1
|
Al-Zikri PNH, Huat TJ, Khan AA, Patar A, Reza MF, Idris FM, Abdullah JM, Jaafar H. Transplantation of IGF-1-induced BMSC-derived NPCs promotes tissue repair and motor recovery in a rat spinal cord injury model. Heliyon 2022; 8:e10384. [PMID: 36090221 PMCID: PMC9449758 DOI: 10.1016/j.heliyon.2022.e10384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/14/2022] [Accepted: 08/15/2022] [Indexed: 12/02/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) have therapeutic potential for spinal cord injury (SCI). We have shown that insulin-like growth factor 1 (IGF-1) enhances the cellular proliferation and survivability of BMSCs-derived neural progenitor cells (NPCs) by downregulating miR-22-3p. However, the functional application of BMSCs-derived NPCs has not been investigated fully. In this study, we demonstrate that knockdown of endogenous miR-22-3p in BMSCs-derived NPCs upregulates Akt1 expression, leading to enhanced cellular proliferation. RNASeq analysis reveals 3,513 differentially expressed genes in NPCs. The upregulated genes in NPCs enrich the gene ontology term associated with nervous system development. Terminally differentiated NPCs generate cells with neuronal-like morphology and phenotypes. Transplantation of NPCs in the SCI rat model results in better recovery in locomotor and sensory functions 4 weeks after transplantation. Altogether, the result of this study demonstrate that NPCs derived with IGF-1 supplementation could be differentiated into functional neural lineage cells and are optimal for stem cell therapy in SCI.
Collapse
|
2
|
Lei L, Meng L, Changqing X, Chen Z, Gang Y, Shiyuan F. Effect of cell receptors in the pathogenesis of osteoarthritis: Current insights. Open Life Sci 2022; 17:695-709. [PMID: 35859614 PMCID: PMC9267313 DOI: 10.1515/biol-2022-0075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/16/2022] [Accepted: 03/21/2022] [Indexed: 11/15/2022] Open
Abstract
Osteoarthritis (OA) is a chronic arthritic disease characterized by cartilage degradation, synovial inflammation, and subchondral bone lesions. The studies on the pathogenesis of OA are complex and diverse. The roles of receptors signaling in chondrocyte anabolism, inflammatory factors expression of synovial fibroblast, and angiogenesis in subchondral bone are particularly important for exploring the pathological mechanism of OA and clinical diagnosis and treatment. By reviewing the relevant literature, this article elaborates on the abnormal expression of receptors and the signaling transduction pathways from different pathological changes of OA anatomical components, aiming to provide new research ideas and clinical therapeutic value for OA pathogenesis.
Collapse
Affiliation(s)
- Li Lei
- Department of Orthopaedics, The First Affiliated Hospital of University of Science and Technology of China, 17 Lujiang Road, Hefei, Anhui, China
| | - Li Meng
- Department of Orthopaedics, The First Affiliated Hospital of University of Science and Technology of China, 17 Lujiang Road, Hefei, Anhui, China
| | - Xu Changqing
- Department of Orthopaedics, Dongxihu District People's Hospital Affiliated to Huazhong University of Science and Technology, Wuhan, China
| | - Zhu Chen
- Department of Orthopaedics, The First Affiliated Hospital of University of Science and Technology of China, 17 Lujiang Road, Hefei, Anhui, China
| | - Yao Gang
- Department of Orthopaedics, The First Affiliated Hospital of University of Science and Technology of China, 17 Lujiang Road, Hefei, Anhui, China
| | - Fang Shiyuan
- Department of Orthopaedics, The First Affiliated Hospital of University of Science and Technology of China, 17 Lujiang Road, Hefei, Anhui, China
| |
Collapse
|
3
|
Dixit M, Poudel SB, Yakar S. Effects of GH/IGF axis on bone and cartilage. Mol Cell Endocrinol 2021; 519:111052. [PMID: 33068640 PMCID: PMC7736189 DOI: 10.1016/j.mce.2020.111052] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
Growth hormone (GH) and its mediator, the insulin-like growth factor-1 (IGF-1) regulate somatic growth, metabolism and many aspects of aging. As such, actions of GH/IGF have been studied in many tissues and organs over decades. GH and IGF-1 are part of the hypothalamic/pituitary somatotrophic axis that consists of many other regulatory hormones, receptors, binding proteins, and proteases. In humans, GH/IGF actions peak during pubertal growth and regulate skeletal acquisition through stimulation of extracellular matrix production and increases in bone mineral density. During aging the activity of these hormones declines, a state called somatopaguss, which associates with deleterious effects on the musculoskeletal system. In this review, we will focus on GH/IGF-1 action in bone and cartilage. We will cover many studies that have utilized congenital ablation or overexpression of members of this axis, as well as cell-specific gene-targeting approaches used to unravel the nature of the GH/IGF-1 actions in the skeleton in vivo.
Collapse
Affiliation(s)
- Manisha Dixit
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, NY, 10010, USA
| | - Sher Bahadur Poudel
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, NY, 10010, USA
| | - Shoshana Yakar
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, NY, 10010, USA.
| |
Collapse
|
4
|
Dual delivery of stem cells and insulin-like growth factor-1 in coacervate-embedded composite hydrogels for enhanced cartilage regeneration in osteochondral defects. J Control Release 2020; 327:284-295. [PMID: 32763434 DOI: 10.1016/j.jconrel.2020.08.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/07/2020] [Accepted: 08/02/2020] [Indexed: 12/31/2022]
Abstract
Exogenous dual delivery of progenitor cell population and therapeutic growth factors (GFs) is one of alternative tissue engineering strategies for osteochondral tissue regeneration. In the present study, an implantable dual delivery platform was developed using coacervates (Coa) (i.e., a tertiary complex of poly(ethylene argininylaspartate diglyceride) (PEAD) polycation, heparin, and cargo insulin-like growth factor-1 (IGF-1), in thiolated gelatin (gelatin-SH)/ poly(ethylene glycol) diacrylate (PEGDA) interpenetrating network (IPN) hydrogels. Since Coa is able to protect cargo GF and maintain its long-term bioactivity, it is speculated that Coa-mediated delivery of chondrogenic factor IGF-1 with the aid of adipose-derived stem cells (ADSCs) would synergistically facilitate osteochondral tissue repair during physiological regeneration process. Our results indicate that gelatin-SH/PEGDA IPN hydrogels demonstrated biocompatibility and mechanical properties for a possible long-term transplantation, and PEAD-base Coa exhibited a sustained release of bioactive IGF-1 over 3 weeks. Subsequently, released IGF-1 from Coa could effectively induce chondrogenic differentiation of embedded ADSCs in the hydrogel, by showing enhanced glycosaminoglycan deposition and expression of chondrogenesis-associated genes. More importantly, at 12 weeks post-implantation in a rabbit full thickness osteochondral defect model, the quality of regenerative tissues in both chondral and subchondral layers was significantly improved in dual delivery of ADSC and IGF-1 in Coa encapsulated in gelatin-SH/PEGDA IPN hydrogels, as compared with a single delivery of ADSC only and a dual delivery without Coa. Therefore, we conclude that our Coa-embedded composite hydrogel platform could effectively augment osteochondral tissue regeneration holds promise for a feasible osteoarthritis therapeutic application.
Collapse
|
5
|
Bianchini E, Mancini F, Di Meo A, Stabile A, Buratta S, Moscati L, Pistilli A, Floridi C, Pepe M, Chiaradia E. Protective effects of platelet-rich plasma against lidocaine cytotoxicity on canine articular chondrocytes. Acta Vet Scand 2018; 60:63. [PMID: 30367652 PMCID: PMC6204030 DOI: 10.1186/s13028-018-0418-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 10/20/2018] [Indexed: 02/06/2023] Open
Abstract
Background Lidocaine (LD) is one of the most commonly used local anesthetics for performing arthroscopic surgery and managing of osteoarthritic pain in both human and veterinary medicine. However, over the last years, several studies have focused on the chondrotoxic effects of LD. In order to ensure that intra-articular lidocaine is safe to use, treatments aimed at mitigating chondrocyte death have recently been investigated. The aim of this study is to evaluate the possible protective effects of platelet-rich plasma (PRP) against LD cytotoxicity on canine articular chondrocytes. Results Articular canine chondrocytes, were exposed to 1% or 1.8% LD alone or in co-presence with 10% PRP for 30 min. In order to evaluate the effects of PRP pre-treatments, experiments were carried out on cells cultured in serum-free medium-or in medium supplemented with 10% PRP or 10% fetal bovine serum. Cell viability was evaluated by methyl thiazolyl tetrazolium assay and cell apoptosis was analyzed by flow cytometry using annexin V-fluorescein isothiocyanate/propidium iodide. The results showed that LD significantly reduced canine chondrocytes viability, probably due to apoptosis induction. Pre-treatment or the co-presence of PRP in the media restored the number of viable chondrocytes. The PRP also seemed to protect the cells from LD-induced apoptosis. Conclusions Pre-treatments and/or the simultaneous administration of PRP reduced LD-induced cytotoxicity in canine chondrocytes. Further in vivo studies are required to determine whether PRP can be used as a save protective treatment for dogs receiving intra-articular LD injections.
Collapse
|
6
|
Yu Q, Zhao B, He Q, Zhang Y, Peng X. microRNA‐206 is required for osteoarthritis development through its effect on apoptosis and autophagy of articular chondrocytes via modulating the phosphoinositide 3‐kinase/protein kinase B‐mTOR pathway by targeting insulin‐like growth factor‐1. J Cell Biochem 2018; 120:5287-5303. [PMID: 30335903 DOI: 10.1002/jcb.27803] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 09/10/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Qian Yu
- Department of Joint Surgery & Sports Medicine Qianfoshan Hospital of Shandong Province Jinan China
| | - Bei Zhao
- Department of Orthopaedics Liaocheng People’s Hospital and Liaocheng Clinical School of Taishan Medical University Liaocheng China
| | - Qi He
- Department of Blood Transfusion Shandong Provincial Hospital Jinan China
| | - Yuan Zhang
- Department of Geriatric Neurology Qianfoshan Hospital of Shandong Province Jinan China
| | - Xian‐Bo Peng
- Department of Joint Surgery & Sports Medicine Qianfoshan Hospital of Shandong Province Jinan China
| |
Collapse
|
7
|
He A, Ning Y, Wen Y, Cai Y, Xu K, Cai Y, Han J, Liu L, Du Y, Liang X, Li P, Fan Q, Hao J, Wang X, Guo X, Ma T, Zhang F. Use of integrative epigenetic and mRNA expression analyses to identify significantly changed genes and functional pathways in osteoarthritic cartilage. Bone Joint Res 2018; 7:343-350. [PMID: 29922454 PMCID: PMC5987683 DOI: 10.1302/2046-3758.75.bjr-2017-0284.r1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Aim Osteoarthritis (OA) is caused by complex interactions between genetic and environmental factors. Epigenetic mechanisms control the expression of genes and are likely to regulate the OA transcriptome. We performed integrative genomic analyses to define methylation-gene expression relationships in osteoarthritic cartilage. Patients and Methods Genome-wide DNA methylation profiling of articular cartilage from five patients with OA of the knee and five healthy controls was conducted using the Illumina Infinium HumanMethylation450 BeadChip (Illumina, San Diego, California). Other independent genome-wide mRNA expression profiles of articular cartilage from three patients with OA and three healthy controls were obtained from the Gene Expression Omnibus (GEO) database. Integrative pathway enrichment analysis of DNA methylation and mRNA expression profiles was performed using integrated analysis of cross-platform microarray and pathway software. Gene ontology (GO) analysis was conducted using the Database for Annotation, Visualization and Integrated Discovery (DAVID). Results We identified 1265 differentially methylated genes, of which 145 are associated with significant changes in gene expression, such as DLX5, NCOR2 and AXIN2 (all p-values of both DNA methylation and mRNA expression < 0.05). Pathway enrichment analysis identified 26 OA-associated pathways, such as mitogen-activated protein kinase (MAPK) signalling pathway (p = 6.25 × 10-4), phosphatidylinositol (PI) signalling system (p = 4.38 × 10-3), hypoxia-inducible factor 1 (HIF-1) signalling pathway (p = 8.63 × 10-3 pantothenate and coenzyme A (CoA) biosynthesis (p = 0.017), ErbB signalling pathway (p = 0.024), inositol phosphate (IP) metabolism (p = 0.025), and calcium signalling pathway (p = 0.032). Conclusion We identified a group of genes and biological pathwayswhich were significantly different in both DNA methylation and mRNA expression profiles between patients with OA and controls. These results may provide new clues for clarifying the mechanisms involved in the development of OA. Cite this article: A. He, Y. Ning, Y. Wen, Y. Cai, K. Xu, Y. Cai, J. Han, L. Liu, Y. Du, X. Liang, P. Li, Q. Fan, J. Hao, X. Wang, X. Guo, T. Ma, F. Zhang. Use of integrative epigenetic and mRNA expression analyses to identify significantly changed genes and functional pathways in osteoarthritic cartilage. Bone Joint Res 2018;7:343–350. DOI: 10.1302/2046-3758.75.BJR-2017-0284.R1.
Collapse
Affiliation(s)
- A He
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Y Ning
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Y Wen
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Y Cai
- Department of Orthopaedics, The First Affiliated Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - K Xu
- Department of Joint Surgery, Xi'an Hong-Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Y Cai
- Department of Joint Surgery, Xi'an Hong-Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - J Han
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - L Liu
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Y Du
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - X Liang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - P Li
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Q Fan
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - J Hao
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - X Wang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - X Guo
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - T Ma
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - F Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
8
|
Diao X, Wang J, Zhu H, He B. Overexpression of programmed cell death 5 in a mouse model of ovalbumin-induced allergic asthma. BMC Pulm Med 2016; 16:149. [PMID: 27846830 PMCID: PMC5109699 DOI: 10.1186/s12890-016-0317-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 11/10/2016] [Indexed: 11/10/2022] Open
Abstract
Background Programmed cell death 5 (PDCD5) was first identified as an apoptosis-promoting protein and involved in some autoimmune diseases and inflammatory processes. Our previous study demonstrated greater expression of serum PDCD5 in asthmatic patients than controls. This study aimed to further explore the significance of PDCD5 in mice with induced allergic asthma. Methods We divided 16 female mice into 2 groups: control (n = 8) and allergen (ovalbumin, OVA)-challenged mice (n = 8). The modified ovalbumin inhalation method was used to generate the allergic asthma mouse model, and the impact of OVA was assessed by histology of lung tissue and morphometry. The number of cells in bronchoalveolar lavage fluid (BALF) was detected. Pulmonary function was measured by pressure sensors. PDCD5 and active caspase-3 levels were detected. Results The expression of PDCD5 was higher with OVA challenge than for controls (p < 0.05). PDCD5 level was correlated with number of inflammatory cells in BALF and lung function. Moreover, active caspase-3 level was increased in the OVA-challenged mice (p < 0.001) and correlated with PDCD5 level (p = 0.000). Conclusions These data demonstrate an association between level of PDCD5 and asthma severity and indicate that PDCD5 may play a role in allergic asthma. Electronic supplementary material The online version of this article (doi:10.1186/s12890-016-0317-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaolin Diao
- Department of Respiratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Juan Wang
- Department of Respiratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Hong Zhu
- Department of Respiratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Bei He
- Department of Respiratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
9
|
Wang W, Song XW, Zhao CH. Roles of programmed cell death protein 5 in inflammation and cancer (Review). Int J Oncol 2016; 49:1801-1806. [PMID: 27826615 DOI: 10.3892/ijo.2016.3706] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 09/19/2016] [Indexed: 11/06/2022] Open
Abstract
PDCD5 (programmed cell death 5) is an apoptosis related gene cloned in 1999 from a human leukemic cell line. PDCD5 protein containing 125 amino acid (aa) residues sharing significant homology to the corresponding proteins of species. Decreased expression of PDCD5 has been found in many human tumors, including breast, gastric cancer, astrocytic glioma, chronic myelogenous leukemia and hepatocellular carcinoma. In recent years, increased number of studies have shown the functions and mechanisms of PDCD5 protein in cancer cells, such as paraptosis, cell cycle and immunoregulation. In the present review, we provide a comprehensive review on the role of PDCD5 in cancer tissues and cells. This review summarizes the recent studies of the roles of PDCD5 in inflammation and cancer. We mainly focus on discoveries related to molecular mechanisms of PDCD5 protein. We also discuss some discrepancies between the current studies. Overall, the current available data will open new perspectives for a better understanding of PDCD5 in cancer.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Xiao-Wen Song
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Cheng-Hai Zhao
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, P.R. China
| |
Collapse
|
10
|
Zhou Q, Li B, Zhao J, Pan W, Xu J, Chen S. IGF-I induces adipose derived mesenchymal cell chondrogenic differentiation in vitro and enhances chondrogenesis in vivo. In Vitro Cell Dev Biol Anim 2016; 52:356-364. [PMID: 26822434 DOI: 10.1007/s11626-015-9969-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 10/16/2015] [Indexed: 12/29/2022]
Abstract
Recent studies have demonstrated that insulin-like growth factor-1 (IGF-I) modulates bone mesenchymal stem cell chondrogenic differentiation independent of transforming growth factor beta (TGF-β) signaling in vitro. However, it is unclear whether IGF-I can solely modulate human adipose-derived mesenchymal cell (hAMC) chondrogenic differentiation, or whether it has additive effects with TGF-β1 to induce chondrogenic differentiation in vitro and development of mature cartilage in vivo. We investigated the effect of IGF-I on the induction of hAMC chondrogenic differentiation in the presence or absence of transforming growth factor beta 1 (TGF-β1) in vitro, and chondrogenesis of the induced hAMC in vivo. The results showed that IGF-I alone induced collagen type II, aggrecan, and Sox9 mRNA expression and collagen type II and aggrecan proteins expressions in hAMCs. Notably, there was greater mRNA expression of collagen type II, aggrecan and Sox9, and greater protein expression of collagen type II and aggrecan following TGF-β1 + IGF-I treatment, compared to either TGF-β1 or IGF-I-treated hAMCs. These results were confirmed in cartilage tissues derived from induced hAMCs. These findings indicate that IGF-I alone has the ability to induce chondrogenic differentiation and has additive effects with TGF-β1 to induce chondrogenic differentiation in vitro and in vivo.
Collapse
Affiliation(s)
- Quan Zhou
- Department of Orthopaedics, Huai'an Hospital Affiliated of Xuzhou Medical College and Huai'an Second Hospital, No. 62 Huaihai Road South, Huai'an, 223002, China
| | - Baojun Li
- Department of Joint Surgery, Second People's Hospital of Hunan Province, Changsha, 410007, China
| | - Jiali Zhao
- Department of Orthopaedics, Huai'an Hospital Affiliated of Xuzhou Medical College and Huai'an Second Hospital, No. 62 Huaihai Road South, Huai'an, 223002, China
| | - Wei Pan
- Department of Orthopaedics, Huai'an Hospital Affiliated of Xuzhou Medical College and Huai'an Second Hospital, No. 62 Huaihai Road South, Huai'an, 223002, China
| | - Jin Xu
- Department of Orthopaedics, Huai'an Hospital Affiliated of Xuzhou Medical College and Huai'an Second Hospital, No. 62 Huaihai Road South, Huai'an, 223002, China
| | - Sumei Chen
- Department of Orthopaedics, Huai'an Hospital Affiliated of Xuzhou Medical College and Huai'an Second Hospital, No. 62 Huaihai Road South, Huai'an, 223002, China.
| |
Collapse
|
11
|
Li G, Ma D, Chen Y. Cellular functions of programmed cell death 5. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:572-80. [PMID: 26775586 DOI: 10.1016/j.bbamcr.2015.12.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 12/24/2015] [Accepted: 12/29/2015] [Indexed: 01/01/2023]
Abstract
Programmed cell death 5 (PDCD5) was originally identified as an apoptosis-accelerating protein that is widely expressed and has been well conserved during the process of evolution. PDCD5 has complex biological functions, including programmed cell death and immune regulation. It can accelerate apoptosis in different type of cells in response to different stimuli. During this process, PDCD5 rapidly translocates from the cytoplasm to the nucleus. PDCD5 regulates the activities of TIP60, HDAC3, MDM2 and TP53 transcription factors. These proteins form part of a signaling network that is disrupted in most, if not all, cancer cells. Recent evidence suggests that PDCD5 participates in immune regulation by promoting regulatory T cell function via the PDCD5-TIP60-FOXP3 pathway. The stability and expression of PDCD5 are finely regulated by other molecules, such as NF-κB p65, OTUD5, YAF2 and DNAJB1. PDCD5 is phosphorylated by CK2 at Ser119, which is required for nuclear translocation in response to genotoxic stress. In this review, we describe what is known about PDCD5 and its cellular functions.
Collapse
Affiliation(s)
- Ge Li
- Department of Immunology, Peking University School of Basic Medical Sciences, 38 Xueyuan Road, Beijing 100191, China; Center for Human Disease Genomics, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Dalong Ma
- Department of Immunology, Peking University School of Basic Medical Sciences, 38 Xueyuan Road, Beijing 100191, China; Center for Human Disease Genomics, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Yingyu Chen
- Department of Immunology, Peking University School of Basic Medical Sciences, 38 Xueyuan Road, Beijing 100191, China; Center for Human Disease Genomics, Peking University, 38 Xueyuan Road, Beijing 100191, China.
| |
Collapse
|
12
|
Xiao J, Li G, Hu J, Qu L, Ma D, Chen Y. Anti-inflammatory effects of recombinant human PDCD5 (rhPDCD5) in a rat collagen-induced model of arthritis. Inflammation 2015; 38:70-8. [PMID: 25178696 PMCID: PMC4312386 DOI: 10.1007/s10753-014-0008-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Programmed cell death 5 (PDCD5) was first identified as a gene upregulated in cells undergoing apoptosis. We recently demonstrated the inhibitory effect of PDCD5 on experimentally induced autoimmune encephalomyelitis. In this study, we investigated the anti-inflammatory effects of recombinant human PDCD5 (rhPDCD5) in a rat collagen-induced arthritis (CIA) model. We find that vaccination of collagen II (CII) induced CIA rats with rhPDCD5 significantly delayed the occurrence and reduced the severity of CIA rats. rhPDCD5 also restored the loss of Foxp3+ regulatory T (Treg) cells and decreased the population of Th1 and Th17 in CIA rats. Simultaneously, rhPDCD5 treatment suppressed the production of pro-inflammatory cytokines (interleukin (IL)-6, IL-17A, tumor necrosis factor-α (TNF-α), and interferon gamma (IFN-γ)) and increased the secretion of anti-inflammatory cytokines (transforming growth factor beta 1 (TGF-β1) and IL-10) in CIA rats. In addition, rhPDCD5 inhibited the ability of CII to induce proliferation of splenocytes and lymph node cells (LNCs) and promoted the CII-activated CD4+ cell apoptosis. These results of rhPDCD5-treated CIA rats were similar with those of recombinant human TNF-α receptor IgG Fc (rhTNFR:Fc). Thus, to our knowledge, we provide the first evidence that rhPDCD5 may be an efficient approach to diminishing exacerbated immune responses in CIA, indicating its therapeutic potential in the treatment of rheumatoid arthritis and other autoimmune diseases.
Collapse
Affiliation(s)
- Juan Xiao
- Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing, 100191, China
| | | | | | | | | | | |
Collapse
|
13
|
Fan GL, Yao Y, Yao L, Li Y. PDCD5 transfection increases cisplatin sensitivity and decreases invasion in hepatic cancer cells. Oncol Lett 2014; 9:411-417. [PMID: 25436001 PMCID: PMC4246700 DOI: 10.3892/ol.2014.2645] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 07/25/2014] [Indexed: 01/08/2023] Open
Abstract
Low expression levels of the programmed cell death 5 (PDCD5) gene have been reported in numerous human cancers, however, PDCD5 expression has not been investigated in hepatic cancer. The present study aims to investigate the biological behavior of PDCD5 overexpression in hepatocellular carcinoma (HCC) cells. The PDCD5 gene was stably transfected into the HepG2 HCC cell line (HepG2-PDCD5), and the expression levels of PDCD5 were examined by quantitative polymerase chain reaction and western blotting. An MTT assay was used to assess the cellular proliferating ability, and propidium iodide (PI) staining was used to evaluate the cell cycle by flow cytometry. The cells were incubated with 2 ng/ml transforming growth factor (TGF)-β for 7 days in order to induce invasion and epithelial-mesenchymal transition (EMT). Apoptosis was measured by Annexin V-fluorescein isothiocyanate and PI double labeling. A Boyden chamber invasion assay was carried out to detect tumor invasion. Western blotting was performed to detect the protein expression levels of PDCD5, insulin-like growth factor (IGF)-1 and the EMT marker, Snail. The results showed that the HepG2-PDCD5 cells exhibited slower proliferation rates and high G2/M cell numbers compared with those of the HepG2 and HepG2-Neo controls (P<0.05). The PDCD5 transfected cells showed higher sensitivity to cisplatin treatment than the HepG2-Neo cells, with a higher p53 protein expression level. PDCD5 overexpression can attenuate tumor invasion, EMT and the level of IGF-1 protein induced by TGF-β treatment. In conclusion, stable transfection of the PDCD5 gene can inhibit growth and induce cell cycle arrest in HepG2 cells, and its also notably improves the apoptosis-inducing effects of cisplatin, and reverses invasion and EMT induced by TGF-β. The use of PDCD5 is a novel strategy for improving the chemotherapeutic effects on HCC.
Collapse
Affiliation(s)
- Gui-Ling Fan
- Department of Gastroenterology, Shandong Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Yong Yao
- Department of General Surgery, The First People's Hospital of Shanxian County, Heze, Shandong 274300, P.R. China
| | - Li Yao
- Department of Dentistry, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250001, P.R. China
| | - Yun Li
- Department of Nursing, The First People's Hospital of Shanxian County, Heze, Shandong 274300, P.R. China
| |
Collapse
|
14
|
Borrisser-Pairó F, Antúnez E, Tobías E, Fernández-Solà J. Insulin-like growth factor 1 myocardial expression decreases in chronic alcohol consumption. Regen Med Res 2013; 1:3. [PMID: 25984322 PMCID: PMC4375930 DOI: 10.1186/2050-490x-1-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Accepted: 04/02/2013] [Indexed: 01/21/2023] Open
Abstract
Background Alcoholic cardiomyopathy (CMP) is one of the major complications of chronic excessive alcohol consumption. The pathogenic mechanisms implicated are diverse, inducing functional and structural changes in the myocardium. Insulin-like Growth Factor 1 (IGF-1) plays an important role in modulating the cell cycle, and helps the differentiation and proliferation of cardiac tissue inhibiting apoptosis. Experimental studies have suggested the role of IGF-1 in alcohol-induced cardiac damage. The aim of the present study was to determine the effect of chronic alcohol consumption on IGF-1 myocardial expression and to compare this expression in cases of hypertension and other cardiac diseases. Methods We studied heart samples from human organ donors: 10 healthy donors, 16 with hypertension, 23 with chronic alcohol consumption and 7 with other causes of cardiac disease. IGF-1 myocardial expression was evaluated with a specific immunohistochemistry assay using a semi-quantitative method. Results A significant decrease in IGF-1 myocardial expression was observed comparing all the cases included with control donors. This decrease in IGF-1 myocardial expression was significantly lower in the group of donors with chronic alcohol consumption compared to controls. On group evaluation according to the presence of CMP, donors with chronic alcohol consumption without CMP presented significantly lower IGF-1 expression than controls, whereas donors with chronic alcohol consumption with CMP showed a downward trend without achieving significance. Conclusions Chronic alcohol consumption significantly reduces IGF-1 myocardial expression. This decrease induced by alcohol is partially compensated in the presence of structural myocardial damage.
Collapse
Affiliation(s)
- Francesc Borrisser-Pairó
- Alcohol Research Unit. Hospital Clínic. Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Emilia Antúnez
- Alcohol Research Unit. Hospital Clínic. Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Ester Tobías
- Alcohol Research Unit. Hospital Clínic. Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Joaquim Fernández-Solà
- Alcohol Research Unit. Hospital Clínic. Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). Department of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
15
|
Xiao J, Liu C, Li G, Peng S, Hu J, Qu L, Lv P, Zhang Y, Ma D, Chen Y. PDCD5 negatively regulates autoimmunity by upregulating FOXP3(+) regulatory T cells and suppressing Th17 and Th1 responses. J Autoimmun 2013; 47:34-44. [PMID: 24012345 DOI: 10.1016/j.jaut.2013.08.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 08/12/2013] [Accepted: 08/12/2013] [Indexed: 10/26/2022]
Abstract
Maintenance of FOXP3 protein expression is crucial for differentiation and maturation of regulatory T (Treg) cells, which play important roles in immune homeostasis and immune tolerance. We demonstrate here that PDCD5 interacts with FOXP3, increases acetylation of FOXP3 in synergy with Tip60 and enhances the repressive function of FOXP3. In PDCD5 transgenic (PDCD5tg) mice, overexpression of PDCD5 enhanced the level of FOXP3 protein and percentage of CD4(+)CD25(+)FOXP3(+) cells. Naïve CD4(+) T cells from PDCD5tg mice were more sensitive to TGF-β-induced Treg polarization and expansion. These induced Tregs retained normal suppressive function in vitro. Severity of experimentally-induced autoimmune encephalomyelitis (EAE) in PDCD5tg mice was significantly reduced relative to that of wild-type mice. The beneficial effect of PDCD5 likely resulted from increases of Treg cell frequency, accompanied by a reduction of the predominant pathogenic Th17/Th1 response. Activation-induced cell death enhanced by PDCD5 was also linked to this process. This is the first report revealing that PDCD5 activity in T cells suppresses autoimmunity by modulating Tregs. This study suggests that PDCD5 serves as a guardian of immunological functions and that the PDCD5-FOXP3-Treg axis may be a therapeutic target for autoimmunity.
Collapse
Affiliation(s)
- Juan Xiao
- Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing 100191, China; Peking University Center for Human Disease Genomics, Peking University, Beijing 100191, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|