1
|
Olejnik A, Radajewska A, Krzywonos-Zawadzka A, Bil-Lula I. Klotho inhibits IGF1R/PI3K/AKT signalling pathway and protects the heart from oxidative stress during ischemia/reperfusion injury. Sci Rep 2023; 13:20312. [PMID: 37985893 PMCID: PMC10662387 DOI: 10.1038/s41598-023-47686-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023] Open
Abstract
Ischemia/reperfusion injury (IRI) of the heart involves the activation of oxidative and proapoptotic pathways. Simultaneously Klotho protein presents anti-aging, antiapoptotic and antioxidative properties. Therefore, this study aimed to evaluate the effect of Klotho protein on oxidative stress in hearts subjected to IRI. Isolated rat hearts perfused with the Langendorff method were subjected to ischemia, followed by reperfusion, in the presence or absence of recombinant rat Klotho protein. The factors involved in the activation of insulin-like growth factor receptor (IGF1R)/phosphoinositide-3-kinase (PI3K)/protein kinase B (AKT) signalling pathway were evaluated. IRI caused activation of the IGF1R (p = 0.0122)/PI3K (p = 0.0022) signalling, as compared to the aerobic control group. Infusion supply of Klotho protein during IRI significantly reduced the level of phospho-IGF1R (p = 0.0436), PI3K (p = 0.0218) and phospho-AKT (p = 0.0020). Transcriptional activity of forkhead box protein O3 (FOXO3) was reduced (p = 0.0207) in hearts subjected to IRI, compared to aerobic control. Administration of Klotho decreased phosphorylation of FOXO3 (p = 0.0355), and enhanced activity of glutathione peroxidase (p = 0.0452) and superoxide dismutase (p = 0.0060) in IRI + Klotho group. The levels of reactive oxygen/nitrogen species (ROS/RNS) (p = 0.0480) and hydrogen peroxide (H2O2) (p = 0.0460), and heart injury (p = 0.0005) were significantly increased in hearts from the IRI group in comparison to the aerobic group. Klotho reduced NADPH oxidase 2 (NOX2) (p = 0.0390), ROS/RNS (p = 0.0435) and H2O2 (p = 0.0392) levels, and heart damage (p = 0.0286) in the hearts subjected to IRI. In conclusion, Klotho contributed to the protection of the heart against IRI and oxidative stress via inhibition of the IGF1R/PI3K/AKT pathway, thus can be recognized as a novel cardiopreventive/cardioprotective agent.
Collapse
Affiliation(s)
- Agnieszka Olejnik
- Division of Clinical Chemistry and Laboratory Haematology, Department of Medical Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A St., 50-556, Wrocław, Poland
| | - Anna Radajewska
- Division of Clinical Chemistry and Laboratory Haematology, Department of Medical Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A St., 50-556, Wrocław, Poland
| | - Anna Krzywonos-Zawadzka
- Division of Clinical Chemistry and Laboratory Haematology, Department of Medical Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A St., 50-556, Wrocław, Poland
| | - Iwona Bil-Lula
- Division of Clinical Chemistry and Laboratory Haematology, Department of Medical Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A St., 50-556, Wrocław, Poland.
| |
Collapse
|
2
|
Zhu M, Miao S, Zhou W, Elnesr SS, Dong X, Zou X. MAPK, AKT/FoxO3a and mTOR pathways are involved in cadmium regulating the cell cycle, proliferation and apoptosis of chicken follicular granulosa cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 214:112091. [PMID: 33706141 DOI: 10.1016/j.ecoenv.2021.112091] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 02/15/2021] [Accepted: 02/19/2021] [Indexed: 06/12/2023]
Abstract
The occurrence of cadmium (Cd) in feed is a major problem in animal health and production. Studies have confirmed that Cd depresses egg production of laying hens, which is closely related to follicular atresia. This study aimed to assess the toxic impacts of Cd on the ovarian tissue, and to examine the mechanism of Cd-induced granulosa cell proliferation and apoptosis. Results from the nitric oxide (NO) and malondialdehyde (MDA) content, total superoxide dismutase (T-SOD), glutathione peroxide (GSH-Px), total nitric oxide synthase (T-NOS) and adenosine triphosphatase (ATPase) activities, terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) assay, and hematoxylin-eosin (H & E) staining indicated that excess Cd induced oxidative stress, granulosa cell apoptosis and follicular atresia in the layer ovary. Low-dose Cd exposure (1 μM) induced the granulosa cell proliferation, upregulated the mRNA levels of RSK1 and RHEB, activated FoxO3a, AKT, ERK1/2, mTOR and p70S6K1 phosphorylation, and promoted cell cycle progression from phase G1 to S. However, high-dose Cd exposure (15 μM) induced reactive oxygen species (ROS) generation and cell apoptosis, upregulated the mRNA levels of the inflammatory factors, ASK1, JNK, p38 and TAK1, downregulated the expressions of RSK1 and RHEB genes, and inhibited the phosphorylation of ERK1/2, mTOR and p70S6K1 proteins, and the cell cycle progression. Rapamycin pre-treatment completely blocked the phosphorylation of mTOR and p70S6K1 proteins, and the cell cycle progression induced by 1 μM Cd, and accelerated 15 μM Cd-induced cell apoptosis and cell cycle arrest. The microRNA sequencing result showed that 15 μM Cd induced differential expression of microRNA genes, which may regulate AKT, ERK1/2 and mTOR signaling and cell cycle progression by regulating the activity of G proteins and cell cycle-related proteins. Conclusively, these results indicated that Cd can cause the ovarian damage and follicular atresia, and regulate cell cycle, cell proliferation or apoptosis of granulosa cells through MAPK, AKT/FoxO3a and mTOR pathways in laying hens.
Collapse
Affiliation(s)
- Mingkun Zhu
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, PR China; School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, PR China
| | - Sasa Miao
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Wenting Zhou
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Shaaban Saad Elnesr
- Department of Poultry Production, Faculty of Agriculture, Fayoum University, 63514 Fayoum, Egypt
| | - Xinyang Dong
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Xiaoting Zou
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, PR China.
| |
Collapse
|
3
|
A miR-511-binding site SNP in the 3'UTR of IGF-1 gene is associated with proliferation and apoptosis of PK-15 cells. In Vitro Cell Dev Biol Anim 2019; 55:323-330. [PMID: 30945114 DOI: 10.1007/s11626-019-00329-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/28/2019] [Indexed: 01/07/2023]
Abstract
Insulin-like growth factor-1 (IGF-1) is a functional candidate gene for pig growth and development due to its crucial role in the growth axis of growth hormone-IGF-1. Considering that the 3' untranslated region (3'UTR) of gene may affect its expression, we analyzed the effect of a single-nucleotide polymorphism (SNP) (rs34142920, c.674C > T) on gene expression, cell proliferation, and apoptosis and the possible related molecular mechanisms in PK-15 cells. The SNP was found in the 3'UTR of IGF-1 in Bama Xiang pig in previous investigations. Results showed that the SNP was located at the target site binding to microRNA (miR-511). The 3'UTR of IGF-1 gene with C allele significantly downregulated the expression of IGF-1 gene compared with that of the gene with T allele by luciferase assay. miR-511 was transfected into porcine kidney cell line (PK-15 cells) to reveal its effects on cells and whether or not it targets IGF-1. The expression levels of IGF-1 at mRNA and protein levels were remarkably downregulated. miR-511 significantly inhibited cell proliferation and promoted cell apoptosis by downregulating the phosphorylation level of AKT and ERK1/2. This finding confirmed that miR-511 inhibits proliferation and promotes apoptosis by downregulating the IGF-1 in PK-15 cells.
Collapse
|
4
|
Genetic and Epigenetic Control of CDKN1C Expression: Importance in Cell Commitment and Differentiation, Tissue Homeostasis and Human Diseases. Int J Mol Sci 2018; 19:ijms19041055. [PMID: 29614816 PMCID: PMC5979523 DOI: 10.3390/ijms19041055] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 03/31/2018] [Accepted: 03/31/2018] [Indexed: 12/28/2022] Open
Abstract
The CDKN1C gene encodes the p57Kip2 protein which has been identified as the third member of the CIP/Kip family, also including p27Kip1 and p21Cip1. In analogy with these proteins, p57Kip2 is able to bind tightly and inhibit cyclin/cyclin-dependent kinase complexes and, in turn, modulate cell division cycle progression. For a long time, the main function of p57Kip2 has been associated only to correct embryogenesis, since CDKN1C-ablated mice are not vital. Accordingly, it has been demonstrated that CDKN1C alterations cause three human hereditary syndromes, characterized by altered growth rate. Subsequently, the p57Kip2 role in several cell phenotypes has been clearly assessed as well as its down-regulation in human cancers. CDKN1C lies in a genetic locus, 11p15.5, characterized by a remarkable regional imprinting that results in the transcription of only the maternal allele. The control of CDKN1C transcription is also linked to additional mechanisms, including DNA methylation and specific histone methylation/acetylation. Finally, long non-coding RNAs and miRNAs appear to play important roles in controlling p57Kip2 levels. This review mostly represents an appraisal of the available data regarding the control of CDKN1C gene expression. In addition, the structure and function of p57Kip2 protein are briefly described and correlated to human physiology and diseases.
Collapse
|
5
|
Sun Y, Xu R, Huang J, Yao Y, Pan X, Chen Z, Ma G. Insulin-like growth factor-1-mediated regulation of miR-193a expression promotes the migration and proliferation of c-kit-positive mouse cardiac stem cells. Stem Cell Res Ther 2018; 9:41. [PMID: 29467020 PMCID: PMC5822561 DOI: 10.1186/s13287-017-0762-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/13/2017] [Accepted: 12/22/2017] [Indexed: 12/13/2022] Open
Abstract
Background C-kit-positive cardiac stem cells (CSCs) have been shown to be a promising candidate treatment for myocardial infarction and heart failure. Insulin-like growth factor (IGF)-1 is an anabolic growth hormone that regulates cellular proliferation, differentiation, senescence, and death in various tissues. Although IGF-1 promotes the migration and proliferation of c-kit-positive mouse CSCs, the underlying mechanism remains unclear. Methods Cells were isolated from adult mouse hearts, and c-kit-positive CSCs were separated using magnetic beads. The cells were cultured with or without IGF-1, and c-kit expression was measured by Western blotting. IGF-1 induced CSC proliferation and migration, as measured through Cell Counting Kit-8 (CCK-8) and Transwell assays, respectively. The miR-193a expression was measured by quantitative real-time PCR (qPCR) assays. Results IGF-1 enhanced c-kit expression in c-kit-positive CSCs. The activities of the phosphoinositol 3-kinase (PI3K)/AKT signaling pathway and DNA methyltransferases (DNMTs) were enhanced, and their respective inhibitors LY294002 and 5-azacytidine (5-AZA) blunted c-kit expression. Based on the results of quantitative real-time PCR (qPCR) assays, the expression of miR-193a, which is embedded in a CpG island, was down-regulated in the IGF-1-stimulated group and negatively correlated with c-kit expression, whereas c-kit-positive CSCs infected with lentivirus carrying micro-RNA193a displayed reduced c-kit expression, migration and proliferation. Conclusions IGF-1 upregulated c-kit expression in c-kit-positive CSCs resulting in enhanced CSC proliferation and migration by activating the PI3K/AKT/DNMT signaling pathway to epigenetically silence miR-193a, which negatively modifies the c-kit expression level. Electronic supplementary material The online version of this article (10.1186/s13287-017-0762-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuning Sun
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, DingjiaQiao No. 87, Hunan Road, Nanjing, 210009, Jiangsu, China
| | - Rongfeng Xu
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, DingjiaQiao No. 87, Hunan Road, Nanjing, 210009, Jiangsu, China
| | - Jia Huang
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, DingjiaQiao No. 87, Hunan Road, Nanjing, 210009, Jiangsu, China
| | - Yuyu Yao
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, DingjiaQiao No. 87, Hunan Road, Nanjing, 210009, Jiangsu, China
| | - Xiaodong Pan
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, DingjiaQiao No. 87, Hunan Road, Nanjing, 210009, Jiangsu, China
| | - Zhongpu Chen
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, DingjiaQiao No. 87, Hunan Road, Nanjing, 210009, Jiangsu, China.
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, DingjiaQiao No. 87, Hunan Road, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
6
|
FOXO1/3: Potential suppressors of fibrosis. Ageing Res Rev 2018; 41:42-52. [PMID: 29138094 DOI: 10.1016/j.arr.2017.11.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/07/2017] [Accepted: 11/08/2017] [Indexed: 02/06/2023]
Abstract
Fibrosis is a universally age-related disease that involves nearly all organs. It is typically initiated by organic injury and eventually results in organ failure. There are still few effective therapeutic strategy targets for fibrogenesis. Forkhead box proteins O1 and O3 (FOXO1/3) have been shown to have favorable inhibitory effects on fibroblast activation and subsequent extracellular matrix production and can ameliorate fibrosis levels in numerous organs, including the heart, liver, lung, and kidney; they are therefore promising targets for anti-fibrosis therapy. Moreover, we can develop appropriate strategies to make the best use of FOXO1/3's anti-fibrosis properties. The information reviewed here should be significant for understanding the roles of FOXO1/3 in fibrosis and should contribute to the design of further studies related to FOXO1/3 and the fibrotic response and shed light on a potential treatment for fibrosis.
Collapse
|
7
|
Yin L, Huang D, Liu X, Wang Y, Liu J, Liu F, Yu B. Omentin-1 effects on mesenchymal stem cells: proliferation, apoptosis, and angiogenesis in vitro. Stem Cell Res Ther 2017; 8:224. [PMID: 29017592 PMCID: PMC5633887 DOI: 10.1186/s13287-017-0676-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 07/01/2017] [Accepted: 09/18/2017] [Indexed: 02/07/2023] Open
Abstract
Background Mesenchymal stem cells (MSCs) are emerging as an extremely promising therapeutic agent for tissue repair. However, limitations exist such as the low numbers of MSCs obtained from donors, and the poor survival and function of donor cells. Omentin-1, a new fat depot-specific secretory adipokine, exerts proproliferation, prosurvival, and proangiogenic functions in certain cells via an Akt-dependent mechanism; however, little is known about the influence of omentin-1 on MSCs. Methods MSCs were isolated from 60–80 g donor rats. Cell proliferation was assessed with CCK-8 and EdU assay. Cell cycle, apoptosis ratio, reactive oxygen species concentration, and mitochondrial membrane potential were detected by flow cytometry. Hoechst 33342 dye was used to assess morphological changes of apoptosis. Expression levels of Akt, FoxO3a, GSK-3β, and apoptosis- and cell cycle-associated proteins were detected by Western blotting. Tube formation assay was used to test the angiogenesis role of conditioned medium from MSCs in vitro. The cytokine secretion was assessed by ELISA. Results After treatment with omentin-1 (100–800 ng/ml), MSCs displayed a higher proliferative capacity with an increasing number of cells in the S and G2 phase of the cell cycle. Moreover, omentin-1 preconditioning for 1 h could protect MSCs against H2O2-induced apoptosis in a concentration-dependent manner. Furthermore, omentin-1 pretreatment reduced the excessive reactive oxygen species. Western blots revealed that increased Bcl-2 and decreased Bax appeared in MSCs after omentin-1 incubation, which inhibited the mitochondrial apoptosis pathways with evidence showing inhibition of caspase-3 cleavage and preservation of mitochondrial membrane potential. Omentin-1 could enhance angiogenic growth factor secretion and elevate the ability of MSCs to stimulate tube formation by human umbilical vein endothelial cells (HUVECs). Furthermore, omentin-1 enhanced Akt phosphorylation; however, blockade of the PI3K/Akt pathway with an inhibitor, LY294002 (20 μM), suppressed the above beneficial effects of omentin-1. Conclusion Omentin-1 can exert beneficial effects on MSCs by promoting proliferation, inhibiting apoptosis, increasing secretion of angiogenic cytokines, and enhancing the ability for stimulating tube formation by HUVECs via the PI3K/Akt signaling pathway. Thus, omentin-1 may be considered a candidate for optimizing MSC-based cell therapy.
Collapse
Affiliation(s)
- Li Yin
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Dan Huang
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Xinxin Liu
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Yongshun Wang
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Jingjin Liu
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Fang Liu
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Bo Yu
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China. .,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.
| |
Collapse
|
8
|
Torán JL, Aguilar S, López JA, Torroja C, Quintana JA, Santiago C, Abad JL, Gomes-Alves P, Gonzalez A, Bernal JA, Jiménez-Borreguero LJ, Alves PM, R-Borlado L, Vázquez J, Bernad A. CXCL6 is an important paracrine factor in the pro-angiogenic human cardiac progenitor-like cell secretome. Sci Rep 2017; 7:12490. [PMID: 28970523 PMCID: PMC5624898 DOI: 10.1038/s41598-017-11976-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 08/29/2017] [Indexed: 12/22/2022] Open
Abstract
Studies in recent years have established that the principal effects in cardiac cell therapy are associated with paracrine/autocrine factors. We combined several complementary techniques to define human cardiac progenitor cell (CPC) secretome constituted by 914 proteins/genes; 51% of these are associated with the exosomal compartment. To define the set of proteins specifically or highly differentially secreted by CPC, we compared human mesenchymal stem cells and dermal fibroblasts; the study defined a group of growth factors, cytokines and chemokines expressed at high to medium levels by CPC. Among them, IL-1, GROa (CXCL1), CXCL6 (GCP2) and IL-8 are examples whose expression was confirmed by most techniques used. ELISA showed that CXCL6 is significantly overexpressed in CPC conditioned medium (CM) (18- to 26-fold) and western blot confirmed expression of its receptors CXCR1 and CXCR2. Addition of anti-CXCL6 completely abolished migration in CPC-CM compared with anti-CXCR2, which promoted partial inhibition, and anti-CXCR1, which was inefficient. Anti-CXCL6 also significantly inhibited CPC CM angiogenic activity. In vivo evaluation also supported a relevant role for angiogenesis. Altogether, these results suggest a notable angiogenic potential in CPC-CM and identify CXCL6 as an important paracrine factor for CPC that signals mainly through CXCR2.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/pharmacology
- Cell Movement
- Chemokine CXCL1/genetics
- Chemokine CXCL1/metabolism
- Chemokine CXCL6/antagonists & inhibitors
- Chemokine CXCL6/genetics
- Chemokine CXCL6/metabolism
- Culture Media, Conditioned/chemistry
- Culture Media, Conditioned/metabolism
- Fibroblasts/cytology
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Gene Expression Regulation
- Human Umbilical Vein Endothelial Cells/cytology
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Interleukin-1/genetics
- Interleukin-1/metabolism
- Interleukin-8/genetics
- Interleukin-8/metabolism
- Male
- Mesenchymal Stem Cells/cytology
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/metabolism
- Mice
- Mice, Inbred C57BL
- Myocardium/cytology
- Myocardium/metabolism
- Neovascularization, Physiologic/genetics
- Paracrine Communication/genetics
- Proteome/genetics
- Proteome/metabolism
- Receptors, Interleukin-8A/antagonists & inhibitors
- Receptors, Interleukin-8A/genetics
- Receptors, Interleukin-8A/metabolism
- Receptors, Interleukin-8B/antagonists & inhibitors
- Receptors, Interleukin-8B/genetics
- Receptors, Interleukin-8B/metabolism
- Signal Transduction
- Stem Cells/cytology
- Stem Cells/drug effects
- Stem Cells/metabolism
Collapse
Affiliation(s)
- José Luis Torán
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Susana Aguilar
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Juan Antonio López
- Cardiovascular Proteomics Laboratory, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernaández Almagro 3, 28029, Madrid, Spain
| | - Carlos Torroja
- Bioinformatics Unit, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Juan Antonio Quintana
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
- Cell and Developmental Biology, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Cesar Santiago
- Department of Macromolecular Structures, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - José Luis Abad
- Coretherapix SLU, Santiago Grisolia 2, 28769, Tres Cantos, Madrid, Spain
| | - Patricia Gomes-Alves
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal
| | - Andrés Gonzalez
- Myocardial pathophysiology, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Juan Antonio Bernal
- Myocardial pathophysiology, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Luis Jesús Jiménez-Borreguero
- Cell and Developmental Biology, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
- Hospital de la Princesa, Diego de León 62, 28006, Madrid, Spain
| | - Paula Marques Alves
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal
| | - Luis R-Borlado
- Coretherapix SLU, Santiago Grisolia 2, 28769, Tres Cantos, Madrid, Spain
| | - Jesús Vázquez
- Cardiovascular Proteomics Laboratory, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernaández Almagro 3, 28029, Madrid, Spain
| | - Antonio Bernad
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain.
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| |
Collapse
|
9
|
Sharma M, Kartha CC, Mukhopadhyay B, Goyal RK, Gupta SK, Ganguly NK, Dhalla NS. India’s March to Halt the Emerging Cardiovascular Epidemic. Circ Res 2017; 121:913-916. [DOI: 10.1161/circresaha.117.310904] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Meenakshi Sharma
- From the Indian Council of Medical Research, New Delhi, India (M.S.); Rajiv Gandhi Center for Biotechnology, Trivandrum, India (C.C.K.); Policy Center for Biomedical Research, Faridabad, India (B.M.); Delhi Pharmaceutical Sciences and Research University, India (R.K.G., S.K.G.); Jawaharlal Nehru University, New Delhi, India (N.K.G.); and Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Center, Winnipeg, Canada (N.S.D.)
| | - Chandrasekharan C. Kartha
- From the Indian Council of Medical Research, New Delhi, India (M.S.); Rajiv Gandhi Center for Biotechnology, Trivandrum, India (C.C.K.); Policy Center for Biomedical Research, Faridabad, India (B.M.); Delhi Pharmaceutical Sciences and Research University, India (R.K.G., S.K.G.); Jawaharlal Nehru University, New Delhi, India (N.K.G.); and Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Center, Winnipeg, Canada (N.S.D.)
| | - Bratati Mukhopadhyay
- From the Indian Council of Medical Research, New Delhi, India (M.S.); Rajiv Gandhi Center for Biotechnology, Trivandrum, India (C.C.K.); Policy Center for Biomedical Research, Faridabad, India (B.M.); Delhi Pharmaceutical Sciences and Research University, India (R.K.G., S.K.G.); Jawaharlal Nehru University, New Delhi, India (N.K.G.); and Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Center, Winnipeg, Canada (N.S.D.)
| | - Ramesh K. Goyal
- From the Indian Council of Medical Research, New Delhi, India (M.S.); Rajiv Gandhi Center for Biotechnology, Trivandrum, India (C.C.K.); Policy Center for Biomedical Research, Faridabad, India (B.M.); Delhi Pharmaceutical Sciences and Research University, India (R.K.G., S.K.G.); Jawaharlal Nehru University, New Delhi, India (N.K.G.); and Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Center, Winnipeg, Canada (N.S.D.)
| | - Suresh K. Gupta
- From the Indian Council of Medical Research, New Delhi, India (M.S.); Rajiv Gandhi Center for Biotechnology, Trivandrum, India (C.C.K.); Policy Center for Biomedical Research, Faridabad, India (B.M.); Delhi Pharmaceutical Sciences and Research University, India (R.K.G., S.K.G.); Jawaharlal Nehru University, New Delhi, India (N.K.G.); and Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Center, Winnipeg, Canada (N.S.D.)
| | - Nirmal K. Ganguly
- From the Indian Council of Medical Research, New Delhi, India (M.S.); Rajiv Gandhi Center for Biotechnology, Trivandrum, India (C.C.K.); Policy Center for Biomedical Research, Faridabad, India (B.M.); Delhi Pharmaceutical Sciences and Research University, India (R.K.G., S.K.G.); Jawaharlal Nehru University, New Delhi, India (N.K.G.); and Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Center, Winnipeg, Canada (N.S.D.)
| | - Naranjan S. Dhalla
- From the Indian Council of Medical Research, New Delhi, India (M.S.); Rajiv Gandhi Center for Biotechnology, Trivandrum, India (C.C.K.); Policy Center for Biomedical Research, Faridabad, India (B.M.); Delhi Pharmaceutical Sciences and Research University, India (R.K.G., S.K.G.); Jawaharlal Nehru University, New Delhi, India (N.K.G.); and Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Center, Winnipeg, Canada (N.S.D.)
| |
Collapse
|
10
|
O'Neill HS, O'Sullivan J, Porteous N, Ruiz-Hernandez E, Kelly HM, O'Brien FJ, Duffy GP. A collagen cardiac patch incorporating alginate microparticles permits the controlled release of hepatocyte growth factor and insulin-like growth factor-1 to enhance cardiac stem cell migration and proliferation. J Tissue Eng Regen Med 2017; 12:e384-e394. [PMID: 27943590 DOI: 10.1002/term.2392] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 11/17/2016] [Accepted: 12/06/2016] [Indexed: 12/25/2022]
Abstract
Cardiac stem cells (CSCs) represent a logical cell type to exploit as a regenerative treatment option for tissue damage accrued as a result of a myocardial infarction. However, the isolation and expansion of CSCs prior to cell transplantation is time consuming, costly and invasive, and the reliability of cell expansion may also prove to be a major obstacle in the clinical application of CSC-based transplantation therapy after a myocardial infarction. In order to overcome this, we propose the incorporation of growth factor-eluting alginate microparticles into collagen-based scaffolds as an implantable biomaterial to promote the recruitment and expansion of CSCs in the myocardium. In order to obtain scaffolds able to enhance the motogenic and proliferative potential of CSCs, the aim of this work was to achieve a sustained delivery of both hepatocyte growth factor and insulin-like growth factor-1. Both proteins were initially encapsulated in alginate microparticles by spray drying and subsequently incorporated into a collagen scaffold. Microparticles were seen to homogeneously distribute through the interconnected scaffold pore structure. The resulting scaffolds were capable of extending the release of both proteins up to 15 days, a three-fold increase over non-encapsulated proteins embedded in the scaffolds. In vitro assays with isolated CSCs demonstrated that the sustained release of both bioactive proteins resulted in an increased motogenic and proliferative effect. As presently practiced, the isolation and expansion of CSCs for autologous cell transplantation is slow, expensive and difficult to attain. Thus, there is a need for strategies to specifically activate in situ the intrinsic cardiac regenerative potential represented by the CSCs using combinations of growth factors obviating the need for cell transplantation. By favouring the natural regenerative capability of CSCs, it is hypothesized that the cardiac patch presented here will result in positive therapeutic outcomes in MI and heart failure patients in the future. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Hugh S O'Neill
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin, Ireland.,School of Pharmacy, RCSI, Dublin, Ireland
| | - Janice O'Sullivan
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin, Ireland
| | - Niamh Porteous
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin, Ireland
| | - Eduardo Ruiz-Hernandez
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin (TCD), Dublin, Ireland
| | - Helena M Kelly
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,School of Pharmacy, RCSI, Dublin, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin, Ireland
| | - Garry P Duffy
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin, Ireland.,Anatomy, School of Medicine, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Ireland
| |
Collapse
|
11
|
Chimenti I, Massai D, Morbiducci U, Beltrami AP, Pesce M, Messina E. Stem Cell Spheroids and Ex Vivo Niche Modeling: Rationalization and Scaling-Up. J Cardiovasc Transl Res 2017; 10:150-166. [PMID: 28289983 DOI: 10.1007/s12265-017-9741-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/27/2017] [Indexed: 02/08/2023]
Abstract
Improved protocols/devices for in vitro culture of 3D cell spheroids may provide essential cues for proper growth and differentiation of stem/progenitor cells (S/PCs) in their niche, allowing preservation of specific features, such as multi-lineage potential and paracrine activity. Several platforms have been employed to replicate these conditions and to generate S/PC spheroids for therapeutic applications. However, they incompletely reproduce the niche environment, with partial loss of its highly regulated network, with additional hurdles in the field of cardiac biology, due to debated resident S/PCs therapeutic potential and clinical translation. In this contribution, the essential niche conditions (metabolic, geometric, mechanical) that allow S/PCs maintenance/commitment will be discussed. In particular, we will focus on both existing bioreactor-based platforms for the culture of S/PC as spheroids, and on possible criteria for the scaling-up of niche-like spheroids, which could be envisaged as promising tools for personalized cardiac regenerative medicine, as well as for high-throughput drug screening.
Collapse
Affiliation(s)
- Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Rome, Italy
| | - Diana Massai
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Umberto Morbiducci
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | | | - Maurizio Pesce
- Tissue Engineering Research Unit, "Centro Cardiologico Monzino", IRCCS, Milan, Italy
| | - Elisa Messina
- Department of Pediatrics and Infant Neuropsychiatry, "Umberto I" Hospital, "La Sapienza" University, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
12
|
Lv T, Wu Y, Mu C, Liu G, Yan M, Xu X, Wu H, Du J, Yu J, Mu J. Insulin-like growth factor 1 promotes the proliferation and committed differentiation of human dental pulp stem cells through MAPK pathways. Arch Oral Biol 2016; 72:116-123. [DOI: 10.1016/j.archoralbio.2016.08.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 06/15/2016] [Accepted: 08/08/2016] [Indexed: 01/07/2023]
|
13
|
Xin Z, Ma Z, Jiang S, Wang D, Fan C, Di S, Hu W, Li T, She J, Yang Y. FOXOs in the impaired heart: New therapeutic targets for cardiac diseases. Biochim Biophys Acta Mol Basis Dis 2016; 1863:486-498. [PMID: 27890702 DOI: 10.1016/j.bbadis.2016.11.023] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/24/2016] [Accepted: 11/23/2016] [Indexed: 01/17/2023]
Abstract
Cardiac diseases have a high morbidity and mortality and affect the global population. Based on recent accumulating evidence, Forkhead box O (FOXOs) play important roles in cardiac diseases. Therefore, a summary of the current literature on the molecular mechanisms and roles of FOXOs in the heart will provide valuable information. In this review, we first briefly introduce the molecular features of FOXOs. Then, we discuss the regulation and cardiac actions of the FOXO pathways. Based on this background, we expand our discussion to the roles of FOXOs in several major cardiac diseases, such as ischemic cardiac diseases, diabetic cardiomyopathy and myocardial hypertrophy. Then, we describe some methodological problems associated with the FOXO gene-modified animal models. Finally, we discuss potential future directions. The information reviewed here may be significant for the design of future studies and may increase the potential of FOXOs as therapeutic targets.
Collapse
Affiliation(s)
- Zhenlong Xin
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Shuai Jiang
- Department of Aerospace Medicine, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Dongjin Wang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Chongxi Fan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Shouyin Di
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Wei Hu
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Junjun She
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, China.
| | - Yang Yang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
14
|
Gude N, Joyo E, Toko H, Quijada P, Villanueva M, Hariharan N, Sacchi V, Truffa S, Joyo A, Voelkers M, Alvarez R, Sussman MA. Notch activation enhances lineage commitment and protective signaling in cardiac progenitor cells. Basic Res Cardiol 2015; 110:29. [PMID: 25893875 DOI: 10.1007/s00395-015-0488-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 03/04/2015] [Accepted: 04/14/2015] [Indexed: 12/12/2022]
Abstract
Phase I clinical trials applying autologous progenitor cells to treat heart failure have yielded promising results; however, improvement in function is modest, indicating a need to enhance cardiac stem cell reparative capacity. Notch signaling plays a crucial role in cardiac development, guiding cell fate decisions that underlie myocyte and vessel differentiation. The Notch pathway is retained in the adult cardiac stem cell niche, where level and duration of Notch signal influence proliferation and differentiation of cardiac progenitors. In this study, Notch signaling promotes growth, survival and differentiation of cardiac progenitor cells into smooth muscle lineages in vitro. Cardiac progenitor cells expressing tamoxifen-regulated intracellular Notch1 (CPCeK) are significantly larger and proliferate more slowly than control cells, exhibit elevated mTORC1 and Akt signaling, and are resistant to oxidative stress. Vascular smooth muscle and cardiomyocyte markers increase in CPCeK and are augmented further upon ligand-mediated induction of Notch signal. Paracrine signals indicative of growth, survival and differentiation increase with Notch activity, while markers of senescence are decreased. Adoptive transfer of CPCeK into infarcted mouse myocardium enhances preservation of cardiac function and reduces infarct size relative to hearts receiving control cells. Greater capillary density and proportion of vascular smooth muscle tissue in CPCeK-treated hearts indicate improved vascularization. Finally, we report a previously undescribed signaling mechanism whereby Notch activation stimulates CPC growth, survival and differentiation via mTORC1 and paracrine factor expression. Taken together, these findings suggest that regulated Notch activation potentiates the reparative capacity of CPCs in the treatment of cardiac disease.
Collapse
Affiliation(s)
- Natalie Gude
- Heart Institute, and Biology Department, SDSU Integrated Regenerative Research Institute, Life Sciences North, Room 426, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|