1
|
Xiang P, Liu Q, Jing W, Wang Y, Yu H. Combined ROS Sensitive PEG-PPS-PEG with Peptide Agonist for Effective Target Therapy in Mouse Model. Int J Nanomedicine 2024; 19:9109-9120. [PMID: 39253061 PMCID: PMC11382658 DOI: 10.2147/ijn.s471036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
Background and Purpose Growth hormone-releasing hormone (GHRH) agonist, a 29-amino acid peptide, shows significant potential in treating myocardial infarction (MI) by aiding the repair of injured heart tissue. The challenge lies in the effective on-site delivery of GHRH agonist. This study explores the use of a targetable delivery system employing ROS-responsive PEG-PPS-PEG polymers to encapsulate and deliver GHRH agonist MR409 for enhanced therapeutic efficacy. Methods We synthesized a self-assembling poly (ethylene glycol)-poly (propylene sulfide)-poly (ethylene glycol) polymer (PEG-PPS-PEG) amphiphilic polymer responsive to reactive oxygen species (ROS). The hydrophilic peptide GHRH agonist MR409 was encapsulated within these polymers to form nano PEG-PPS-PEG@MR409 vesicles (NPs). Cardiomyocyte apoptosis was induced under hypoxia and serum-free culture condition for 24 hours, and their production of ROS was detected by fluorescence dye staining. The cellular uptake of PEG-PPS-PEG@MR409 NPs was observed using fluorescence-labeled MR409. Targeting ability and therapeutic efficacy were evaluated using a mouse MI model. Results PEG-PPS-PEG@MR409 NPs were efficiently internalized by cardiomyocytes, reducing ROS levels and apoptosis. These NPs exhibited superior targeting to the infarcted heart compared to naked MR409 peptide. With a reduced injection frequency (once every three days), PEG-PPS-PEG@MR409 NPs significantly promoted cardiac function recovery post-MI, matching the efficacy of daily MR409 injections. Conclusion ROS-responsive PEG-PPS-PEG polymers provide a novel and effective platform for the targeted delivery of GHRH agonist peptides, improving cardiac function and offering a new approach for peptide therapy in MI treatment.
Collapse
Affiliation(s)
- Pingping Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310009, People's Republic of China
- Key Laboratory of Multiple Organ Failure (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang Province, People's Republic of China
| | - Qi Liu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310009, People's Republic of China
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Wangwei Jing
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310009, People's Republic of China
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, People's Republic of China
| | - Yaping Wang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310009, People's Republic of China
| | - Hong Yu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310009, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, People's Republic of China
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, People's Republic of China
| |
Collapse
|
2
|
Yellon DM, Beikoghli Kalkhoran S, Davidson SM. The RISK pathway leading to mitochondria and cardioprotection: how everything started. Basic Res Cardiol 2023; 118:22. [PMID: 37233787 PMCID: PMC10220132 DOI: 10.1007/s00395-023-00992-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 05/27/2023]
Abstract
Ischaemic heart disease, which often manifests clinically as myocardial infarction (MI), remains a major cause of mortality worldwide. Despite the development of effective pre-clinical cardioprotective therapies, clinical translation has been disappointing. Nevertheless, the 'reperfusion injury salvage kinase' (RISK) pathway appears to be a promising target for cardioprotection. This pathway is crucial for the induction of cardioprotection by numerous pharmacological and non-pharmacological interventions, such as ischaemic conditioning. An important component of the cardioprotective effects of the RISK pathway involves the prevention of mitochondrial permeability transition pore (MPTP) opening and subsequent cardiac cell death. Here, we will review the historical perspective of the RISK pathway and focus on its interaction with mitochondria in the setting of cardioprotection.
Collapse
Affiliation(s)
- Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK.
| | | | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| |
Collapse
|
3
|
Datta A, Saha C, Godse P, Sharma M, Sarmah D, Bhattacharya P. Neuroendocrine regulation in stroke. Trends Endocrinol Metab 2023; 34:260-277. [PMID: 36922255 DOI: 10.1016/j.tem.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 03/14/2023]
Abstract
The neuroendocrine system, a crosstalk between the central nervous system and endocrine glands, balances and controls hormone secretion and their functions. Neuroendocrine pathways and mechanisms often get dysregulated following stroke, leading to altered hormone secretion and aberrant receptor expression. Dysregulation of the hypothalamus-pituitary-thyroid (HPT) axis and hypothalamus-pituitary-adrenal (HPA) axis often led to severe stroke outcomes. Post-stroke complications such as cognitive impairment, depression, infection etc. are directly or indirectly influenced by the altered neuroendocrine activity that plays a crucial role in stroke vulnerability and susceptibility. Therefore, it is imperative to explore various neurohormonal inter-relationships in regulating stroke, its outcome, and prognosis. Here, we review the biology of different hormones associated with stroke and explore their regulation with a view towards prospective therapeutics.
Collapse
Affiliation(s)
- Aishika Datta
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat-382355, India
| | - Chandrima Saha
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat-382355, India
| | - Pratiksha Godse
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat-382355, India
| | - Muskaan Sharma
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat-382355, India
| | - Deepaneeta Sarmah
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat-382355, India
| | - Pallab Bhattacharya
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat-382355, India.
| |
Collapse
|
4
|
Dulce RA, Kanashiro-Takeuchi RM, Takeuchi LM, Salerno AG, Wanschel ACBA, Kulandavelu S, Balkan W, Zuttion MSSR, Cai R, Schally AV, Hare JM. Synthetic growth hormone-releasing hormone agonist ameliorates the myocardial pathophysiology characteristic of heart failure with preserved ejection fraction. Cardiovasc Res 2023; 118:3586-3601. [PMID: 35704032 DOI: 10.1093/cvr/cvac098] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 05/06/2022] [Accepted: 05/25/2022] [Indexed: 02/07/2023] Open
Abstract
AIMS To test the hypothesis that the activation of the growth hormone-releasing hormone (GHRH) receptor signalling pathway within the myocardium both prevents and reverses diastolic dysfunction and pathophysiologic features consistent with heart failure with preserved ejection fraction (HFpEF). Impaired myocardial relaxation, fibrosis, and ventricular stiffness, among other multi-organ morbidities, characterize the phenotype underlying the HFpEF syndrome. Despite the rapidly increasing prevalence of HFpEF, few effective therapies have emerged. Synthetic agonists of the GHRH receptors reduce myocardial fibrosis, cardiomyocyte hypertrophy, and improve performance in animal models of ischaemic cardiomyopathy, independently of the growth hormone axis. METHODS AND RESULTS CD1 mice received 4- or 8-week continuous infusion of angiotensin-II (Ang-II) to generate a phenotype with several features consistent with HFpEF. Mice were administered either vehicle or a potent synthetic agonist of GHRH, MR-356 for 4-weeks beginning concurrently or 4-weeks following the initiation of Ang-II infusion. Ang-II-treated animals exhibited diastolic dysfunction, ventricular hypertrophy, interstitial fibrosis, and normal ejection fraction. Cardiomyocytes isolated from these animals exhibited incomplete relaxation, depressed contractile responses, altered myofibrillar protein phosphorylation, and disturbed calcium handling mechanisms (ex vivo). MR-356 both prevented and reversed the development of the pathological phenotype in vivo and ex vivo. Activation of the GHRH receptors increased cAMP and cGMP in cardiomyocytes isolated from control animals but only cAMP in cardiac fibroblasts, suggesting that GHRH-A exert differential effects on cardiomyocytes and fibroblasts. CONCLUSION These findings indicate that the GHRH receptor signalling pathway(s) represents a new molecular target to counteract dysfunctional cardiomyocyte relaxation by targeting myofilament phosphorylation and fibrosis. Accordingly, activation of GHRH receptors with potent, synthetic GHRH agonists may provide a novel therapeutic approach to management of the myocardial alterations associated with the HFpEF syndrome.
Collapse
Affiliation(s)
- Raul A Dulce
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA
| | - Rosemeire M Kanashiro-Takeuchi
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA.,Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lauro M Takeuchi
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA
| | - Alessandro G Salerno
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA
| | - Amarylis C B A Wanschel
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA
| | - Shathiyah Kulandavelu
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA.,Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Marilia S S R Zuttion
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA
| | - Renzhi Cai
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, FL 33125, USA
| | - Andrew V Schally
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA.,Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, FL 33125, USA.,Division of Hematology/Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
5
|
Verma H, Bhattacharjee A, Shivavedi N, Nayak PK. Evaluation of rosmarinic acid against myocardial infarction in maternally separated rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:1189-1207. [PMID: 35876905 DOI: 10.1007/s00210-022-02273-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/10/2022] [Indexed: 12/07/2022]
Abstract
Depression and coronary heart diseases are the common comorbid disorder affecting humans globally. The present study evaluated the effectiveness of rosmarinic acid (RA) against myocardial infarction (MI) in comorbid depression induced by maternal separation in rats. Maternal stress is one of the childhood crises that may be a potential risk factor for coronary heart disease in later part of life. As per protocol, 70-80% of pups were separated daily for 3 h between postnatal day 1 (PND1) and postnatal day 21 (PND21). Forced-swim test, sucrose preference test, and electrocardiography were performed during the experiment. Body weight was measured on PND0, PND35, and PND55. Orally rosmarinic acid (25 mg/kg and 50 mg/kg) and fluoxetine (10 mg/kg) was done from PND35 to PND55. On PND53 and PND54, isoproterenol (100 mg/kg, subcutaneously) was administered to induce myocardial infarction. On PND55, blood was collected and animals sacrificed, and plasma corticosterone, brain-derived neurotrophic factor, cardiac biomarkers, interleukine-10, and anti-oxidant parameters were measured. Rosmarinic acid and fluoxetine ameliorated the maternal separation-induced increase in immobility period, anhedonia, body weight, ST elevation, corticosterone, creatine kinase-MB (CK-MB), and lactate dehydrogenase (LDH). At the same time, both drugs elevated the tissue levels of BDNF, IL-10, glutathione, and superoxide dismutase activity. This study provides the first experimental evidence that maternal stress is an independent risk factor of cardiac abnormalities in rats. Moreover, maternal stress synergistically increases the severity of cardiac abnormalities induced by isoproterenol. Interestingly, fluoxetine and rosmarinic acid effectively ameliorated behavioral anomalies and myocardial infarction in maternally separated rats. Schematic representation of possible molecular mechanism of action of rosmarinic acid against MS-induced myocardial infarction. RA, rosmarinic acid; MS, maternal separation; PND, postnatal days; ISO, isoproterenol; BDNF, brain-derived neurotrophic factor; GSH, glutathione; SOD, superoxide dismutase; IL-10, interleukin-10; MI, myocardial infarction.
Collapse
Affiliation(s)
- Himanshu Verma
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University (BHU), Uttar Pradesh, Varanasi, 221005, India
| | - Anindita Bhattacharjee
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Uttar Pradesh, Varanasi, 221005, India
| | - Naveen Shivavedi
- Shri Ram Group of Institutions, Faculty of Pharmacy, Jabalpur, Madhya Pradesh, India
| | - Prasanta Kumar Nayak
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University (BHU), Uttar Pradesh, Varanasi, 221005, India.
| |
Collapse
|
6
|
Antagonist of Growth Hormone-Releasing Hormone Potentiates the Antitumor Effect of Pemetrexed and Cisplatin in Pleural Mesothelioma. Int J Mol Sci 2022; 23:ijms231911248. [PMID: 36232554 PMCID: PMC9569772 DOI: 10.3390/ijms231911248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Pleural mesothelioma (PM) is an aggressive cancer with poor prognosis and no effective therapies, mainly caused by exposure to asbestos. Antagonists of growth hormone-releasing hormone (GHRH) display strong antitumor effects in many experimental cancers, including lung cancer and mesothelioma. Here, we aimed to determine whether GHRH antagonist MIA-690 potentiates the antitumor effect of cisplatin and pemetrexed in PM. In vitro, MIA-690, in combination with cisplatin and pemetrexed, synergistically reduced cell viability, restrained cell proliferation and enhanced apoptosis, compared with drugs alone. In vivo, the same combination resulted in a strong growth inhibition of MSTO-211H xenografts, decreased tumor cell proliferation and increased apoptosis. Mechanistically, MIA-690, particularly with chemotherapeutic drugs, inhibited proliferative and oncogenic pathways, such as MAPK ERK1/2 and cMyc, and downregulated cyclin D1 and B1 mRNAs. Inflammatory pathways such as NF-kB and STAT3 were also reduced, as well as oxidative, angiogenic and tumorigenic markers (iNOS, COX-2, MMP2, MMP9 and HMGB1) and growth factors (VEGF and IGF-1). Overall, these findings strongly suggest that GHRH antagonists of MIA class, such as MIA-690, could increase the efficacy of standard therapy in PM.
Collapse
|
7
|
LI Z, LI Y. Effect of growth hormone releasing hormone on chondrocytes of osteoarthritis. Korean J Intern Med 2022; 37:222-229. [PMID: 31875669 PMCID: PMC8747918 DOI: 10.3904/kjim.2018.399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/07/2019] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND/AIMS To evaluate the effect and possible mechanism of growth hormone releasing hormone (GHRH) on chondrocytes of osteoarthritis (OA). METHODS Articular chondrocytes were cultured and the expression of GHRH receptor in chondrocytes was detected. Then recombinant adenovirus GHRH (Ad-GHRH) was transfected to one group of chondrocytes. The expression of collagen type II, matrix metalloproteinase 13 (MMP-13) and signal transducer and activator of transcription 3 (STAT3) in each experimental group was determined by Western blot. RESULTS The GHRH receptor was expressed in chondrocytes, and this provided a basis for further study of the role of GHRH in chondrocytes. Cell proliferation of the Ad-GHRH group was significantly higher than that of the OA group by CCK-8 assay. Compared with the OA-group, the protein expression of MMP‑13 was decreased in the Ad-GHRH group. Compared with the OA-group, the protein expression of collagen type II, phosphorylated STAT3 (P-STAT3) were increased in the Ad-GHRH group. CONCLUSION Our results show that the GHRH receptor is expressed in chondrocytes. GHRH can promote the proliferation of chondrocytes and the synthesis of type II collagen, and increase the extracellular matrix, which is achieved by phosphorylated STAT3 protein.
Collapse
Affiliation(s)
- Zhuoran LI
- Dalian Medical University, Dalian, China
| | - Yao LI
- Department of Physiology, Jinzhou Medical University, Jinzhou, China
- Correspondence to Yao Li, Ph.D. Department of Physiology, Jinzhou Medical University, No 40, SongPo Street, Jinzhou 121000, China Tel: +86-135-9129-8092 Fax: +86-135-9129-8092 E-mail:
| |
Collapse
|
8
|
Cinar I, Yayla M, Tavaci T, Toktay E, Ugan RA, Bayram P, Halici H. In Vivo and In Vitro Cardioprotective Effect of Gossypin Against Isoproterenol-Induced Myocardial Infarction Injury. Cardiovasc Toxicol 2022; 22:52-62. [PMID: 34599475 DOI: 10.1007/s12012-021-09698-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/08/2021] [Indexed: 10/20/2022]
Abstract
The aim of the study was to examine the protective effects and possible mechanism of gossypin against isoproterenol (ISO)-mediated myocardial damage in vivo and H9c2 cell damage in vitro. H9c2 cells were categorized into five groups. Viability was evaluated with MTT and LDH release in H9c2 cells. Apoptotic parameter analysis was performed with cytochrome c (Cyt-c), caspase-3 (CASP-3), and BCL2/Bax mRNA expression levels. In vivo, gossypin was administered orally to mice at doses of 5, 10, and 20 mg/kg for 7 days. ISO groups were injected with isoproterenol (150 mg/kg) subcutaneously (on 8th and 9th) for 2 days. Afterward, lactate dehydrogenase (LDH), creatine kinase-MB (CK-MB) levels and Troponin-I (Tn-I) amount from their serum, oxidative stress parameters superoxide dismutase (SOD) activity, glutathione (GSH) and malondialdehyde (MDA) levels, and tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin-1β (IL-1 β), and NF-kB mRNA expression levels with inflammatory markers from heart tissue were evaluated. In addition, IL-1B, BCL-2, and cas-3 immunohistochemical staining was performed from heart tissue and TNF-a level was measured by ELISA method. Administration of Gossypin protected the cells by dose-dependent, eliminating the reduced cell viability and increased LDH release of ISO in H9c2 cells. In mice serum analyses, increased LDH, CK-MB levels, and Tn-I levels were normalized by gossypin. ISO administration in heart tissue is regulated by gossypin with increased SOD activity, GSH amount, TNF-α, IL-6, IL-1β, and NF-kB mRNA expression levels and decreased MDA amount. Overall, the present results demonstrated that gossypin has a potential cardioprotective treatment for ischemic heart disease on in vivo and in vitro.
Collapse
Affiliation(s)
- Irfan Cinar
- Department of Pharmacology, Faculty of Medicine, Kastamonu University, 3700, Kastamonu, Turkey.
| | - Muhammed Yayla
- Faculty of Medicine, Department of Pharmacology, Kafkas University, Kars, Turkey
| | - Taha Tavaci
- Faculty of Medicine, Department of Pharmacology, Atatürk University, Erzurum, Turkey
| | - Erdem Toktay
- Faculty of Medicine, Department of Histology and Embriology, Kafkas University, Kars, Turkey
| | - Rustem Anil Ugan
- Faculty of Pharmacy, Department of Pharmacology, Atatürk University, Erzurum, Turkey
| | - Pınar Bayram
- Faculty of Medicine, Department of Histology and Embriology, Kafkas University, Kars, Turkey
| | - Hamza Halici
- Faculty of Medicine, Department of Pharmacology, Atatürk University, Erzurum, Turkey
| |
Collapse
|
9
|
Agonistic analog of growth hormone-releasing hormone promotes neurofunctional recovery and neural regeneration in ischemic stroke. Proc Natl Acad Sci U S A 2021; 118:2109600118. [PMID: 34782465 DOI: 10.1073/pnas.2109600118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 12/13/2022] Open
Abstract
Ischemic stroke can induce neurogenesis. However, most stroke-generated newborn neurons cannot survive. It has been shown that MR-409, a potent synthetic agonistic analog of growth hormone-releasing hormone (GHRH), can protect against some life-threatening pathological conditions by promoting cell proliferation and survival. The present study shows that long-term treatment with MR-409 (5 or 10 μg/mouse/d) by subcutaneous (s.c.) injection significantly reduces the mortality, ischemic insult, and hippocampal atrophy, and improves neurological functional recovery in mice operated on for transient middle cerebral artery occlusion (tMCAO). Besides, MR-409 can stimulate endogenous neurogenesis and improve the tMCAO-induced loss of neuroplasticity. MR-409 also enhances the proliferation and inhibits apoptosis of neural stem cells treated with oxygen and glucose deprivation-reperfusion. The neuroprotective effects of MR-409 are closely related to the activation of AKT/CREB and BDNF/TrkB pathways. In conclusion, the present study demonstrates that GHRH agonist MR-409 has remarkable neuroprotective effects through enhancing endogenous neurogenesis in cerebral ischemic mice.
Collapse
|
10
|
Recinella L, Chiavaroli A, Orlando G, Ferrante C, Gesmundo I, Granata R, Cai R, Sha W, Schally AV, Brunetti L, Leone S. Growth hormone-releasing hormone antagonistic analog MIA-690 stimulates food intake in mice. Peptides 2021; 142:170582. [PMID: 34051291 DOI: 10.1016/j.peptides.2021.170582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/20/2021] [Accepted: 05/23/2021] [Indexed: 12/26/2022]
Abstract
In addition to its metabolic and endocrine effects, growth hormone-releasing hormone (GHRH) was found to modulate feeding behavior in mammals. However, the role of recently synthetized GHRH antagonist MIA-690 and MR-409, a GHRH agonist, on feeding regulation remains to be evaluated. We investigated the effects of chronic subcutaneous administration of MIA-690 and MR-409 on feeding behavior and energy metabolism, in mice. Compared to vehicle, MIA-690 increased food intake and body weight, while MR-409 had no effect. Both analogs did not modify locomotor activity, as well as subcutaneous, visceral and brown adipose tissue (BAT) mass. A significant increase of hypothalamic agouti-related peptide (AgRP) gene expression and norepinephrine (NE) levels, along with a reduction of serotonin (5-HT) levels were found after MIA-690 treatment. MIA-690 was also found able to decrease gene expression of leptin in visceral adipose tissue. By contrast, MR-409 had no effect on the investigated markers. Concluding, chronic peripheral administration of MIA-690 could play an orexigenic role, paralleled by an increase in body weight. The stimulation of feeding could be mediated, albeit partially, by elevation of AgRP gene expression and NE levels and decreased 5-HT levels in the hypothalamus, along with reduced leptin gene expression, in the visceral adipose tissue.
Collapse
Affiliation(s)
- Lucia Recinella
- Department of Pharmacy, G. d'Annunzio University, Chieti, Italy.
| | | | - Giustino Orlando
- Department of Pharmacy, G. d'Annunzio University, Chieti, Italy.
| | - Claudio Ferrante
- Department of Pharmacy, G. d'Annunzio University, Chieti, Italy.
| | - Iacopo Gesmundo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin and Città Della Salute e Della Scienza Hospital, Turin, 10126, Italy.
| | - Riccarda Granata
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin and Città Della Salute e Della Scienza Hospital, Turin, 10126, Italy.
| | - Renzhi Cai
- Veterans Affairs Medical Center, Miami, FL, 33125, United States; Division of Endocrinology, Diabetes and Metabolism, and Division of Medical/Oncology, Department of Medicine, and Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL33136 and Sylvester Comprehensive Cancer Center, Miami, FL, 33136, United States.
| | - Wei Sha
- Veterans Affairs Medical Center, Miami, FL, 33125, United States; Division of Endocrinology, Diabetes and Metabolism, and Division of Medical/Oncology, Department of Medicine, and Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL33136 and Sylvester Comprehensive Cancer Center, Miami, FL, 33136, United States.
| | - Andrew V Schally
- Veterans Affairs Medical Center, Miami, FL, 33125, United States; Division of Endocrinology, Diabetes and Metabolism, and Division of Medical/Oncology, Department of Medicine, and Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL33136 and Sylvester Comprehensive Cancer Center, Miami, FL, 33136, United States.
| | - Luigi Brunetti
- Department of Pharmacy, G. d'Annunzio University, Chieti, Italy.
| | - Sheila Leone
- Department of Pharmacy, G. d'Annunzio University, Chieti, Italy.
| |
Collapse
|
11
|
Pathomechanisms and therapeutic opportunities in radiation-induced heart disease: from bench to bedside. Clin Res Cardiol 2021; 110:507-531. [PMID: 33591377 PMCID: PMC8055626 DOI: 10.1007/s00392-021-01809-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/16/2021] [Indexed: 12/14/2022]
Abstract
Cancer management has undergone significant improvements, which led to increased long-term survival rates among cancer patients. Radiotherapy (RT) has an important role in the treatment of thoracic tumors, including breast, lung, and esophageal cancer, or Hodgkin's lymphoma. RT aims to kill tumor cells; however, it may have deleterious side effects on the surrounding normal tissues. The syndrome of unwanted cardiovascular adverse effects of thoracic RT is termed radiation-induced heart disease (RIHD), and the risk of developing RIHD is a critical concern in current oncology practice. Premature ischemic heart disease, cardiomyopathy, heart failure, valve abnormalities, and electrical conduct defects are common forms of RIHD. The underlying mechanisms of RIHD are still not entirely clear, and specific therapeutic interventions are missing. In this review, we focus on the molecular pathomechanisms of acute and chronic RIHD and propose preventive measures and possible pharmacological strategies to minimize the burden of RIHD.
Collapse
|
12
|
Protective effects of growth hormone-releasing hormone analogs in DSS-induced colitis in mice. Sci Rep 2021; 11:2530. [PMID: 33510215 PMCID: PMC7844299 DOI: 10.1038/s41598-021-81778-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/08/2021] [Indexed: 02/07/2023] Open
Abstract
Besides its metabolic and endocrine effects, growth hormone (GH)-releasing hormone (GHRH) is involved in the modulation of inflammation. Recently synthetized GHRH antagonist MIA-690 and MR-409, GHRH agonist, developed by us have shown potent pharmacological effects in various experimental paradigms. However, whether their administration modify resistance to chronic inflammatory stimuli in colon is still unknown. Ex vivo results demonstrated that MIA-690 and MR-409 inhibited production of pro-inflammatory and oxidative markers induced by lipopolysaccharide on isolated mouse colon specimens. In vivo, both MIA-690 and MR-409 have also been able to decrease the responsiveness to nociceptive stimulus, in hot plate test. Additionally, both peptides also induced a decreased sensitivity to acute and persistent inflammatory stimuli in male mice, in formalin test and dextran sodium sulfate (DSS)-induced colitis model, respectively. MIA-690 and MR-409 attenuate DSS-induced colitis with particular regard to clinical manifestations, histopathological damage and release of pro-inflammatory and oxidative markers in colon specimens. Respect to MR-409, MIA-690 showed higher efficacy in inhibiting prostaglandin (PG)E2, 8-iso-PGF2α and serotonin (5-HT) levels, as well as tumor necrosis factor (TNF)-α, interleukin (IL)-6 and nitric oxide synthase gene expression in colon specimens of DSS-induced colitis. Furthermore, MIA-690 decreased serum insulin-like growth factor (IGF)-1 levels in mice DSS-treated, respect to MR-409. Thus, our findings highlight the protective effects of MIA-690 and MR-409 on inflammation stimuli. The higher antinflammatory and antioxidant activities observed with MIA-690 could be related to decreased serum IGF-1 levels.
Collapse
|
13
|
Gong Y, Luo S, Fan P, Zhu H, Li Y, Huang W. Growth hormone activates PI3K/Akt signaling and inhibits ROS accumulation and apoptosis in granulosa cells of patients with polycystic ovary syndrome. Reprod Biol Endocrinol 2020; 18:121. [PMID: 33287836 PMCID: PMC7720521 DOI: 10.1186/s12958-020-00677-x] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND It is reported that growth hormone (GH) can alleviate oxidative stress (OS) induced apoptosis in some types of cells by activating the PI3K/Akt signaling pathway. This study investigated the role and underlying mechanism of GH in OS and apoptosis in granulosa cells (GCs) of patients with polycystic ovary syndrome (PCOS). METHODS Primary GCs were collected from patients with and without PCOS (controls, n = 32) during oocyte retrieval. The patients with PCOS were randomly assigned to take GH treatment (PCOS-GH, n = 30) or without GH treatment (PCOS-C, n = 31). Reactive oxygen species (ROS) level was determined by spectrophotometry and fluorescence microscopy. GC apoptosis and mitochondrial membrane potential (MMP) were detected by Annexin V-FITC/PI double-staining and JC-1 staining, respectively (flow cytometry). The expression of apoptosis-related genes and proteins involved in PI3K/Akt signaling was determined by quantitative reverse-transcription polymerase chain reaction and western blotting, while active caspase-9 and caspase-3 levels of GCs were determined by enzyme-linked immunosorbent assay. RESULTS Our study found that in GCs of the PCOS-GH group, the ROS levels and apoptotic rates were significantly decreased, whereas MMP was significantly increased when compared to those in the PCOS-C group (P < 0.05). The mRNA levels of FOXO1, Bax, caspase-9, and caspase-3 were significantly decreased, whereas Bcl-2 was increased in GCs of the PCOS-GH group than those in the PCOS-C group (P < 0.05). The protein levels of FOXO1, Bax, cleaved caspase-9/caspase-9 and cleaved caspase-3/caspase-3 were decreased, whereas p-PI3K/PI3K, p-Akt/Akt, p-FOXO1 and Bcl-2 were increased in GCs of the PCOS-GH group, compared with those in the PCOS-C group (P < 0.05). CONCLUSION OS induced apoptosis and downregulated the PI3K/Akt signaling pathway in patients with PCOS. GH could alleviate apoptosis and activate the PI3K/Akt signaling pathway. CLINICAL TRIAL REGISTRATION NUMBER Chinese Clinical Trial Registry. ChiCTR1800019437 . Prospectively registered on October 20, 2018.
Collapse
Affiliation(s)
- Yan Gong
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Chengdu, Sichuan, People's Republic of China
- Reproductive Medicine Center, Sichuan Provincial Women's and Children's Hospital, The Affiliated Women's and children's Hospital of Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Shan Luo
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Chengdu, Sichuan, People's Republic of China
| | - Ping Fan
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Chengdu, Sichuan, People's Republic of China
| | - Huili Zhu
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Chengdu, Sichuan, People's Republic of China
| | - Yujing Li
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Chengdu, Sichuan, People's Republic of China
| | - Wei Huang
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Chengdu, Sichuan, People's Republic of China.
- Department of Reproductive Medicine, West China Second University Hospital of Sichuan University, #1416 Chenglong Road, JinJiang District, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
14
|
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: 27] [Impact Index Per Article: 6.8] [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
|
15
|
Li Z, Zhang N, Zhu L, Nan J, Shen J, Wang Z, Lin Y. Growth hormone-releasing hormone promotes therapeutic effects of peripheral blood endothelial progenitor cells in ischemic repair. J Endocrinol Invest 2020; 43:315-328. [PMID: 31506908 DOI: 10.1007/s40618-019-01109-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 08/30/2019] [Indexed: 11/26/2022]
Abstract
PURPOSE In peripheral artery disease, blockage of the blood supply to the limbs leads to blood flow attenuation and tissue ischemia. We investigated whether growth hormone-releasing hormone (GHRH) could enhance the biological functions and therapeutic effects of endothelial progenitor cells (EPCs) derived from adult human peripheral blood (PB). METHODS EPCs were isolated from human PB (PB-EPCs) and cord blood and expanded in vitro. PB-EPCs incubated with or without GHRH were evaluated for proliferation, migration, and angiogenesis capacity and apoptosis rates under oxidative stress conditions. Activation of STAT3 and Akt pathways was evaluated using Western blot. A hind-limb ischemia (HLI) mouse model was used to study the efficacy of GHRH in improving EPC therapy in vivo. RESULTS GHRH enhanced the proliferation, migration, and angiogenesis capacity of PB-EPCs and reduced apoptosis under H2O2 stimulation. These beneficial effects were GHRH receptor-dependent and were paralleled by increased phosphorylation of STAT3 and Akt. Transplantation of GHRH-preconditioned EPCs into HLI model mice enhanced blood flow recovery by increasing vascular formation density and enhanced tissue regeneration at the lesion site. CONCLUSION Our studies demonstrate a novel role for GHRH in dramatically improving therapeutic angiogenesis in HLI by enhancing the biological functions of EPCs. These findings support additional studies to explore the full potential of GHRH in augmenting cell therapy for the management of ischemia.
Collapse
Affiliation(s)
- Z Li
- Research Institute of Experimental Neurobiology, Department of Neurology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - N Zhang
- Research Institute of Experimental Neurobiology, Department of Neurology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - L Zhu
- Provincial Key Cardiovascular Research Laboratory, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - J Nan
- Provincial Key Cardiovascular Research Laboratory, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - J Shen
- Provincial Key Cardiovascular Research Laboratory, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Z Wang
- Wenzhou Municipal Key Cardiovascular Research Laboratory, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Y Lin
- Wenzhou Municipal Key Cardiovascular Research Laboratory, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China.
| |
Collapse
|
16
|
Stimulation of neuroendocrine differentiation in prostate cancer cells by GHRH and its blockade by GHRH antagonists. Invest New Drugs 2019; 38:746-754. [PMID: 31312936 DOI: 10.1007/s10637-019-00831-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/01/2019] [Indexed: 02/06/2023]
Abstract
Prostate cancer is the second leading cause of cancer-related deaths among men in developed countries. Neuroendocrine prostate cancer, in particular, is associated with an aggressive phenotype and a poor prognosis. Neuroendocrine cells produce and secrete peptide hormones and growth factors in a paracrine/autocrine manner which promote the progression of the disease. Recent studies have demonstrated that extracellular vesicles or exosomes are released by prostate cancer cells, supporting the spread of prostate cancer. Hence, the aim of this study was to investigate the effect of growth hormone-releasing hormone (GHRH) on neuroendocrine differentiation (NED) in the androgen-dependent prostate cancer cell line LNCaP and the molecular mechanisms underlying these effects. GHRH induced an increase in the percentage of neurite-bearing cells and in the protein levels of Neuron-Specific Enolase. Both effects were blocked by the GHRH receptor antagonist MIA-690. In addition, pretreatment of these cells with the calcium chelator BAPTA, the EGFR inhibitor AG-1478 or the HER2 inhibitor AG-825 reduced the effect of GHRH, suggesting that the GHRH-induced stimulation of NED involves calcium channel activation and EGFR/HER2 transactivation. Finally, PC3-derived exosomes led to an increase in NED, cell proliferation and cell adhesion. Altogether, these findings suggest that GHRH antagonists should be considered for in the management of neuroendocrine prostate cancer.
Collapse
|
17
|
Schally AV, Zhang X, Cai R, Hare JM, Granata R, Bartoli M. Actions and Potential Therapeutic Applications of Growth Hormone-Releasing Hormone Agonists. Endocrinology 2019; 160:1600-1612. [PMID: 31070727 DOI: 10.1210/en.2019-00111] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/03/2019] [Indexed: 11/19/2022]
Abstract
In this article, we briefly review the identification of GHRH, provide an abridged overview of GHRH antagonists, and focus on studies with GHRH agonists. Potent GHRH agonists of JI and MR class were synthesized and evaluated biologically. Besides the induction of the release of pituitary GH, GHRH analogs promote cell proliferation and exert stimulatory effects on various tissues, which express GHRH receptors (GHRH-Rs). A large body of work shows that GHRH agonists, such as MR-409, improve pancreatic β-cell proliferation and metabolic functions and facilitate engraftment of islets after transplantation in rodents. Accordingly, GHRH agonists offer a new therapeutic approach to treating diabetes. Various studies demonstrate that GHRH agonists promote repair of cardiac tissue, producing improvement of ejection fraction and reduction of infarct size in rats, reduction of infarct scar in swine, and attenuation of cardiac hypertrophy in mice, suggesting clinical applications. The presence of GHRH-Rs in ocular tissues and neuroprotective effects of GHRH analogs in experimental diabetic retinopathy indicates their possible therapeutic applications for eye diseases. Other effects of GHRH agonists, include acceleration of wound healing, activation of immune cells, and action on the central nervous system. As GHRH might function as a growth factor, we examined effects of GHRH agonists on tumors. In vitro, GHRH agonists stimulate growth of human cancer cells and upregulate GHRH-Rs. However, in vivo, GHRH agonists inhibit growth of human cancers xenografted into nude mice and downregulate pituitary and tumoral GHRH-Rs. Therapeutic applications of GHRH analogs are discussed. The development of GHRH analogs should lead to their clinical use.
Collapse
Affiliation(s)
- Andrew V Schally
- Veterans Affairs Medical Center, Miami, Florida
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Xianyang Zhang
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, Florida
| | - Renzhi Cai
- Veterans Affairs Medical Center, Miami, Florida
| | - Joshua M Hare
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, Florida
| | - Riccarda Granata
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Manuela Bartoli
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
18
|
Antagonists of growth hormone-releasing hormone (GHRH) inhibit the growth of human malignant pleural mesothelioma. Proc Natl Acad Sci U S A 2019; 116:2226-2231. [PMID: 30659154 PMCID: PMC6369772 DOI: 10.1073/pnas.1818865116] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive cancer with poor prognosis and limited treatment options. MPM remains a serious public health problem, and novel therapeutic strategies are urgently needed. The antitumor properties of growth hormone-releasing hormone (GHRH) antagonists have been demonstrated in different cancers; however, their influence in MPM remains unexplored. Our work shows that GHRH antagonists MIA-602 and MIA-690 reduce survival, proliferation, and migration of human MPM cell lines and primary MPM cells in vitro by modulating apoptotic and oncogenic pathways. In vivo, GHRH antagonists inhibited the growth of MPM xenografts and blunted the production of growth factors in tumors. Overall, the inhibitory activities described in this study suggest that GHRH antagonists may be considered for development of therapies for MPM. Malignant pleural mesothelioma (MPM) is an aggressive malignancy associated with exposure to asbestos, with poor prognosis and no effective therapies. The strong inhibitory activities of growth hormone-releasing hormone (GHRH) antagonists have been demonstrated in different experimental human cancers, including lung cancer; however, their role in MPM remains unknown. We assessed the effects of the GHRH antagonists MIA-602 and MIA-690 in vitro in MPM cell lines and in primary MPM cells, and in vivo in MPM xenografts. GHRH, GHRH receptor, and its main splice variant SV1 were found in all the MPM cell types examined. In vitro, MIA-602 and MIA-690 reduced survival and proliferation in both MPM cell lines and primary cells and showed synergistic inhibitory activity with the chemotherapy drug pemetrexed. In MPM cells, GHRH antagonists also regulated activity and expression of apoptotic molecules, inhibited cell migration, and reduced the expression of matrix metalloproteinases. These effects were accompanied by impairment of mitochondrial activity and increased production of reactive oxygen species. In vivo, s.c. administration of MIA-602 and MIA-690 at the dose of 5 μg/d for 4 wk strongly inhibited the growth of MPM xenografts in mice, along with reduction of tumor insulin-like growth factor-I and vascular endothelial growth factor. Overall, these results suggest that treatment with GHRH antagonists, alone or in association with chemotherapy, may offer an approach for the treatment of MPM.
Collapse
|
19
|
Saad NS, Elnakish MT, Ahmed AAE, Janssen PML. Protein Kinase A as a Promising Target for Heart Failure Drug Development. Arch Med Res 2018; 49:530-537. [PMID: 30642654 PMCID: PMC6451668 DOI: 10.1016/j.arcmed.2018.12.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/13/2018] [Indexed: 12/24/2022]
Abstract
Heart failure (HF) is a clinical syndrome characterized by impaired ability of the heart to fill or eject blood. HF is rather prevalent and it represents the foremost reason of hospitalization in the United States. The costs linked to HF overrun those of all other causes of disabilities, and death in the United States and all over the developed as well as the developing countries which amplify the supreme significance of its prevention. Protein kinase (PK) A plays multiple roles in heart functions including, contraction, metabolism, ion fluxes, and gene transcription. Altered PKA activity is likely to cause the progression to cardiomyopathy and HF. Thus, this review is intended to focus on the roles of PKA and PKA-mediated signal transduction in the healthy heart as well as during the development of HF. Furthermore, the impact of cardiac PKA inhibition/activation will be highlighted to identify PKA as a potential target for the HF drug development.
Collapse
Affiliation(s)
- Nancy S Saad
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Mohammad T Elnakish
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Amany A E Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Paul M L Janssen
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
20
|
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: 0.5] [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
|
21
|
Shen J, Zhang N, Lin YN, Xiang P, Liu XB, Shan PF, Hu XY, Zhu W, Tang YL, Webster KA, Cai R, Schally AV, Wang J, Yu H. Regulation of Vascular Calcification by Growth Hormone-Releasing Hormone and Its Agonists. Circ Res 2018; 122:1395-1408. [PMID: 29618597 DOI: 10.1161/circresaha.117.312418] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
RATIONALE Vascular calcification (VC) is a marker of the severity of atherosclerotic disease. Hormones play important roles in regulating calcification; estrogen and parathyroid hormones exert opposing effects, the former alleviating VC and the latter exacerbating it. To date no treatment strategies have been developed to regulate clinical VC. OBJECTIVE The objective of this study was to investigate the effect of growth hormone-releasing hormone (GHRH) and its agonist (GHRH-A) on the blocking of VC in a mouse model. METHODS AND RESULTS Young adult osteoprotegerin-deficient mice were given daily subcutaneous injections of GHRH-A (MR409) for 4 weeks. Significant reductions in calcification of the aortas of MR409-treated mice were paralleled by markedly lower alkaline phosphatase activity and a dramatic reduction in the expression of transcription factors, including the osteogenic marker gene Runx2 and its downstream factors, osteonectin and osteocalcin. The mechanism of action of GHRH-A was dissected in smooth muscle cells isolated from human and mouse aortas. Calcification of smooth muscle cells induced by osteogenic medium was inhibited in the presence of GHRH or MR409, as evidenced by reduced alkaline phosphatase activity and Runx2 expression. Inhibition of calcification by MR409 was partially reversed by MIA602, a GHRH antagonist, or a GHRH receptor-selective small interfering RNA. Treatment with MR409 induced elevated cytosolic cAMP and its target, protein kinase A which in turn blocked nicotinamide adenine dinucleotide phosphate oxidase activity and reduced production of reactive oxygen species, thus blocking the phosphorylation of nuclear factor κB (p65), a key intermediate in the ligand of receptor activator for nuclear factor-κ B-Runx2/alkaline phosphatase osteogenesis program. A protein kinase A-selective small interfering RNA or the chemical inhibitor H89 abolished these beneficial effects of MR409. CONCLUSIONS GHRH-A controls osteogenesis in smooth muscle cells by targeting cross talk between protein kinase A and nuclear factor κB (p65) and through the suppression of reactive oxygen species production that induces the Runx2 gene and alkaline phosphatase. Inflammation-mediated osteogenesis is thereby blocked. GHRH-A may represent a new pharmacological strategy to regulate VC.
Collapse
Affiliation(s)
- Jian Shen
- From the Departments of Cardiology (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
- Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
| | - Ning Zhang
- From the Departments of Cardiology (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
- Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
| | - Yi-Nuo Lin
- From the Departments of Cardiology (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
- Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
| | - PingPing Xiang
- From the Departments of Cardiology (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
- Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
| | - Xian-Bao Liu
- From the Departments of Cardiology (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
- Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
| | | | - Xin-Yang Hu
- From the Departments of Cardiology (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
- Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
| | - Wei Zhu
- From the Departments of Cardiology (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
- Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
| | - Yao-Liang Tang
- Vascular Biology Center, Georgia Regents University, Augusta (Y.-l.T.)
| | - Keith A Webster
- Department of Molecular and Cellular Pharmacology and the Vascular Biology Institute (K.A.W., R.C., A.V.S.)
| | - Renzhi Cai
- Department of Molecular and Cellular Pharmacology and the Vascular Biology Institute (K.A.W., R.C., A.V.S.)
- Divisions of Hematology/Oncology, Department of Medicine (R.C., A.V.S.)
- Miller School of Medicine, University of Miami, FL; and Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL (R.C., A.V.S.)
| | - Andrew V Schally
- Department of Molecular and Cellular Pharmacology and the Vascular Biology Institute (K.A.W., R.C., A.V.S.)
- Divisions of Hematology/Oncology, Department of Medicine (R.C., A.V.S.)
- Miller School of Medicine, University of Miami, FL; and Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL (R.C., A.V.S.)
| | - Jian'an Wang
- From the Departments of Cardiology (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
- Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
| | - Hong Yu
- From the Departments of Cardiology (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
- Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (J.S., N.Z., Y.-N.L., P.P.X., X.-b.L., X.-y.H., W.Z., J.W., H.Y.)
| |
Collapse
|
22
|
Caicedo D, Devesa P, Arce VM, Requena J, Devesa J. Chronic limb-threatening ischemia could benefit from growth hormone therapy for wound healing and limb salvage. Ther Adv Cardiovasc Dis 2018; 12:53-72. [PMID: 29271292 PMCID: PMC5772430 DOI: 10.1177/1753944717745494] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/12/2017] [Indexed: 01/20/2023] Open
Abstract
Revascularization for chronic limb-threatening ischemia (CLTI) is necessary to alleviate symptoms and wound healing. When it fails or is not possible, there are few alternatives to avoid limb amputation in these patients. Although experimental studies with stem cells and growth factors have shown promise, clinical trials have demonstrated inconsistent results because CLTI patients generally need arteriogenesis rather than angiogenesis. Moreover, in addition to the perfusion of the limb, there is the need to improve the neuropathic response for wound healing, especially in diabetic patients. Growth hormone (GH) is a pleiotropic hormone capable of boosting the aforementioned processes and adds special benefits for the redox balance. This hormone has the potential to mitigate symptoms in ischemic patients with no other options and improves the cardiovascular complications associated with the disease. Here, we discuss the pros and cons of using GH in such patients, focus on its effects on peripheral arteries, and analyze the possible benefits of treating CLTI with this hormone.
Collapse
Affiliation(s)
- Diego Caicedo
- Scientific Direction, Medical Center Foltra. Travesía Montouto, 24; 15710-Teo, A Coruña, 15886, Spain
| | - Pablo Devesa
- Scientific Direction, Medical Center Foltra. Travesía Montouto, 24; 15710-Teo, A Coruña, 15886, Spain
| | - Víctor M. Arce
- Scientific Direction, Medical Center Foltra. Travesía Montouto, 24; 15710-Teo, A Coruña, 15886, Spain
| | - Julia Requena
- Scientific Direction, Medical Center Foltra. Travesía Montouto, 24; 15710-Teo, A Coruña, 15886, Spain
| | - Jesús Devesa
- Scientific Direction, Medical Center Foltra. Travesía Montouto, 24; 15710-Teo, A Coruña, 15886, Spain
| |
Collapse
|
23
|
Growth hormone-releasing hormone attenuates cardiac hypertrophy and improves heart function in pressure overload-induced heart failure. Proc Natl Acad Sci U S A 2017; 114:12033-12038. [PMID: 29078377 PMCID: PMC5692579 DOI: 10.1073/pnas.1712612114] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pathological cardiac hypertrophy, characterized by heart growth in response to pressure or volume overload, such as in the setting of hypertension, is the main risk factor for heart failure (HF). The identification of therapeutic strategies to prevent or reverse cardiac hypertrophy is therefore a priority for curing HF. It is known that growth hormone-releasing hormone (GHRH) displays cardioprotective functions; however, its therapeutic potential in hypertrophy and HF is unknown. Here we show that GHRH reduces cardiomyocyte hypertrophy in vitro through inhibition of hypertrophic pathways. In vivo, the GHRH analog MR-409 attenuates cardiac hypertrophy in mice subjected to transverse aortic constriction and improves cardiac function. These findings suggest therapeutic use of GHRH analogs for treatment of pathological cardiac hypertrophy and HF. It has been shown that growth hormone-releasing hormone (GHRH) reduces cardiomyocyte (CM) apoptosis, prevents ischemia/reperfusion injury, and improves cardiac function in ischemic rat hearts. However, it is still not known whether GHRH would be beneficial for life-threatening pathological conditions, like cardiac hypertrophy and heart failure (HF). Thus, we tested the myocardial therapeutic potential of GHRH stimulation in vitro and in vivo, using GHRH or its agonistic analog MR-409. We show that in vitro, GHRH(1-44)NH2 attenuates phenylephrine-induced hypertrophy in H9c2 cardiac cells, adult rat ventricular myocytes, and human induced pluripotent stem cell-derived CMs, decreasing expression of hypertrophic genes and regulating hypertrophic pathways. Underlying mechanisms included blockade of Gq signaling and its downstream components phospholipase Cβ, protein kinase Cε, calcineurin, and phospholamban. The receptor-dependent effects of GHRH also involved activation of Gαs and cAMP/PKA, and inhibition of increase in exchange protein directly activated by cAMP1 (Epac1). In vivo, MR-409 mitigated cardiac hypertrophy in mice subjected to transverse aortic constriction and improved cardiac function. Moreover, CMs isolated from transverse aortic constriction mice treated with MR-409 showed improved contractility and reversal of sarcolemmal structure. Overall, these results identify GHRH as an antihypertrophic regulator, underlying its therapeutic potential for HF, and suggest possible beneficial use of its analogs for treatment of pathological cardiac hypertrophy.
Collapse
|
24
|
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: 3.1] [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
|
25
|
Growth Hormone-Releasing Hormone and Its Analogues: Significance for MSCs-Mediated Angiogenesis. Stem Cells Int 2016; 2016:8737589. [PMID: 27774107 PMCID: PMC5059609 DOI: 10.1155/2016/8737589] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 06/19/2016] [Accepted: 07/03/2016] [Indexed: 02/08/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are promising candidates for regenerative medicine because of their multipotency, immune-privilege, and paracrine properties including the potential to promote angiogenesis. Accumulating evidence suggests that the inherent properties of cytoprotection and tissue repair by native MSCs can be enhanced by various preconditioning stimuli implemented prior to cell transplantation. Growth hormone-releasing hormone (GHRH), a stimulator in extrahypothalamus systems including tumors, has attracted great attentions in recent years because GHRH and its agonists could promote angiogenesis in various tissues. GHRH and its agonists are proangiogenic in responsive tissues including tumors, and GHRH antagonists have been tested as antitumor agents through their ability to suppress angiogenesis and cell growth. GHRH-R is expressed by MSCs and evolving work from our laboratory indicates that treatment of MSCs with GHRH agonists prior to cell transplantation markedly enhanced the angiogenic potential and tissue reparative properties of MSCs through a STAT3 signaling pathway. In this review we summarized the possible effects of GHRH analogues on cell growth and development, as well as on the proangiogenic properties of MSCs. We also discussed the relationship between GHRH analogues and MSC-mediated angiogenesis. The analyses provide new insights into molecular pathways of MSCs-based therapies and their augmentation by GHRH analogues.
Collapse
|
26
|
Kiscsatári L, Varga Z, Schally AV, Gáspár R, Nagy CT, Giricz Z, Ferdinandy P, Fábián G, Kahán Z, Görbe A. Protection of neonatal rat cardiac myocytes against radiation-induced damage with agonists of growth hormone-releasing hormone. Pharmacol Res 2016; 111:859-866. [PMID: 27480202 DOI: 10.1016/j.phrs.2016.07.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 07/05/2016] [Accepted: 07/25/2016] [Indexed: 12/15/2022]
Abstract
Despite the great clinical significance of radiation-induced cardiac damage, experimental investigation of its mechanisms is an unmet need in medicine. Beneficial effects of growth hormone-releasing hormone (GHRH) agonists in regeneration of the heart have been demonstrated. The aim of this study was the evaluation of the potential of modern GHRH agonistic analogs in prevention of radiation damage in an in vitro cardiac myocyte-based model. Cultures of cardiac myocytes isolated from newborn rats (NRVM) were exposed to a radiation dose of 10Gy. The effects of the agonistic analogs, JI-34 and MR-356, of human GHRH on cell viability, proliferation, their mechanism of action and the protein expression of the GHRH/SV1 receptors were studied. JI-34 and MR-356, had no effect on cell viability or proliferation in unirradiated cultures. However, in irradiated cells JI-34 showed protective effects on cell viability at concentrations of 10 and 100nM, and MR-356 at 500nM; but no such protective effect was detected on cell proliferation. Both agonistic analogs decreased radiation-induced ROS level and JI-34 interfered with the activation of SAFE/RISK pathways. Using Western blot analysis, a 52kDa protein isoform of GHRHR was detected in the samples in both irradiated and unirradiated cells. Since GHRH agonistic analogs, JI-34 and MR-356 alleviated radiation-induced damage of cardiac myocytes, they should be tested in vivo as potential protective agents against radiogenic heart damage.
Collapse
Affiliation(s)
- Laura Kiscsatári
- Department of Oncotherapy, University of Szeged, Szeged, Hungary
| | - Zoltán Varga
- Department of Oncotherapy, University of Szeged, Szeged, Hungary
| | - Andrew V Schally
- Veterans Affairs Medical Center, Miami FL, USA and Departments of Pathology and Medicine, Divisions of Hematology/Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Renáta Gáspár
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Csilla Terézia Nagy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Péter Ferdinandy
- Pharmahungary Group, Szeged, Hungary; Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Gabriella Fábián
- Department of Oncotherapy, University of Szeged, Szeged, Hungary
| | - Zsuzsanna Kahán
- Department of Oncotherapy, University of Szeged, Szeged, Hungary
| | - Anikó Görbe
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged, Hungary; Pharmahungary Group, Szeged, Hungary; Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
27
|
Granata R. Peripheral activities of growth hormone-releasing hormone. J Endocrinol Invest 2016; 39:721-7. [PMID: 26891937 DOI: 10.1007/s40618-016-0440-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/03/2016] [Indexed: 12/21/2022]
Abstract
Growth hormone (GH)-releasing hormone (GHRH) is produced by the hypothalamus and stimulates GH synthesis and release in the anterior pituitary gland. In addition to its endocrine role, GHRH exerts a wide range of extrapituitary effects which include stimulation of cell proliferation, survival and differentiation, and inhibition of apoptosis. Accordingly, expression of GHRH, as well as the receptor GHRH-R and its splice variants, has been demonstrated in different peripheral tissues and cell types. Among the direct peripheral activities, GHRH regulates pancreatic islet and β-cell survival and function and endometrial cell proliferation, promotes cardioprotection and wound healing, influences the immune and reproductive systems, reduces inflammation, indirectly increases lifespan and adiposity and acts on skeletal muscle cells to inhibit cell death and atrophy. Therefore, it is becoming increasingly clear that GHRH exerts important extrapituitary functions, suggesting potential therapeutic use of the peptide and its analogs in a wide range of medical settings.
Collapse
Affiliation(s)
- R Granata
- Lab of Molecular and Cellular Endocrinology, Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Torino, Corso Dogliotti, 14, 10126, Turin, Italy.
| |
Collapse
|
28
|
Kanashiro-Takeuchi RM, Szalontay L, Schally AV, Takeuchi LM, Popovics P, Jaszberenyi M, Vidaurre I, Zarandi M, Cai RZ, Block NL, Hare JM, Rick FG. New therapeutic approach to heart failure due to myocardial infarction based on targeting growth hormone-releasing hormone receptor. Oncotarget 2016; 6:9728-39. [PMID: 25797248 PMCID: PMC4496393 DOI: 10.18632/oncotarget.3303] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 02/08/2015] [Indexed: 12/21/2022] Open
Abstract
Background We previously showed that growth hormone-releasing hormone (GHRH) agonists are cardioprotective following myocardial infarction (MI). Here, our aim was to evaluate the in vitro and in vivo activities of highly potent new GHRH agonists, and elucidate their mechanisms of action in promoting cardiac repair. Methods and Results H9c2 cells were cultured in serum-free medium, mimicking nutritional deprivation. GHRH agonists decreased calcium influx and significantly improved cell survival. Rats with cardiac infarction were treated with GHRH agonists or placebo for four weeks. MI size was reduced by selected GHRH agonists (JI-38, MR-356, MR-409); this accompanied an increased number of cardiac c-kit+ cells, cellular mitotic divisions, and vascular density. One week post-MI, MR-409 significantly reduced plasma levels of IL-2, IL-6, IL-10 and TNF-α compared to placebo. Gene expression studies revealed favorable outcomes of MR-409 treatment partially result from inhibitory activity on pro-apoptotic molecules and pro-fibrotic systems, and by elevation of bone morphogenetic proteins. Conclusions Treatment with GHRH agonists appears to reduce the inflammatory responses post-MI and may consequently improve mechanisms of healing and cardiac remod eling by regulating pathways involved in fibrosis, apoptosis and cardiac repair. Patients with cardiac dysfunction could benefit from treatment with novel GHRH agonists.
Collapse
Affiliation(s)
- Rosemeire M Kanashiro-Takeuchi
- Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, Florida, United States of America.,Department of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Florida, United States of America
| | - Luca Szalontay
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America
| | - Andrew V Schally
- Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, Florida, United States of America.,Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America.,Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, United States of America.,Department of Medicine, Divisions of Hematology/Oncology and Endocrinology, University of Miami, Miller School of Medicine, Miami, Florida, United States of America.,Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Lauro M Takeuchi
- Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Petra Popovics
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America.,Department of Medicine III, Medical Faculty Carl Gustav Carus, TU Dresden, Germany.,Department of Medicine, Division of Cardiology, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Miklos Jaszberenyi
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America.,Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Irving Vidaurre
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America
| | - Marta Zarandi
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America
| | - Ren-Zhi Cai
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America.,Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Norman L Block
- Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, Florida, United States of America.,Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America.,Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, United States of America.,Department of Medicine, Divisions of Hematology/Oncology and Endocrinology, University of Miami, Miller School of Medicine, Miami, Florida, United States of America.,Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, Florida, United States of America.,Department of Medicine, Division of Cardiology, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Ferenc G Rick
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America.,Department of Urology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, United States of America
| |
Collapse
|
29
|
Ma Q, Xia X, Tao Q, Lu K, Shen J, Xu Q, Hu X, Tang Y, Block NL, Webster KA, Schally AV, Wang J, Yu H. Profound Actions of an Agonist of Growth Hormone-Releasing Hormone on Angiogenic Therapy by Mesenchymal Stem Cells. Arterioscler Thromb Vasc Biol 2016; 36:663-672. [PMID: 26868211 DOI: 10.1161/atvbaha.116.307126] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 01/21/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The efficiency of cell therapy is limited by poor cell survival and engraftment. Here, we studied the effect of the growth hormone-releasing hormone agonist, JI-34, on mesenchymal stem cell (MSC) survival and angiogenic therapy in a mouse model of critical limb ischemia. APPROACH AND RESULTS Mouse bone marrow-derived MSCs were incubated with or without 10(-8) mol/L JI-34 for 24 hours. MSCs were then exposed to hypoxia and serum deprivation to detect the effect of preconditioning on cell apoptosis, migration, and tube formation. For in vivo tests, critical limb ischemia was induced by femoral artery ligation. After surgery, mice received 50 μL phosphate-buffered saline or with 1×10(6) MSCs or with 1×10(6) JI-34-reconditioned MSCs. Treatment of MSCs with JI-34 improved MSC viability and mobility and markedly enhanced their capability to promote endothelial tube formation in vitro. These effects were paralleled by an increased phosphorylation and nuclear translocation of signal transducer and activator of transcription 3. In vivo, JI-34 pretreatment enhanced the engraftment of MSCs into ischemic hindlimb muscles and augmented reperfusion and limb salvage compared with untreated MSCs. Significantly more vasculature and proliferating CD31(+) and CD34(+) cells were detected in ischemic muscles that received MSCs treated with JI-34. CONCLUSIONS Our studies demonstrate a novel role for JI-34 to markedly improve therapeutic angiogenesis in hindlimb ischemia by increasing the viability and mobility of MSCs. These findings support additional studies to explore the full potential of growth hormone-releasing hormone agonists to augment cell therapy in the management of ischemia.
Collapse
MESH Headings
- Active Transport, Cell Nucleus
- Animals
- Antigens, CD34/metabolism
- Apoptosis/drug effects
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Cells, Cultured
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Female
- Growth Hormone-Releasing Hormone/agonists
- Growth Hormone-Releasing Hormone/analogs & derivatives
- Growth Hormone-Releasing Hormone/metabolism
- Growth Hormone-Releasing Hormone/pharmacology
- Hindlimb
- Ischemia/metabolism
- Ischemia/physiopathology
- Ischemia/therapy
- Male
- Mesenchymal Stem Cell Transplantation
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/metabolism
- Mice, Inbred C57BL
- Muscle, Skeletal/blood supply
- Neovascularization, Physiologic
- Peptide Fragments/pharmacology
- Phosphorylation
- Platelet Endothelial Cell Adhesion Molecule-1/metabolism
- Receptors, Neuropeptide/agonists
- Receptors, Neuropeptide/metabolism
- Receptors, Pituitary Hormone-Regulating Hormone/agonists
- Receptors, Pituitary Hormone-Regulating Hormone/metabolism
- STAT3 Transcription Factor/metabolism
- Time Factors
Collapse
|
30
|
Fridlyand LE, Tamarina NA, Schally AV, Philipson LH. Growth Hormone-Releasing Hormone in Diabetes. Front Endocrinol (Lausanne) 2016; 7:129. [PMID: 27777568 PMCID: PMC5056186 DOI: 10.3389/fendo.2016.00129] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/06/2016] [Indexed: 12/13/2022] Open
Abstract
Growth hormone-releasing hormone (GHRH) is produced by the hypothalamus and stimulates growth hormone synthesis and release in the anterior pituitary gland. In addition, GHRH is an important regulator of cellular functions in many cells and organs. Expression of GHRH G-Protein Coupled Receptor (GHRHR) has been demonstrated in different peripheral tissues and cell types, including pancreatic islets. Among the peripheral activities, recent studies demonstrate a novel ability of GHRH analogs to increase and preserve insulin secretion by beta-cells in isolated pancreatic islets, which makes them potentially useful for diabetes treatment. This review considers the role of GHRHR in the beta-cell and addresses the unique engineered GHRH agonists and antagonists for treatment of type 2 diabetes mellitus. We discuss the similarity of signaling pathways activated by GHRHR in pituitary somatotrophs and in pancreatic beta-cells and possible ways as to how the GHRHR pathway can interact with glucose and other secretagogues to stimulate insulin secretion. We also consider the hypothesis that novel GHRHR agonists can improve glucose metabolism in Type 2 diabetes by preserving the function and survival of pancreatic beta-cells. Wound healing and cardioprotective action with new GHRH agonists suggest that they may prove useful in ameliorating certain diabetic complications. These findings highlight the future potential therapeutic effectiveness of modulators of GHRHR activity for the development of new therapeutic approaches in diabetes and its complications.
Collapse
Affiliation(s)
- Leonid E. Fridlyand
- Department of Medicine, Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
- *Correspondence: Leonid E. Fridlyand,
| | - Natalia A. Tamarina
- Department of Medicine, Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Andrew V. Schally
- VA Medical Center, Miami, FL, USA
- Department of Pathology and Medicine, Division of Endocrinology and Hematology-Oncology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Louis H. Philipson
- Department of Medicine, Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
31
|
Pasqua T, Tota B, Penna C, Corti A, Cerra MC, Loh Y P, Angelone T. pGlu-serpinin protects the normotensive and hypertensive heart from ischemic injury. J Endocrinol 2015; 227:167-178. [PMID: 26400960 PMCID: PMC4651656 DOI: 10.1530/joe-15-0199] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/23/2015] [Indexed: 12/15/2022]
Abstract
Serpinin peptides derive from proteolytic cleavage of Chromogranin-A at C-terminus. Serpinin and the more potent pyroglutaminated-serpinin (pGlu-Serp) are positive cardiac β-adrenergic-like modulators, acting through β1-AR/AC/cAMP/PKA pathway. Because in some conditions this pathway and/or other pro-survival pathways, activated by other Chromogranin-A fragments, may cross-talk and may be protective, here we explored whether pGlu-Serp cardioprotects against ischemia/reperfusion injury under normotensive and hypertensive conditions. In the latter condition, cardioprotection is often blunted because of the limitations on pro-survival Reperfusion Injury Salvage Kinases (RISK) pathway activation. The effects of pGlu-Serp were evaluated on infarct size (IS) and cardiac function by using the isolated and Langendorff perfused heart of normotensive (Wistar Kyoto, WKY) and spontaneously hypertensive (SHR) rats exposed to ischemic pre-conditioning (PreC) and post-conditioning (PostC). In both WKY and SHR rat, pGlu-Serp induced mild cardioprotection in both PreC and PostC. pGlu-Serp administered at the reperfusion (Serp-PostC) significantly reduced IS, being more protective in SHR than in WKY. Conversely, left ventricular developed pressure (LVDevP) post-ischemic recovery was greater in WKY than in SHR. pGlu-Serp-PostC reduced contracture in both strains. Co-infusion with specific RISK inhibitors (PI3K/Akt, MitoKATP channels and PKC) blocked the pGlu-Serp-PostC protective effects. To show direct effect on cardiomyocytes, we pre-treated H9c2 cells with pGlu-Serp, which were thus protected against hypoxia/reoxygenation. These results suggest pGlu-Serp as a potential modulatory agent implicated in the protective processes that can limit infarct size and overcome the hypertension-induced failure of PostC.
Collapse
Affiliation(s)
- T Pasqua
- Dept of Biology, Ecology, and E.S., University of Calabria, Rende (CS), Italy
| | - B Tota
- Dept of Biology, Ecology, and E.S., University of Calabria, Rende (CS), Italy
| | - C Penna
- Dept of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - A Corti
- Tumor Biology and Vascular Targeting Unit, Division of Molecular Oncology, San Raffaele Scientific Institute, Milan, Italy
| | - M C Cerra
- Dept of Biology, Ecology, and E.S., University of Calabria, Rende (CS), Italy
| | - P Loh Y
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Md 20892, USA
| | - T Angelone
- Dept of Biology, Ecology, and E.S., University of Calabria, Rende (CS), Italy
| |
Collapse
|
32
|
Gallo D, Gesmundo I, Trovato L, Pera G, Gargantini E, Minetto MA, Ghigo E, Granata R. GH-Releasing Hormone Promotes Survival and Prevents TNF-α-Induced Apoptosis and Atrophy in C2C12 Myotubes. Endocrinology 2015; 156:3239-52. [PMID: 26110916 DOI: 10.1210/en.2015-1098] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Skeletal muscle atrophy is a consequence of different chronic diseases, including cancer, heart failure, and diabetes, and also occurs in aging and genetic myopathies. It results from an imbalance between anabolic and catabolic processes, and inflammatory cytokines, such as TNF-α, have been found elevated in muscle atrophy and implicated in its pathogenesis. GHRH, in addition to stimulating GH secretion from the pituitary, exerts survival and antiapoptotic effects in different cell types. Moreover, we and others have recently shown that GHRH displays antiapoptotic effects in isolated cardiac myocytes and protects the isolated heart from ischemia/reperfusion injury and myocardial infarction in vivo. On these bases, we investigated the effects of GHRH on survival and apoptosis of TNF-α-treated C2C12 myotubes along with the underlying mechanisms. GHRH increased myotube survival and prevented TNF-α-induced apoptosis through GHRH receptor-mediated mechanisms. These effects involved activation of phosphoinositide 3-kinase/Akt pathway and inactivation of glycogen synthase kinase-3β, whereas mammalian target of rapamycin was unaffected. GHRH also increased the expression of myosin heavy chain and the myogenic transcription factor myogenin, which were both reduced by the cytokine. Furthermore, GHRH inhibited TNF-α-induced expression of nuclear factor-κB, calpain, and muscle ring finger1, which are all involved in muscle protein degradation. In summary, these results indicate that GHRH exerts survival and antiapoptotic effects in skeletal muscle cells through the activation of anabolic pathways and the inhibition of proteolytic routes. Overall, our findings suggest a novel therapeutic role for GHRH in the treatment of muscle atrophy-associated diseases.
Collapse
Affiliation(s)
- Davide Gallo
- Laboratory of Molecular and Cellular Endocrinology (D.G., I.G., L.T., G.P., E.Ga., R.G.), and Division of Endocrinology, Diabetes, and Metabolism (D.G., I.G., L.T., G.P., E.Ga., M.A.M., E.Gh., R.G.), Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Iacopo Gesmundo
- Laboratory of Molecular and Cellular Endocrinology (D.G., I.G., L.T., G.P., E.Ga., R.G.), and Division of Endocrinology, Diabetes, and Metabolism (D.G., I.G., L.T., G.P., E.Ga., M.A.M., E.Gh., R.G.), Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Letizia Trovato
- Laboratory of Molecular and Cellular Endocrinology (D.G., I.G., L.T., G.P., E.Ga., R.G.), and Division of Endocrinology, Diabetes, and Metabolism (D.G., I.G., L.T., G.P., E.Ga., M.A.M., E.Gh., R.G.), Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Giulia Pera
- Laboratory of Molecular and Cellular Endocrinology (D.G., I.G., L.T., G.P., E.Ga., R.G.), and Division of Endocrinology, Diabetes, and Metabolism (D.G., I.G., L.T., G.P., E.Ga., M.A.M., E.Gh., R.G.), Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Eleonora Gargantini
- Laboratory of Molecular and Cellular Endocrinology (D.G., I.G., L.T., G.P., E.Ga., R.G.), and Division of Endocrinology, Diabetes, and Metabolism (D.G., I.G., L.T., G.P., E.Ga., M.A.M., E.Gh., R.G.), Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Marco Alessandro Minetto
- Laboratory of Molecular and Cellular Endocrinology (D.G., I.G., L.T., G.P., E.Ga., R.G.), and Division of Endocrinology, Diabetes, and Metabolism (D.G., I.G., L.T., G.P., E.Ga., M.A.M., E.Gh., R.G.), Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Ezio Ghigo
- Laboratory of Molecular and Cellular Endocrinology (D.G., I.G., L.T., G.P., E.Ga., R.G.), and Division of Endocrinology, Diabetes, and Metabolism (D.G., I.G., L.T., G.P., E.Ga., M.A.M., E.Gh., R.G.), Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Riccarda Granata
- Laboratory of Molecular and Cellular Endocrinology (D.G., I.G., L.T., G.P., E.Ga., R.G.), and Division of Endocrinology, Diabetes, and Metabolism (D.G., I.G., L.T., G.P., E.Ga., M.A.M., E.Gh., R.G.), Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| |
Collapse
|
33
|
Zhou D, You J, Li QY, Li HZ, Wu WF, Zhang XD, Zhang JH, Tang SS, Wang YK, Liu T. Synthesis and biological evaluation of novel structure-related hGHRH agonistic analogs. Growth Factors 2015; 33:160-8. [PMID: 25798996 DOI: 10.3109/08977194.2015.1010644] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Activity and half-life play key roles in the application of GHRH analogues. The GHRH monomers produced in a solid synthesizer were incubated, respectively, in NH4OH solution and lyophilized to obtain their dimers. The activities, specificities, and receptor affinities of the GHRH dimers were evaluated in rGH release/inhibition, rACTH/LH/PRL release, pituitary homogenate binding, and fluorescent staining. Compared to hGHRH(1-44)NH2 (S), PP-hGHRH(1-44)-GGC-CGG-hGHRH(44-1)-PP (2D), P-hGHRH(1-44)-GGC-CGG-hGHRH(44-1)-P (2E), (1)P-hGHRH(2-44)-GGC-CGG-hGHRH(44-2)-(1)P (2F), or hGHRH(1-44)-GGC-CGG-hGHRH(44-1) (2Y) had potency of 104 ± 16.7%, 94 ± 32.6%, 114 ± 16.6%, or 122 ± 14.5% and similar specificities. The inhibition effect of GHIH on rGH stimulated by GHRH dimer was in dose-/time-dependent manner. The staining of FITC-labeled dimer showed cytomembrane distribution and the binding ranking was 2F>2D>2Y>2E>S. 2F presents the strongest activity and the highest affinity to pituitary cells. The dimer with (1)Pro-GHRH stimulates stronger rGH release than that with (1)Tyr-GHRH and the N-terminal single cyclic amino acid is required for the stimulation.
Collapse
Affiliation(s)
- Dong Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Courses, Guangdong Pharmaceutical University , Guangzhou , China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Bagno LL, Kanashiro-Takeuchi RM, Suncion VY, Golpanian S, Karantalis V, Wolf A, Wang B, Premer C, Balkan W, Rodriguez J, Valdes D, Rosado M, Block NL, Goldstein P, Morales A, Cai RZ, Sha W, Schally AV, Hare JM. Growth hormone-releasing hormone agonists reduce myocardial infarct scar in swine with subacute ischemic cardiomyopathy. J Am Heart Assoc 2015; 4:jah3883. [PMID: 25827134 PMCID: PMC4579962 DOI: 10.1161/jaha.114.001464] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Growth hormone-releasing hormone agonists (GHRH-As) stimulate cardiac repair following myocardial infarction (MI) in rats through the activation of the GHRH signaling pathway within the heart. We tested the hypothesis that the administration of GHRH-As prevents ventricular remodeling in a swine subacute MI model. METHODS AND RESULTS Twelve female Yorkshire swine (25 to 30 kg) underwent transient occlusion of the left anterior descending coronary artery (MI). Two weeks post MI, swine were randomized to receive injections of either 30 μg/kg GHRH-A (MR-409) (GHRH-A group; n=6) or vehicle (placebo group; n=6). Cardiac magnetic resonance imaging and pressure-volume loops were obtained at multiple time points. Infarct, border, and remote (noninfarcted) zones were assessed for GHRH receptor by immunohistochemistry. Four weeks of GHRH-A treatment resulted in reduced scar mass (GHRH-A: -21.9 ± 6.42%; P=0.02; placebo: 10.9 ± 5.88%; P=0.25; 2-way ANOVA; P=0.003), and scar size (percentage of left ventricular mass) (GHRH-A: -38.38 ± 4.63; P=0.0002; placebo: -14.56 ± 6.92; P=0.16; 2-way ANOVA; P=0.02). This was accompanied by improved diastolic strain. Unlike in rats, this reduced infarct size in swine was not accompanied by improved cardiac function as measured by serial hemodynamic pressure-volume analysis. GHRH receptors were abundant in cardiac tissue, with a greater density in the border zone of the GHRH-A group compared with the placebo group. CONCLUSIONS Daily subcutaneous administration of GHRH-A is feasible and safe in a large animal model of subacute ischemic cardiomyopathy. Furthermore, GHRH-A therapy significantly reduced infarct size and improved diastolic strain, suggesting a local activation of the GHRH pathway leading to the reparative process.
Collapse
Affiliation(s)
- Luiza L Bagno
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL (L.L.B., R.M.K.T., V.Y.S., S.G., V.K., A.W., B.W., C.P., W.B., J.R., D.V., M.R., J.M.H.)
| | - Rosemeire M Kanashiro-Takeuchi
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL (L.L.B., R.M.K.T., V.Y.S., S.G., V.K., A.W., B.W., C.P., W.B., J.R., D.V., M.R., J.M.H.) Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL (R.M.K.T., C.P., J.M.H.)
| | - Viky Y Suncion
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL (L.L.B., R.M.K.T., V.Y.S., S.G., V.K., A.W., B.W., C.P., W.B., J.R., D.V., M.R., J.M.H.)
| | - Samuel Golpanian
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL (L.L.B., R.M.K.T., V.Y.S., S.G., V.K., A.W., B.W., C.P., W.B., J.R., D.V., M.R., J.M.H.)
| | - Vasileios Karantalis
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL (L.L.B., R.M.K.T., V.Y.S., S.G., V.K., A.W., B.W., C.P., W.B., J.R., D.V., M.R., J.M.H.)
| | - Ariel Wolf
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL (L.L.B., R.M.K.T., V.Y.S., S.G., V.K., A.W., B.W., C.P., W.B., J.R., D.V., M.R., J.M.H.)
| | - Bo Wang
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL (L.L.B., R.M.K.T., V.Y.S., S.G., V.K., A.W., B.W., C.P., W.B., J.R., D.V., M.R., J.M.H.)
| | - Courtney Premer
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL (L.L.B., R.M.K.T., V.Y.S., S.G., V.K., A.W., B.W., C.P., W.B., J.R., D.V., M.R., J.M.H.) Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL (R.M.K.T., C.P., J.M.H.)
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL (L.L.B., R.M.K.T., V.Y.S., S.G., V.K., A.W., B.W., C.P., W.B., J.R., D.V., M.R., J.M.H.)
| | - Jose Rodriguez
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL (L.L.B., R.M.K.T., V.Y.S., S.G., V.K., A.W., B.W., C.P., W.B., J.R., D.V., M.R., J.M.H.)
| | - David Valdes
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL (L.L.B., R.M.K.T., V.Y.S., S.G., V.K., A.W., B.W., C.P., W.B., J.R., D.V., M.R., J.M.H.)
| | - Marcos Rosado
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL (L.L.B., R.M.K.T., V.Y.S., S.G., V.K., A.W., B.W., C.P., W.B., J.R., D.V., M.R., J.M.H.)
| | - Norman L Block
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL (N.L.B., A.M., R.Z.C., W.S., A.V.S., J.M.H.) Bruce A. Carter Miami Veterans Affairs Healthcare System, Miami, FL (N.L.B., R.Z.C., W.S., A.V.S.)
| | | | - Azorides Morales
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL (N.L.B., A.M., R.Z.C., W.S., A.V.S., J.M.H.)
| | - Ren-Zhi Cai
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL (N.L.B., A.M., R.Z.C., W.S., A.V.S., J.M.H.) Bruce A. Carter Miami Veterans Affairs Healthcare System, Miami, FL (N.L.B., R.Z.C., W.S., A.V.S.)
| | - Wei Sha
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL (N.L.B., A.M., R.Z.C., W.S., A.V.S., J.M.H.) Bruce A. Carter Miami Veterans Affairs Healthcare System, Miami, FL (N.L.B., R.Z.C., W.S., A.V.S.)
| | - Andrew V Schally
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL (N.L.B., A.M., R.Z.C., W.S., A.V.S., J.M.H.) Bruce A. Carter Miami Veterans Affairs Healthcare System, Miami, FL (N.L.B., R.Z.C., W.S., A.V.S.)
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL (L.L.B., R.M.K.T., V.Y.S., S.G., V.K., A.W., B.W., C.P., W.B., J.R., D.V., M.R., J.M.H.) Department of Medicine, University of Miami Miller School of Medicine, Miami, FL (N.L.B., A.M., R.Z.C., W.S., A.V.S., J.M.H.) Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL (R.M.K.T., C.P., J.M.H.)
| |
Collapse
|
35
|
Antagonist of GH-releasing hormone receptors alleviates experimental ocular inflammation. Proc Natl Acad Sci U S A 2014; 111:18303-8. [PMID: 25489106 DOI: 10.1073/pnas.1421815112] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Disruptions in immunity and occurrence of inflammation cause many eye diseases. The growth hormone-releasing hormone-growth hormone-insulin-like growth factor-1 (GHRH-GH-IGF1) axis exerts regulatory effects on the immune system. Its involvement in ocular inflammation remains to be investigated. Here we studied this signaling in endotoxin-induced uveitis (EIU) generated by LPS. The increase in GHRH receptor (GHRH-R) protein levels was parallel to the increase in mRNA levels of pituitary-specific transcription factor-1, GHRH-R splice variant 1, GHRH, and GH following LPS insult. Elevation of GHRH-R and GH receptor was localized on the epithelium of the iris and ciliary body, and GHRH-R was confined to the infiltrating macrophages and leukocytes in aqueous humor but not to those in stroma. Treatment with GHRH-R antagonist decreased LPS-stimulated surges of GH and IGF1 in aqueous humor and alleviated inflammation by reducing the infiltration of macrophages and leukocytes and the production of TNF-α, IL-1β, and monocyte chemotactic protein-1. Our results indicate that inflammation in the iris and ciliary body involves the activation of GHRH signaling, which affects the recruitment of immune cells and the production of proinflammatory mediators that contribute to EIU pathogenesis. Moreover, the results suggest that GHRH-R antagonists are potential therapeutic agents for the treatment of acute ocular inflammation.
Collapse
|
36
|
Agonists of growth hormone-releasing hormone stimulate self-renewal of cardiac stem cells and promote their survival. Proc Natl Acad Sci U S A 2014; 111:17260-5. [PMID: 25404316 DOI: 10.1073/pnas.1420375111] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The beneficial effects of agonists of growth hormone-releasing hormone receptor (GHRH-R) in heart failure models are associated with an increase in the number of ckit(+) cardiac stem cells (CSCs). The goal of the present study was to determine the presence of GHRH-R in CSCs, the effect of GHRH-R agonists on their proliferation and survival, and the mechanisms involved. We investigated the expression of GHRH-R in CSCs of different species and the effect of GHRH-R agonists on their cell proliferation and survival. GHRH-R is expressed in ckit(+) CSCs isolated from mouse, rat, and pig. Treatment of porcine CSCs with the GHRH-R agonist JI-38 significantly increased the rate of cell division. Similar results were observed with other GHRH-R agonists, MR-356 and MR-409. JI-38 exerted a protective effect on survival of porcine CSCs under conditions of oxidative stress induced by exposure to hydrogen peroxide. Treatment with JI-38 before exposure to peroxide significantly reduced cell death. A similar effect was observed with MR-356. Addition of GHRH-R agonists to porcine CSCs induced activation of ERK and AKT pathways as determined by increased expression of phospho-ERK and phospho-AKT. Inhibitors of ERK and AKT pathways completely reversed the effect of GHRH-R agonists on CSC proliferation. Our findings extend the observations of the expression of GHRH-R by CSCs and demonstrate that GHRH-R agonists have a direct effect on proliferation and survival of CSCs. These results support the therapeutic use of GHRH-R agonists for stimulating endogenous mechanisms for myocardial repair or for preconditioning of stem cells before transplantation.
Collapse
|
37
|
Kiaris H, Block NL, Papavassiliou AG, Schally AV. GHRH and wound healing. Commun Integr Biol 2014. [DOI: 10.4161/cib.14002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
38
|
Yuan MJ, Huang H, Quan L, Tang YH, Wang X, Jiang H, Huang CX. Expression of ghrelin and its receptor in rats after coronary artery ligation. ACTA ACUST UNITED AC 2014; 192-193:1-5. [PMID: 25058156 DOI: 10.1016/j.regpep.2014.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 06/18/2014] [Accepted: 07/14/2014] [Indexed: 11/26/2022]
Abstract
Ghrelin is a novel growth hormone-releasing peptide, which has been shown to exert beneficial effects on cardiac function and ventricular remodeling. The present study aimed to investigate the expression of ghrelin and the growth hormone (GH) secretagogue receptor 1a (GHSR-1a), and the association with cardiac remodeling in rats with myocardial infarction (MI). Twenty-four hours after ligation of the anterior descending artery (LAD), adult male Sprague-Dawley rats were randomized to 3 d, 7 d and 28 d group. Sham animals underwent thoracotomy and pericardiotomy, but not LAD ligation. Expression of both ghrelin and GHSR-1a was assessed by means of immunohistochemistry and real-time PCR. Plasma ghrelin levels were measured by ELISA kit. In addition, cardiac remodeling was assessed by echocardiographic and hemodynamic measurements. Plasma and cardiac expression of ghrelin decreased on days 3, 7 and 28 compared with the sham group (P<0.05). In contrast the GHSR-1a mRNA levels increased during the same days (P<0.05). Decreased positive immunoreaction for ghrelin and increased positive GHSR-1a were also observed in the infarcted heart. Interestingly, plasma ghrelin correlated negatively with left ventricular end-diastolic pressure (r=-0.59, P=0.002) and left ventricular end-diastolic dimension (r=-0.73, P<0.01). The ghrelin system may play an important role regulating cardiac remodeling after MI and present as a potential significant target for pharmacological modulation and treating cardiac remodeling.
Collapse
Affiliation(s)
- Ming-Jie Yuan
- Dept of Cardiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei Province, PR China
| | - He Huang
- Dept of Cardiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei Province, PR China
| | - Li Quan
- Dept of Cardiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei Province, PR China
| | - Yan-Hong Tang
- Dept of Cardiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei Province, PR China
| | - Xi Wang
- Dept of Cardiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei Province, PR China
| | - Hong Jiang
- Dept of Cardiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei Province, PR China
| | - Cong-Xin Huang
- Dept of Cardiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei Province, PR China.
| |
Collapse
|
39
|
Hua K, Nie Y, Hou J, Zheng Z, Hu S. Cardiomyocyte cytokinesis score: a potential method for cardiomyocyte proliferation. Cell Biol Int 2014; 38:1032-40. [PMID: 24800698 DOI: 10.1002/cbin.10307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 03/31/2014] [Indexed: 01/05/2023]
Abstract
One of the most important indicators of myocardial regeneration is cardiomyocyte proliferation. However, it is difficult to distinguish cardiomyocytes in the regenerating stage from binucleated or multinucleated myocytes by conventional morphometric techniques. As cell cycle progression (CCP) scores have been successfully applied to the evaluation of the proliferation of cancer cells, we sought to establish a multi-gene score to evaluate cardiomyocyte proliferation in this study. Given the disturbances of nuclear division without cell division that occurs in cardiomyocytes, ten cytokinesis-correlated genes (Anln, Aurkb, Cenpa, Kif4, Kif23, Prc1, RhoA, Spin1, TACC2, and CDC42) were chosen to establish the cardiomyocyte cytokinesis score (CC-Score). The expression levels of these genes in H9C2 rat cardiomyoblast cells, the proliferation of which were stimulated or inhibited, were detected using qRT-PCR. To confirm the feasibility of the CC-Score system, four conventional methods for evaluating cardiomyocyte proliferation, including the MTT assay, BrdU assay, immunofluorescence, and flow cytometry analysis, were used in each group. The results of the CC-Score in the assessment of the proliferation of H9C2 cells were consistent with those of four commonly used proliferative assay methods. We conclude that the CC-Score can be used to assess the proliferation status of H9C2 cells, and suggest that the CC-Score may be a potential method for the assessment of cardiomyocyte proliferation in myocardial regeneration. However, validation studies utilizing primary cultured rat cardiomyocytes and heart tissue are warranted.
Collapse
Affiliation(s)
- Kun Hua
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | | | | | | | | |
Collapse
|
40
|
Mezey G, Treszl A, Schally AV, Block NL, Vízkeleti L, Juhász A, Klekner Á, Nagy J, Balázs M, Halmos G, Bognár L. Prognosis in human glioblastoma based on expression of ligand growth hormone-releasing hormone, pituitary-type growth hormone-releasing hormone receptor, its splicing variant receptors, EGF receptor and PTEN genes. J Cancer Res Clin Oncol 2014; 140:1641-9. [DOI: 10.1007/s00432-014-1716-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Accepted: 05/14/2014] [Indexed: 12/13/2022]
|
41
|
Ginsenoside rb1 reduces isoproterenol-induced cardiomyocytes apoptosis in vitro and in vivo. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:454389. [PMID: 24363767 PMCID: PMC3852087 DOI: 10.1155/2013/454389] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 09/30/2013] [Accepted: 10/07/2013] [Indexed: 01/08/2023]
Abstract
Cardiomyocytes apoptosis can lead to heart failure. Conventional and alternative drugs, such as Chinese herbal remedies, have been developed to target cardiomyoblast cells apoptosis. In this study, we investigated the effects of ginsenoside Rb1 (Rb1), an active compound, which is isolated from Notoginseng and Ginseng on isoproterenol-(ISO-) induced apoptosis in rat cardiomyocytes and its mechanism in vivo and in vitro. Rb1 reduced the ISO-induced apoptosis in rat cardiomyocytes and H9c2 cells. The effect of Rb1 was significantly suppressed by H89 (inhibitor for PKA), but not by C-1 (inhibitor for PKC). Based on in-cell blot analysis, the ISO-induced PKA and PKC expressions were decreased by Rb1, which was inhibited by H89, but not by C-1. The expressions of caspase-3 and caspase-9 were decreased after treatment with both ISO and Rb1, but with no change for caspase-8. Our results indicated that Rb1 reducing ISO-induced rat cardiomyocytes apoptosis may be involved in PKA and caspase-9 pathways.
Collapse
|
42
|
Redox balance and cardioprotection. Basic Res Cardiol 2013; 108:392. [DOI: 10.1007/s00395-013-0392-7] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 09/24/2013] [Accepted: 10/14/2013] [Indexed: 12/11/2022]
|
43
|
Katsushima Y, Sato T, Yamada C, Ito M, Suzuki Y, Ogawa E, Sukegawa I, Sukegawa J, Fukunaga K, Yanagisawa T. Interaction of PICK1 with C-terminus of growth hormone-releasing hormone receptor (GHRHR) modulates trafficking and signal transduction of human GHRHR. J Pharmacol Sci 2013; 122:193-204. [PMID: 23823934 DOI: 10.1254/jphs.12287fp] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Release of growth hormone (GH) from the somatotroph is regulated by binding GH-releasing hormone (GHRH) to its cognate receptor (GHRHR), one of the members of the G protein-coupled receptor (GPCR) superfamily. Proteins bound to the carboxy (C)-terminus of GPCR have been reported to regulate intracellular trafficking and function of the receptor; however, no functionally significant protein associated with GHRHR has been reported. We have identified a protein interacting with C-kinase 1 (PICK1) as a binding partner of GHRHR. In vitro binding assay revealed the PDZ-domain of PICK1 and the last four amino acid residues of GHRHR were prerequisite for the interaction. Further, in vivo association of these proteins was confirmed. Immunostaining data of a stable cell line expressing GHRHR with or without PICK1 suggested the C-terminus of GHRHR promoted cell surface expression of GHRHR and PICK1 affected the kinetics of the cell surface expression of GHRHR. Furthermore, cAMP production assay showed the C-terminus of GHRHR is involved in the regulation of receptor activation, and the interaction of GHRHR with PICK1 may influence intensities of the signal response after ligand stimulation. Thus, the interaction of the C-terminus of GHRHR with PICK1 has a profound role in regulating the trafficking and the signaling of GHRHR. [Supplementary Figure: available only at http://dx.doi.org/10.1254/jphs.12287FP].
Collapse
Affiliation(s)
- Yuriko Katsushima
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
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: 2.2] [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
|
45
|
Ceylan-Isik AF, Dong M, Zhang Y, Dong F, Turdi S, Nair S, Yanagisawa M, Ren J. Cardiomyocyte-specific deletion of endothelin receptor A rescues aging-associated cardiac hypertrophy and contractile dysfunction: role of autophagy. Basic Res Cardiol 2013; 108:335. [PMID: 23381122 DOI: 10.1007/s00395-013-0335-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 12/27/2012] [Accepted: 01/24/2013] [Indexed: 11/25/2022]
Abstract
Cardiac aging is manifested as cardiac remodeling and contractile dysfunction although precise mechanisms remain elusive. This study was designed to examine the role of endothelin-1 (ET-1) in aging-associated myocardial morphological and contractile defects. Echocardiographic and cardiomyocyte contractile properties were evaluated in young (5-6 months) and old (26-28 months) C57BL/6 wild-type and cardiomyocyte-specific ET(A) receptor knockout (ETAKO) mice. Cardiac ROS production and histology were examined. Our data revealed that ETAKO mice displayed an improved survival. Aging increased plasma levels of ET-1 and Ang II, compromised cardiac function (fractional shortening, cardiomyocyte peak shortening, maximal velocity of shortening/relengthening and prolonged relengthening) and intracellular Ca(2+) handling (reduced intracellular Ca(2+) release and decay), the effects of which with the exception of ET-1 and Ang II levels was improved by ETAKO. Histological examination displayed cardiomyocyte hypertrophy and interstitial fibrosis associated with cardiac remodeling in aged C57 mice, which were alleviated in ETAKO mice. Aging promoted ROS generation, protein damage, ER stress, upregulated GATA4, ANP, NFATc3 and the autophagosome cargo protein p62, downregulated intracellular Ca(2+) regulatory proteins SERCA2a and phospholamban as well as the autophagic markers Beclin-1, Atg7, Atg5 and LC3BII, which were ablated by ETAKO. ET-1 triggered a decrease in autophagy and increased hypertrophic markers in vitro, the effect of which were reversed by the ET(A) receptor antagonist BQ123 and the autophagy inducer rapamycin. Antagonism of ET(A), but not ET(B) receptor, rescued cardiac aging, which was negated by autophagy inhibition. Taken together, our data suggest that cardiac ET(A) receptor ablation protects against aging-associated myocardial remodeling and contractile dysfunction possibly through autophagy regulation.
Collapse
Affiliation(s)
- Asli F Ceylan-Isik
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Down-regulation of replication factor C-40 (RFC40) causes chromosomal missegregation in neonatal and hypertrophic adult rat cardiac myocytes. PLoS One 2012; 7:e39009. [PMID: 22720015 PMCID: PMC3375256 DOI: 10.1371/journal.pone.0039009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 05/15/2012] [Indexed: 11/25/2022] Open
Abstract
Background Adult mammalian cardiac myocytes are generally assumed to be terminally differentiated; nonetheless, a small fraction of cardiac myocytes have been shown to replicate during ventricular remodeling. However, the expression of Replication Factor C (RFC; RFC140/40/38/37/36) and DNA polymerase δ (Pol δ) proteins, which are required for DNA synthesis and cell proliferation, in the adult normal and hypertrophied hearts has been rarely studied. Methods We performed qRT-PCR and Western blot analysis to determine the levels of RFC and Pol δ message and proteins in the adult normal cardiac myocytes and cardiac fibroblasts, as well as in adult normal and pulmonary arterial hypertension induced right ventricular hypertrophied hearts. Immunohistochemical analyses were performed to determine the localization of the re-expressed DNA replication and cell cycle proteins in adult normal (control) and hypertrophied right ventricle. We determined right ventricular cardiac myocyte polyploidy and chromosomal missegregation/aneuploidy using Fluorescent in situ hybridization (FISH) for rat chromosome 12. Results RFC40-mRNA and protein was undetectable, whereas Pol δ message was detectable in the cardiac myocytes isolated from control adult hearts. Although RFC40 and Pol δ message and protein significantly increased in hypertrophied hearts as compared to the control hearts; however, this increase was marginal as compared to the fetal hearts. Immunohistochemical analyses revealed that in addition to RFC40, proliferative and mitotic markers such as cyclin A, phospho-Aurora A/B/C kinase and phospho-histone 3 were also re-expressed/up-regulated simultaneously in the cardiac myocytes. Interestingly, FISH analyses demonstrated cardiac myocytes polyploidy and chromosomal missegregation/aneuploidy in these hearts. Knock-down of endogenous RFC40 caused chromosomal missegregation/aneuploidy and decrease in the rat neonatal cardiac myocyte numbers. Conclusion Our novel findings suggest that transcription of RFC40 is suppressed in the normal adult cardiac myocytes and its insufficient re-expression may be responsible for causing chromosomal missegregation/aneuploidy and in cardiac myocytes during right ventricular hypertrophy.
Collapse
|
47
|
Dioufa N, Farmaki E, Schally AV, Kiaris H, Vlahodimitropoulos D, Papavassiliou AG, Kittas C, Block NL, Chatzistamou I. Growth hormone-releasing hormone receptor splice variant 1 is frequently expressed in oral squamous cell carcinomas. Discov Oncol 2012; 3:172-80. [PMID: 22441816 DOI: 10.1007/s12672-012-0108-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Accepted: 03/12/2012] [Indexed: 01/03/2023] Open
Abstract
The expression of growth hormone-releasing hormone (GHRH) splice variant 1 (SV1) receptor in neoplastic lesions of the oral cavity was assessed. The sensitivity of HaCaT keratinocytes to GHRH analogs was also evaluated. Thirty-three benign precancerous oral lesions and 27 squamous cell carcinomas of the oral cavity were evaluated by immunohistochemistry for SV1 expression. SV1 expression in HaCaT keratinocytes was assessed by western blot. HaCaT proliferation was evaluated by cell counting. Anti-SV1 immunoreactivity was detected in only 9% (three of 33) precancerous lesions (one hyperplasia and two dysplasias), while 44% (12 of 27) carcinomas were positive for SV1 (p<0.002). GHRH(1-29)NH(2) and GHRH agonist JI-38 stimulated HaCaT proliferation in vitro, and this effect was blocked by GHRH antagonists. These results indicate that SV1 expression may be associated with the transition of precancerous lesions to carcinomas of the oral epithelium. GHRH antagonists may be useful for the management of the disease.
Collapse
Affiliation(s)
- Nikolina Dioufa
- Department of Biological Chemistry, University of Athens Medical School, M. Asias 75, 115 27 Athens, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Agonist of growth hormone-releasing hormone reduces pneumolysin-induced pulmonary permeability edema. Proc Natl Acad Sci U S A 2012; 109:2084-9. [PMID: 22308467 DOI: 10.1073/pnas.1121075109] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Aggressive treatment with antibiotics in patients infected with Streptococcus pneumoniae induces release of the bacterial virulence factor pneumolysin (PLY). Days after lungs are sterile, this pore-forming toxin can still induce pulmonary permeability edema in patients, characterized by alveolar/capillary barrier dysfunction and impaired alveolar liquid clearance (ALC). ALC is mainly regulated through Na(+) transport by the apically expressed epithelial sodium channel (ENaC) and the basolaterally expressed Na(+)/K(+)-ATPase in type II alveolar epithelial cells. Because no standard treatment is currently available to treat permeability edema, the search for novel therapeutic candidates is of high priority. We detected mRNA expression for the active receptor splice variant SV1 of the hypothalamic polypeptide growth hormone-releasing hormone (GHRH), as well as for GHRH itself, in human lung microvascular endothelial cells (HL-MVEC). Therefore, we have evaluated the effect of the GHRH agonist JI-34 on PLY-induced barrier and ALC dysfunction. JI-34 blunts PLY-mediated endothelial hyperpermeability in monolayers of HL-MVEC, in a cAMP-dependent manner, by means of reducing the phosphorylation of myosin light chain and vascular endothelial (VE)-cadherin. In human airway epithelial H441 cells, PLY significantly impairs Na(+) uptake, but JI-34 restores it to basal levels by means of increasing cAMP levels. Intratracheal instillation of PLY into C57BL6 mice causes pulmonary alveolar epithelial and endothelial hyperpermeability as well as edema formation, all of which are blunted by JI-34. These findings point toward a protective role of the GHRH signaling pathway in PLY-induced permeability edema.
Collapse
|
49
|
Activation of growth hormone releasing hormone (GHRH) receptor stimulates cardiac reverse remodeling after myocardial infarction (MI). Proc Natl Acad Sci U S A 2011; 109:559-63. [PMID: 22203988 DOI: 10.1073/pnas.1119203109] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Both cardiac myocytes and cardiac stem cells (CSCs) express the receptor of growth hormone releasing hormone (GHRH), activation of which improves injury responses after myocardial infarction (MI). Here we show that a GHRH-agonist (GHRH-A; JI-38) reverses ventricular remodeling and enhances functional recovery in the setting of chronic MI. This response is mediated entirely by activation of GHRH receptor (GHRHR), as demonstrated by the use of a highly selective GHRH antagonist (MIA-602). One month after MI, animals were randomly assigned to receive: placebo, GHRH-A (JI-38), rat recombinant GH, MIA-602, or a combination of GHRH-A and MIA-602, for a 4-wk period. We assessed cardiac performance and hemodynamics by using echocardiography and micromanometry derived pressure-volume loops. Morphometric measurements were carried out to determine MI size and capillary density, and the expression of GHRHR was assessed by immunofluorescence and quantitative RT-PCR. GHRH-A markedly improved cardiac function as shown by echocardiographic and hemodynamic parameters. MI size was substantially reduced, whereas myocyte and nonmyocyte mitosis was markedly increased by GHRH-A. These effects occurred without increases in circulating levels of growth hormone and insulin-like growth factor I and were, at least partially, nullified by GHRH antagonism, confirming a receptor-mediated mechanism. GHRH-A stimulated CSCs proliferation ex vivo, in a manner offset by MIA-602. Collectively, our findings reveal the importance of the GHRH signaling pathway within the heart. Therapy with GHRH-A although initiated 1 mo after MI substantially improved cardiac performance and reduced infarct size, suggesting a regenerative process. Therefore, activation of GHRHR provides a unique therapeutic approach to reverse remodeling after MI.
Collapse
|
50
|
Kiaris H, Chatzistamou I, Papavassiliou AG, Schally AV. Growth hormone-releasing hormone: not only a neurohormone. Trends Endocrinol Metab 2011; 22:311-7. [PMID: 21530304 DOI: 10.1016/j.tem.2011.03.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 03/14/2011] [Accepted: 03/16/2011] [Indexed: 11/25/2022]
Abstract
Growth hormone-releasing hormone (GHRH) is mostly thought to act by stimulating the production and release of growth hormone from the pituitary. However, this neuropeptide emerges as a rather pleiotropic hormone in view of the identification of various extrapituitary sources for GHRH production, as well as the demonstration of a direct action of GHRH on several tissues other than the pituitary. Non-pituitary GHRH has a wide spectrum of activity, exemplified by its ability to modulate cell proliferation, especially in malignant tissues, to regulate differentiation of some cell types, and to promote healing of skin wounds. These findings extend the role of GHRH and its analogs beyond its accepted regulation of somatotropic activity and indicate new possibilities for therapeutic intervention.
Collapse
Affiliation(s)
- Hippokratis Kiaris
- Department of Biochemistry, University of Athens Medical School, Mikras Asias 75, 11527 Athens, Greece.
| | | | | | | |
Collapse
|