1
|
Bonfield TL, Lazarus HM. Human mesenchymal stem cell therapy: Potential advances for reducing cystic fibrosis infection and organ inflammation. Best Pract Res Clin Haematol 2025; 38:101602. [PMID: 40274338 DOI: 10.1016/j.beha.2025.101602] [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: 11/20/2024] [Revised: 02/11/2025] [Accepted: 03/04/2025] [Indexed: 04/26/2025]
Abstract
Innovation in cystic fibrosis (CF) supportive care, including implementing new antimicrobial agents, improved physiotherapy, and highly effective modulators therapy, has advanced patient survival into the 4th and 5th decades of life. However, even with these remarkable improvements in therapy, CF patients continue to suffer from pulmonary infection and other visceral organ complications associated with long-term deficient cystic fibrosis transmembrane conductance regulator (CFTR) expression. Human mesenchymal stem cells (MSCs) have been utilized in tissue engineering based upon their capacity to provide structural components of mesenchymal tissues. An alternative role of MSCs, however is their versatile utilization as cell-based infusion powerhouses due to the unique capacity to deliver milieu specific soluble biologic factors, promoting immune supportive antimicrobial and anti-inflammatory potency. MSCs derived from umbilical cord blood, bone marrow, adipose and other tissues can be expanded in ex vivo using good manufacturing procedure facilities for a safe, unique therapeutic to reduce and limit CF infection and facilitate the resolution of multi-organ inflammation. In our efforts, we conducted extensive preclinical development and validation of an allogeneic derived bone marrow derived MSC product in preparation for a clinical trial in CF. In this process, potency models were developed to ensure the functional capacity of the MSC product to provide clinical benefit. In vitro, murine in vivo and patient tissue ex vivo potency models were utilized to follow MSC anti-infective and anti-inflammatory potency associated with the CFTR deficient environment. We showed in our "First in CF" clinical trial that the allogeneic MSCs obtained from healthy volunteer bone marrow samples were safe. The advent of improved CF care measures and exciting new small molecules has changed the survival and morbidity phenotype of patients with CF, however, there are CF patients who cannot tolerate or have genotypes that are non-responsive to modulators. Additionally, even with the small molecule therapy, CF patients are living longer, but without genetic correction, with the CF disease manifestation aggravated by the continuance of pre-existing CFTR-associated clinical issues such as ongoing inflammation. MSCs secrete bio-active factors that enhance and protect tissue function and can promote "self-immune" regulation. These properties can provide therapeutic support for the traditional and changing face of CF disease clinical complications. Further, MSC-derived bio-active factors can directly mitigate colonizing pathogens' survival by producing antimicrobial peptides (AMPs) which change the pathogen surface and increase host recognition, elimination, and sensitivity to antibiotics. Herein, we review the potential of MSC therapeutics for treating many facets of CF, emphasizing the potential for providing great additive therapeutics for managing morbidity and quality of life.
Collapse
Affiliation(s)
- Tracey L Bonfield
- Genetics and Genome Sciences, National Center for Regenerative Medicine, Pediatrics and Pathology, Case Western Reserve University, Cleveland, Ohio, 44106, USA.
| | - Hillard M Lazarus
- Department of Medicine, Hematology and National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| |
Collapse
|
2
|
Zhang R, Zhang Y, Xiao H, Liu Q, Zhao M. Knockout IL4I1 affects macrophages to improve poor efficacy of CD19 CAR-T combined with PD-1 inhibitor in relapsed/refractory diffuse large B-cell lymphoma. J Transl Med 2025; 23:105. [PMID: 39844281 PMCID: PMC11752997 DOI: 10.1186/s12967-024-06028-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/23/2024] [Indexed: 01/30/2025] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy plays a critical role in the treatment of B-cell hematologic malignancies. The combination of PD-1 inhibitors and CAR-T has shown encouraging results in treating patients with relapsed/refractory (R/R) diffuse large B-cell lymphoma (DLBCL). However, there are still cases where treatment is ineffective. This study aimed to investigate the role of IL4I1 in the poor efficacy of CD19 CAR-T combined with PD-1 inhibitors in R/R DLBCL and to explore potential mechanisms. Transcriptomic and metabolomic correlation analyses were performed on tumor tissue from DLBCL patients. We employed an in vitro co-culture system consisting of Pfeiffer cells, CD19 CAR-T and macrophages to investigate the underlying mechanisms. It was found that IL4I1 levels were significantly increased in the tumor tissues of R/R DLBCL patients compared to responders. Correlation analysis revealed a positive association between IL4I1 and tryptophan (Trp)-kynurenic acid (Kyn) related metabolites. In the in vitro co-culture model, the presence of IL4I1 inhibited the cytotoxicity of CAR-T cells. Depletion of IL4I1 disrupted the IDO-AHR-Kyn signaling pathway, thereby enhancing the effectiveness of PD-1 inhibitors in combination with CD19 CAR-T for DLBCL treatment. CAR-T-mediated cytotoxicity was significantly inhibited when IL4I1 was present in the in vitro co-culture model. These findings suggest that IL4I1 may be a contributing factor to poor prognosis in R/R DLBCL patients. IL4I1 expression enhances immunosuppression via the IDO-AHR-Kyn pathway, inhibiting the effectiveness of PD-1 inhibitors combined with CD19 CAR-T. Therefore, suppression of IL4I1 may represent a potential target for combination therapy in DLBCL.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Antigens, CD19/metabolism
- Antigens, CD19/immunology
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/metabolism
- Macrophages/metabolism
- Macrophages/drug effects
- Immunotherapy, Adoptive
- Receptors, Chimeric Antigen/metabolism
- Cell Line, Tumor
- Immune Checkpoint Inhibitors/therapeutic use
- Immune Checkpoint Inhibitors/pharmacology
- Female
- Recurrence
- Male
- Coculture Techniques
- Neoplasm Recurrence, Local
- Middle Aged
Collapse
Affiliation(s)
- Rui Zhang
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Yi Zhang
- Department of Hematology, First Center Clinic College of Tianjin Medical University, Tianjin, China
| | | | | | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China.
- Department of Hematology, First Center Clinic College of Tianjin Medical University, Tianjin, China.
| |
Collapse
|
3
|
Szychowski KA, Skóra B. Elastin-derived peptides (EDPs) affect gene and protein expression in human mesenchymal stem cells (hMSCs) - preliminary study. Cytokine 2024; 182:156725. [PMID: 39106575 DOI: 10.1016/j.cyto.2024.156725] [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: 06/07/2024] [Revised: 07/25/2024] [Accepted: 08/01/2024] [Indexed: 08/09/2024]
Abstract
During the aging process, elastin is degraded and the level of elastin-derived peptides (EDPs) successively increases. The main peptide released from elastin during its degradation is a peptide with the VGVAPG sequence. To date, several papers have described that EDPs or elastin-like peptides (ELPs) affect human mesenchymal stem cells (hMSCs) derived from different tissues. Unfortunately, despite the described effect of EDPs or ELPs on the hMSC differentiation process, the mechanism of action of these peptides has not been elucidated. Therefore, the aim of the present study was to evaluate the impact of the VGVAPG and VVGPGA peptides on the hMSC stemness marker and elucidation of the mechanism of action of these peptides. Our data show that both studied peptides (VGVAPG and VVGPGA) act with the involvement of ERK1/2 and c-SRC kinases. However, their mechanism of activation is probably different in hMSCs derived from adipose tissue. Both studied peptides increase the KI67 protein level in hMSCs, but this is not accompanied with cell proliferation. Moreover, the changes in the NANOG and c-MYC protein expression and in the SOX2 and POU5F1 mRNA expression suggest that EDPs reduced the hMSC stemness properties and could initiate cell differentiation. The initiation of differentiation was evidenced by changes in the expression of AhR and PPARγ protein as well as specific genes (ACTB, TUBB3) and proteins (β-actin, RhoA) involved in cytoskeleton remodeling. Our data suggest that the presence of EDPs in tissue can initiate hMSC differentiation into more tissue-specific cells.
Collapse
Affiliation(s)
- Konrad A Szychowski
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, St. Sucharskiego 2, 35-225 Rzeszow, Poland.
| | - Bartosz Skóra
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, St. Sucharskiego 2, 35-225 Rzeszow, Poland
| |
Collapse
|
4
|
Lai Z, Shu Q, Song Y, Tang A, Tian J. Effect of DNA methylation on the osteogenic differentiation of mesenchymal stem cells: concise review. Front Genet 2024; 15:1429844. [PMID: 39015772 PMCID: PMC11250479 DOI: 10.3389/fgene.2024.1429844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have promising potential for bone tissue engineering in bone healing and regeneration. They are regarded as such due to their capacity for self-renewal, multiple differentiation, and their ability to modulate the immune response. However, changes in the molecular pathways and transcription factors of MSCs in osteogenesis can lead to bone defects and metabolic bone diseases. DNA methylation is an epigenetic process that plays an important role in the osteogenic differentiation of MSCs by regulating gene expression. An increasing number of studies have demonstrated the significance of DNA methyltransferases (DNMTs), Ten-eleven translocation family proteins (TETs), and MSCs signaling pathways about osteogenic differentiation in MSCs. This review focuses on the progress of research in these areas.
Collapse
Affiliation(s)
- Zhihao Lai
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qing Shu
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yue Song
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- College of Sports Medicine, Wuhan Sports University, Wuhan, China
| | - Ao Tang
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- College of Sports Medicine, Wuhan Sports University, Wuhan, China
| | - Jun Tian
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
5
|
Wu X, Tang Y, Lu X, Liu Y, Liu X, Sun Q, Wang L, Huang W, Liu A, Liu L, Chao J, Zhang X, Qiu H. Endothelial cell-derived extracellular vesicles modulate the therapeutic efficacy of mesenchymal stem cells through IDH2/TET pathway in ARDS. Cell Commun Signal 2024; 22:293. [PMID: 38802896 PMCID: PMC11129421 DOI: 10.1186/s12964-024-01672-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a severe and fatal disease. Although mesenchymal stem cell (MSC)-based therapy has shown remarkable efficacy in treating ARDS in animal experiments, clinical outcomes have been unsatisfactory, which may be attributed to the influence of the lung microenvironment during MSC administration. Extracellular vesicles (EVs) derived from endothelial cells (EC-EVs) are important components of the lung microenvironment and play a crucial role in ARDS. However, the effect of EC-EVs on MSC therapy is still unclear. In this study, we established lipopolysaccharide (LPS) - induced acute lung injury model to evaluate the impact of EC-EVs on the reparative effects of bone marrow-derived MSC (BM-MSC) transplantation on lung injury and to unravel the underlying mechanisms. METHODS EVs were isolated from bronchoalveolar lavage fluid of mice with LPS - induced acute lung injury and patients with ARDS using ultracentrifugation. and the changes of EC-EVs were analysed using nanoflow cytometry analysis. In vitro assays were performed to establish the impact of EC-EVs on MSC functions, including cell viability and migration, while in vivo studies were performed to validate the therapeutic effect of EC-EVs on MSCs. RNA-Seq analysis, small interfering RNA (siRNA), and a recombinant lentivirus were used to investigate the underlying mechanisms. RESULTS Compared with that in non-ARDS patients, the quantity of EC-EVs in the lung microenvironment was significantly greater in patients with ARDS. EVs derived from lipopolysaccharide-stimulated endothelial cells (LPS-EVs) significantly decreased the viability and migration of BM-MSCs. Furthermore, engrafting BM-MSCs pretreated with LPS-EVs promoted the release of inflammatory cytokines and increased pulmonary microvascular permeability, aggravating lung injury. Mechanistically, LPS-EVs reduced the expression level of isocitrate dehydrogenase 2 (IDH2), which catalyses the formation of α-ketoglutarate (α-KG), an intermediate product of the tricarboxylic acid (TCA) cycle, in BM-MSCs. α-KG is a cofactor for ten-eleven translocation (TET) enzymes, which catalyse DNA hydroxymethylation in BM-MSCs. CONCLUSIONS This study revealed that EC-EVs in the lung microenvironment during ARDS can affect the therapeutic efficacy of BM-MSCs through the IDH2/TET pathway, providing potential strategies for improving the therapeutic efficacy of MSC-based therapy in the clinic.
Collapse
Affiliation(s)
- Xiao Wu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Ying Tang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Xinxing Lu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Yigao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Xu Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Qin Sun
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Lu Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Wei Huang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Airan Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Ling Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Jie Chao
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
- Department of Physiology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xiwen Zhang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China.
| | - Haibo Qiu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| |
Collapse
|
6
|
Nguyen TM, Geng X, Wei Y, Ye L, Garry DJ, Zhang J. Single-cell RNA sequencing analysis identifies one subpopulation of endothelial cells that proliferates and another that undergoes the endothelial-mesenchymal transition in regenerating pig hearts. Front Bioeng Biotechnol 2024; 11:1257669. [PMID: 38288246 PMCID: PMC10823534 DOI: 10.3389/fbioe.2023.1257669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/04/2023] [Indexed: 01/31/2024] Open
Abstract
Background: In our previous work, we demonstrated that when newborn pigs undergo apical resection (AR) on postnatal day 1 (P1), the animals' hearts were completely recover from a myocardial infarction (MI) that occurs on postnatal day 28 (P28); single-nucleus RNA sequencing (snRNAseq) data suggested that this recovery was achieved by regeneration of pig cardiomyocyte subpopulations in response to MI. However, coronary vasculature also has a key role in promoting cardiac repair. Method: Thus, in this report, we used autoencoder algorithms to analyze snRNAseq data from endothelial cells (ECs) in the hearts of the same animals. Main results: Our results identified five EC clusters, three composed of vascular ECs (VEC1-3) and two containing lymphatic ECs (LEC1-2). Cells from VEC1 expressed elevated levels of each of five cell-cyclespecific markers (Aurora Kinase B [AURKB], Marker of Proliferation Ki-67 [MKI67], Inner Centromere Protein [INCENP], Survivin [BIRC5], and Borealin [CDCA8]), as well as a number of transcription factors that promote EC proliferation, while (VEC3 was enriched for genes that regulate intercellular junctions, participate in transforming growth factor β (TGFβ), bone morphogenic protein (BMP) signaling, and promote the endothelial mesenchymal transition (EndMT). The remaining VEC2 did not appear to participate directly in the angiogenic response to MI, but trajectory analyses indicated that it may serve as a reservoir for the generation of VEC1 and VEC3 ECs in response to MI. Notably, only the VEC3 cluster was more populous in regenerating (i.e., ARP1MIP28) than non-regenerating (i.e., MIP28) hearts during the 1-week period after MI induction, which suggests that further investigation of the VEC3 cluster could identify new targets for improving myocardial recovery after MI. Histological analysis of KI67 and EndMT marker PDGFRA demonstrated that while the expression of proliferation of endothelial cells was not significantly different, expression of EndMT markers was significantly higher among endothelial cells of ARP1MIP28 hearts compared to MIP28 hearts, which were consistent with snRNAseq analysis of clusters VEC1 and VEC3. Furthermore, upregulated secrete genes by VEC3 may promote cardiomyocyte proliferation via the Pi3k-Akt and ERBB signaling pathways, which directly contribute to cardiac muscle regeneration. Conclusion: In regenerative heart, endothelial cells may express EndMT markers, and this process could contribute to regeneration via a endothelial-cardiomyocyte crosstalk that supports cardiomyocyte proliferation.
Collapse
Affiliation(s)
- Thanh Minh Nguyen
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Xiaoxiao Geng
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yuhua Wei
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Lei Ye
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Daniel J. Garry
- Department of Medicine, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Jianyi Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Medicine, Cardiovascular Diseases, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
7
|
Zhang R, Liu Q, Lyu C, Gao X, Ma W. Knockdown SENP1 Suppressed the Angiogenic Potential of Mesenchymal Stem Cells by Impacting CXCR4-Regulated MRTF-A SUMOylation and CCN1 Expression. Biomedicines 2023; 11:biomedicines11030914. [PMID: 36979893 PMCID: PMC10046070 DOI: 10.3390/biomedicines11030914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/18/2023] [Accepted: 02/22/2023] [Indexed: 03/18/2023] Open
Abstract
The angiogenic potential of mesenchymal stem cells (MSCs) is critical for adult vascular regeneration and repair, which is regulated by various growth factors and cytokines. In the current study, we report that knockdown SUMO-specific peptidase 1 (SENP1) stimulated the SUMOylation of MRTF-A and prevented its translocation into the nucleus, leading to downregulation of the cytokine and angiogenic factor CCN1, which significantly impacted MSC-mediated angiogenesis and cell migration. Further studies showed that SENP1 knockdown also suppressed the expression of a chemokine receptor CXCR4, and overexpression of CXCR4 could partially abrogate MRTF-A SUMOylation and reestablish the CCN1 level. Mutation analysis confirmed that SUMOylation occurred on three lysine residues (Lys-499, Lys-576, and Lys-624) of MRTF-A. In addition, SENP1 knockdown abolished the synergistic co-activation of CCN1 between MRTF-A and histone acetyltransferase p300 by suppressing acetylation on histone3K9, histone3K14, and histone4. These results revealed an important signaling pathway to regulate MSC differentiation and angiogenesis by MRTF-A SUMOylation involving cytokine/chemokine activities mediated by CCN1 and CXCR4, which may potentially impact a variety of cellular processes such as revascularization, wound healing, and progression of cancer.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Hematology, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Qingxi Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
- Department of Chemical and Biological Engineering, Qilu Institute of Technology, Jinan 250200, China
- Correspondence: (Q.L.); (W.M.)
| | - Cuicui Lyu
- Department of Hematology, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Xing Gao
- Department of Chemical and Biological Engineering, Qilu Institute of Technology, Jinan 250200, China
| | - Wenjian Ma
- Department of Chemical and Biological Engineering, Qilu Institute of Technology, Jinan 250200, China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Correspondence: (Q.L.); (W.M.)
| |
Collapse
|
8
|
Zhang R, Liu Q, Zhou S, He H, Zhao M, Ma W. Mesenchymal stem cell suppresses the efficacy of CAR-T toward killing lymphoma cells by modulating the microenvironment through stanniocalcin-1. eLife 2023; 12:82934. [PMID: 36779699 PMCID: PMC10019890 DOI: 10.7554/elife.82934] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 02/12/2023] [Indexed: 02/14/2023] Open
Abstract
Stem cells play critical roles both in the development of cancer and therapy resistance. Although mesenchymal stem cells (MSCs) can actively migrate to tumor sites, their impact on chimeric antigen receptor modified T cell (CAR-T) immunotherapy has been little addressed. Using an in vitro cell co-culture model including lymphoma cells and macrophages, here we report that CAR-T cell-mediated cytotoxicity was significantly inhibited in the presence of MSCs. MSCs caused an increase of CD4+ T cells and Treg cells but a decrease of CD8+ T cells. In addition, MSCs stimulated the expression of indoleamine 2,3-dioxygenase and programmed cell death-ligand 1 which contributes to the immune-suppressive function of tumors. Moreover, MSCs suppressed key components of the NLRP3 inflammasome by modulating mitochondrial reactive oxygen species release. Interestingly, all these suppressive events hindering CAR-T efficacy could be abrogated if the stanniocalcin-1 (STC1) gene, which encodes the glycoprotein hormone STC-1, was knockdown in MSC. Using xenograft mice, we confirmed that CAR-T function could also be inhibited by MSC in vivo, and STC1 played a critical role. These data revealed a novel function of MSC and STC-1 in suppressing CAR-T efficacy, which should be considered in cancer therapy and may also have potential applications in controlling the toxicity arising from the excessive immune response.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Hematology, Tianjin First Central Hospital, School of Medicine, Nankai UniversityTianjinChina
| | - Qingxi Liu
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai UniversityTianjinChina
- Qilu Institute of TechnologyShandongChina
| | - Sa Zhou
- College of Biotechnology, Tianjin University of Science and TechnologyTianjinChina
| | - Hongpeng He
- College of Biotechnology, Tianjin University of Science and TechnologyTianjinChina
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, School of Medicine, Nankai UniversityTianjinChina
| | - Wenjian Ma
- Qilu Institute of TechnologyShandongChina
- College of Biotechnology, Tianjin University of Science and TechnologyTianjinChina
| |
Collapse
|
9
|
Epigenetic therapy targeting bone marrow mesenchymal stem cells for age-related bone diseases. Stem Cell Res Ther 2022; 13:201. [PMID: 35578312 PMCID: PMC9109405 DOI: 10.1186/s13287-022-02852-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/14/2022] [Indexed: 02/08/2023] Open
Abstract
As global aging accelerates, the prevention and treatment of age-related bone diseases are becoming a critical issue. In the process of senescence, bone marrow mesenchymal stem cells (BMSCs) gradually lose the capability of self-renewal and functional differentiation, resulting in impairment of bone tissue regeneration and disorder of bone tissue homeostasis. Alteration in epigenetic modification is an essential factor of BMSC dysfunction during aging. Its transferability and reversibility provide the possibility to combat BMSC aging by reversing age-related modifications. Emerging evidence demonstrates that epigenetic therapy based on aberrant epigenetic modifications could alleviate the senescence and dysfunction of stem cells. This review summarizes potential therapeutic targets for BMSC aging, introduces some potential approaches to alleviating BMSC aging, and analyzes its prospect in the clinical application of age-related bone diseases.
Collapse
|
10
|
Liu Y, Gan L, Cui DX, Yu SH, Pan Y, Zheng LW, Wan M. Epigenetic regulation of dental pulp stem cells and its potential in regenerative endodontics. World J Stem Cells 2021; 13:1647-1666. [PMID: 34909116 PMCID: PMC8641018 DOI: 10.4252/wjsc.v13.i11.1647] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/07/2021] [Accepted: 11/03/2021] [Indexed: 02/06/2023] Open
Abstract
Regenerative endodontics (RE) therapy means physiologically replacing damaged pulp tissue and regaining functional dentin–pulp complex. Current clinical RE procedures recruit endogenous stem cells from the apical papilla, periodontal tissue, bone marrow and peripheral blood, with or without application of scaffolds and growth factors in the root canal space, resulting in cementum-like and bone-like tissue formation. Without the involvement of dental pulp stem cells (DPSCs), it is unlikely that functional pulp regeneration can be achieved, even though acceptable repair can be acquired. DPSCs, due to their specific odontogenic potential, high proliferation, neurovascular property, and easy accessibility, are considered as the most eligible cell source for dentin–pulp regeneration. The regenerative potential of DPSCs has been demonstrated by recent clinical progress. DPSC transplantation following pulpectomy has successfully reconstructed neurovascularized pulp that simulates the physiological structure of natural pulp. The self-renewal, proliferation, and odontogenic differentiation of DPSCs are under the control of a cascade of transcription factors. Over recent decades, epigenetic modulations implicating histone modifications, DNA methylation, and noncoding (nc)RNAs have manifested as a new layer of gene regulation. These modulations exhibit a profound effect on the cellular activities of DPSCs. In this review, we offer an overview about epigenetic regulation of the fate of DPSCs; in particular, on the proliferation, odontogenic differentiation, angiogenesis, and neurogenesis. We emphasize recent discoveries of epigenetic molecules that can alter DPSC status and promote pulp regeneration through manipulation over epigenetic profiles.
Collapse
Affiliation(s)
- Ying Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Lu Gan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Di-Xin Cui
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Si-Han Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yue Pan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Li-Wei Zheng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Mian Wan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
11
|
Xu Z, Cao J, Zhao Z, Qiao Y, Liu X, Zhong J, Wang B, Suo G. A functional extracellular matrix biomaterial enriched with VEGFA and bFGF as vehicle of human umbilical cord mesenchymal stem cells in skin wound healing. Biomed Mater 2021; 17. [PMID: 34749352 DOI: 10.1088/1748-605x/ac37b0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/08/2021] [Indexed: 11/12/2022]
Abstract
The construction of microvascular network is one of the greatest challenges for tissue engineering and cell therapy. Endothelial cells are essential for the construction of network of blood vessels. However, their application meets challenges in clinic due to the limited resource of autologous endothelium. Mesenchymal stem cells can effectively promote the angiogenesis in ischemic tissues for their abilities of endothelial differentiation and paracrine, and abundant sources. Extracellular matrix (ECM) has been widely used as an ideal biomaterial to mimic cellular microenvironment for tissue engineering due to its merits of neutrality, good biocompatibility, degradability, and controllability. In this study, a functional cell derived ECM biomaterial enriched with VEGFA and bFGF by expressing the collagen-binding domain fused factor genes in host cells was prepared. This material could induce endothelial differentiation of human umbilical cord mesenchymal stem cells (hUCMSCs) and promote angiogenesis, which may improve the healing effect of skin injury. Our research not only provides a functional ECM material to inducing angiogenesis by inducing endothelial differentiation of hUCMSCs, but also shed light on the ubiquitous approaches to endow ECM materials different functions by enriching different factors. This study will benefit tissue engineering and regenerative medicine researches.
Collapse
Affiliation(s)
- Zhongjuan Xu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, Anhui, People's Republic of China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, People's Republic of China
| | - Junjun Cao
- Livingchip Lnc., Nanjing 211112, Jiangsu, People's Republic of China
| | - Zhe Zhao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, People's Republic of China
| | - Yong Qiao
- Livingchip Lnc., Nanjing 211112, Jiangsu, People's Republic of China
| | - Xingzhi Liu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, Anhui, People's Republic of China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, People's Republic of China
| | - Junjie Zhong
- Department of Neurosurgery, Fudan University Huashan Hospital, National Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200041, People's Republic of China
| | - Bin Wang
- Center for Clinic Stem Cell Research, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, People's Republic of China
| | - Guangli Suo
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, People's Republic of China
| |
Collapse
|
12
|
Lian WS, Wu RW, Chen YS, Ko JY, Wang SY, Jahr H, Wang FS. MicroRNA-29a Mitigates Osteoblast Senescence and Counteracts Bone Loss through Oxidation Resistance-1 Control of FoxO3 Methylation. Antioxidants (Basel) 2021; 10:antiox10081248. [PMID: 34439496 PMCID: PMC8389244 DOI: 10.3390/antiox10081248] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/01/2021] [Accepted: 08/01/2021] [Indexed: 12/20/2022] Open
Abstract
Senescent osteoblast overburden accelerates bone mass loss. Little is understood about microRNA control of oxidative stress and osteoblast senescence in osteoporosis. We revealed an association between microRNA-29a (miR-29a) loss, oxidative stress marker 8-hydroxydeoxyguanosine (8-OHdG), DNA hypermethylation marker 5-methylcystosine (5mC), and osteoblast senescence in human osteoporosis. miR-29a knockout mice showed low bone mass, sparse trabecular microstructure, and osteoblast senescence. miR-29a deletion exacerbated bone loss in old mice. Old miR-29a transgenic mice showed fewer osteoporosis signs, less 5mC, and less 8-OHdG formation than age-matched wild-type mice. miR-29a overexpression reversed age-induced senescence and osteogenesis loss in bone-marrow stromal cells. miR-29a promoted transcriptomic landscapes of redox reaction and forkhead box O (FoxO) pathways, preserving oxidation resistance protein-1 (Oxr1) and FoxO3 in old mice. In vitro, miR-29a interrupted DNA methyltransferase 3b (Dnmt3b)-mediated FoxO3 promoter methylation and senescence-associated β-galactosidase activity in aged osteoblasts. Dnmt3b inhibitor 5'-azacytosine, antioxidant N-acetylcysteine, or Oxr1 recombinant protein attenuated loss in miR-29a and FoxO3 to mitigate oxidative stress, senescence, and mineralization matrix underproduction. Taken together, miR-29a promotes Oxr1, compromising oxidative stress and FoxO3 loss to delay osteoblast aging and bone loss. This study sheds light on a new antioxidation mechanism by which miR-29a protects against osteoblast aging and highlights the remedial effects of miR-29a on osteoporosis.
Collapse
Affiliation(s)
- Wei-Shiung Lian
- Core Laboratory for Phenomics and Diagnostic, Department of Medical Research, College of Medicine, Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (W.-S.L.); (Y.-S.C.); (S.-Y.W.)
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Re-Wen Wu
- Department of Orthopedic Surgery, College of Medicine, Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (R.-W.W.); (J.-Y.K.)
| | - Yu-Shan Chen
- Core Laboratory for Phenomics and Diagnostic, Department of Medical Research, College of Medicine, Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (W.-S.L.); (Y.-S.C.); (S.-Y.W.)
| | - Jih-Yang Ko
- Department of Orthopedic Surgery, College of Medicine, Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (R.-W.W.); (J.-Y.K.)
| | - Shao-Yu Wang
- Core Laboratory for Phenomics and Diagnostic, Department of Medical Research, College of Medicine, Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (W.-S.L.); (Y.-S.C.); (S.-Y.W.)
| | - Holger Jahr
- Department of Anatomy and Cell Biology, University Hospital RWTH Aachen, 52074 Aachen, Germany;
- Department of Orthopedic Surgery, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands
| | - Feng-Sheng Wang
- Core Laboratory for Phenomics and Diagnostic, Department of Medical Research, College of Medicine, Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (W.-S.L.); (Y.-S.C.); (S.-Y.W.)
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Correspondence: ; Tel.: +886-7-731-7123
| |
Collapse
|
13
|
Chakraborty S, Sinha S, Sengupta A. Emerging trends in chromatin remodeler plasticity in mesenchymal stromal cell function. FASEB J 2020; 35:e21234. [PMID: 33337557 DOI: 10.1096/fj.202002232r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/10/2020] [Accepted: 11/13/2020] [Indexed: 12/13/2022]
Abstract
Emerging evidences highlight importance of epigenetic regulation and their integration with transcriptional and cell signaling machinery in determining tissue resident adult pluripotent mesenchymal stem/stromal cell (MSC) activity, lineage commitment, and multicellular development. Histone modifying enzymes and large multi-subunit chromatin remodeling complexes and their cell type-specific plasticity remain the central defining features of gene regulation and establishment of tissue identity. Modulation of transcription factor expression gradient ex vivo and concomitant flexibility of higher order chromatin architecture in response to signaling cues are exciting approaches to regulate MSC activity and tissue rejuvenation. Being an important constituent of the adult bone marrow microenvironment/niche, pathophysiological perturbation in MSC homeostasis also causes impaired hematopoietic stem/progenitor cell function in a non-cell autonomous mechanism. In addition, pluripotent MSCs can function as immune regulatory cells, and they reside at the crossroad of innate and adaptive immune response pathways. Research in the past few years suggest that MSCs/stromal fibroblasts significantly contribute to the establishment of immunosuppressive microenvironment in shaping antitumor immunity. Therefore, it is important to understand mesenchymal stromal epigenome and transcriptional regulation to leverage its applications in regenerative medicine, epigenetic memory-guided trained immunity, immune-metabolic rewiring, and precision immune reprogramming. In this review, we highlight the latest developments and prospects in chromatin biology in determining MSC function in the context of lineage commitment and immunomodulation.
Collapse
Affiliation(s)
- Sayan Chakraborty
- Stem Cell & Leukemia Laboratory, Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India.,Translational Research Unit of Excellence (TRUE), Kolkata, India
| | - Sayantani Sinha
- Stem Cell & Leukemia Laboratory, Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India.,Translational Research Unit of Excellence (TRUE), Kolkata, India
| | - Amitava Sengupta
- Stem Cell & Leukemia Laboratory, Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India.,Translational Research Unit of Excellence (TRUE), Kolkata, India
| |
Collapse
|
14
|
Su K, Lin N, Xie S, Han Y, Yang Z, Zhang H, He H, Zhou SA, Ma W, Zhang T, Wang N. DNMT3A inhibits E2F1-induced arterial marker expression and impairs angiogenesis in human umbilical artery endothelial cells. Acta Biochim Biophys Sin (Shanghai) 2020; 52:1236-1246. [PMID: 33079978 DOI: 10.1093/abbs/gmaa109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 06/05/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022] Open
Abstract
Arterial marker genes EphrinB2 and HEY2 are essential for cardiovascular development and postnatal neovascularization. Our previous study confirmed that E2F1 could activate the transcription of EphrinB2 and HEY2 in human mesenchymal stem cells; however, the detailed mechanism has not been resolved yet. In this study, we focused on the interaction between E2F1 and DNMT3A, a de novo DNA methyltransferase, on regulating the expression of EphrinB2 and HEY2, and explored the potential mechanisms. Gain- and loss-of-function experiments implicated the positive effect of E2F1 on the expression of EphrinB2 and HEY2 and tube formation in human umbilical artery endothelial cells. Accumulation of DNMT3A decreased the levels of EphrinB2 and HEY2, and impaired tube formation induced by E2F1, while inhibiting DNMT3A by RNA interference augmented their expression and angiogenesis in E2F1-trasfected cells. We then asked whether the low expressions of EphrinB2 and HEY2 induced by DNMT3A are related to the methylation status of their promoters. Surprisingly, the methylation status of the CpG islands in the promoter region was not significantly affected by overexpression of exogenous DNMT3A. Furthermore, the interaction between E2F1 and DNMT3A was confirmed by co-immunoprecipitation. DNMT3A could inhibit the transcription of EphrinB2 and HEY2 promoters by affecting the binding of E2F1 to its recognition sequences as revealed by luciferase reporter assay and chromatin immunoprecipitation. These results identified a novel mechanism underlying the cooperation of DNMT3A with E2F1 on regulating target gene expression, and revealed their roles in the angiogenic process.
Collapse
Affiliation(s)
- Kaiyue Su
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, China
| | - Ningning Lin
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, China
| | - Shouqiang Xie
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, China
| | - Yabo Han
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, China
| | - Zaiming Yang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, China
| | - Hongmin Zhang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, China
| | - Hongpeng He
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, China
| | - S a Zhou
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, China
| | - Wenjian Ma
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, China
| | - Tongcun Zhang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, China
| | - Nan Wang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, China
| |
Collapse
|
15
|
Xue WL, Chen RQ, Zhang QQ, Li XH, Cao L, Li MY, Li Y, Lin G, Chen Y, Wang MJ, Zhu YC. Hydrogen sulfide rescues high glucose-induced migration dysfunction in HUVECs by upregulating miR-126-3p. Am J Physiol Cell Physiol 2020; 318:C857-C869. [PMID: 32186933 DOI: 10.1152/ajpcell.00406.2019] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diabetes (especially Type II) is one of the primary threats to cardiovascular health. Wound healing defects and vascular dysfunction are common in diabetic patients, and the primary cause of deterioration is sustained high plasma glucose. microRNA, a noncoding RNA, has regulatory functions that are critical to maintaining homeostasis. MicroRNA (miR)-126-3p is a potential diabetes biomarker and a proangiogenic factor, and its plasma level decreases in diabetic patients. Previous studies have revealed the proangiogenic character of the gasotransmitter hydrogen sulfide (H2S). However, little is known about the relationship between H2S and miR-126-3p when the extracellular glucose level is high, let alone their influences on deteriorated endothelial cell migration, a key component of angiogenesis, which is crucial for wound healing. Human umbilical vein endothelial cells (HUVECs) were treated with high glucose (33.3 mmol/L) or normal glucose (5.5 mmol/L) for 48 h. Affymetrix miRNA profiling and real-time PCR were used to validate the miRNA expression. An H2S probe (HSip-1) was used to detect endogenous H2S. Scratch wound-healing assays were used to evaluate HUVEC migration. The protein levels were quantified by Western blot. Both exogenous and endogenous H2S could upregulate the miR-126-3p levels in HUVECs or muscle tissue. High glucose decreased the H2S level and the protein expression of the H2S-producing enzyme cystathionine γ-lyase (CSE) in HUVECs; however, the DNA methyltransferase 1 (DNMT1) protein level was upregulated. CSE overexpression not only increased the miR-126-3p level by decreasing the DNMT1 protein level but also rescued the deteriorated cell migration in HUVECs treated with high glucose. DNMT1 overexpression decreased the miR-126-3p level and inhibited the migration of HUVECs, whereas silencing DNMT1 improved cell migration. High glucose decreased the endogenous H2S and miR-126-3p levels and increased the DNMT1 expression, thus inducing the migration dysfunction of HUVECs. Treatment with exogenous H2S or the overexpression of the endogenously produced enzyme CSE would rescue this migration dysfunction through H2S-DNMT1-miR-126-3p.
Collapse
Affiliation(s)
- Wen-Long Xue
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Rui-Qin Chen
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qing-Qing Zhang
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xing-Hui Li
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lei Cao
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Meng-Yao Li
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ye Li
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ge Lin
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ying Chen
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ming-Jie Wang
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yi-Chun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Sui BD, Zheng CX, Li M, Jin Y, Hu CH. Epigenetic Regulation of Mesenchymal Stem Cell Homeostasis. Trends Cell Biol 2020; 30:97-116. [DOI: 10.1016/j.tcb.2019.11.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/19/2019] [Accepted: 11/26/2019] [Indexed: 12/24/2022]
|
17
|
Jiang W, Agrawal DK, Boosani CS. Non-coding RNAs as Epigenetic Gene Regulators in Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:133-148. [PMID: 32285409 DOI: 10.1007/978-981-15-1671-9_7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Epigenetic gene regulations can be considered as de-novo initiation of abnormal molecular signaling events whose regulation is otherwise required during normal or specific developmental stages of the organisms. Primarily, three different mechanisms have been identified to participate in epigenetic gene regulations which include, DNA methylation, non-coding RNA species (microRNAs [miRNA], and long non-coding RNAs [LNC-RNA]) and histone modifications. These de-novo epigenetic mechanisms have been associated with altered normal cellular functions which eventually facilitate normal cells to transition into an abnormal phenotype. Among the three modes of regulation, RNA species which are usually considered to be less stable, can be speculated to initiate instant alterations in gene expression compared to DNA methylation or histone modifications. However, LNC-RNAs appear to be more stable in the cells than the other RNA species. Moreover, there is increasing literature which clearly suggests that a single specific LNC-RNA can regulate multiple mechanisms and disease phenotypes. With specific focus on cardiovascular diseases, here we attempt to provide UpToDate information on the functional role of miRNAs and LNC-RNAs. Here we discuss the role of these epigenetic mediators in different components of cardiovascular disease which include physiopathological heart development, athersclerosis, retenosis, diabetic hearts, myocardial infarction, ischemia-reperfusion, heart valve disease, aortic aneurysm, osteogenesis, angiogenesis and hypoxia in the heart. While there is abundant literature support that shows the involvement of many LNC-RNAs and miRNAs in cardiovascular diseases, very few RNA species have been identified which regulate epigenetic mechanisms which is the current focus in this article. Understanding the role of these RNA species in regulating epigenetic mechanisms in different cell types causing cardiovascular disease, would advance the field and promote disease prevention approaches that are aimed to target epigenetic mechanisms.
Collapse
Affiliation(s)
- Wanlin Jiang
- Department of Clinical & Translational Research, Western University of Health Sciences, Pomona, CA, USA
| | - Devendra K Agrawal
- Department of Clinical & Translational Research, Western University of Health Sciences, Pomona, CA, USA
| | - Chandra Shekhar Boosani
- Department of Clinical & Translational Research, Western University of Health Sciences, Pomona, CA, USA.
| |
Collapse
|
18
|
Gomes AM, Pinto TS, Costa Fernandes CJ, Silva RA, Zambuzzi WF. Wortmannin targeting phosphatidylinositol 3‐kinase suppresses angiogenic factors in shear‐stressed endothelial cells. J Cell Physiol 2019; 235:5256-5269. [DOI: 10.1002/jcp.29412] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 12/05/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Anderson M. Gomes
- Bioassays and Cell Dynamics Laboratory, Department of Chemistry and BiochemistryBioscience Institute UNESP Botucatu Sao Paulo Brazil
| | - Thais S. Pinto
- Bioassays and Cell Dynamics Laboratory, Department of Chemistry and BiochemistryBioscience Institute UNESP Botucatu Sao Paulo Brazil
| | - Célio J. Costa Fernandes
- Bioassays and Cell Dynamics Laboratory, Department of Chemistry and BiochemistryBioscience Institute UNESP Botucatu Sao Paulo Brazil
| | - Rodrigo A. Silva
- Bioassays and Cell Dynamics Laboratory, Department of Chemistry and BiochemistryBioscience Institute UNESP Botucatu Sao Paulo Brazil
- Department of Biology, Dental SchoolUniversity of Taubaté Taubaté São Paulo Brazil
| | - Willian F. Zambuzzi
- Bioassays and Cell Dynamics Laboratory, Department of Chemistry and BiochemistryBioscience Institute UNESP Botucatu Sao Paulo Brazil
| |
Collapse
|
19
|
Belloni E, Di Matteo A, Pradella D, Vacca M, Wyatt CDR, Alfieri R, Maffia A, Sabbioneda S, Ghigna C. Gene Expression Profiles Controlled by the Alternative Splicing Factor Nova2 in Endothelial Cells. Cells 2019; 8:cells8121498. [PMID: 31771184 PMCID: PMC6953062 DOI: 10.3390/cells8121498] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/11/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023] Open
Abstract
Alternative splicing (AS) plays an important role in expanding the complexity of the human genome through the production of specialized proteins regulating organ development and physiological functions, as well as contributing to several pathological conditions. How AS programs impact on the signaling pathways controlling endothelial cell (EC) functions and vascular development is largely unknown. Here we identified, through RNA-seq, changes in mRNA steady-state levels in ECs caused by the neuro-oncological ventral antigen 2 (Nova2), a key AS regulator of the vascular morphogenesis. Bioinformatics analyses identified significant enrichment for genes regulated by peroxisome proliferator-activated receptor-gamma (Ppar-γ) and E2F1 transcription factors. We also showed that Nova2 in ECs controlled the AS profiles of Ppar-γ and E2F dimerization partner 2 (Tfdp2), thus generating different protein isoforms with distinct function (Ppar-γ) or subcellular localization (Tfdp2). Collectively, our results supported a mechanism whereby Nova2 integrated splicing decisions in order to regulate Ppar-γ and E2F1 activities. Our data added a layer to the sequential series of events controlled by Nova2 in ECs to orchestrate vascular biology.
Collapse
Affiliation(s)
- Elisa Belloni
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Anna Di Matteo
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Davide Pradella
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Margherita Vacca
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Christopher D. R. Wyatt
- Centre for Biodiversity and Environment Research, University College London, Gower Street, London WC1E 6BT, UK
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr Aiguader 88, 08003 Barcelona, Spain
- Universitat Pompeu Fabra, Plaça de la Mercè, 10-12, 08002 Barcelona, Spain
| | - Roberta Alfieri
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Antonio Maffia
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Simone Sabbioneda
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Claudia Ghigna
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
- Correspondence:
| |
Collapse
|
20
|
Shang T, Li S, Zhang Y, Lu L, Cui L, Guo FF. Hypoxia promotes differentiation of adipose-derived stem cells into endothelial cells through demethylation of ephrinB2. Stem Cell Res Ther 2019; 10:133. [PMID: 31109374 PMCID: PMC6528245 DOI: 10.1186/s13287-019-1233-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/06/2019] [Accepted: 04/10/2019] [Indexed: 01/22/2023] Open
Abstract
Background Delivery of endothelial cells into the ischemic tissue is emerging as an alternative approach in revascularization of injured tissues by means of angiogenesis to restore organ function. Adipose-derived stem cells (ASCs) are a readily accessible source of the mesenchymal stem cell with rapid expansion and multidifferentiation potential. The view has emerged that endothelial cells (ECs) differentiated from ASCs is a step forward for adult vascular repair in regenerative medicine and construction of the blood vessel by tissue engineering approach. Methods In this study, differentiation of human ASCs (hASCs) into vascular EC lineage was induced by combined treatment of vascular endothelial growth factor (VEGF) and bone morphogenetic protein-4 (BMP4) under hypoxia condition. The expression of CD31, VEGF-R2, and VE-cadherin was determined by immunofluorescent staining, real-time PCR, and western blot analysis. These differentiated cells acquired functional characteristics of mature ECs as determined by their tube formation ability, DiI-ac-LDL uptake, and nitric oxide secretion in vitro. The methylation status in the proximal promoter CpGs was determined by bisulfite sequencing. Results hASCs expressed endothelial cell markers including CD31, VEGF-R2, and VE-cadherin by combined treatment of VEGF and BMP4 under hypoxia condition. These differentiated cells exhibited the angiogenesis potential in vitro, and injection of these differentiated cells enhanced angiogenesis in the ischemic hindlimb of diabetic mice. Furthermore, it was found that hypoxia increased significantly EphrinB2 expression EC differentiation, which is greatly downregulated with EphrinB2 blockage. The methylation status in the proximal promoter CpG results showed that methylation of EphrinB2 promoter decreased in hASCs with exposure to hypoxia. Conclusion Our data demonstrate that hASCs can be efficiently induced to differentiate into vascular EC lineages which are mediated by demethylation of ephrinB2 under hypoxia condition.
Collapse
Affiliation(s)
- Ting Shang
- Department of Plastic Surgery, Beijing Shijitan Hospital affiliated to Capital Medical University, 10 Tieyi Road, Beijing, China
| | - Shuaijun Li
- Department of Plastic Surgery, Beijing Shijitan Hospital affiliated to Capital Medical University, 10 Tieyi Road, Beijing, China
| | - Yun Zhang
- Department of Plastic Surgery, Beijing Shijitan Hospital affiliated to Capital Medical University, 10 Tieyi Road, Beijing, China
| | - Laiya Lu
- Department of Orthopedics, Shanghai Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai, China
| | - Lei Cui
- Department of Plastic Surgery, Beijing Shijitan Hospital affiliated to Capital Medical University, 10 Tieyi Road, Beijing, China. .,Department of Orthopedics, Shanghai Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai, China.
| | - Fang Fang Guo
- Department of Plastic and Reconstructive surgery, Zhongda Hospital, Southeast University, 87 Dingjiaqiao street, Nanjing, Jiangsu Province, China.
| |
Collapse
|
21
|
Bandara N, Gurusinghe S, Kong A, Mitchell G, Wang LX, Lim SY, Strappe P. Generation of a nitric oxide signaling pathway in mesenchymal stem cells promotes endothelial lineage commitment. J Cell Physiol 2019; 234:20392-20407. [PMID: 30997675 DOI: 10.1002/jcp.28640] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 12/22/2022]
Abstract
Enhancing differentiation of mesenchymal stem cells (MSCs) to endothelial cells may improve their ability to vascularize tissue and promote wound healing. This study describes a novel role for nitric oxide (NO) in reprogramming MSCs towards an endothelial lineage and highlights the role of Wnt signaling and epigenetic modification by NO. Rat MSCs were transduced with lentiviral vectors expressing endothelial nitric oxide synthase (pLV-eNOS) and a mutated caveolin gene (pLV-CAV-1F92A ) to enhance NO generation resulting in increased in vitro capillary tubule formation and endothelial marker gene expression. An exogenous source of NO could also stimulate CD31 expression in MSCs. NO was associated with an arterial-specific endothelial gene expression profile of Notch1, Dll4, and Hey2 and significantly reduced expression of venous markers. Wnt signaling associated with NO was evident through increased gene expression of Wnt3a and β-catenin protein, and expression of the endothelial marker Pecam-1 could be significantly reduced by treatment with the Wnt signaling inhibitor Dkk-1. The role of NO as an epigenetic modifier was evident with reduced gene expression of the methyltransferase, DNMT1, and bisulfite sequencing of the endothelial Flt1 promoter region in NO-producing MSCs showed significant demethylation compared to control cells. Finally, subcutaneous implantation of NO-producing MSCs seeded in a biomaterial scaffold (NovoSorb®) resulted in survival of transplanted cells and the formation of blood vessels. In summary, this study describes, NO as a potent endothelial programming factor which acts as an epigenetic modifier in MSCs and may provide a novel platform for vascular regenerative therapy.
Collapse
Affiliation(s)
- Nadeeka Bandara
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Saliya Gurusinghe
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia.,School of Agricultural and Wine Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Anne Kong
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Geraldine Mitchell
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,Department of Surgery, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia.,Faculty of Health Sciences, Australian Catholic University, Fitzroy, Victoria, Australia
| | - Le-Xin Wang
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Shiang Y Lim
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,Department of Surgery, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| | - Padraig Strappe
- School of Health, Medicine and Applied Sciences, Central Queensland University, Rockhampton, Queensland, Australia
| |
Collapse
|
22
|
Balakrishnan A, Guruprasad KP, Satyamoorthy K, Joshi MB. Interleukin-6 determines protein stabilization of DNA methyltransferases and alters DNA promoter methylation of genes associated with insulin signaling and angiogenesis. J Transl Med 2018; 98:1143-1158. [PMID: 29955086 DOI: 10.1038/s41374-018-0079-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 04/09/2018] [Accepted: 04/22/2018] [Indexed: 02/03/2023] Open
Abstract
Individuals with type 2 diabetes (T2D) display vascular insulin resistance and decreased nitric oxide production leading to vasoconstriction and atherosclerosis. Soluble factors such as pro-inflammatory molecules, and various genetic and epigenetic mechanisms have been implicated to induce insulin resistance in vascular endothelial cells. Epigenetic mechanisms such as altered promoter DNA methylation have been demonstrated in development and progression of metabolic disorders and atherosclerosis. However, underlying precise epigenetic mechanisms regulating cross talk between insulin signaling genes and inflammation in vascular cells remains to be fully understood. Human endothelial cells when (a) treated with interleukin-6 (IL-6) and insulin together, (b) pretreated with IL-6, and (c) under hyperinsulinemic conditions led to a state of vascular insulin resistance resulting in decreased Akt/eNOS activation and subsequent stabilization of STAT3 phosphorylation. IL-6 abrogated insulin effects on angiogenesis in 3D spheroid and matrigel assays. IL-6-induced insulin resistance was associated with decreased activity of DNA methyltransferase isoforms and global DNA hypomethylation, which inversely correlated with S-phase of cell cycle. CpG microarray analysis in IL-6 treated endothelial cells revealed promoters associated hypo- and hypermethylation of 199 and 98 genes respectively. Promoter DNA methylation status of genes associated with insulin signaling and angiogenesis such as RPS6KA2, PIK3R2, FOXD3, EXOC7, MAP3K8, ITPKB, EPHA6, IGF1R, and FOXC2 were validated by bisulfite DNA sequencing. Concentration and time-dependent analysis revealed that IL-6 reduced DNMT1 and DNMT3B but not DNMT3A protein levels. Our data indicate a causal link between IL-6-induced changes in global and promoter-specific DNA methylation, due to reduced DNMT1 and DNMT3B protein levels leading to altered expression of critical genes involved in insulin signaling and angiogenesis.
Collapse
Affiliation(s)
- Aswath Balakrishnan
- School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | | | | | - Manjunath B Joshi
- School of Life Sciences, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
23
|
Jiang C, Gong F. MiR-148a promotes myocardial differentiation of human bone mesenchymal stromal cells via DNA methyltransferase 1 (DNMT1). Cell Biol Int 2018; 42:913-922. [PMID: 28656724 DOI: 10.1002/cbin.10813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 06/24/2017] [Indexed: 11/08/2022]
Abstract
MicroRNAs have potential to modulate the differentiation of stem cells. In previous study, we found that miR-148a was up-regulated in myocardial differentiation of human bone mesenchymal stromal cells (hBMSCs) induced by 5'-azacytidine. However, the role of miR-148a in regulating this process still remains unclear. In this study, we investigated the function and molecular mechanism of miR-148a in myocardial differentiation of hBMSCs. We found that miR-148a was significantly increased while DNA methyltransferase 1 (DNMT1) was significantly decreased in myocardial differentiation of hBMSCs. Then, the dual luciferase reporter assays method indicated that DNMT1 was the direct target of miR-148a. In addition, we showed that up-regulation of miR-148a could enhance myocardial differentiation of hBMSCs, while down-regulation of miR-148a could inhibit myocardial differentiation process. Moreover, knockdown of DNMT1 could block the role of miR-148a in promoting myocardial differentiation of hBMSCs. Finally, MiR-148a acted on methylation level of GATA-4 and knockdown of DNMT1 could block this function. Therefore, our results indicate that miR-148a plays a vital role in regulating myocardial differentiation of hBMSCs by targeting DNMT1.
Collapse
Affiliation(s)
- Changke Jiang
- Department of Pediatrics, Yongchuan Hospital of Chongqing Medical University, 439 Xuanhua Road, Yongchuan, Chongqing, 402160, China
| | - Fang Gong
- Department of Pediatrics, Yongchuan Hospital of Chongqing Medical University, 439 Xuanhua Road, Yongchuan, Chongqing, 402160, China
| |
Collapse
|
24
|
Sun H, Zhao X, Zhang C, Zhang Z, Lun J, Liao W, Zhang Z. MiR-455-3p inhibits the degenerate process of chondrogenic differentiation through modification of DNA methylation. Cell Death Dis 2018; 9:537. [PMID: 29748607 PMCID: PMC5945650 DOI: 10.1038/s41419-018-0565-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 03/19/2018] [Accepted: 04/05/2018] [Indexed: 12/21/2022]
Abstract
The aim of this work was to determine whether miR-455-3p regulates DNA methylation during chondrogenic differentiation of hMSCs. The expression of miR-455-3p and de novo methyltransferase DNMT3A was assessed in micromass culture of hBMSCs, which induced chondrogenic differentiation in vitro, and in E16.5 mice in vivo. A luciferase reporter assay was used to confirm whether miR-455-3p directly targets DNMT3A by interaction with the 3′-UTR. Using an Illumina Infinium Methylation EPIC microarray, genome-wide DNA methylation of hBMSCs with or without overexpressed miR-455-3p was examined for 28 days during induced chondrogenic differentiation. Here, we showed that miR-455-3p was more expressed during the middle stage of hBMSC chondrogenic differentiation, and less expressed in the late stage. DNMT3A was less expressed in the middle stage and more expressed in the late stage, and was also more expressed in the palms of miR-455-3p deletion mice compared to those of wild-type mice. The luciferase reporter assay demonstrated that miR-455-3p directly targets DNMT3A 3′-UTR. miR-455-3p overexpression inhibits the degenerate process during chondrogenic differentiation, while deletion of miR-455-3p in mice accelerated cartilage degeneration. Genome-wide DNA methylation analysis showed miR-455-3p overexpression regulates DNA methylation of cartilage-specific genes. GO analysis revealed PI3K-Akt signaling pathway was most hypomethylated. Our data show that miR-455-3p can regulate hMSC chondrogenic differentiation by affecting DNA methylation. Overexpression of miR-455-3p and DNA methylation inhibitors can thus potentially be utilized to optimize chondrogenic differentiation.
Collapse
Affiliation(s)
- Hao Sun
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China
| | - Xiaoyi Zhao
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China
| | - Chengyun Zhang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China
| | - Ziji Zhang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China
| | - Jiayong Lun
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China
| | - Weiming Liao
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China.
| | - Zhiqi Zhang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China.
| |
Collapse
|
25
|
The Methods and Mechanisms to Differentiate Endothelial-Like Cells and Smooth Muscle Cells from Mesenchymal Stem Cells for Vascularization in Vaginal Reconstruction. Mol Biotechnol 2018; 60:396-411. [DOI: 10.1007/s12033-018-0079-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
26
|
Marquez J, Fernandez-Piñeiro I, Araúzo-Bravo MJ, Poschmann G, Stühler K, Khatib AM, Sanchez A, Unda F, Ibarretxe G, Bernales I, Badiola I. Targeting liver sinusoidal endothelial cells with miR-20a-loaded nanoparticles reduces murine colon cancer metastasis to the liver. Int J Cancer 2018; 143:709-719. [PMID: 29492958 DOI: 10.1002/ijc.31343] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 02/02/2018] [Accepted: 02/19/2018] [Indexed: 12/15/2022]
Abstract
Phenotypic transformation of liver sinusoidal endothelial cells is one of the most important stages of liver metastasis progression. The miRNA effects on liver sinusoidal endothelial cells during liver metastasis have not yet been studied. Herein, whole genome analysis of miRNA expression in these cells during colorectal liver metastasis revealed repressed expression of microRNA-20a. Importantly, downregulation of miR-20a occurs in parallel with upregulation of its known protein targets. To restore normal miR-20a levels in liver sinusoidal endothelial cells, we developed chondroitin sulfate-sorbitan ester nanoparticles conjugated with miR-20a in a delivery system that specifically targets liver sinusoidal endothelial cells. The restoration of normal mir-20a levels in these cells induced downregulation of the expression of its protein targets, and this also resulted in a reduction of in vitro LSEC migration and a reduction of in vivo activation and tumor-infiltrating capacity and ability of the tumor decreased by ∼80% in a murine liver metastasis model.
Collapse
Affiliation(s)
- Joana Marquez
- Department of Cell Biology and Histology, Faculty of Medicine and Nursery, University of Basque Country, UPV/EHU, Leioa, Spain
| | - Ines Fernandez-Piñeiro
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Marcos J Araúzo-Bravo
- Computational Biology and Systems Biomedicine Research Group.Computational Biology Data Analysis Platform. Biodonostia Health Research Institute, San Sebastián, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Gereon Poschmann
- Molecular Proteomics Laboratory (MPL), Biologisch-Medizinisches Forschungszentrum (BMFZ),Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory (MPL), Biologisch-Medizinisches Forschungszentrum (BMFZ),Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Abdel-Majid Khatib
- Université Bordeaux, Pessac, France.,INSERM, LAMC, UMR 1029, Pessac, France
| | - Alejandro Sanchez
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, Spain.,Genetics and Biology of the Development of Kidney Diseases Unit, Sanitary Research Institute (IDIS) of the University Hospital Complex of Santiago de Compostela, Santiago de Compostela, Spain
| | - Fernando Unda
- Department of Cell Biology and Histology, Faculty of Medicine and Nursery, University of Basque Country, UPV/EHU, Leioa, Spain
| | - Gaskon Ibarretxe
- Department of Cell Biology and Histology, Faculty of Medicine and Nursery, University of Basque Country, UPV/EHU, Leioa, Spain
| | - Irantzu Bernales
- Gene Expression Unit, Genomics Facility of General Research Services (SGIker), University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Iker Badiola
- Department of Cell Biology and Histology, Faculty of Medicine and Nursery, University of Basque Country, UPV/EHU, Leioa, Spain
| |
Collapse
|
27
|
Xu R, Chen W, Zhang Z, Qiu Y, Wang Y, Zhang B, Lu W. Integrated data analysis identifies potential inducers and pathways during the endothelial differentiation of bone-marrow stromal cells by DNA methyltransferase inhibitor, 5-aza-2'-deoxycytidine. Gene 2018. [PMID: 29514045 DOI: 10.1016/j.gene.2018.03.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Bone-Marrow Stromal Cells (BMSCs)-derived vascular endothelial cells (VECs) is regarded as an important therapeutic strategy for spinal cord injury, disc degeneration, cerebral ischemic disease and diabetes. The change in DNA methylation level is essential for stem cell differentiation. However, the DNA methylation related mechanisms underlying the endothelial differentiation of BMSCs are not well understood. In this study, DNA methyltransferase inhibitor, 5-aza-2'-deoxycytidine (5-aza-dC) significantly elevated the endothelial markers expression (CD31/PECAM1, CD105/ENG, eNOS and VE-cadherin), as well as promoted the capacity of angiogenesis on Matrigel. The result of Alexa 488-Ac-LDL uptake assay indicated that the differentiation ratio of BMSCs into VECs was 68.7% in 5-azaz-dC induced differentiation. And then we screened differentiation inducers with altered expression patterns and DNA methylation levels in four important families (VEGF, ANG, FGF and ETS). By integrating these data, five endothelial differentiation inducers (VEGFA, ANGPT2, FGF2, FGF9 and ETS1) which were directly upregulated by 5-aza-dC and five indirect factors (FGF1, FGF3, ETS2, ETV1 and ETV4) were identified. These data suggested that 5-aza-dC is an excellent chemical molecule for BMSCs differentiation into functional VECs and also provided essential clues for DNA methylation related signaling during 5-aza-dC induced endothelial differentiation of BMSCs.
Collapse
Affiliation(s)
- Rui Xu
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Wenbin Chen
- Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Zhifen Zhang
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Yang Qiu
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Yong Wang
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Bingchang Zhang
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Wei Lu
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
28
|
De Pauw A, Andre E, Sekkali B, Bouzin C, Esfahani H, Barbier N, Loriot A, De Smet C, Vanhoutte L, Moniotte S, Gerber B, di Mauro V, Catalucci D, Feron O, Hilfiker-Kleiner D, Balligand JL. Dnmt3a-mediated inhibition of Wnt in cardiac progenitor cells improves differentiation and remote remodeling after infarction. JCI Insight 2017; 2:91810. [PMID: 28614798 DOI: 10.1172/jci.insight.91810] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 05/15/2017] [Indexed: 12/28/2022] Open
Abstract
Adult cardiac progenitor cells (CPCs) display a low capacity to differentiate into cardiomyocytes in injured hearts, strongly limiting the regenerative capacity of the mammalian myocardium. To identify new mechanisms regulating CPC differentiation, we used primary and clonally expanded Sca-1+ CPCs from murine adult hearts in homotypic culture or coculture with cardiomyocytes. Expression kinetics analysis during homotypic culture differentiation showed downregulation of Wnt target genes concomitant with increased expression of the Wnt antagonist, Wnt inhibitory factor 1 (Wif1), which is necessary to stimulate CPC differentiation. We show that the expression of the Wif1 gene is repressed by DNA methylation and regulated by the de novo DNA methyltransferase Dnmt3a. In addition, miR-29a is upregulated early during CPC differentiation and downregulates Dnmt3a expression, thereby decreasing Wif1 gene methylation and increasing the efficiency of differentiation of Sca-1+ CPCs in vitro. Extending these findings in vivo, transient silencing of Dnmt3a in CPCs subsequently injected in the border zone of infarcted mouse hearts improved CPC differentiation in situ and remote cardiac remodeling. In conclusion, miR-29a and Dnmt3a epigenetically regulate CPC differentiation through Wnt inhibition. Remote effects on cardiac remodeling support paracrine signaling beyond the local injection site, with potential therapeutic interest for cardiac repair.
Collapse
Affiliation(s)
- Aurelia De Pauw
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique, and Department of Medicine, Cliniques Saint-Luc, and
| | - Emilie Andre
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique, and Department of Medicine, Cliniques Saint-Luc, and
| | - Belaid Sekkali
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique, and Department of Medicine, Cliniques Saint-Luc, and
| | - Caroline Bouzin
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique, and Department of Medicine, Cliniques Saint-Luc, and
| | - Hrag Esfahani
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique, and Department of Medicine, Cliniques Saint-Luc, and
| | - Nicolas Barbier
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique, and Department of Medicine, Cliniques Saint-Luc, and
| | - Axelle Loriot
- Group of Genetics and Epigenetics, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Charles De Smet
- Group of Genetics and Epigenetics, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Laetitia Vanhoutte
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique, and Department of Medicine, Cliniques Saint-Luc, and.,Division of Paediatric Cardiology and
| | | | - Bernhard Gerber
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique and Cliniques Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Vittoria di Mauro
- Humanitas Clinical and Research Center, National Research Council, Institute of Genetic and Biomedical Research, Milan, Italy
| | - Daniele Catalucci
- Humanitas Clinical and Research Center, National Research Council, Institute of Genetic and Biomedical Research, Milan, Italy
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique, and Department of Medicine, Cliniques Saint-Luc, and
| | | | - Jean-Luc Balligand
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique, and Department of Medicine, Cliniques Saint-Luc, and
| |
Collapse
|
29
|
Flores-Figueroa E, Gratzinger D. Beyond the Niche: Myelodysplastic Syndrome Topobiology in the Laboratory and in the Clinic. Int J Mol Sci 2016; 17:553. [PMID: 27089321 PMCID: PMC4849009 DOI: 10.3390/ijms17040553] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 03/26/2016] [Accepted: 04/07/2016] [Indexed: 12/18/2022] Open
Abstract
We review the murine and human microenvironment and hematopoietic stem cell niche in the context of intact bone marrow architecture in man and mouse, both in normal and in myelodysplastic syndrome marrow. We propose that the complexity of the hematopoietic stem cell niche can usefully be approached in the context of its topobiology, and we provide a model that incorporates in vitro and in vivo models as well as in situ findings from intact human marrow to explain the changes seen in myelodysplastic syndrome patients. We highlight the clinical application of the study of the bone marrow microenvironment and its topobiology in myelodysplastic syndromes.
Collapse
Affiliation(s)
- Eugenia Flores-Figueroa
- Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Avenida Cuauhtémoc 330, Colonia Doctores, c.p. 06720 Mexico City, Mexico.
| | - Dita Gratzinger
- Department of Pathology, Stanford University School of Medicine 300 Pasteur Dr., L235, Stanford, CA 94305, USA.
| |
Collapse
|