was reported to exert opposite effects to those of ghrelin on food intake and body weight gain, through interaction with GPR39; however, these findings are still strongly debated and obestatin biological role remains largely unknown. Interestingly, binding of obestatin to the glucagon-like peptide 1 receptor has been recently suggested. Despite being a controversial peptide, recent findings have clearly indicated that obestatin is indeed a multifunctional peptide, exerting a variety of effects, such as stimulation of cell proliferation, survival and differentiation, influence on glucose and lipid metabolism, as well as anti-inflammatory and cardioprotective actions. Its positive effects on glucose and lipid metabolism candidate this peptide as a potential therapeutic tool in pathological conditions such as insulin resistance and diabetes.
Collapse
Affiliation(s)
- Iacopo Gesmundo
- Division of Endocrinology, Diabetology and Metabolism, Department of Medical Sciences, University of Torino, Torino, Italy
| | | | | | | | | |
Collapse
|
44
|
Zhang MQ, Zheng YL, Chen H, Tu JF, Shen Y, Guo JP, Yang XH, Yuan SR, Chen LZ, Chai JJ, Lu JH, Zhai CL. Sodium tanshinone IIA sulfonate protects rat myocardium against ischemia-reperfusion injury via activation of PI3K/Akt/FOXO3A/Bim pathway. Acta Pharmacol Sin 2013; 34:1386-96. [PMID: 24077633 DOI: 10.1038/aps.2013.91] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Accepted: 06/24/2013] [Indexed: 02/06/2023]
Abstract
AIM To investigate the mechanisms underlying the protective effects of sodium tanshinone IIA sulfonate (STS) in an ischemia-reperfusion (I/R)-induced rat myocardial injury model. METHODS Male SD rats were iv injected with STS, STS+LY294002 or saline (NS) for 15 d. Then the hearts were subjected to 30 min of global ischemia followed by 2 h of reperfusion. Cardiac function, infarction size and area at risk were assessed. Cell apoptosis was evaluated with TUNEL staining, DNA laddering and measuring caspase-3 activity. In addition, isolated cardiomyocytes of neonatal rats were pretreated with the above drugs, then exposed to H2O2 (200 mol/L) for 1 h. Cell apoptosis was detected using flow cytometric assay. The levels of p-Akt, p-FOXO3A and Bim were examined with immunoblotting. RESULTS Compared to NS group, administration of STS (20 mg/kg) significantly reduced myocardial infarct size (40.28%±5.36% in STS group vs 59.52%±7.28% in NS group), and improved the myocardial function as demonstrated by the increased values of dp/dtmax, LVDP and coronary flow at different reperfusion time stages. Furthermore, STS significantly decreased the rate of apoptotic cells (15.11%±3.71% in STS group vs 38.21%±7.83% in NS group), and reduced caspase-3 activity to nearly a quarter of that in NS group. Moreover, STS significantly increased the phosphorylation of Akt and its downstream target FOXO3A, and decreased the expression of pro-apoptotic gene Bim. Co-treatment with the PI3K inhibitor LY294002 (40 mg/kg) partially countered the protective effects induced by STS treatment. In isolated cardiomyocytes, STS exerted similar protective effects as shown in the ex vivo I/R model. CONCLUSION STS pretreatment reduces infarct size and improves cardiac function in an I/R-induced rat myocardial injury model via activation of Akt/FOXO3A/Bim-mediated signal pathway.
Collapse
|
45
|
Low serum concentration of obestatin as a predictor of mortality in maintenance hemodialysis patients. BIOMED RESEARCH INTERNATIONAL 2013; 2013:796586. [PMID: 24102059 PMCID: PMC3786512 DOI: 10.1155/2013/796586] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 07/19/2013] [Indexed: 12/04/2022]
Abstract
Obestatin, a proposed anorexigenic gut hormone, has been shown to have a number of beneficial cardiotropic effects in experimental studies. We hypothesized that obestatin alteration in hemodialysis patients may link to clinical outcomes. This cross-sectional study with prospective followup for almost 4 years was performed on 94 prevalent hemodialysis patients. Obestatin, leptin, proinflammatory cytokines (tumor necrosis factor-α [TNF-α], interleukin-6, and various nutritional markers were measured. Patients with low obestatin levels, defined as a level less than median, had a worse all-cause mortality and cardiovascular mortality. The crude all-cause (HR 2.23, 95% CI 1.17 to 4.24) and cardiovascular mortality hazard ratios (HR 4.03, 95% CI 1.27 to 12.76) in these patients continued to be significant after adjustment for various confounders for all-cause mortality. Across the four obestatin-TNF-α categories, the group with low obestatin and high TNF-α (above median level) exhibited a worse outcome in both all-cause mortality and cardiovascular mortality. Clinical characteristics of patients in low obestatin high TNF-α group did not differ from other obestatin-TNF-α categorized groups. In summary, low serum obestatin concentration is an independent predictor of mortality in prevalent hemodialysis patients. Novel interactions were observed between obestatin and TNF-α, which were associated with mortality risk, especially those due to cardiovascular causes.
Collapse
|
46
|
Penna C, Settanni F, Tullio F, Trovato L, Pagliaro P, Alloatti G, Ghigo E, Granata R. GH-releasing hormone induces cardioprotection in isolated male rat heart via activation of RISK and SAFE pathways. Endocrinology 2013; 154:1624-35. [PMID: 23417421 DOI: 10.1210/en.2012-2064] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
GHRH stimulates GH synthesis and release from the pituitary and exerts direct effects in extrapituitary tissues. We have previously shown that pretreatment with GHRH reduces cardiomyocyte apoptosis and improves heart function in isolated rat hearts subjected to ischemia/reperfusion (I/R). Here, we determined whether GHRH given at reperfusion reduces myocardial reperfusion injury and investigated the molecular mechanisms involved in GHRH effects. Isolated rat hearts subjected to I/R were treated at the onset of reperfusion with: 1) GHRH; 2) GHRH+GHRH antagonist JV-1-36; 3) GHRH+mitochondrial ATP-dependent potassium channel inhibitor 5-hydroxydecanoate; 4) GHRH+mitochondrial permeability transition pore opener atractyloside; 5) GHRH+ phosphoinositide 3-kinase/Akt inhibitor Wortmannin (WM); and 6) GHRH+signal transducer and activator of transcription-3 inhibitor tyrphostin-AG490 (AG490). GHRH reduced infarct size at the end of reperfusion and reverted contractility dysfunction in I/R hearts. These effects were inhibited by either JV-1-36, 5-hydroxydecanoate, atractylosid, WM, or AG490. Western blot analysis on left ventricles showed GHRH-induced phosphorylation of either the reperfusion injury salvage kinases (RISK), phosphoinositide 3-kinase/Akt, ERK1/2, and glycogen synthase kinase-3β or signal transducer and activator of transcription-3, as part of the survivor activating factor enhancement (SAFE) pathway. GHRH-induced activation of RISK and SAFE pathways was blocked by JV-1-36, WM, and AG490. Furthermore, GHRH increased the phosphorylation of endothelial nitric oxide synthase and AMP-activated protein kinase and preserved postischemic nicotinamide adenine dinucleotide (NAD(+)) levels. These results suggest that GHRH protects the heart from I/R injury through receptor-mediated mechanisms, leading to activation of RISK and SAFE pathways, which converge on mitochondria and possibly on AMP-activated protein kinase.
Collapse
Affiliation(s)
- Claudia Penna
- Department of Clinical and Biological Sciences, University of Torino, 10126 Torino, Italy
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Cao Y, Tang J, Yang T, Ma H, Yi D, Gu C, Yu S. Cardioprotective effect of ghrelin in cardiopulmonary bypass involves a reduction in inflammatory response. PLoS One 2013; 8:e55021. [PMID: 23359315 PMCID: PMC3554674 DOI: 10.1371/journal.pone.0055021] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 12/18/2012] [Indexed: 01/04/2023] Open
Abstract
Background Ghrelin has been reported to protect the cardiovascular system; however, the cardioprotective effect of ghrelin against cardiopulmonary bypass (CPB) induced myocardial injury are unclear. In this study, the protective effect of ghrelin on CPB induced myocardial injury and the underlying mechanisms were investigated. Methods and Results Adult male rats were subjected to CPB and randomly to receive vehicle (n = 8), ghrelin (n = 8), ghrelin plus [D-Lys3]-GHRP-6, a GHSR-1a inhibitor (n = 8), or ghrelin plus wortmannin, a phosphoinositide 3′-kinase (PI3K) inhibitor (n = 8). In vitro study was performed on cultured cardiomyocytes subjected to simulated cardiopulmonary bypass (SCPB). Ghrelin attenuated the inflammatory response, as evidenced by reduced induction of TNF-α, IL-6 and myocardial myeloperoxidase activity and concurrent reduction in apoptosis, oxidative stress, and levels of myocardial injury markers following CPB. Moreover, ghrelin significantly increased cardiac function after CPB. In cultured cardiomyocytes subjected to simulated CPB, ghrelin increased cell viability and decreased the percentage of apoptotic myocytes. Inhibition of ghrelin downstream signaling blocked the cardioprotective effects both in vivo and vitro. Conclusions Ghrelin could provide an effective approach to the attenuation of CPB induced myocardial injury. The cardioprotective effects elicited by ghrelin may contribute to the inhibition of inflammatory response through the Akt-activated pathway.
Collapse
Affiliation(s)
- Yukun Cao
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi Province, China
| | - Jun Tang
- Department of Anesthesiology, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi Province, China
| | - Ting Yang
- Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi Province, China
| | - Heng Ma
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi Province, China
| | - Dinghua Yi
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi Province, China
- * E-mail: (DHY); (CHG); (SQY)
| | - Chunhu Gu
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi Province, China
- * E-mail: (DHY); (CHG); (SQY)
| | - Shiqiang Yu
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi Province, China
- * E-mail: (DHY); (CHG); (SQY)
| |
Collapse
|
48
|
Low Serum Concentration of Obestatin as a Predictor of Mortality in Maintenance Hemodialysis Patients. BIOMED RESEARCH INTERNATIONAL 2013. [DOI: 10.1155/2013/796586 www.ncbi.nlm.nih.gov/pmc/articles/pmc3786512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Obestatin, a proposed anorexigenic gut hormone, has been shown to have a number of beneficial cardiotropic effects in experimental studies. We hypothesized that obestatin alteration in hemodialysis patients may link to clinical outcomes. This cross-sectional study with prospective followup for almost 4 years was performed on 94 prevalent hemodialysis patients. Obestatin, leptin, proinflammatory cytokines (tumor necrosis factor-α[TNF-α], interleukin-6, and various nutritional markers were measured. Patients with low obestatin levels, defined as a level less than median, had a worse all-cause mortality and cardiovascular mortality. The crude all-cause (HR 2.23, 95% CI 1.17 to 4.24) and cardiovascular mortality hazard ratios (HR 4.03, 95% CI 1.27 to 12.76) in these patients continued to be significant after adjustment for various confounders for all-cause mortality. Across the four obestatin-TNF-αcategories, the group with low obestatin and high TNF-α(above median level) exhibited a worse outcome in both all-cause mortality and cardiovascular mortality. Clinical characteristics of patients in low obestatin high TNF-αgroup did not differ from other obestatin-TNF-αcategorized groups. In summary, low serum obestatin concentration is an independent predictor of mortality in prevalent hemodialysis patients. Novel interactions were observed between obestatin and TNF-α, which were associated with mortality risk, especially those due to cardiovascular causes.
Collapse
|
49
|
Chopin LK, Seim I, Walpole CM, Herington AC. The ghrelin axis--does it have an appetite for cancer progression? Endocr Rev 2012; 33:849-91. [PMID: 22826465 DOI: 10.1210/er.2011-1007] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ghrelin, the endogenous ligand for the GH secretagogue receptor (GHSR), is a peptide hormone with diverse physiological roles. Ghrelin regulates GH release, appetite and feeding, gut motility, and energy balance and also has roles in the cardiovascular, immune, and reproductive systems. Ghrelin and the GHSR are expressed in a wide range of normal and tumor tissues, and a fluorescein-labeled, truncated form of ghrelin is showing promise as a biomarker for prostate cancer. Plasma ghrelin levels are generally inversely related to body mass index and are unlikely to be useful as a biomarker for cancer, but may be useful as a marker for cancer cachexia. Some single nucleotide polymorphisms in the ghrelin and GHSR genes have shown associations with cancer risk; however, larger studies are required. Ghrelin regulates processes associated with cancer, including cell proliferation, apoptosis, cell migration, cell invasion, inflammation, and angiogenesis; however, the role of ghrelin in cancer is currently unclear. Ghrelin has predominantly antiinflammatory effects and may play a role in protecting against cancer-related inflammation. Ghrelin and its analogs show promise as treatments for cancer-related cachexia. Further studies using in vivo models are required to determine whether ghrelin has a role in cancer progression.
Collapse
Affiliation(s)
- Lisa K Chopin
- Ghrelin Research Group, Institute of Health and Biomedical Innovation, Queensland University of Technology and Australian Prostate Cancer Research Centre-Queensland, Brisbane, Queensland 4001, Australia.
| | | | | | | |
Collapse
|
50
|
Aragno M, Mastrocola R, Ghé C, Arnoletti E, Bassino E, Alloatti G, Muccioli G. Obestatin induced recovery of myocardial dysfunction in type 1 diabetic rats: underlying mechanisms. Cardiovasc Diabetol 2012; 11:129. [PMID: 23066908 PMCID: PMC3537569 DOI: 10.1186/1475-2840-11-129] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 09/21/2012] [Indexed: 01/17/2023] Open
Abstract
Background The aim of this study was to investigate whether obestatin (OB), a peptide mediator encoded by the ghrelin gene exerting a protective effect in ischemic reperfused heart, is able to reduce cardiac dysfunctions in adult diabetic rats. Methods Diabetes was induced by STZ injection (50 mg/kg) in Wistar rats (DM). OB was administered (25 μg/kg) twice a day for 6 weeks. Non-diabetic (ND) rats and DM rats were distributed into four groups: untreated ND, OB-treated ND, untreated DM, OB-treated DM. Cardiac contractility and ß-adrenergic response were studied on isolated papillary muscles. Phosphorylation of AMPK, Akt, ERK1/2 and GSK3ß as well ß-1 adrenoreceptors levels were detected by western blot, while α-MHC was measured by RT-PCR. Results OB preserved papillary muscle contractility (85 vs 27% of ND), ß-adrenergic response (103 vs 65% of ND), as well ß1-adrenoreceptors and α-MHC levels in diabetic myocardial tissue. Moreover, OB up-regulated the survival kinases Akt and ERK1/2, and enhanced AMPK and GSK3ß phosphorylation. OB corrected oxidative unbalance, reduced pro-inflammatory cytokine TNF-α plasma levels, NFkB translocation and pro-fibrogenic factors expression in diabetic myocardium. Conclusions OB displays a significant beneficial effect against the alterations of contractility and ß-adrenergic response in the heart of STZ-treated diabetic rats, which was mainly associated with the ability of OB to up-regulate the transcription of ß1-adrenergic receptors and α-MHC; this protective effect was accompanied by the ability to restore oxidative balance and to promote phosphorylation/modulation of AMPK and pro-survival kinases such as Akt, ERK1/2 and GSK3ß.
Collapse
Affiliation(s)
- Manuela Aragno
- Department of Experimental Medicine and Oncology, University of Turin, Corso Raffaello 30, Turin, 10125, Italy.
| | | | | | | | | | | | | |
Collapse
|