1
|
Ferreira‐Marques M, Carvalho A, Franco AC, Leal A, Botelho M, Carmo‐Silva S, Águas R, Cortes L, Lucas V, Real AC, López‐Otín C, Nissan X, de Almeida LP, Cavadas C, Aveleira CA. Ghrelin delays premature aging in Hutchinson-Gilford progeria syndrome. Aging Cell 2023; 22:e13983. [PMID: 37858983 PMCID: PMC10726901 DOI: 10.1111/acel.13983] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 10/21/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare and fatal genetic condition that arises from a single nucleotide alteration in the LMNA gene, leading to the production of a defective lamin A protein known as progerin. The accumulation of progerin accelerates the onset of a dramatic premature aging phenotype in children with HGPS, characterized by low body weight, lipodystrophy, metabolic dysfunction, skin, and musculoskeletal age-related dysfunctions. In most cases, these children die of age-related cardiovascular dysfunction by their early teenage years. The absence of effective treatments for HGPS underscores the critical need to explore novel safe therapeutic strategies. In this study, we show that treatment with the hormone ghrelin increases autophagy, decreases progerin levels, and alleviates other cellular hallmarks of premature aging in human HGPS fibroblasts. Additionally, using a HGPS mouse model (LmnaG609G/G609G mice), we demonstrate that ghrelin administration effectively rescues molecular and histopathological progeroid features, prevents progressive weight loss in later stages, reverses the lipodystrophic phenotype, and extends lifespan of these short-lived mice. Therefore, our findings uncover the potential of modulating ghrelin signaling offers new treatment targets and translational approaches that may improve outcomes and enhance the quality of life for patients with HGPS and other age-related pathologies.
Collapse
Affiliation(s)
- Marisa Ferreira‐Marques
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- Faculty of PharmacyUniversity of CoimbraCoimbraPortugal
| | - André Carvalho
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Ana Catarina Franco
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- Faculty of PharmacyUniversity of CoimbraCoimbraPortugal
| | - Ana Leal
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Mariana Botelho
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Sara Carmo‐Silva
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
| | - Rodolfo Águas
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Luísa Cortes
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
| | - Vasco Lucas
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Ana Carolina Real
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Carlos López‐Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de OncologíaUniversidad de OviedoOviedoSpain
| | - Xavier Nissan
- CECS, I‐StemCorbeil‐EssonnesFrance
- INSERM U861, I‐StemCorbeil‐EssonnesFrance
- UEVE U861, I‐StemCorbeil‐EssonnesFrance
| | - Luís Pereira de Almeida
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- Faculty of PharmacyUniversity of CoimbraCoimbraPortugal
| | - Cláudia Cavadas
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- Faculty of PharmacyUniversity of CoimbraCoimbraPortugal
| | - Célia A. Aveleira
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- MIA‐Portugal – Multidisciplinar Institute of AgeingUniversity of CoimbraCoimbraPortugal
| |
Collapse
|
2
|
Bi S, Tang J, Zhang L, Huang L, Chen J, Wang Z, Chen D, Du L. Fine particulate matter reduces the pluripotency and proliferation of human embryonic stem cells through ROS induced AKT and ERK signaling pathway. Reprod Toxicol 2020; 96:231-240. [PMID: 32745510 DOI: 10.1016/j.reprotox.2020.07.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/23/2020] [Accepted: 07/27/2020] [Indexed: 02/08/2023]
Abstract
Epidemiological investigations have found that air fine particulate matter (PM) exposure not only causes respiratory and cardiovascular diseases in adults and children, but also affects embryonic development during pregnancy, leading to poor pregnancy outcomes. However, its exact molecular mechanism is still unclear. In this study, human embryonic stem cells (hESCs) were treated with PM at different concentrations then the morphology and proliferation capacity were measured. The mRNA and protein expression of NANOG and OCT4 were detected using quantitative PCR, immunofluorescence, western blotting, and flow cytometry. Reactive oxygen species (ROS) generation and AKT/ERK activation were also measured. Meanwhile, changes in ROS, the expression of NANOG, OCT4, and the AKT/ERK pathways were measured in the hESCs with or without pretreatment of ROS scavenger N-acetylcysteine (NAC) prior to PM exposure. After PM exposure, the proliferation capacity and expression of OCT4 and NANOG at the mRNA and protein levels were downregulated. The ROS level in the hESCs increased after PM exposure, but this increase in ROS was attenuated by pretreatment with NAC. Further analysis showed that the levels of phosphorylated AKT and ERK increased after PM exposure. After pretreatment with NAC, the phosphorylation levels of AKT and ERK, which are crucial for regulating the proliferation, pluripotency, and differentiation of hESC, were significantly attenuated compared with the non-NAC pretreated exposure group. These results suggest that PM exposure may reduce the proliferation and pluripotency of hESC through ROS-mediated AKT/ERK pathways, thereby affecting the long-term development of embryos.
Collapse
Affiliation(s)
- Shilei Bi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Jingman Tang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Lizi Zhang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Lijun Huang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Jingsi Chen
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China; Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, PR China; Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, PR China
| | - Zhijian Wang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Dunjin Chen
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China; Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, PR China; Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, PR China.
| | - Lili Du
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China; Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, PR China; Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, PR China.
| |
Collapse
|
3
|
Miao H, Pan H, Wang L, Yang H, Zhu H, Gong F. Ghrelin Promotes Proliferation and Inhibits Differentiation of 3T3-L1 and Human Primary Preadipocytes. Front Physiol 2019; 10:1296. [PMID: 31681009 PMCID: PMC6798085 DOI: 10.3389/fphys.2019.01296] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/27/2019] [Indexed: 12/11/2022] Open
Abstract
Objective Ghrelin is a 28-amino-acid peptide that regulates energy hemostasis, glucose and lipid metabolism. We aimed to explore the effects of ghrelin on the proliferation and differentiation of 3T3-L1 and human primary preadipocytes. Methods 3-(4,5-Dimethylthiazol-2-yl) 2,5-diphenyl tetrazolium bromide (MTT) spectrophotometry, Oil Red O staining, intracellular glycerol-3-phosphate dehydrogenase (G-3-PDH) assays and semiquantitative reverse transcription polymerase chain reaction were used to investigate the action of ghrelin. Results Ghrelin (0.01–1000 ng/ml) significantly increased the numbers of 3T3-L1 cells, and the maximum stimulatory effect was observed with the 10 ng/ml ghrelin treatment for 24 h (p < 0.05). Ghrelin also promoted the proliferation of human primary preadipocytes from 24 h (p < 0.05) to 48 h (p < 0.05) at a concentration of 1000 ng/ml. Further investigation showed that IGF-1 levels were notably increased in ghrelin-treated 3T3-L1 and human preadipocytes, and IGF-1 antibody was capable to attenuate this stimulatory action of ghrelin (all p < 0.05). Additionally, ghrelin significantly suppressed the differentiation of 3T3-L1 and human primary preadipocytes; 10 ng/ml ghrelin notably downregulated G-3-PDH activities in 3T3-L1 cells on day 3 and in human cells from days 4 to 12 following differentiation (all p < 0.05), and the intracellular lipoprotein lipase mRNA levels were lower than that of the controls (p < 0.05). Further investigation showed that the mRNA levels of peroxisome proliferator-activated receptor γ2 (PPARγ2) and CCAAT/enhancer binding protein α (C/EBPs) were also suppressed in ghrelin-treated human differentiating adipocytes. Conclusion Ghrelin promotes the proliferation of 3T3-L1 and human primary preadipocytes by increasing the expression of IGF-1. Ghrelin inhibits murine and human adipocyte differentiation by downregulating PPARγ2 and C/EBPα levels, consequently leading to decreased lipid accumulation and lipogenic enzymes expression.
Collapse
Affiliation(s)
- Hui Miao
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui Pan
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Linjie Wang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongbo Yang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huijuan Zhu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fengying Gong
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Liang ZT, Li J, Rong R, Wang YJ, Xiao LG, Yang GT, Zhang HQ. Ghrelin up-regulates cartilage-specific genes via the ERK/STAT3 pathway in chondrocytes of patients with adolescent idiopathic scoliosis. Biochem Biophys Res Commun 2019; 518:259-265. [PMID: 31421834 DOI: 10.1016/j.bbrc.2019.08.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/08/2019] [Indexed: 01/19/2023]
Abstract
Adolescent idiopathic scoliosis (AIS) is a severe spinal deformity that often occurs during puberty. The occurrence of AIS is suggested to be related to abnormal development of cartilage. Our previous study found increased serum ghrelin levels in AIS patients that may linked to the development of AIS. However, whether ghrelin affects cartilage in AIS patients is unclear. We used quantitative real-time PCR (qRT-PCR) and immunohistochemistry to detect the expression of cartilage-specific genes and the ghrelin receptor, growth hormone secretagogue receptor (GHSR). The mRNA and protein levels of collagen II (COLII), SOX9, AGGRECAN (ACAN) and GHSR were higher in AIS patients than in controls. In addition, the protein levels of GHSR downstream signaling pathway members p-STAT3 (Ser727), and p-ERK1/2 were increased. Furthermore, we treated chondrocytes from AIS patients with 100 nM ghrelin, the cell proliferation assay and Western blotting showed that ghrelin promotes chondrocyte proliferation and enhances COLII, SOX9, ACAN, p-ERK1/2 and p-STAT3 expression, respectively. Interestingly, all these observed alterations were abolished by ghrelin + [D-Lys3]-GHRP-6 (a ghrelin receptor inhibitor) treatment. And after U0126 (an inhibitor of ERK1/2 phosphorylation) treatment, ERK1/2 and STAT3 (Ser727) phosphorylation was simultaneously suppressed indicating that ERK1/2 is an upstream pathway protein of STAT3 (Ser727). In conclusion, ghrelin plays an important role in upregulating cartilage-specific genes on AIS primary chondrocytes by activating ERK/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Zhuo-Tao Liang
- Department of Spine Surgery, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, China
| | - Jiong Li
- Department of Spine Surgery, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, China
| | - Rong- Rong
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yun-Jia Wang
- Department of Spine Surgery, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, China
| | - Li-Ge Xiao
- Department of Spine Surgery, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, China
| | - Guan-Teng Yang
- Department of Spine Surgery, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, China
| | - Hong-Qi Zhang
- Department of Spine Surgery, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, China.
| |
Collapse
|
5
|
Çetin E. Protective effect of ghrelin against tilmicosin-induced left ventricular dysfunction in rats. Can J Physiol Pharmacol 2019; 97:407-412. [DOI: 10.1139/cjpp-2018-0511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
This study was conducted to investigate the possible protective effects of ghrelin against tilmicosin-induced acute ventricular dysfunction in rats. Forty adult male Sprague Dawley rats were randomly divided into 4 equal groups: control, ghrelin, tilmicosin, and ghrelin + tilmicosin. The left ventricular structural and functional parameters together with cardiac biomarker levels were evaluated. The results showed that tilmicosin treatment alone significantly decreased the left ventricular fractional shortening, left ventricular ejection fraction, left ventricular stroke volume, and cardiac output when compared with control group. In addition, tilmicosin led to a significant increase in left ventricular internal dimension in systole and left ventricular fractional end-systolic volume. At the same time, serum lactate dehydrogenase, creatine kinase, and creatine kinase-myocardial B fraction levels were significantly increased in tilmicosin-treated group when compared with control group. However, ghrelin pretreatment significantly prevented the left ventricular internal dimension in systole, left ventricular fractional end-systolic volume, left ventricular stroke volume, left ventricular ejection fraction, left ventricular fractional shortening, and cardiac output changes caused by tilmicosin. Moreover, ghrelin pretreatment could reduce significantly serum lactate dehydrogenase, creatine kinase, and creatine kinase-myocardial B fraction levels. These data indicated that ghrelin treatment may provide a protective effect against tilmicosin-induced left ventricular systolic dysfunction.
Collapse
Affiliation(s)
- Ebru Çetin
- Department of Physiology, Faculty of Veterinary Medicine, Erciyes University, 38039, Kayseri, Turkey
- Department of Physiology, Faculty of Veterinary Medicine, Erciyes University, 38039, Kayseri, Turkey
| |
Collapse
|
6
|
Abdanipour A, Shahsavandi B, Alipour M, Feizi H. Ghrelin Upregulates Hoxb4 Gene Expression in Rat Bone Marrow Stromal Cells. CELL JOURNAL 2018; 20:183-187. [PMID: 29633595 PMCID: PMC5893289 DOI: 10.22074/cellj.2018.5164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 07/12/2017] [Indexed: 01/18/2023]
Abstract
OBJECTIVES Ghrelin is a peptide which has a proliferative and antiapoptotic effect in many cells including bone marrow stromal cells (BMSCs). Homeobox protein B4 (HOXB4) is a transcription factor involved in stem cell regeneration and survival. The aim of the study was to find out the efect of ghrelin on Hoxb4 expression in BMSCs. MATERIALS AND METHODS In this experimental study, rat BMSCs were cultivated in Dulbecco's Modified Eagle Medium (DMEM). Passage three BMSCs were treated with ghrelin 100 μM for 48 hours. Real-time polymerase chain reaction (PCR) was carried out from the untreated BMSCs (B), BMSCs treated with 125 μM H2O2 (BH), BMSCs treated with 100 μM ghrelin then 125 μM H2O2 (BGH) and BMSCs treated with 100 μM ghrelin (BG) groups. For immunofluorescence, cells were incubated with an anti-HOXB4 monoclonal antibody. Primary antibodies were visualized using the Fluorescein isothiocyanate (FITC) method. All data are presented as mean ± SEM and P<0.05 was considered as statistical significant. RESULTS Hoxb4 expression significantly increased in the BG compared with BH and BGH groups. Furthermore, 100 μM ghrelin, increased the mean of HOXB4 positive immunoreactive cells compared to the BH group. CONCLUSIONS Ghrelin probably enhances proliferation and viability of BMSCs through Hoxb4 upregulation. However, the signaling pathway and other biological outcomes of this effect should be elucidated in different stem cells.
Collapse
Affiliation(s)
- Alireza Abdanipour
- Department of Anatomical Sciences, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Behnaz Shahsavandi
- Department of Physiology and Pharmacology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohsen Alipour
- Department of Physiology and Pharmacology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hadi Feizi
- Department of Physiology and Pharmacology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
7
|
Ye N, Wang L, Dou Z, Huang J. Ghrelin accelerates the cartilagic differentiation of rabbit mesenchymal stem cells through the ERK1/2 pathway. Cytotechnology 2017; 70:415-421. [PMID: 29230632 DOI: 10.1007/s10616-017-0156-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/20/2017] [Indexed: 01/04/2023] Open
Abstract
Mesenchymal stem cells (MSCs) can differentiate into chondroblasts, adipocytes, or cartilage under appropriate stimulation. Identifying a mechanism triggering the differentiation of MSCs into cartilage may help develop novel therapeutic approaches for treating heterotopic ossification, the pathological formation of lamellar bone in soft tissue outside the skeleton that can lead to debilitating immobility. Ghrelin, an endogenous ligand for the growth hormone secretagogue receptor, stimulates growth hormone secretion, and has both orexigenic and adipogenic effects. This study sought to understand the potential involvement of the ERK1/2 signaling pathway in the ghrelin-induced growth of rat MSCs (rMSCs). We applied various concentrations of ghrelin to cultured rMSCs by observing the changes in the phosphorylation state of ERK1/2, p38, JNK as well as the type II collagen expression levels by western blot. The highest expression level for both type II collagen was obtained with 600 ng/mL ghrelin at 24 h. We found that the ghrelin-induced differentiation of rMSCs into cartilage was promoted primarily by the ERK1/2 pathway. Our study suggests that ghrelin induced differentiation of rMSCs into cartilage primarily through the ERK1/2 pathway.
Collapse
Affiliation(s)
- Nan Ye
- Department of Cervical Surgery, The Second Affiliated Hospital of Inner Mongolia Medical University, Muslims Camp Square Road No 1, Hohhot, China
| | - Lin Wang
- Department of Cervical Surgery, The Second Affiliated Hospital of Inner Mongolia Medical University, Muslims Camp Square Road No 1, Hohhot, China
| | - Zhe Dou
- Department of Cervical Surgery, The Second Affiliated Hospital of Inner Mongolia Medical University, Muslims Camp Square Road No 1, Hohhot, China
| | - Jian Huang
- Department of Cervical Surgery, The Second Affiliated Hospital of Inner Mongolia Medical University, Muslims Camp Square Road No 1, Hohhot, China.
| |
Collapse
|
8
|
Zhang F, Wang C, Lin J, Wang X. Oxidized low-density lipoprotein (ox-LDL) promotes cardiac differentiation of bone marrow mesenchymal stem cells via activating ERK1/2 signaling. Cardiovasc Ther 2017; 35. [PMID: 28880487 DOI: 10.1111/1755-5922.12305] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/16/2017] [Accepted: 09/02/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND/AIMS The differentiation efficiency of bone marrow mesenchymal stem cells (BM-MSCs) is low in vivo after transplantation. Therefore, it is necessary to look for effective reagents for enhancing cardiac differentiation of BM-MSCs. It has been reported that cardiac differentiation of stem cells depends on the activation of extracellular signal-regulated protein 1/2 (ERK1/2) signaling. Oxidized low-density lipoprotein (ox-LDL) is a potent reagent for ERK1/2 activation. This indicates that ox-LDL may be a potential reagent to stimulate cardiac differentiation of stem cells. In this study, we investigated the effect of ox-LDL on cardiac differentiation of BM-MSCs and its relationship with ERK1/2 signaling. METHODS BM-MSCs were isolated from mouse bone marrow, cultured in DMEM supplemented with 15% FBS, and passaged up to the 3rd passage. Following culture with 5 μg/mL ox-LDL for 3 weeks, the cardiac differentiation of the 3rd passage BM-MSCs was identified by immunostaining, Western blotting, and RT-PCR assays for measuring the expression of cardiac-specific markers. To further explore the role of ERK1/2 signaling in cardiac differentiation of BM-MSCs, we simultaneously exposed BM-MSCs to ERK1/2 inhibitor (U0126) and ox-LDL, and identified the cardiac differentiation again. RESULTS The expressions of cardiac-specific markers including α-cardiac actin, α-MHC, β-MHC, ANP, and BNP were markedly increased in BM-MSCs following treatment with ox-LDL (P < .05), which indicates a directional differentiation of BM-MSCs to cardiac cells. Further, ox-LDL could also activate ERK1/2 in BM-MSCs, and application of U0126 markedly inhibited ox-LDL-induced cardiac transformation of BM-MSCs. CONCLUSIONS Ox-LDL induces cardiac differentiation of BM-MSCs via activation of ERK1/2 signaling.
Collapse
Affiliation(s)
- Fenxi Zhang
- Stem Cell and Biotheraphy Technology Research Center, Xinxiang Medical University, Xinxiang, China
| | - Congrui Wang
- Stem Cell and Biotheraphy Technology Research Center, Xinxiang Medical University, Xinxiang, China
| | - Juntang Lin
- Stem Cell and Biotheraphy Technology Research Center, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Xianwei Wang
- Stem Cell and Biotheraphy Technology Research Center, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
9
|
Ghrelin protected neonatal rat cardiomyocyte against hypoxia/reoxygenation injury by inhibiting apoptosis through Akt-mTOR signal. Mol Biol Rep 2017; 44:219-226. [PMID: 28281036 DOI: 10.1007/s11033-017-4098-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 01/02/2017] [Indexed: 12/17/2022]
Abstract
Reducing reperfusion period myocardial cell damage is efficient to reduce myocardial ischemia-reperfusion injury. Ghrelin can increase myocardial contractility, improve heart failure caused by myocardial infarction. This study aimed to investigate the protective effect of Ghrelin on myocardial hypoxia/reoxygenation (H/R) injury of neonatal rat cardiomyocytes (NRCMs) and to explore the mechanisms. We isolated the NRCMs, established myocardial H/R model, blocked growth hormone secretagogue receptor (GHSR) by siRNA technique, examined cell activity by MTT and LDH assay, detected apoptosis by Hoechst 33258 staining and flow cytometry and determined the expression levels of apoptosis related proteins and signaling pathway proteins by western blot. We found that Ghrelin can significantly improve cell activity and decrease apoptosis after H/R, however this effect was abolished by GHSR-siRNA. In addition, we found that Ghrelin can significantly increase the expression of Bcl-2 but inhibit the level of Bax and caspase-3. Further mechanism study found that the phosphorylation level of signaling pathway protein Akt and mTOR in Ghrelin treated group were significantly higher than that in other groups. In conclusion, Ghrelin can reduce the H/R damage on NRCMs and inhibit the apoptosis by activating Akt-mTOR signaling pathway.
Collapse
|
10
|
Wei R, Yang J, Gao M, Wang H, Hou W, Mu Y, Chen G, Hong T. Infarcted cardiac microenvironment may hinder cardiac lineage differentiation of human embryonic stem cells. Cell Biol Int 2016; 40:1235-1246. [PMID: 27600481 DOI: 10.1002/cbin.10679] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 09/04/2016] [Indexed: 11/07/2022]
Abstract
Microenvironment regulates cell fate and function. In this study, we investigated the effects of the infarcted cardiac microenvironment on cardiac differentiation of human embryonic stem cells (hESCs). hESCs were intramyocardially transplanted into infarcted or uninjured rat hearts. After 4 weeks, mesodermal and cardiac lineage markers were detected by immunofluorescence. Cardiac function was assessed by echocardiography. hESCs were differentiated in vitro under hypoxic (5% O2 ), low-nutrient (5% FBS), or control condition. The numbers of beating clusters, proportions of cardiac troponin T (cTnT)-positive cells, and relative levels of cardiac-specific markers were determined. Results showed that in both uninjured and infarcted hearts, hESCs survived, underwent development, and formed intracardiac grafts, with a higher proportion in the uninjured hearts. However, cells that were double positive for human fetal liver kinase 1 (Flk1), a marker of cardiac progenitors, and human β-tubulin, a marker for labeling human cells, were found in the uninjured hearts but not in the infarcted hearts. hESC transplantation did not restore the cardiac function of acutely infarcted rats. In vitro, low FBS treatment was associated with fewer beating clusters, a lower proportion of cTnT-positive cells and lower levels of cardiac troponin I (cTnI) and α-myosin heavy chain (α-MHC) expression than those in the control. Conversely, hypoxia treatment was associated with a higher proportion of cTnT-positive cells and higher levels of cTnI expression. In conclusion, transplanted hESCs differentiate toward Flk1-positive cardiac progenitors in the uninjured but not infarcted hearts. The infarcted cardiac microenvironment recapitulated is unsuitable for cardiac differentiation of hESCs, likely due to nutrient deprivation.
Collapse
Affiliation(s)
- Rui Wei
- Department of Endocrinology and Metabolism, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.,Clinical Stem Cell Research Centre, Peking University Third Hospital, Beijing, China
| | - Jin Yang
- Department of Endocrinology and Metabolism, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.,Clinical Stem Cell Research Centre, Peking University Third Hospital, Beijing, China
| | - Meijuan Gao
- Department of Endocrinology and Metabolism, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.,Clinical Stem Cell Research Centre, Peking University Third Hospital, Beijing, China
| | - Haining Wang
- Department of Endocrinology and Metabolism, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.,Clinical Stem Cell Research Centre, Peking University Third Hospital, Beijing, China
| | - Wenfang Hou
- Department of Endocrinology and Metabolism, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.,Clinical Stem Cell Research Centre, Peking University Third Hospital, Beijing, China
| | - Yiming Mu
- Department of Endocrinology, Chinese PLA General Hospital, Beijing, China
| | - Guian Chen
- Clinical Stem Cell Research Centre, Peking University Third Hospital, Beijing, China
| | - Tianpei Hong
- Department of Endocrinology and Metabolism, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China. .,Clinical Stem Cell Research Centre, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
11
|
Li J, Zhang M, Wang M, Wang Z, Liu Y, Zhang W, Wang N. GHSR deficiency suppresses neointimal formation in injured mouse arteries. Biochem Biophys Res Commun 2016; 479:125-131. [PMID: 27404127 DOI: 10.1016/j.bbrc.2016.06.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 06/07/2016] [Indexed: 01/25/2023]
Abstract
Growth hormone secretagogue receptor (GHSR) is involved in appetite regulation and energy homeostasis. In the present study, we examined the role of GHSR in neointimal formation following vascular injury. In the mouse model of femoral artery wire injury, we found that vessel intima-to-media ratio was significantly reduced in GHSR deficiency (GHSR-/-) mice compared with that in wild-type mice. Immunohistochemical staining showed that the smooth muscle cell (SMCs) in the neointima were significantly decreased in the injured arteries of GHSR-/- mice which was associated with decreased SMC proliferation and migration. Furthermore, immunoblotting demonstrated that, in cultured rat aortic SMCs, small interfering RNA-mediated GHSR knockdown suppressed the activation of Akt and ERK1/2 signaling pathway. These findings suggested a novel role of GHSR in neointimal formation likely via promoting the proliferation and migration of SMCs involving Akt and ERK1/2 signaling. Therefore, GHSR may be a potential therapeutic target in restenosis and vascular remodeling.
Collapse
Affiliation(s)
- Jing Li
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Man Zhang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Mo Wang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Zhipeng Wang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yahan Liu
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Weizhen Zhang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Nanping Wang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China; The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
12
|
Szebényi K, Péntek A, Erdei Z, Várady G, Orbán TI, Sarkadi B, Apáti Á. Efficient generation of human embryonic stem cell-derived cardiac progenitors based on tissue-specific enhanced green fluorescence protein expression. Tissue Eng Part C Methods 2015; 21:35-45. [PMID: 24734786 DOI: 10.1089/ten.tec.2013.0646] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cardiac progenitor cells (CPCs) are committed to the cardiac lineage but retain their proliferative capacity before becoming quiescent mature cardiomyocytes (CMs). In medical therapy and research, the use of human pluripotent stem cell-derived CPCs would have several advantages compared with mature CMs, as the progenitors show better engraftment into existing heart tissues, and provide unique potential for cardiovascular developmental as well as for pharmacological studies. Here, we demonstrate that the CAG promoter-driven enhanced green fluorescence protein (EGFP) reporter system enables the identification and isolation of embryonic stem cell-derived CPCs. Tracing of CPCs during differentiation confirmed up-regulation of surface markers, previously described to identify cardiac precursors and early CMs. Isolated CPCs express cardiac lineage-specific transcripts, still have proliferating capacity, and can be re-aggregated into embryoid body-like structures (CAG-EGFP(high) rEBs). Expression of troponin T and NKX2.5 mRNA is up-regulated in long-term cultured CAG-EGFP(high) rEBs, in which more than 90% of the cells become Troponin I positive mature CMs. Moreover, about one third of the CAG-EGFP(high) rEBs show spontaneous contractions. The method described here provides a powerful tool to generate expandable cultures of pure human CPCs that can be used for exploring early markers of the cardiac lineage, as well as for drug screening or tissue engineering applications.
Collapse
Affiliation(s)
- Kornélia Szebényi
- 1 Institute of Molecular Pharmacology , Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | | | | | | | | | | | | |
Collapse
|
13
|
Ye N, Jiang D. Ghrelin accelerates the growth and osteogenic differentiation of rabbit mesenchymal stem cells through the ERK1/2 pathway. BMC Biotechnol 2015; 15:51. [PMID: 26054524 PMCID: PMC4460755 DOI: 10.1186/s12896-015-0176-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/29/2015] [Indexed: 02/07/2023] Open
Abstract
Background Mesenchymal stem cells (MSCs) can differentiate into chondroblasts, adipocytes, or osteoblasts under appropriate stimulation. Ghrelin, an endogenous ligand for the growth hormone secretagogue receptor (GHSR), stimulates growth hormone (GH) secretion, and has both orexigenic and adipogenic effects. This study sought to understand the potential involvement of members of MAPK serine/threonine kinases in the ghrelin-induced growth of rabbit MSCs ( rBMSC). Methods We applied various concentrations of ghrelin to cultured rBMSC and observed the growth rate of the cells by MTT, changes in the phosphorylation state of ERK1/2, JNK and p38, and the expression levels of ALP, Runx2, and Osterix by wetern blot. Results We found that the growth and osteogenic differentiation of ghrelin-treated rBMSC are promoted primarily by phosphorylated ERK1/2, and that this phosphorylation, as well p38 phosphorylation, is mediated by GHSR. Conclusions Our study suggests that ghrelin promotes the growth and osteogenic differentiation of rBMSC primarily through the ERK1/2 pathway.
Collapse
Affiliation(s)
- Nan Ye
- Department of Orthopedic, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District Yueyuan Road No 1, Chongqing, China. .,Dpartment of Cervical Surgery, The second Affiliated Hospital of Inner Mongolia Medical University, Muslims camp Square Road No 1, Hohhot, China.
| | - Dianming Jiang
- Department of Orthopedic, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District Yueyuan Road No 1, Chongqing, China.
| |
Collapse
|
14
|
Liu T, Fang Y, Liu S, Yu X, Zhang H, Liang M, Ding X. Limb ischemic preconditioning protects against contrast-induced acute kidney injury in rats via phosphorylation of GSK-3β. Free Radic Biol Med 2015; 81:170-82. [PMID: 25451640 DOI: 10.1016/j.freeradbiomed.2014.10.509] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 10/16/2014] [Accepted: 10/17/2014] [Indexed: 12/31/2022]
Abstract
Contrast-induced acute kidney injury (CI-AKI) resulting from the use of intravascular iodinated contrast media for diagnostic and interventional cardiovascular procedures is associated with substantial morbidity and mortality. Despite preventative measures intended to mitigate the risk of CI-AKI, there remains a need for a novel and effective therapeutic approach. Limb ischemic preconditioning (LIPC), where short-term ischemia/reperfusion is applied to an arm prior to administration of the contrast agent, has been shown in several trials to preserve renal function in patients at high risk for CI-AKI. However, the underlying mechanism by which this procedure provides renoprotection against contrast media insults is not known. Here, we explored the molecular mechanism(s) of LIPC-induced protection of the kidneys from CI-AKI, particularly the role of phosphorylated glycogen synthase kinase-3β (GSK-3β). We used a novel CI-AKI model consisting of 5/6 nephrectomized (NE) rats at 6 weeks after the ablative surgery. LIPC- or sham-treated rats were administered iohexol (10 ml/kg, 3.5 gI) via the tail vein. The results showed that LIPC protected the kidneys against iohexol-induced injury. This protective effect was accompanied by the attenuation of renal dysfunction, tubular damage, apoptosis, mitochondrial swelling, oxidative stress, and inflammation. Furthermore, LIPC-induced renoprotection was blocked via treatment with inhibitors of PI3K (wortmannin or LY294002), but not ERK (U0126 or PD98059). LIPC also increased the protein expression levels of phospho-Akt, phospho-GSK-3β, and nuclear Nrf2, and decreased the levels of nuclear NF-κB. A specific GSK-3β inhibitor (SB216763) mimicked this effect of LIPC, by inhibiting the opening of the mitochondrial permeability transition pore and reducing the levels of oxidative stress and inflammation via activation of Nrf2 and suppression of NF-κB. The above results demonstrate that LIPC induces protection against CI-AKI, making this procedure a promising strategy for preventing CI-AKI. In particular, this renoprotective effect involves the phosphorylation of GSK-3β.
Collapse
Affiliation(s)
- Tongqiang Liu
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Division of Nephrology, the Affiliated Changzhou No. 2 Hospital of Nanjing Medical College, Changzhou 213003, Jiangsu, China
| | - Yi Fang
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Kidney and Dialysis, Shanghai 200032, China
| | - Shaopeng Liu
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaofang Yu
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Kidney and Dialysis, Shanghai 200032, China
| | - Hui Zhang
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Mingyu Liang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53201, USA
| | - Xiaoqiang Ding
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Kidney and Dialysis, Shanghai 200032, China.
| |
Collapse
|
15
|
Angelino E, Reano S, Ferrara M, Agosti E, Graziani A, Filigheddu N. Antifibrotic activity of acylated and unacylated ghrelin. Int J Endocrinol 2015; 2015:385682. [PMID: 25960743 PMCID: PMC4415458 DOI: 10.1155/2015/385682] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 04/01/2015] [Indexed: 12/15/2022] Open
Abstract
Fibrosis can affect almost all tissues and organs, it often represents the terminal stage of chronic diseases, and it is regarded as a major health issue for which efficient therapies are needed. Tissue injury, by inducing necrosis/apoptosis, triggers inflammatory response that, in turn, promotes fibroblast activation and pathological deposition of extracellular matrix. Acylated and unacylated ghrelin are the main products of the ghrelin gene. The acylated form, through its receptor GHSR-1a, stimulates appetite and growth hormone (GH) release. Although unacylated ghrelin does not bind or activate GHSR-1a, it shares with the acylated form several biological activities. Ghrelin peptides exhibit anti-inflammatory, antioxidative, and antiapoptotic activities, suggesting that they might represent an efficient approach to prevent or reduce fibrosis. The aim of this review is to summarize the available evidence regarding the effects of acylated and unacylated ghrelin on different pathologies and experimental models in which fibrosis is a predominant characteristic.
Collapse
Affiliation(s)
- Elia Angelino
- Department of Translational Medicine, University of Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | - Simone Reano
- Department of Translational Medicine, University of Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | - Michele Ferrara
- Department of Translational Medicine, University of Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | - Emanuela Agosti
- Department of Translational Medicine, University of Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | - Andrea Graziani
- Department of Translational Medicine, University of Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | - Nicoletta Filigheddu
- Department of Translational Medicine, University of Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
- *Nicoletta Filigheddu:
| |
Collapse
|
16
|
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: 2.8] [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
|
17
|
Zhao H, Wang L, Wei R, Xiu D, Tao M, Ke J, Liu Y, Yang J, Hong T. Activation of glucagon-like peptide-1 receptor inhibits tumourigenicity and metastasis of human pancreatic cancer cells via PI3K/Akt pathway. Diabetes Obes Metab 2014; 16:850-60. [PMID: 24641303 DOI: 10.1111/dom.12291] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 01/14/2014] [Accepted: 03/11/2014] [Indexed: 01/18/2023]
Abstract
AIMS It has been reported that glucagon-like peptide-1 (GLP-1) agents are associated with an increased risk of pancreatic cancer in patients with type 2 diabetes. Reports have indicated that GLP-1 promotes pancreatic metaplasia and premalignant lesions. The aims of this study were to determine the effects of GLP-1-based therapy on pancreatic cancer cells. METHODS Immunohistochemistry was used to investigate GLP-1 receptor (GLP-1R) expression in 30 human pancreatic cancer tissues. We also analysed associated clinicopathological data and each patient's prognosis. Two human pancreatic cancer cell lines were used to evaluate the in vitro effects of the GLP-1R agonist liraglutide on cell growth, migration and invasion. Mouse xenograft models of human pancreatic cancer were established to evaluate the effects of liraglutide in vivo. RESULTS Human pancreatic cancer tissues showed lower levels or a lack of GLP-1R expression when compared with levels in the tumour-adjacent pancreatic tissues. Negative GLP-1R expression occurred more frequently in advanced tumours with larger diameters and lymphatic metastasis, and was associated with a poor prognosis. GLP-1R activation with liraglutide inhibited tumourigenicity and metastasis of human pancreatic cancer cells in vitro and in vivo. Akt activation was dose-dependently inhibited by liraglutide, and the PI3K inhibitors, LY294002 and wortmannin, displayed similar suppressive effects to liraglutide in human pancreatic cancer cells. CONCLUSIONS GLP-1R activation has an antitumour effect on human pancreatic cancers via inhibition of the PI3K/Akt pathway. This finding suggests that GLP-1-based therapies may be beneficial, rather than harmful, in treating type 2 diabetic patients with pancreatic cancer.
Collapse
Affiliation(s)
- H Zhao
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Lee TJ, Park S, Bhang SH, Yoon JK, Jo I, Jeong GJ, Hong BH, Kim BS. Graphene enhances the cardiomyogenic differentiation of human embryonic stem cells. Biochem Biophys Res Commun 2014; 452:174-80. [PMID: 25152405 DOI: 10.1016/j.bbrc.2014.08.062] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 08/15/2014] [Indexed: 12/11/2022]
Abstract
Graphene has drawn attention as a substrate for stem cell culture and has been reported to stimulate the differentiation of multipotent adult stem cells. Here, we report that graphene enhances the cardiomyogenic differentiation of human embryonic stem cells (hESCs) at least in part, due to nanoroughness of graphene. Large-area graphene on glass coverslips was prepared via the chemical vapor deposition method. The coating of the graphene with vitronectin (VN) was required to ensure high viability of the hESCs cultured on the graphene. hESCs were cultured on either VN-coated glass (glass group) or VN-coated graphene (graphene group) for 21 days. The cells were also cultured on glass coated with Matrigel (Matrigel group), which is a substrate used in conventional, directed cardiomyogenic differentiation systems. The culture of hESCs on graphene promoted the expression of genes involved in the stepwise differentiation into mesodermal and endodermal lineage cells and subsequently cardiomyogenic differentiation compared with the culture on glass or Matrigel. In addition, the culture on graphene enhanced the gene expression of cardiac-specific extracellular matrices. Culture on graphene may provide a new platform for the development of stem cell therapies for ischemic heart diseases by enhancing the cardiomyogenic differentiation of hESCs.
Collapse
Affiliation(s)
- Tae-Jin Lee
- Engineering Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Subeom Park
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Jeong-Kee Yoon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Insu Jo
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Gun-Jae Jeong
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Byung Hee Hong
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea.
| | - Byung-Soo Kim
- Engineering Research Institute, Seoul National University, Seoul, Republic of Korea; School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea; Institute of Bioengineering, Institute of Chemical Processes, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
19
|
Prodam F, Filigheddu N. Ghrelin gene products in acute and chronic inflammation. Arch Immunol Ther Exp (Warsz) 2014; 62:369-84. [PMID: 24728531 DOI: 10.1007/s00005-014-0287-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 03/21/2014] [Indexed: 12/27/2022]
Abstract
Ghrelin gene products--the peptides ghrelin, unacylated ghrelin, and obestatin--have several actions on the immune system, opening new perspectives within neuroendocrinology, metabolism and inflammation. The aim of this review is to summarize the available evidence regarding the less known role of these peptides in the machinery of inflammation and autoimmunity, outlining some of their most promising therapeutic applications.
Collapse
Affiliation(s)
- Flavia Prodam
- Departmant of Health Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | | |
Collapse
|
20
|
Douglas GAF, McGirr R, Charlton CL, Kagan DB, Hoffman LM, Luyt LG, Dhanvantari S. Characterization of a far-red analog of ghrelin for imaging GHS-R in P19-derived cardiomyocytes. Peptides 2014; 54:81-8. [PMID: 24468548 DOI: 10.1016/j.peptides.2014.01.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 01/16/2014] [Accepted: 01/16/2014] [Indexed: 11/29/2022]
Abstract
Ghrelin and its receptor, the growth hormone secretagogue receptor (GHS-R), are expressed in the heart, and may function to promote cardiomyocyte survival, differentiation and contractility. Previously, we had generated a truncated analog of ghrelin conjugated to fluorescein isothiocyanate for the purposes of determining GHS-R expression in situ. We now report the generation and characterization of a far-red ghrelin analog, [Dpr(3)(octanoyl), Lys(19)(Cy5)]ghrelin (1-19), and show that it can be used to image changes in GHS-R in developing cardiomyocytes. We also generated the des-acyl analog, des-acyl [Lys(19)(Cy5)]ghrelin (1-19) and characterized its binding to mouse heart sections. Receptor binding affinity of Cy5-ghrelin as measured in HEK293 cells overexpressing GHS-R1a was within an order of magnitude of that of fluorescein-ghrelin and native human ghrelin, while the des-acyl Cy5-ghrelin did not bind GHS-R1a. Live cell imaging in HEK293/GHS-R1a cells showed cell surface labeling that was displaced by excess ghrelin. Interestingly, Cy5-ghrelin, but not the des-acyl analog, showed concentration-dependent binding in mouse heart tissue sections. We then used Cy5-ghrelin to track GHS-R expression in P19-derived cardiomyocytes. Live cell imaging at different time points after DMSO-induced differentiation showed that GHS-R expression preceded that of the differentiation marker aMHC and tracked with the contractility marker SERCA 2a. Our far-red analog of ghrelin adds to the tools we are developing to map GHS-R in developing and diseased cardiac tissues.
Collapse
Affiliation(s)
- Gregory A F Douglas
- Departments of Pathology, Chemistry, Medical Imaging, Medical Biophysics, and Oncology, Western University, Richmond Street, London, Ontario, Canada; Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
| | - Rebecca McGirr
- Departments of Pathology, Chemistry, Medical Imaging, Medical Biophysics, and Oncology, Western University, Richmond Street, London, Ontario, Canada; Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
| | - Carlie L Charlton
- Departments of Pathology, Chemistry, Medical Imaging, Medical Biophysics, and Oncology, Western University, Richmond Street, London, Ontario, Canada; Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
| | - Dov B Kagan
- Departments of Pathology, Chemistry, Medical Imaging, Medical Biophysics, and Oncology, Western University, Richmond Street, London, Ontario, Canada; Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
| | - Lisa M Hoffman
- Departments of Pathology, Chemistry, Medical Imaging, Medical Biophysics, and Oncology, Western University, Richmond Street, London, Ontario, Canada; Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
| | - Leonard G Luyt
- Departments of Pathology, Chemistry, Medical Imaging, Medical Biophysics, and Oncology, Western University, Richmond Street, London, Ontario, Canada; Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
| | - Savita Dhanvantari
- Departments of Pathology, Chemistry, Medical Imaging, Medical Biophysics, and Oncology, Western University, Richmond Street, London, Ontario, Canada; Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada.
| |
Collapse
|
21
|
The growth hormone secretagogue receptor: its intracellular signaling and regulation. Int J Mol Sci 2014; 15:4837-55. [PMID: 24651458 PMCID: PMC3975427 DOI: 10.3390/ijms15034837] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/06/2014] [Accepted: 03/11/2014] [Indexed: 01/29/2023] Open
Abstract
The growth hormone secretagogue receptor (GHSR), also known as the ghrelin receptor, is involved in mediating a wide variety of biological effects of ghrelin, including: stimulation of growth hormone release, increase of food intake and body weight, modulation of glucose and lipid metabolism, regulation of gastrointestinal motility and secretion, protection of neuronal and cardiovascular cells, and regulation of immune function. Dependent on the tissues and cells, activation of GHSR may trigger a diversity of signaling mechanisms and subsequent distinct physiological responses. Distinct regulation of GHSR occurs at levels of transcription, receptor interaction and internalization. Here we review the current understanding on the intracellular signaling pathways of GHSR and its modulation. An overview of the molecular structure of GHSR is presented first, followed by the discussion on its signaling mechanisms. Finally, potential mechanisms regulating GHSR are reviewed.
Collapse
|