151
|
Jin ZX, Liao XY, Da WW, Zhao YJ, Li XF, Tang DZ. Osthole enhances the bone mass of senile osteoporosis and stimulates the expression of osteoprotegerin by activating β-catenin signaling. Stem Cell Res Ther 2021; 12:154. [PMID: 33640026 PMCID: PMC7912492 DOI: 10.1186/s13287-021-02228-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/14/2021] [Indexed: 02/08/2023] Open
Abstract
Introduction Osthole has a potential therapeutic application for anti-osteoporosis. The present study verified whether osthole downregulates osteoclastogenesis via targeting OPG. Methods In vivo, 12-month-old male mice were utilized to evaluate the effect of osthole on bone mass. In vitro, bone marrow stem cells (BMSCs) were isolated and extracted from 3-month-old OPG−/− mice and the littermates of OPG+/+ mice. Calvaria osteoblasts were extracted from 3-day-old C57BL/6J mice or 3-day-old OPG−/− mice and the littermates of OPG+/+ mice. Results Osthole significantly increased the gene and protein levels of OPG in primary BMSCs in a dose-dependent manner. The deletion of the OPG gene did not affect β-catenin expression. The deletion of the β-catenin gene inhibited OPG expression in BMSCs, indicating that osthole stimulates the expression of OPG via activation of β-catenin signaling. Conclusion Osthole attenuates osteoclast formation by stimulating the activation of β-catenin-OPG signaling and could be a potential drug for the senile osteoporosis.
Collapse
Affiliation(s)
- Zhen-Xiong Jin
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Xin-Yuan Liao
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Navy Medical University, Shanghai, 201705, China
| | - Wei-Wei Da
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yong-Jian Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Xiao-Feng Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - De-Zhi Tang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China. .,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
152
|
Lam ATL, Lee AP, Jayaraman P, Tan KY, Raghothaman D, Lim HL, Cheng H, Zhou L, Tan AHM, Reuveny S, Oh S. Multiomics analyses of cytokines, genes, miRNA, and regulatory networks in human mesenchymal stem cells expanded in stirred microcarrier-spinner cultures. Stem Cell Res 2021; 53:102272. [PMID: 33676128 DOI: 10.1016/j.scr.2021.102272] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/19/2021] [Accepted: 02/21/2021] [Indexed: 01/09/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are of great clinical interest as a form of allogenic therapy due to their excellent regenerative and immunomodulatory effects for various therapeutic indications. Stirred suspension bioreactors using microcarriers (MC) have been used for large-scale production of MSCs compared to planar cultivation systems. Previously, we have demonstrated that expansion of MSCs in MC-spinner cultures improved chondrogenic, osteogenic, and cell migration potentials as compared to monolayer-static cultures. In this study, we sought to address this by analyzing global gene expression patterns, miRNA profiles and secretome under both monolayer-static and MC-spinner cultures in serum-free medium at different growth phases. The datasets revealed differential expression patterns that correlated with potentially improved MSC properties in cells from MC-spinner cultures compared to those of monolayer-static cultures. Transcriptome analysis identified a unique expression signature for cells from MC-spinner cultures, which correlated well with miRNA expression, and cytokine secretion involved in key MSC functions. Importantly, MC-spinner cultures and conditioned medium showed increased expression of factors that possibly enhance pathways of extracellular matrix dynamics, cellular metabolism, differentiation potential, immunoregulatory function, and wound healing. This systematic analysis provides insights for the efficient optimization of stem cell bioprocessing and infers that MC-based bioprocess manufacturing could improve post-expansion cellular properties for stem cell therapies.
Collapse
Affiliation(s)
- Alan Tin-Lun Lam
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore.
| | - Alison P Lee
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Premkumar Jayaraman
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Kah Yong Tan
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Deepak Raghothaman
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Hsueh Lee Lim
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - He Cheng
- MiRXES, 2 Tukang Innovation Grove, JTC MedTech Hub, Singapore
| | - Lihan Zhou
- MiRXES, 2 Tukang Innovation Grove, JTC MedTech Hub, Singapore
| | - Andy Hee-Meng Tan
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Shaul Reuveny
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Steve Oh
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore.
| |
Collapse
|
153
|
Pouikli A, Tessarz P. Metabolism and chromatin: A dynamic duo that regulates development and ageing: Elucidating the metabolism-chromatin axis in bone-marrow mesenchymal stem cell fate decisions. Bioessays 2021; 43:e2000273. [PMID: 33629755 DOI: 10.1002/bies.202000273] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/12/2022]
Abstract
Bone-marrow mesenchymal stem cell (BM-MSC) proliferation and lineage commitment are under the coordinated control of metabolism and epigenetics; the MSC niche contains low oxygen, which is an important determinant of the cellular metabolic state. In turn, metabolism drives stem cell fate decisions via alterations of the chromatin landscape. Due to the fundamental role of BM-MSCs in the development of adipose tissue, bones and cartilage, age-associated changes in metabolism and the epigenome perturb the balance between stem cell proliferation and differentiation leading to stem cell depletion, fat accumulation and bone-quality related diseases. Therefore, understanding the dynamics of the metabolism-chromatin interplay is crucial for maintaining the stem cell pool and delaying the development and progression of ageing. This review summarizes the current knowledge on the role of metabolism in stem cell identity and highlights the impact of the metabolic inputs on the epigenome, with regards to stemness and pluripotency.
Collapse
Affiliation(s)
- Andromachi Pouikli
- Max-Planck Research Group Chromatin and Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Peter Tessarz
- Max-Planck Research Group Chromatin and Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany.,Cologne Excellence Cluster on Stress Responses in ageing-associated Diseases (CECAD), Cologne, Germany
| |
Collapse
|
154
|
Chen X, Li N, Weng J, Du X. Senescent Mesenchymal Stem Cells in Myelodysplastic Syndrome: Functional Alterations, Molecular Mechanisms, and Therapeutic Strategies. Front Cell Dev Biol 2021; 8:617466. [PMID: 33644035 PMCID: PMC7905046 DOI: 10.3389/fcell.2020.617466] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/31/2020] [Indexed: 01/01/2023] Open
Abstract
Myelodysplastic syndrome (MDS) is a group of clonal hematopoietic disorders related to hematopoietic stem and progenitor cell dysfunction. However, therapies that are currently used to target hematopoietic stem cells are not effective. These therapies are able to slow the evolution toward acute myeloid leukemia but cannot eradicate the disease. Mesenchymal stem cells (MSCs) have been identified as one of the main cellular components of the bone marrow microenvironment, which plays an indispensable role in normal hematopoiesis. When functional and regenerative capacities of aging MSCs are diminished, some enter replicative senescence, which promotes inflammation and disease progression. Recent studies that investigated the contribution of bone marrow microenvironment and MSCs to the initiation and progression of the disease have offered new insights into the MDS. This review presents the latest updates on the role of MSCs in the MDS and discusses potential targets for the treatment of MDS.
Collapse
Affiliation(s)
- Xiaofang Chen
- Department of Hematology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ningyu Li
- Department of Hematology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Jianyu Weng
- Department of Hematology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Xin Du
- Department of Hematology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
155
|
Podestà MA, Remuzzi G, Casiraghi F. Mesenchymal Stromal Cell Therapy in Solid Organ Transplantation. Front Immunol 2021; 11:618243. [PMID: 33643298 PMCID: PMC7902912 DOI: 10.3389/fimmu.2020.618243] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/29/2020] [Indexed: 12/29/2022] Open
Abstract
Transplantation is the gold-standard treatment for the failure of several solid organs, including the kidneys, liver, heart, lung and small bowel. The use of tailored immunosuppressive agents has improved graft and patient survival remarkably in early post-transplant stages, but long-term outcomes are frequently unsatisfactory due to the development of chronic graft rejection, which ultimately leads to transplant failure. Moreover, prolonged immunosuppression entails severe side effects that severely impact patient survival and quality of life. The achievement of tolerance, i.e., stable graft function without the need for immunosuppression, is considered the Holy Grail of the field of solid organ transplantation. However, spontaneous tolerance in solid allograft recipients is a rare and unpredictable event. Several strategies that include peri-transplant administration of non-hematopoietic immunomodulatory cells can safely and effectively induce tolerance in pre-clinical models of solid organ transplantation. Mesenchymal stromal cells (MSC), non-hematopoietic cells that can be obtained from several adult and fetal tissues, are among the most promising candidates. In this review, we will focus on current pre-clinical evidence of the immunomodulatory effect of MSC in solid organ transplantation, and discuss the available evidence of their safety and efficacy in clinical trials.
Collapse
Affiliation(s)
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Bergamo, Italy
| | - Federica Casiraghi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Bergamo, Italy
| |
Collapse
|
156
|
Jing D, Li C, Yao K, Xie X, Wang P, Zhao H, Feng JQ, Zhao Z, Wu Y, Wang J. The vital role of Gli1 + mesenchymal stem cells in tissue development and homeostasis. J Cell Physiol 2021; 236:6077-6089. [PMID: 33533019 DOI: 10.1002/jcp.30310] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 01/04/2021] [Accepted: 01/21/2021] [Indexed: 02/05/2023]
Abstract
The hedgehog (Hh) signaling pathway plays an essential role in both tissue development and homeostasis. Glioma-associated oncogene homolog 1 (Gli1) is one of the vital transcriptional factors as well as the direct target gene in the Hh signaling pathway. The cells expressing the Gli1 gene (Gli1+ cells) have been identified as mesenchymal stem cells (MSCs) that are responsible for various tissue developments, homeostasis, and injury repair. This review outlines some recent discoveries on the crucial roles of Gli1+ MSCs in the development and homeostasis of varieties of hard and soft tissues.
Collapse
Affiliation(s)
- Dian Jing
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chaoyuan Li
- Department of Oral Implantology, School and Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China
| | - Ke Yao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xudong Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Peiqi Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hu Zhao
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yafei Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
157
|
Aramini B, Masciale V, Grisendi G, Banchelli F, D'Amico R, Maiorana A, Morandi U, Dominici M, Haider KH. Cancer stem cells and macrophages: molecular connections and future perspectives against cancer. Oncotarget 2021; 12:230-250. [PMID: 33613850 PMCID: PMC7869576 DOI: 10.18632/oncotarget.27870] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) have been considered the key drivers of cancer initiation and progression due to their unlimited self-renewal capacity and their ability to induce tumor formation. Macrophages, particularly tumor-associated macrophages (TAMs), establish a tumor microenvironment to protect and induce CSCs development and dissemination. Many studies in the past decade have been performed to understand the molecular mediators of CSCs and TAMs, and several studies have elucidated the complex crosstalk that occurs between these two cell types. The aim of this review is to define the complex crosstalk between these two cell types and to highlight potential future anti-cancer strategies.
Collapse
Affiliation(s)
- Beatrice Aramini
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Valentina Masciale
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Grisendi
- Division of Oncology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Federico Banchelli
- Center of Statistic, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto D'Amico
- Center of Statistic, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonino Maiorana
- Institute of Pathology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Uliano Morandi
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Division of Oncology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | |
Collapse
|
158
|
Banjanin B, Schneider RK. Mesenchymal Stromal Cells as a Cellular Target in Myeloid Malignancy: Chances and Challenges in the Genome Editing of Stromal Alterations. Front Genome Ed 2021; 2:618308. [PMID: 34713241 PMCID: PMC8525402 DOI: 10.3389/fgeed.2020.618308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/22/2020] [Indexed: 11/13/2022] Open
Abstract
The contribution of bone marrow stromal cells to the pathogenesis and therapy response of myeloid malignancies has gained significant attention over the last decade. Evidence suggests that the bone marrow stroma should not be neglected in the design of novel, targeted-therapies. In terms of gene-editing, the focus of gene therapies has mainly been on correcting mutations in hematopoietic cells. Here, we outline why alterations in the stroma should also be taken into consideration in the design of novel therapeutic strategies but also outline the challenges in specifically targeting mesenchymal stromal cells in myeloid malignancies caused by somatic and germline mutations.
Collapse
Affiliation(s)
- Bella Banjanin
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, Netherlands
- Oncode Institute, Erasmus Medical Center Cancer Institute, Rotterdam, Netherlands
| | - Rebekka K. Schneider
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, Netherlands
- Oncode Institute, Erasmus Medical Center Cancer Institute, Rotterdam, Netherlands
- Department of Cell Biology, Faculty of Medicine, Institute for Biomedical Engineering, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| |
Collapse
|
159
|
Dupard SJ, Bourgine PE. 3D Engineering of Human Hematopoietic Niches in Perfusion Bioreactor. Methods Mol Biol 2021; 2308:253-262. [PMID: 34057728 DOI: 10.1007/978-1-0716-1425-9_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The hematopoietic microenvironment, also referred to as hematopoietic niche, is a functional three-dimensional (3D) unit of the bone marrow (BM) that planar culture systems cannot recapitulate. Existing limitations of 2D protocols are driving the development of advanced 3D methodologies, capable of superior modeling of the native organization and interactions between hematopoietic cells and their niche.Hereafter we describe the use of a 3D perfusion bioreactor for in vitro generation of human hematopoietic niches. The approach enables the recapitulation of the interactions between hematopoietic stem and progenitor cells (HSPCs), mesenchymal cells (MSCs), and their extracellular matrix in a 3D relevant setting. This was shown to support the functional maintenance of blood populations, self-distributing in the system compartments depending on their differentiation status. Such 3D niche modeling represents an advanced tool toward uncovering human hematopoiesis in relation to its host microenvironment , for both fundamental hematopoiesis and personalized medicine applications.
Collapse
Affiliation(s)
- Steven J Dupard
- Cell, Tissue & Organ engineering laboratory, BMC B11, 221 84, Department of Clinical Sciences Lund, Stem Cell Center, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Paul E Bourgine
- Cell, Tissue & Organ engineering laboratory, BMC B11, 221 84, Department of Clinical Sciences Lund, Stem Cell Center, Lund University, Lund, Sweden. .,Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
160
|
Ambrosi TH, Chan CKF. Skeletal Stem Cells as the Developmental Origin of Cellular Niches for Hematopoietic Stem and Progenitor Cells. Curr Top Microbiol Immunol 2021; 434:1-31. [PMID: 34850280 PMCID: PMC8864730 DOI: 10.1007/978-3-030-86016-5_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The skeletal system is a highly complex network of mesenchymal, hematopoietic, and vasculogenic stem cell lineages that coordinate the development and maintenance of defined microenvironments, so-called niches. Technological advancements in recent years have allowed for the dissection of crucial cell types as well as their autocrine and paracrine signals that regulate these niches during development, homeostasis, regeneration, and disease. Ingress of blood vessels and bone marrow hematopoiesis are initiated by skeletal stem cells (SSCs) and their more committed downstream lineage cell types that direct shape and form of skeletal elements. In this chapter, we focus on the role of SSCs as the developmental origin of niches for hematopoietic stem and progenitor cells. We discuss latest updates in the definition of SSCs, cellular processes establishing and maintaining niches, as well as alterations of stem cell microenvironments promoting malignancies. We conclude with an outlook on future studies that could take advantage of SSC-niche engineering as a basis for the development of new therapeutic tools to not only treat bone-related diseases but also maladies stemming from derailed niche dynamics altering hematopoietic output.
Collapse
Affiliation(s)
- Thomas H Ambrosi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Charles K F Chan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
161
|
Zorina T, Black L. Mesenchymal–Hematopoietic Stem Cell Axis: Applications for Induction of Hematopoietic Chimerism and Therapies for Malignancies. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
162
|
Current Understanding of Myelomatous Mesenchymal Stromal Cells Extended through Advances in Experimental Methods. Cancers (Basel) 2020; 13:cancers13010025. [PMID: 33374627 PMCID: PMC7793501 DOI: 10.3390/cancers13010025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 01/03/2023] Open
Abstract
Simple Summary As the amount of information available has grown, now it is known that many types of non-hematopoietic cells, including mesenchymal stem/progenitor cells, mature mesenchymal cells, and endothelial cells, as well as mature hematopoietic cells such as monocytes, macrophages, T-cells, and B-cells, have roles in the pathogenesis of multiple myeloma. This review focuses on the role of mesenchymal cells in the microenvironment of multiple myeloma. We summarize the experimental strategies and current understanding of the biological roles in the pathogenesis of myeloma. Furthermore, we discuss the possible clinical applications targeting mesenchymal cells. Abstract Multiple myeloma is an incurable cancer formed by malignant plasma cells. For the proliferation and survival of myeloma cells, as well as the occurrence of the complications, numerous intra- and extra-cellular mechanisms are involved. The interaction of myeloma cells with the microenvironment is known to be one of the most critical mechanisms. A specific microenvironment could affect the progression and growth of tumor cells, as well as drug resistance. Among various microenvironment components, such as hematological and non-hematological cells, and soluble factors (cytokines, chemokines, and extracellular matrix (ECM) proteins), in this review, we focus on the role of mesenchymal cells. We aimed to summarize the experimental strategies used for conducting studies and current understanding of the biological roles in the pathogenesis of myeloma. Furthermore, we discuss the possible clinical applications targeting mesenchymal cells.
Collapse
|
163
|
Zorina TD. New Insights on the Role of the Mesenchymal-Hematopoietic Stem Cell Axis in Autologous and Allogeneic Hematopoiesis. Stem Cells Dev 2020; 30:2-16. [PMID: 33231142 DOI: 10.1089/scd.2020.0148] [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] [Indexed: 11/13/2022] Open
Abstract
Cytoreductive protocols are integral both as conditioning regimens for bone marrow (BM) transplantation and as part of therapies for malignancies, but their associated comorbidities represent a long-standing clinical problem. In particular, they cause myeloablation that debilitates the physiological role of mesenchymal stem and precursor cells (MSPCs) in sustaining hematopoiesis. This review addresses the damaging impact of cytoreductive regimens on MSPCs. In addition, it discusses prospects for alleviating the resulting iatrogenic comorbidities. New insights into the structural and functional dynamics of hematopoietic stem cell (HSC) niches reveal the existence of "empty" niches and the ability of the donor-derived healthy HSCs to outcompete the defective HSCs in occupying these niches. These findings support the notion that conditioning regimens, conventionally used to ablate the recipient hematopoiesis to create space for engraftment of the donor-derived HSCs, may not be a necessity for allogeneic BM transplantation. In addition, the capacity of the MSPCs to cross-talk with HSCs, despite major histocompatibility complex disparity, and suppress graft versus host disease indicates the possibility for development of a conditioning-free, MSPCs-enhanced protocol for BM transplantation. The clinical advantage of supplementing cytoreductive protocols with MSPCs to improve autologous hematopoiesis reconstitution and alleviate cytopenia associated with chemo and radiation therapies for cancer is also discussed.
Collapse
Affiliation(s)
- Tatiana D Zorina
- Department of Medical Laboratory Science and Biotechnology, Jefferson College of Health Professions, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
164
|
Lu S, Qiao X. Single-cell profiles of human bone marrow-derived mesenchymal stromal cells after IFN-γ and TNF-α licensing. Gene 2020; 771:145347. [PMID: 33333228 DOI: 10.1016/j.gene.2020.145347] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/12/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Pre-licensing mesenchymal stromal cells (MSCs) with IFN-γ and TNF-α can empower their immune fate and induce a more effective immune regulation. However, the cellular heterogeneity of MSCs limits our understanding of this inflammatory licensing. METHODS The publicly available Gene Expression Omnibus single-cell RNA sequencing (scRNA-seq) data of human bone marrow-derived MSCs with or without IFN-γ and TNF-α licensing were analyzed. Based on the scRNA-seq data and related marker genes, the cell-cycle, stemness, differentiative potencies, and immunomodulate capability of unlicensed and licensed MSCs were compared. RESULTS After removing low-quality cells and regressing out the ribosomal gene effects, high-quality data reflecting IFN-γ and TNF-α effect on MSCs were chosen for further analysis. Despite the heterogeneity, pre-licensing didn't influence the cell-cycle and stemness of human bone marrow-derived MSCs. The osteogenesis potencies were decreased, the chondrogenesis potencies were increased while the adipogenesis potencies were stable in licensed MSCs. Licensed MSCs also showed more effective immunomodulate capability including expression of related chemokines, cytokines, surface molecules, and receptors. CONCLUSION Collectively, our study showed the expression profiles of human bone marrow-derived unlicensed and licensed MSCs about the cell cycle, stemness, differentiative potencies, and immunomodulate capability at single-cell resolution, which may help the comprehensive understanding about the inflammatory licensing of human bone marrow-derived MSCs and their further clinical application.
Collapse
Affiliation(s)
- Shuanglong Lu
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai 200065, China
| | - Xiaohong Qiao
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai 200065, China.
| |
Collapse
|
165
|
Abstract
Haematopoietic stem and progenitor cell (HSPC) gene therapy has emerged as an effective treatment modality for monogenic disorders of the blood system such as primary immunodeficiencies and β-thalassaemia. Medicinal products based on autologous HSPCs corrected using lentiviral and gammaretroviral vectors have now been approved for clinical use, and the site-specific genome modification of HSPCs using gene editing techniques such as CRISPR-Cas9 has shown great clinical promise. Preclinical studies have shown engineered HSPCs could also be used to cross-correct non-haematopoietic cells in neurodegenerative metabolic diseases. Here, we review the most recent advances in HSPC gene therapy and discuss emerging strategies for using HSPC gene therapy for a range of diseases.
Collapse
|
166
|
Thompson ER, Connelly C, Ali S, Sheerin NS, Wilson CH. Cell therapy during machine perfusion. Transpl Int 2020; 34:49-58. [PMID: 33131097 DOI: 10.1111/tri.13780] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/03/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022]
Abstract
There has been increasing use of organs from extended criteria or donation after circulatory death donors to meet the demands of the transplant waiting list. Over the past decade, there has been considerable progress in technologies to preserve organs prior to transplantation to improve the function of these marginal organs. This has led to the development of normothermic machine perfusion, whereby an organ is perfused with warmed, oxygenated blood and nutrients to resume normal physiological function in an isolated ex-vivo platform. With this advance in preservation comes significant opportunities to recondition, repair and regenerate organs prior to transplantation using cellular therapies. This review aims to discuss the possibilities of machine perfusion technology; highlighting the potential for organ-directed reconditioning and the future avenues for investigation in this field.
Collapse
Affiliation(s)
- Emily R Thompson
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Chloe Connelly
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Simi Ali
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Neil S Sheerin
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Colin H Wilson
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
167
|
Camacho V, Matkins VR, Patel SB, Lever JM, Yang Z, Ying L, Landuyt AE, Dean EC, George JF, Yang H, Ferrell PB, Maynard CL, Weaver CT, Turnquist HR, Welner RS. Bone marrow Tregs mediate stromal cell function and support hematopoiesis via IL-10. JCI Insight 2020; 5:135681. [PMID: 33208555 PMCID: PMC7710301 DOI: 10.1172/jci.insight.135681] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 10/07/2020] [Indexed: 12/31/2022] Open
Abstract
The nonimmune roles of Tregs have been described in various tissues, including the BM. In this study, we comprehensively phenotyped marrow Tregs, elucidating their key features and tissue-specific functions. We show that marrow Tregs are migratory and home back to the marrow. For trafficking, marrow Tregs use S1P gradients, and disruption of this axis allows for specific targeting of the marrow Treg pool. Following Treg depletion, the function and phenotype of both mesenchymal stromal cells (MSCs) and hematopoietic stem cells (HSCs) was impaired. Transplantation also revealed that a Treg-depleted niche has a reduced capacity to support hematopoiesis. Finally, we found that marrow Tregs are high producers of IL-10 and that Treg-secreted IL-10 has direct effects on MSC function. This is the first report to our knowledge revealing that Treg-secreted IL-10 is necessary for stromal cell maintenance, and our work outlines an alternative mechanism by which this cytokine regulates hematopoiesis.
Collapse
Affiliation(s)
| | | | | | - Jeremie M. Lever
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, and
| | - Zhengqin Yang
- Division of Cardiothoracic Surgery, Department of Surgery, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Li Ying
- Cancer Science Institute of Singapore & Department of Biochemistry, National University of Singapore, Singapore
| | - Ashley E. Landuyt
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Emma C. Dean
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James F. George
- Division of Cardiothoracic Surgery, Department of Surgery, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Henry Yang
- Cancer Science Institute of Singapore & Department of Biochemistry, National University of Singapore, Singapore
| | - Paul Brent Ferrell
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Craig L. Maynard
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Casey T. Weaver
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Heth R. Turnquist
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
168
|
Brennen WN, J Thorek DL, Jiang W, Krueger TE, Antony L, Denmeade SR, Isaacs JT. Overcoming stromal barriers to immuno-oncological responses via fibroblast activation protein-targeted therapy. Immunotherapy 2020; 13:155-175. [PMID: 33148078 DOI: 10.2217/imt-2020-0066] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The tumor microenvironment contributes to disease progression through multiple mechanisms, including immune suppression mediated in part by fibroblast activation protein (FAP)-expressing cells. Herein, a review of FAP biology is presented, supplemented with primary data. This includes FAP expression in prostate cancer and activation of latent reservoirs of TGF-β and VEGF to produce a positive feedback loop. This collectively suggests a normal wound repair process subverted during cancer pathophysiology. There has been immense interest in targeting FAP for diagnostic, monitoring and therapeutic purposes. Until recently, this development has outpaced an understanding of the biology; impeding optimal translation into the clinic. A summary of these applications is provided with an emphasis on eliminating tumor-infiltrating FAP-positive cells to overcome stromal barriers to immuno-oncological responses.
Collapse
Affiliation(s)
- W Nathaniel Brennen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, MD 21287, USA
| | - Daniel L J Thorek
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO 63310, USA.,Department of Biomedical Engineering, Washington University School of Medicine, Saint Louis, MO 63310, USA
| | - Wen Jiang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Timothy E Krueger
- Department of Pharmacology & Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Lizamma Antony
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, MD 21287, USA
| | - Samuel R Denmeade
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, MD 21287, USA
| | - John T Isaacs
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, MD 21287, USA
| |
Collapse
|
169
|
Abney KK, Galipeau J. Aryl hydrocarbon receptor in mesenchymal stromal cells: new frontiers in AhR biology. FEBS J 2020; 288:3962-3972. [PMID: 33064873 DOI: 10.1111/febs.15599] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/30/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022]
Abstract
Mesenchymal stromal cells (MSCs) are nonhematopoietic cells that have been clinically explored as investigational cellular therapeutics for tissue injury regeneration and immune-mediated diseases. Their pharmaceutical properties arise from activation of endogenous receptors and transcription factors leading to a paracrine effect which mirror the biology of progenitors from which they arise. The aryl hydrocarbon receptor (AhR) is a transcription factor that has been extensively studied as an environmental sensor for xenobiotics, but recent findings suggest it can modulate immunological functions. Both genetic and pharmacological investigations revealed that MSCs express AhR and that it plays roles in inflammation, immunomodulation, and mesodermal plasticity of endogenous MSCs. Further, AhR has been shown to interact with key signaling cascades associated with these conditions. Therefore, AhR has potential to be an attractive target in both endogenous and culture-adapted MSCs for novel therapeutics to treat inflammation and other age-related disorders.
Collapse
Affiliation(s)
- Kristopher K Abney
- Department of Medicine and Carbone Cancer Center, University of Wisconsin in Madison, WI, USA
| | - Jacques Galipeau
- Department of Medicine and Carbone Cancer Center, University of Wisconsin in Madison, WI, USA
| |
Collapse
|
170
|
Hou H, Zhang L, Duan L, Liu Y, Han Z, Li Z, Cao X. Spatio-Temporal Metabolokinetics and Efficacy of Human Placenta-Derived Mesenchymal Stem/Stromal Cells on Mice with Refractory Crohn's-like Enterocutaneous Fistula. Stem Cell Rev Rep 2020; 16:1292-1304. [PMID: 33011925 DOI: 10.1007/s12015-020-10053-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 12/14/2022]
Abstract
Crohn's disease (CD) with externally fistulizing openings indicates the aggressive and relapsing manifestation and results in undesirable long-term outcomes of patients. MSC-based approach combined with multidisciplinary strategy has mandated a redefinition of the administration and management of numerous recurrent and refractory diseases whereas the spatio-temporal evaluation of the metabolokinetics and efficacy of MSCs on intractable CD with enterocutaneous fistula (EF) are largely inaccessible and dauntingly complex. Herein, we primitively established dual-fluorescence expressing placenta-derived MSCs (DF-MSCs) and explored their multidimensional attributes, including cytomorphology, immunophenotying, multilineage differentiation and long-term proliferation, together with the recognition of bifluorescence intensity (BLI). Then, with the aid of in vivo living imaging, clinicopathological or inflammatory cytokine examinations and in vitro analyses, we systematically and meticulously dissected the metabolokinetics and curative effect of MSCs on mice with refractory Crohn's-like EF (EF mice), together with revealing the underlying mechanism including reactive oxygen species (ROS) and neovascularization. Strikingly, the DF-MSCs exhibited stabilized BLI and biological properties. The spatio-temporal distribution and therapeutic process of MSCs in EF mice were intuitively delineated. Meanwhile, our data indicated the curative mechanisms of DF-MSCs by simultaneously downregulating ROS and accelerating neovascularization. Collectively, we systematically illuminated the spatio-temporal biofunction and mechanism of DF-MSCs on EF mice. Our findings have supplied new references for safety and effectiveness assessments as well as the establishment of guidelines for optimal administrations of MSC-based cytotherapy in preclinical studies, which collectively indicates the prospect of P-MSC administration in clinical trials during a wide spectrum of disease remodeling including the fistulizing CD. Graphical abstract.
Collapse
Affiliation(s)
- Huixing Hou
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Leisheng Zhang
- The Postdoctoral Research Station, School of Medicine, Nankai University, Tianjin, 300071, China. .,State Key Laboratory of Experimental Hematology & National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, People's Republic of China. .,Precision Medicine Division, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd, Tianjin, 301700, China.
| | - Liyun Duan
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Yuanyuan Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Zhongchao Han
- State Key Laboratory of Experimental Hematology & National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, People's Republic of China.,Precision Medicine Division, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd, Tianjin, 301700, China
| | - Zongjin Li
- The Postdoctoral Research Station, School of Medicine, Nankai University, Tianjin, 300071, China.
| | - Xiaocang Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, 300052, China.
| |
Collapse
|
171
|
Aimaiti A, Wahafu T, Keremu A, Yicheng L, Li C. Strontium Ameliorates Glucocorticoid Inhibition of Osteogenesis Via the ERK Signaling Pathway. Biol Trace Elem Res 2020; 197:591-598. [PMID: 31832923 DOI: 10.1007/s12011-019-02009-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 12/05/2019] [Indexed: 02/06/2023]
Abstract
Glucocorticoid (GC) has been widely used in clinical work due to its anti-inflammatory and immune-inhibitory properties. However, long-term or high-dose administration is associated with side effects, such as GC-induced osteoporosis (GIOP), which causes great pain for and poses a heavy financial burden on patients. We sought to investigate the potential effects of strontium on GIOP and further explore its underlying mechanisms, including its reversal of the inhibitory effect of GC on osteogenesis of bone marrow-derived mesenchymal stem cells (BMSCs). We incubated BMSCs with Dexamethasone (DEX) in combination with or without strontium and then measured osteogenic and adipogenic gene expression levels by RT-qPCR and Western blot. We added a specific ERK signaling pathway inhibitor, U0126, to evaluate the involvement of that pathway. Strontium promoted osteogenic differentiation and matrix mineralization in DEX-treated BMSCs, accompanied by upregulation of RUNX2, Osx, ALP, BSP, COL1A1, and OCN. DEX blocked the expression of several osteogenesis-related marker genes by activating the ERK signaling pathway. U0126 attenuated the suppression of osteogenesis in DEX-treated BMSCs. These results suggested that strontium could enhance osteogenic differentiation and matrix mineralization by counteracting DEX's inhibitory effect on osteogenesis via the ERK signaling pathway. Therefore, strontium might be a promising therapeutic agent for GIOP.
Collapse
Affiliation(s)
- Abudousaimi Aimaiti
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, 137 South LiYuShan Road, Urumqi, 830054, Xinjiang, China
| | - Tuerhongjiang Wahafu
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, 137 South LiYuShan Road, Urumqi, 830054, Xinjiang, China
| | - Ajimu Keremu
- Orthopedic Center, First People's Hospital of Kashgar, Kashgar, 844000, Xinjiang, China
| | - Li Yicheng
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, 137 South LiYuShan Road, Urumqi, 830054, Xinjiang, China
| | - Cao Li
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, 137 South LiYuShan Road, Urumqi, 830054, Xinjiang, China.
| |
Collapse
|
172
|
Bone marrow aspirate clot: A feasible orthobiologic. J Clin Orthop Trauma 2020; 11:S789-S794. [PMID: 32999557 PMCID: PMC7503156 DOI: 10.1016/j.jcot.2020.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 07/05/2020] [Indexed: 12/29/2022] Open
Abstract
Musculoskeletal disorders are one of the major health burdens and a leading source of disability worldwide, affecting both juvenile and elderly populations either as a consequence of ageing or extrinsic factors such as physical injuries. This condition often involves a group of locomotor structures such as the bones, joints and muscles and may therefore cause significant economic and emotional impact. Some pharmacological and non-pharmacological treatments have been considered as potential solutions, however, these alternatives have provided quite limited efficacy due to the short-term effect on pain management and inability to restore damaged tissue. The emergence of novel therapeutic alternatives such as the application of orthobiologics, particularly bone marrow aspirate (BMA) clot, have bestowed medical experts with considerable optimism as evidenced by the significant results found in numerous studies addressed in this manuscript. Although other products have been proposed for the treatment of musculoskeletal injuries, the peculiar interest in BMA, fibrin clot and associated fibrinolytic mechanisms continues to expand. BMA is a rich source of various cellular and molecular components which have demonstrated positive effects on tissue regeneration in many in vitro and in vivo models of musculoskeletal injuries. In addition to being able to undergo self-renewal and differentiation, the hematopoietic and mesenchymal stem cells present in this orthobiologic elicit key immunomodulatory and paracrine roles in inflammatory responses in tissue injury and drive the coagulation cascade towards tissue repair via different mechanisms. Although promising, these complex regenerative mechanisms have not yet been fully elucidated.
Collapse
|
173
|
Helbling PM, Piñeiro-Yáñez E, Gerosa R, Boettcher S, Al-Shahrour F, Manz MG, Nombela-Arrieta C. Global Transcriptomic Profiling of the Bone Marrow Stromal Microenvironment during Postnatal Development, Aging, and Inflammation. Cell Rep 2020; 29:3313-3330.e4. [PMID: 31801092 DOI: 10.1016/j.celrep.2019.11.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/03/2019] [Accepted: 10/31/2019] [Indexed: 12/22/2022] Open
Abstract
Bone marrow (BM) stromal cells provide the regulatory framework for hematopoiesis and contribute to developmental stage-specific niches, such as those preserving hematopoietic stem cells. Despite advances in our understanding of stromal function, little is known about the transcriptional changes that this compartment undergoes throughout lifespan and during adaptation to stress. Using RNA sequencing, we perform transcriptional analyses of four principal stromal subsets, namely CXCL12-abundant reticular, platelet-derived growth factor receptor (PDGFR)-α+Sca1+, sinusoidal, and arterial endothelial cells, from early postnatal, adult, and aged mice. Our data reveal (1) molecular fingerprints defining cell-specific anatomical and functional features, (2) a radical reprogramming of pro-hematopoietic, immune, and matrisomic transcriptional programs during the transition from juvenile stages to adulthood, and (3) the aging-driven progressive upregulation of pro-inflammatory gene expression in stroma. We further demonstrate that transcriptomic pathways elicited in vivo by prototypic microbial molecules are largely recapitulated during aging, thereby supporting the inflammatory basis of age-related adaptations of BM hematopoietic function.
Collapse
Affiliation(s)
- Patrick M Helbling
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Elena Piñeiro-Yáñez
- Bioinformatics Unit, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
| | - Rahel Gerosa
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Steffen Boettcher
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Fátima Al-Shahrour
- Bioinformatics Unit, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - César Nombela-Arrieta
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, 8091 Zurich, Switzerland.
| |
Collapse
|
174
|
Lou B, Hu Y, Lu X, Zhang X, Li Y, Pi J, Xu Y. Long-isoform NRF1 protects against arsenic cytotoxicity in mouse bone marrow-derived mesenchymal stem cells by suppressing mitochondrial ROS and facilitating arsenic efflux. Toxicol Appl Pharmacol 2020; 407:115251. [PMID: 32980394 DOI: 10.1016/j.taap.2020.115251] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 09/02/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022]
Abstract
Acute exposure to arsenic is known to cause bone marrow depression and result in anemia, in which the dusfunction of cells in the bone marrow niche such as mesenchymal stem cells (MSCs) is vital. However, the mechanism underlying response of MSCs to arsenic challange is not fully understood. In the present study, we investigated the role of nuclear factor erythroid 2-related factor (NRF) 1 (NRF1), a sister member of the well-known master regulator in antioxidative response NRF2, in arsenite-induced cytotoxicity in mouse bone marrow-derived MSCs (mBM-MSCs). We found that arsenite exposure induced significant increase in the protein level of long-isoform NRF1 (L-NRF1). Though short-isoform NRF1 (S-NRF1) was induced by arsenite at mRNA level, its protein level was not obviously altered. Silencing L-Nrf1 sensitized the cells to arsenite-induced cytotoxicity. L-Nrf1-silenced mBM-MSCs showed decreased arsenic efflux with reduced expression of arsenic transporter ATP-binding cassette subfamily C member 4 (ABCC4), as well as compromised NRF2-mediated antioxidative defense with elevated level of mitochondrial reactive oxygen species (mtROS) under arsenite-exposed conditions. A specific mtROS scavenger (Mito-quinone) alleviated cell apoptosis induced by arsenite in L-Nrf1-silenced mBM-MSCs. Taken together, these findings suggest that L-NRF1 protects mBM-MSCs from arsenite-induced cytotoxicity via suppressing mtROS in addition to facilitating cellular arsenic efflux.
Collapse
Affiliation(s)
- Bin Lou
- Laboratory of Chronic Diseases and Environmental Genetics, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Yuxin Hu
- Experimental Teaching Center, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Xiaoyu Lu
- Laboratory of Chronic Diseases and Environmental Genetics, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Xinyu Zhang
- Laboratory of Chronic Diseases and Environmental Genetics, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Yongfang Li
- The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China; Research Center of Environment and Non-Communicable Diseases, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Jingbo Pi
- Experimental Teaching Center, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China; Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Yuanyuan Xu
- Laboratory of Chronic Diseases and Environmental Genetics, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China; Experimental Teaching Center, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China.
| |
Collapse
|
175
|
Novoseletskaya E, Grigorieva O, Nimiritsky P, Basalova N, Eremichev R, Milovskaya I, Kulebyakin K, Kulebyakina M, Rodionov S, Omelyanenko N, Efimenko A. Mesenchymal Stromal Cell-Produced Components of Extracellular Matrix Potentiate Multipotent Stem Cell Response to Differentiation Stimuli. Front Cell Dev Biol 2020; 8:555378. [PMID: 33072743 PMCID: PMC7536557 DOI: 10.3389/fcell.2020.555378] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022] Open
Abstract
Extracellular matrix (ECM) provides both structural support and dynamic microenvironment for cells regulating their behavior and fate. As a critical component of stem cell niche ECM maintains stem cells and activates their proliferation and differentiation under specific stimuli. Mesenchymal stem/stromal cells (MSCs) regulate tissue-specific stem cell functions locating in their immediate microenvironment and producing various bioactive factors, including ECM components. We evaluated the ability of MSC-produced ECM to restore stem and progenitor cell microenvironment in vitro and analyzed the possible mechanisms of its effects. Human MSC cell sheets were decellularized by different agents (detergents, enzymes, and apoptosis inductors) to select the optimized combination (CHAPS and DNAse I) based on the conservation of decellularized ECM (dECM) structure and effectiveness of DNA removal. Prepared dECM was non-immunogenic, supported MSC proliferation and formation of larger colonies in colony-forming unit-assay. Decellularized ECM effectively promoted MSC trilineage differentiation (adipogenic, osteogenic, and chondrogenic) compared to plastic or plastic covered by selected ECM components (collagen, fibronectin, laminin). Interestingly, dECM produced by human fibroblasts could not enhance MSC differentiation like MSC-produced dECM, indicating cell-specific functionality of dECM. We demonstrated the significant integrin contribution in dECM-cell interaction by blocking the stimulatory effects of dECM with RGD peptide and suggested the involvement of key intracellular signaling pathways activation (pERK/ERK and pFAK/FAK axes, pYAP/YAP and beta-catenin) in the observed processes based on the results of inhibitory analysis. Taken together, we suppose that MSC-produced dECM may mimic stem cell niche components in vitro and maintain multipotent progenitor cells to insure their effective response to external differentiating stimuli upon activation. The obtained data provide more insights into the possible role of MSC-produced ECM in stem and progenitor cell regulation within their niches. Our results are also useful for the developing of dECM-based cell-free products for regenerative medicine.
Collapse
Affiliation(s)
- Ekaterina Novoseletskaya
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Olga Grigorieva
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Peter Nimiritsky
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Nataliya Basalova
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Roman Eremichev
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Irina Milovskaya
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Konstantin Kulebyakin
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Maria Kulebyakina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Sergei Rodionov
- N.N. Priorov National Medical Research Center of Traumatology and Orthopedics, Moscow, Russia
| | - Nikolai Omelyanenko
- N.N. Priorov National Medical Research Center of Traumatology and Orthopedics, Moscow, Russia
| | - Anastasia Efimenko
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
176
|
Paradoxical Regulation of Allogeneic Bone Marrow Engraftment and Immune Privilege by Mesenchymal Cells and Adenosine. Transplant Cell Ther 2020; 27:92.e1-92.e5. [PMID: 32961376 DOI: 10.1016/j.bbmt.2020.09.017] [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: 07/09/2019] [Revised: 08/04/2020] [Accepted: 09/07/2020] [Indexed: 12/26/2022]
Abstract
Although mesenchymal stromal cell (MSC) transfer has long drawn attention owing to its immunosuppressive potential to treat immune-mediated diseases, the role of endogenous MSCs in immune regulation in vivo has remained largely unclear. MSCs constitute the hematopoietic stem cell (HSC) niche, perhaps contributing to immune protection of HSCs, termed immune privilege. Our recent study demonstrates that immune privilege of HSCs is endowed by niche-residential regulatory T cells (Tregs), which promote allogeneic HSC engraftment. This immune privilege depends on cell surface ectoenzymes CD39 and CD73 on niche Tregs, which generate extracellular adenosine, a nucleotide known to suppress immunity and potentiate Tregs. Another niche constituent, leptin receptor-expressing (lepr+) perivascular MSCs, also highly express CD39 and CD73, prompting us to study their roles in immune privilege. This work demonstrates an unexpected negative regulation of immune privilege by MSC-derived adenosine. CD39 deletion in lepr+ cells increased and potentiated effector memory-like niche Tregs, promoting allogeneic HSC engraftment. CD39 deletion in Tregs also activated niche Tregs, while abrogating engraftment. These observations demonstrate paradoxical effects of MSC-derived adenosine to activate immunity, revealing a previously undescribed dual roles of adenosine. Adenosine from both Tregs and MSCs inhibits niche Tregs, whereas adenosine from Tregs, but not that from MSCs, acts as an effector molecule of immune privilege.
Collapse
|
177
|
Ruan Y, Kim HN, Ogana H, Kim YM. Wnt Signaling in Leukemia and Its Bone Marrow Microenvironment. Int J Mol Sci 2020; 21:ijms21176247. [PMID: 32872365 PMCID: PMC7503842 DOI: 10.3390/ijms21176247] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/16/2020] [Accepted: 08/24/2020] [Indexed: 12/19/2022] Open
Abstract
Leukemia is an aggressive hematologic neoplastic disease. Therapy-resistant leukemic stem cells (LSCs) may contribute to the relapse of the disease. LSCs are thought to be protected in the leukemia microenvironment, mainly consisting of mesenchymal stem/stromal cells (MSC), endothelial cells, and osteoblasts. Canonical and noncanonical Wnt pathways play a critical role in the maintenance of normal hematopoietic stem cells (HSC) and LSCs. In this review, we summarize recent findings on the role of Wnt signaling in leukemia and its microenvironment and provide information on the currently available strategies for targeting Wnt signaling.
Collapse
Affiliation(s)
- Yongsheng Ruan
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90027, USA; (Y.R.); (H.N.K.); (H.O.)
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hye Na Kim
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90027, USA; (Y.R.); (H.N.K.); (H.O.)
| | - Heather Ogana
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90027, USA; (Y.R.); (H.N.K.); (H.O.)
| | - Yong-Mi Kim
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90027, USA; (Y.R.); (H.N.K.); (H.O.)
- Correspondence:
| |
Collapse
|
178
|
Häussinger D, Kordes C. Space of Disse: a stem cell niche in the liver. Biol Chem 2020; 401:81-95. [PMID: 31318687 DOI: 10.1515/hsz-2019-0283] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023]
Abstract
Recent evidence indicates that the plasticity of preexisting hepatocytes and bile duct cells is responsible for the appearance of intermediate progenitor cells capable of restoring liver mass after injury without the need of a stem cell compartment. However, mesenchymal stem cells (MSCs) exist in all organs and are associated with blood vessels which represent their perivascular stem cell niche. MSCs are multipotent and can differentiate into several cell types and are known to support regenerative processes by the release of immunomodulatory and trophic factors. In the liver, the space of Disse constitutes a stem cell niche that harbors stellate cells as liver resident MSCs. This perivascular niche is created by extracellular matrix proteins, sinusoidal endothelial cells, liver parenchymal cells and sympathetic nerve endings and establishes a microenvironment that is suitable to maintain stellate cells and to control their fate. The stem cell niche integrity is important for the behavior of stellate cells in the normal, regenerative, aged and diseased liver. The niche character of the space of Disse may further explain why the liver can become an organ of extra-medullar hematopoiesis and why this organ is frequently prone to tumor metastasis.
Collapse
Affiliation(s)
- Dieter Häussinger
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Moorenstraße 5, D-40225 Düsseldorf, Germany
| | - Claus Kordes
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Moorenstraße 5, D-40225 Düsseldorf, Germany
| |
Collapse
|
179
|
Wang L, Zhang L, Liang X, Zou J, Liu N, Liu T, Wang G, Ding X, Liu Y, Zhang B, Liang R, Wang S. Adipose Tissue-Derived Stem Cells from Type 2 Diabetics Reveal Conservative Alterations in Multidimensional Characteristics. Int J Stem Cells 2020; 13:268-278. [PMID: 32587133 PMCID: PMC7378902 DOI: 10.15283/ijsc20028] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/14/2020] [Accepted: 05/23/2020] [Indexed: 12/17/2022] Open
Abstract
Background and Objectives Adipose tissue-derived mesenchymal stem cells (ASCs) are recognized as an advantaged source for the prevention and treatment of diverse diseases including type 2 diabetes mellitus (T2DM). However, alterations in characteristics of ASCs from the aforementioned T2DM patients are still obscure, which also hinder the rigorous and systematic illumination of progression and pathogenesis. Methods and Results In this study, we originally isolated peripancreatic adipose tissue-derived mesenchymal stem cells from both human type 2 diabetic and non-diabetic donors (T2DM-ASCs, ND-ASCs) with the parental consent, respectively. We noticed that T2DM-ASCs exhibited indistinguishable immunophenotype, cell vitality, chondrogenic differentiation and stemness as ND-ASCs. Simultaneously, there’s merely alterations in migration and immunoregulatory capacities in T2DM-ASCs. However, differing from ND-ASCs, T2DM-ASCs exhibited deficiency in adipogenic and osteogenic differentiation, and in particular, the delayed cell cycle and different cytokine expression spectrum. Conclusions The conservative alterations of T2DM-ASCs in multifaceted characteristics indicated the possibility of autologous application of ASCs for cell-based T2DM treatment in the future.
Collapse
Affiliation(s)
- Le Wang
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China.,NHC Key Laboratory for Critical Care Medicine, Tianjin, China.,Tianjin Clinical Research Center for Organ Transplantation, Tianjin, China
| | - Leisheng Zhang
- The Postdoctoral Research Station, School of Medicine, Nankai University, Tianjin, China
| | - Xue Liang
- NHC Key Laboratory for Critical Care Medicine, Tianjin, China
| | - Jiaqi Zou
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China.,NHC Key Laboratory for Critical Care Medicine, Tianjin, China
| | - Na Liu
- NHC Key Laboratory for Critical Care Medicine, Tianjin, China
| | - Tengli Liu
- NHC Key Laboratory for Critical Care Medicine, Tianjin, China
| | - Guanqiao Wang
- NHC Key Laboratory for Critical Care Medicine, Tianjin, China
| | - Xuejie Ding
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China.,NHC Key Laboratory for Critical Care Medicine, Tianjin, China
| | - Yaojuan Liu
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China.,Tianjin Clinical Research Center for Organ Transplantation, Tianjin, China
| | - Boya Zhang
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China.,Tianjin Clinical Research Center for Organ Transplantation, Tianjin, China
| | - Rui Liang
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China.,NHC Key Laboratory for Critical Care Medicine, Tianjin, China
| | - Shusen Wang
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China.,Tianjin Clinical Research Center for Organ Transplantation, Tianjin, China
| |
Collapse
|
180
|
Mesenchymal Stem Cells in Aplastic Anemia and Myelodysplastic Syndromes: The "Seed and Soil" Crosstalk. Int J Mol Sci 2020; 21:ijms21155438. [PMID: 32751628 PMCID: PMC7432231 DOI: 10.3390/ijms21155438] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/28/2020] [Accepted: 07/28/2020] [Indexed: 12/20/2022] Open
Abstract
There is growing interest in the contribution of the marrow niche to the pathogenesis of bone marrow failure syndromes, i.e., aplastic anemia (AA) and myelodysplastic syndromes (MDSs). In particular, mesenchymal stem cells (MSCs) are multipotent cells that contribute to the organization and function of the hematopoietic niche through their repopulating and supporting abilities, as well as immunomodulatory properties. The latter are of great interest in MDSs and, particularly, AA, where an immune attack against hematopoietic stem cells is the key pathogenic player. We, therefore, conducted Medline research, including all available evidence from the last 10 years concerning the role of MSCs in these two diseases. The data presented show that MSCs display morphologic, functional, and genetic alterations in AA and MDSs and contribute to immune imbalance, ineffective hematopoiesis, and leukemic evolution. Importantly, adoptive MSC infusion from healthy donors can be exploited to heal the "sick" niche, with even better outcomes if cotransplanted with allogeneic hematopoietic stem cells. Finally, future studies on MSCs and the whole microenvironment will further elucidate AA and MDS pathogenesis and possibly improve treatment.
Collapse
|
181
|
Ryk modulates the niche activity of mesenchymal stromal cells by fine-tuning canonical Wnt signaling. Exp Mol Med 2020; 52:1140-1151. [PMID: 32724069 PMCID: PMC8080773 DOI: 10.1038/s12276-020-0477-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 04/02/2020] [Accepted: 04/02/2020] [Indexed: 12/12/2022] Open
Abstract
The importance of modulating the intensity of Wnt signaling has been highlighted in various biological models, but their mechanisms remain unclear. In this study, we found that Ryk—an atypical Wnt receptor with a pseudokinase domain—has a Wnt-modulating effect in bone marrow stromal cells to control hematopoiesis-supporting activities. We first found that Ryk is predominantly expressed in the mesenchymal stromal cells (MSCs) of the bone marrow (BM) compared with hematopoietic cells. Downregulation of Ryk in MSCs decreased their clonogenic activity and ability to support self-renewing expansion of primitive hematopoietic progenitors (HPCs) in response to canonical Wnt ligands. In contrast, under high concentrations of Wnt, Ryk exerted suppressive effects on the transactivation of target genes and HPC-supporting effects in MSCs, thus fine-tuning the signaling intensity of Wnt in BM stromal cells. This ability of Ryk to modulate the HPC-supporting niche activity of MSCs was abrogated by induction of deletion mutants of Ryk lacking the intracellular domain or extracellular domain, indicating that the pseudokinase-containing intracellular domain mediates the Wnt-modulating effects in response to extracellular Wnt ligands. These findings indicate that the ability of the BM microenvironment to respond to extracellular signals and support hematopoiesis may be fine-tuned by Ryk via modulation of Wnt signaling intensity to coordinate hematopoietic activity. Steady production of immune and blood cells depends on a signaling protein that helps maintain stable stem cell populations within the bone marrow. Hematopoietic stem cells (HSCs), which give rise to blood cells, reside within a supportive “niche” surrounded by mesenchymal stromal cells (MSCs), with extensive communication between the two populations. Researchers led by Il-Hoan Oh at The Catholic University of Korea, Seoul, have now identified a mechanism that MSCs employ to stabilize the niche environment through fine-tuning the signaling intensity of Wnt. Oh and colleagues focused on a signaling pathway that controls the undifferentiated state of HSCs, and showed that these signals are specifically modulated by an MSC protein known as Ryk. Without Ryk, MSCs can no longer promote HSC proliferation. However, when these signals are excessively strong, Ryk helps suppress proliferation to keep HSC numbers under control.
Collapse
|
182
|
Chen L, Wang CT, Forsyth NR, Wu P. Transcriptional profiling reveals altered biological characteristics of chorionic stem cells from women with gestational diabetes. Stem Cell Res Ther 2020; 11:319. [PMID: 32711583 PMCID: PMC7382800 DOI: 10.1186/s13287-020-01828-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 06/30/2020] [Accepted: 07/13/2020] [Indexed: 12/17/2022] Open
Abstract
Background Gestational diabetes (GDM) is a common complication of pregnancy. The impact of pregnancy complications on placental function suggests that extraembryonic stem cells in the placenta may also be affected during pregnancy. Neonatal tissue-derived stem cells, with the advantages of their differentiation capacity and non-invasive isolation processes, have been proposed as a promising therapeutic avenue for GDM management through potential cell therapy approaches. However, the influence of GDM on autologous stem cells remains unclear. Thus, studies that provide comprehensive understanding of stem cells isolated from women with GDM are essential to guide future clinical applications. Methods Human chorionic membrane-derived stem cells (CMSCs) were isolated from placentas of healthy and GDM pregnancies. Transcriptional profiling was performed by DNA microarray, and differentially regulated genes between GDM- and Healthy-CMSCs were used to analyse molecular functions, differentiation, and pathway enrichment. Altered genes and biological functions were validated via real-time PCR and in vitro assays. Results GDM-CMSCs displayed, vs. Healthy-CMSCs, 162 upregulated genes associated with increased migration ability, epithelial development, and growth factor-associated signal transduction while the 269 downregulated genes were strongly linked to angiogenesis and cellular metabolic processes. Notably, significantly reduced expression of detoxification enzymes belonging to the aldehyde dehydrogenase gene families (ALDH1A1/1A2, ALDH2, ALDH3) accounted for downregulation across several metabolic pathways. ALDH activity and inhibitor assays indicated that reduced gene expression of ALDHs affected ALDH enzymatic functions and resulted in oxidative stress dysregulation in GDM-CMSCs. Conclusion Our combined transcriptional analysis and in vitro functional characterisation have provided novel insights into fundamental biological differences in GDM- and Healthy-CMSCs. Enhanced mobility of GDM-CMSCs may promote MSC migration toward injured sites; however, impaired cellular metabolic activity may negatively affect any perceived benefit.
Collapse
Affiliation(s)
- Liyun Chen
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Thornburrow Drive, Stoke-on-Trent, UK.,Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - Chung-Teng Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Nicholas R Forsyth
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Thornburrow Drive, Stoke-on-Trent, UK. .,School of Life Science, Guangzhou University, Guangzhou, 510006, China.
| | - Pensee Wu
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Thornburrow Drive, Stoke-on-Trent, UK.,Academic Unit of Obstetrics and Gynaecology, University Hospital of North Midlands, Stoke-on-Trent, UK.,Keele Cardiovascular Research Group, School of Primary, Community, and Social Care, Keele University, Stoke-on-Trent, UK
| |
Collapse
|
183
|
Sonoda S, Murata S, Nishida K, Kato H, Uehara N, Kyumoto YN, Yamaza H, Takahashi I, Kukita T, Yamaza T. Extracellular vesicles from deciduous pulp stem cells recover bone loss by regulating telomerase activity in an osteoporosis mouse model. Stem Cell Res Ther 2020; 11:296. [PMID: 32680564 PMCID: PMC7367365 DOI: 10.1186/s13287-020-01818-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/23/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Systemic transplantation of stem cells from human exfoliated deciduous teeth (SHED) recovers bone loss in animal models of osteoporosis; however, the mechanisms underlying this remain unclear. Here, we hypothesized that trophic factors within SHED-releasing extracellular vesicles (SHED-EVs) rescue osteoporotic phenotype. METHODS EVs were isolated from culture supernatant of SHED. SHED-EVs were treated with or without ribonuclease and systemically administrated into ovariectomized mice, followed by the function of recipient bone marrow mesenchymal stem cells (BMMSCs) including telomerase activity, osteoblast differentiation, and sepmaphorine-3A (SEMA3A) secretion. Subsequently, human BMMSCs were stimulated by SHED-EVs with or without ribonuclease treatment, and then human BMMSCs were examined regarding the function of telomerase activity, osteoblast differentiation, and SEMA3A secretion. Furthermore, SHED-EV-treated human BMMSCs were subcutaneously transplanted into the dorsal skin of immunocompromised mice with hydroxyapatite tricalcium phosphate (HA/TCP) careers and analyzed the de novo bone-forming ability. RESULTS We revealed that systemic SHED-EV-infusion recovered bone volume in ovariectomized mice and improved the function of recipient BMMSCs by rescuing the mRNA levels of Tert and telomerase activity, osteoblast differentiation, and SEMA3A secretion. Ribonuclease treatment depleted RNAs, including microRNAs, within SHED-EVs, and these RNA-depleted SHED-EVs attenuated SHED-EV-rescued function of recipient BMMSCs in the ovariectomized mice. These findings were supported by in vitro assays using human BMMSCs incubated with SHED-EVs. CONCLUSION Collectively, our findings suggest that SHED-secreted RNAs, such as microRNAs, play a crucial role in treating postmenopausal osteoporosis by targeting the telomerase activity of recipient BMMSCs.
Collapse
Affiliation(s)
- Soichiro Sonoda
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Sara Murata
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Section of Orthodontics and Dentofacial Orthopedics, Division of Oral Health, Growth & Development, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Kento Nishida
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroki Kato
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Norihisa Uehara
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yukari N Kyumoto
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Haruyoshi Yamaza
- Department of Pediatric Dentistry, Division of Oral Health, Growth & Development, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Ichiro Takahashi
- Section of Orthodontics and Dentofacial Orthopedics, Division of Oral Health, Growth & Development, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Toshio Kukita
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takayoshi Yamaza
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
184
|
Anselmi G, Helft J, Guermonprez P. Development and function of human dendritic cells in humanized mice models. Mol Immunol 2020; 125:151-161. [PMID: 32688117 DOI: 10.1016/j.molimm.2020.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 04/06/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) are sentinel cells of the immune system arising from hematopoietic stem cells. DCs play a key role in the regulation of both adaptive and innate lymphocyte responses. As such, experimental models enabling a thorough analysis of human DCs development and function are needed. Humanized mice models (termed collectively as HIS mice, or human immune system mice models) provide unique opportunities to model human hematopoiesis and tackle the function of human immune cell types in vivo. Here, we review experimental approaches enabling to recapitulate the ontogeny of DC subsets in HIS mice and discuss studies addressing the biology of human DC subsets implementing HIS mice models.
Collapse
Affiliation(s)
- Giorgio Anselmi
- King's College London, Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immunobiology, United Kingdom
| | - Julie Helft
- PSL Research University, Institut Curie Research Center, Immunity and Cancer department, INSERM U932, Paris, France
| | - Pierre Guermonprez
- King's College London, Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immunobiology, United Kingdom; Université de Paris, Centre for Inflammation Research, CNRS ERL8252, INSERM 1149, Hopital Bichat Claude Bernard, France.
| |
Collapse
|
185
|
Hu B, Lv X, Chen H, Xue P, Gao B, Wang X, Zhen G, Crane JL, Pan D, Liu S, Ni S, Wu P, Su W, Liu X, Ling Z, Yang M, Deng R, Li Y, Wang L, Zhang Y, Wan M, Shao Z, Chen H, Yuan W, Cao X. Sensory nerves regulate mesenchymal stromal cell lineage commitment by tuning sympathetic tones. J Clin Invest 2020; 130:3483-3498. [PMID: 32191640 PMCID: PMC7324175 DOI: 10.1172/jci131554] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 03/11/2020] [Indexed: 12/30/2022] Open
Abstract
The sensory nerve was recently identified as being involved in regulation of bone mass accrual. We previously discovered that prostaglandin E2 (PGE2) secreted by osteoblasts could activate sensory nerve EP4 receptor to promote bone formation by inhibiting sympathetic activity. However, the fundamental units of bone formation are active osteoblasts, which originate from mesenchymal stromal/stem cells (MSCs). Here, we found that after sensory denervation, knockout of the EP4 receptor in sensory nerves, or knockout of COX-2 in osteoblasts, could significantly promote adipogenesis and inhibit osteogenesis in adult mice. Furthermore, injection of SW033291 (a small molecule that locally increases the PGE2 level) or propranolol (a beta blocker) significantly promoted osteogenesis and inhibited adipogenesis. This effect of SW033291, but not propranolol, was abolished in conditional EP4-KO mice under normal conditions or in the bone repair process. We conclude that the PGE2/EP4 sensory nerve axis could regulate MSC differentiation in bone marrow of adult mice.
Collapse
Affiliation(s)
- Bo Hu
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
- Section of Spine Surgery, Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiao Lv
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Chen
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Peng Xue
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Bo Gao
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Xiao Wang
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Gehua Zhen
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Janet L. Crane
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Dayu Pan
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Shen Liu
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Shuangfei Ni
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Panfeng Wu
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Weiping Su
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Xiaonan Liu
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Zemin Ling
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mi Yang
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ruoxian Deng
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yusheng Li
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Lei Wang
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ying Zhang
- Section of Spine Surgery, Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Mei Wan
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huajiang Chen
- Section of Spine Surgery, Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Wen Yuan
- Section of Spine Surgery, Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xu Cao
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
186
|
Drela K, Stanaszek L, Snioch K, Kuczynska Z, Wrobel M, Sarzynska S, Legosz P, Maldyk P, Lukomska B. Bone marrow-derived from the human femoral shaft as a new source of mesenchymal stem/stromal cells: an alternative cell material for banking and clinical transplantation. Stem Cell Res Ther 2020; 11:262. [PMID: 32605638 PMCID: PMC7328271 DOI: 10.1186/s13287-020-01697-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/16/2020] [Accepted: 04/28/2020] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Mesenchymal stem/stromal cells (MSC) are commonly used in regenerative medicine. Among different tissues, iliac crest bone marrow (BM) represents the most exploited source, but its disadvantages are a painful aspiration procedure and low cell number. An alternative, readily available source of MSC for research would be beneficial for regenerative medicine development. This work aimed to propose a new source of bone marrow isolation in which the femoral shaft is taken during total hip arthroplasty (THA). METHODS In preliminary experiments, three different gradient methods for cell separation (Ficoll-Paque 1.078 g/mL, 17% sucrose gradient, BM seeding fraction) were tested with regard to the time of primary culture, initial cell number, the phenotype, and morphology of MSC. Then human bone marrow MSC derived from two different sources, iliac crest aspirate (BM-MSCi) or femoral shaft (BM-MSCt), were analyzed in terms of cell number and colony-forming ability followed by differentiation potential of MSC into osteo-, chondro-, and adipogenic lineages as well as mRNA expression of a variety of cytokines and growth factors. RESULTS Our studies showed that MSC isolated from the bone marrow of two different sources and cultured under appropriate conditions had similar characteristics and comparable propensity to differentiate into mesodermal cells. MSC derived from BM-MSCi or BM-MSCt expressed various growth factors. Interestingly, the expression of EGF, FGF, IGF, and PDGF-A was much higher in BM-MSCt than BM-MSCi. CONCLUSIONS The results of our study demonstrate that human MSC isolated from the BM of the femoral shaft have similar biological characteristics as MSC derived from the iliac crest, suggesting the femoral shaft as a possible alternative source for mesenchymal stem/stromal cells.
Collapse
Affiliation(s)
- Katarzyna Drela
- Mossakowski Medical Research Centre, Polish Academy of Sciences, 5, Pawinskiego, 02-106, Warsaw, Poland
| | - Luiza Stanaszek
- Mossakowski Medical Research Centre, Polish Academy of Sciences, 5, Pawinskiego, 02-106, Warsaw, Poland
| | - Konrad Snioch
- Mossakowski Medical Research Centre, Polish Academy of Sciences, 5, Pawinskiego, 02-106, Warsaw, Poland
| | - Zuzanna Kuczynska
- Mossakowski Medical Research Centre, Polish Academy of Sciences, 5, Pawinskiego, 02-106, Warsaw, Poland
| | - Mikolaj Wrobel
- Ortopedika, Centre for Specialized Surgery, Warsaw, Poland
| | - Sylwia Sarzynska
- Department of Orthopedics and Traumatology, 1st Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Pawel Legosz
- Department of Orthopedics and Traumatology, 1st Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Pawel Maldyk
- Department of Orthopedics and Traumatology, 1st Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Barbara Lukomska
- Mossakowski Medical Research Centre, Polish Academy of Sciences, 5, Pawinskiego, 02-106, Warsaw, Poland.
| |
Collapse
|
187
|
Understanding the regulatory mechanisms of endometrial cells on activities of endometrial mesenchymal stem-like cells during menstruation. Stem Cell Res Ther 2020; 11:239. [PMID: 32552749 PMCID: PMC7302161 DOI: 10.1186/s13287-020-01750-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/25/2020] [Accepted: 05/29/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The identification of endometrial stem/progenitor cells in a high turnover rate tissue suggests that a well-orchestrated underlying network controls the behaviour of these stem cells. The thickness of the endometrium can grow from 0.5-1 mm to 5-7 mm within a week indicating the need of stem cells for self-renewal and differentiation during this period. The cyclical regeneration of the endometrium suggests specific signals can activate the stem cells during or shortly after menstruation. METHODS Endometrial mesenchymal stem-like cells (eMSCs) were cocultured with endometrial epithelial or stromal cells from different phases of the menstrual cycle; the clonogenicity and the phenotypic expression of eMSC markers (CD140b and CD146) were assessed. The functional role of WNT/β-catenin signalling on eMSC was determined by western blot analysis, immunofluorescent staining, flow cytometry, quantitative real-time PCR and small interfering RNA. The cytokine levels in the conditioned medium of epithelial or stromal cells cocultured with eMSCs were evaluated by enzyme-linked immunosorbent assays. RESULTS Coculture of endometrial cells (epithelial or stromal) from the menstrual phase enhanced the clonogenicity and self-renewal activities of eMSCs. Such phenomenon was not observed in niche cells from the proliferative phase. Coculture with endometrial cells from the menstrual phase confirmed an increase in expression of active β-catenin in the eMSCs. Treatment with IWP-2, a WNT inhibitor, suppressed the observed effects. Anti-R-spondin-1 antibody reduced the stimulatory action of endometrial niche cells on WNT/β-catenin activation in the T cell factor/lymphoid enhancer-binding factor luciferase reporter assay. Moreover, the mRNA level and protein immunoreactivities of leucine-rich repeat-containing G-protein coupled receptor 5 were higher in eMSCs than unfractionated stromal cells. Conditioned media of endometrial niche cells cocultured with eMSCs contained increased levels of C-X-C motif ligand 1 (CXCL1), CXCL5 and interleukin 6. Treatment with these cytokines increased the clonogenic activity and phenotypic expression of eMSCs. CONCLUSIONS Our findings indicate a role of WNT/β-catenin signalling in regulating activities of endometrial stem/progenitor cells during menstruation. Certain cytokines at menstruation can stimulate the proliferation and self-renewal activities of eMSCs. Understanding the mechanism in the regulation of eMSCs may contribute to treatments of endometrial proliferative disorders such as Asherman's syndrome.
Collapse
|
188
|
Zarniko N, Skorska A, Steinhoff G, David R, Gaebel R. Dose-Independent Therapeutic Benefit of Bone Marrow Stem Cell Transplantation after MI in Mice. Biomedicines 2020; 8:biomedicines8060157. [PMID: 32545336 PMCID: PMC7345933 DOI: 10.3390/biomedicines8060157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023] Open
Abstract
Several cell populations derived from bone marrow (BM) have been shown to possess cardiac regenerative potential. Among these are freshly isolated CD133+ hematopoietic as well as culture-expanded mesenchymal stem cells. Alternatively, by purifying CD271+ cells from BM, mesenchymal progenitors can be enriched without an ex vivo cultivation. With regard to the limited available number of freshly isolated BM-derived stem cells, the effect of the dosage on the therapeutic efficiency is of particular interest. Therefore, in the present pre-clinical study, we investigated human BM-derived CD133+ and CD271+ stem cells for their cardiac regenerative potential three weeks post-myocardial infarction (MI) in a dose-dependent manner. The improvement of the hemodynamic function as well as cardiac remodeling showed no therapeutic difference after the transplantation of both 100,000 and 500,000 stem cells. Therefore, beneficial stem cell transplantation post-MI is widely independent of the cell dose and detrimental stem cell amplification in vitro can likely be avoided.
Collapse
Affiliation(s)
- Nicole Zarniko
- Department of Cardiac Surgery, Rostock University Medical Center, 18059 Rostock, Germany; (N.Z.); (A.S.); (G.S.); (R.G.)
| | - Anna Skorska
- Department of Cardiac Surgery, Rostock University Medical Center, 18059 Rostock, Germany; (N.Z.); (A.S.); (G.S.); (R.G.)
- Department Life, Light & Matter (LL&M), University of Rostock, A.-Einstein-Str. 25, 18057 Rostock, Germany
| | - Gustav Steinhoff
- Department of Cardiac Surgery, Rostock University Medical Center, 18059 Rostock, Germany; (N.Z.); (A.S.); (G.S.); (R.G.)
- Department Life, Light & Matter (LL&M), University of Rostock, A.-Einstein-Str. 25, 18057 Rostock, Germany
| | - Robert David
- Department of Cardiac Surgery, Rostock University Medical Center, 18059 Rostock, Germany; (N.Z.); (A.S.); (G.S.); (R.G.)
- Department Life, Light & Matter (LL&M), University of Rostock, A.-Einstein-Str. 25, 18057 Rostock, Germany
- Correspondence: ; Tel.: +49-381-4988973; Fax: +49-381-4988970
| | - Ralf Gaebel
- Department of Cardiac Surgery, Rostock University Medical Center, 18059 Rostock, Germany; (N.Z.); (A.S.); (G.S.); (R.G.)
- Department Life, Light & Matter (LL&M), University of Rostock, A.-Einstein-Str. 25, 18057 Rostock, Germany
| |
Collapse
|
189
|
Desterke C, Petit L, Sella N, Chevallier N, Cabeli V, Coquelin L, Durand C, Oostendorp RAJ, Isambert H, Jaffredo T, Charbord P. Inferring Gene Networks in Bone Marrow Hematopoietic Stem Cell-Supporting Stromal Niche Populations. iScience 2020; 23:101222. [PMID: 32535025 PMCID: PMC7300160 DOI: 10.1016/j.isci.2020.101222] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/19/2020] [Accepted: 05/27/2020] [Indexed: 02/07/2023] Open
Abstract
The cardinal property of bone marrow (BM) stromal cells is their capacity to contribute to hematopoietic stem cell (HSC) niches by providing mediators assisting HSC functions. In this study we first contrasted transcriptomes of stromal cells at different developmental stages and then included large number of HSC-supportive and non-supportive samples. Application of a combination of algorithms, comprising one identifying reliable paths and potential causative relationships in complex systems, revealed gene networks characteristic of the BM stromal HSC-supportive capacity and of defined niche populations of perivascular cells, osteoblasts, and mesenchymal stromal cells. Inclusion of single-cell transcriptomes enabled establishing for the perivascular cell subset a partially oriented graph of direct gene-to-gene interactions. As proof of concept we showed that R-spondin-2, expressed by the perivascular subset, synergized with Kit ligand to amplify ex vivo hematopoietic precursors. This study by identifying classifiers and hubs constitutes a resource to unravel candidate BM stromal mediators. A correlation network with predictor genes for the BM HSPC-supportive stromal niche An information theoretic network for the supportive perivascular stromal niche Wnt facilitator Rspo2 together with SCF to amplify ex vivo hematopoietic precursors Resource combining bioinformatics algorithms to search for novel stromal mediators
Collapse
Affiliation(s)
| | - Laurence Petit
- Sorbonne Université, UPMC Université Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement; Paris 75005, France
| | - Nadir Sella
- Institut Curie, PSL Research University, CNRS UMR168, Paris, France
| | - Nathalie Chevallier
- IMRB U955-E10, INSERM, Unité d'Ingenierie et de Thérapie Cellulaire- EFS, Université Paris-EST, Créteil, France
| | - Vincent Cabeli
- Institut Curie, PSL Research University, CNRS UMR168, Paris, France
| | - Laura Coquelin
- IMRB U955-E10, INSERM, Unité d'Ingenierie et de Thérapie Cellulaire- EFS, Université Paris-EST, Créteil, France
| | - Charles Durand
- Sorbonne Université, UPMC Université Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement; Paris 75005, France
| | - Robert A J Oostendorp
- Clinic and Polyclinic for Internal Medicine III, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany
| | - Hervé Isambert
- Institut Curie, PSL Research University, CNRS UMR168, Paris, France
| | - Thierry Jaffredo
- Sorbonne Université, UPMC Université Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement; Paris 75005, France
| | - Pierre Charbord
- Sorbonne Université, UPMC Université Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement; Paris 75005, France.
| |
Collapse
|
190
|
Budgude P, Kale V, Vaidya A. Mesenchymal stromal cell‐derived extracellular vesicles as cell‐free biologics for the ex vivo expansion of hematopoietic stem cells. Cell Biol Int 2020; 44:1078-1102. [DOI: 10.1002/cbin.11313] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 01/31/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Pallavi Budgude
- Symbiosis Centre for Stem Cell ResearchSymbiosis International (Deemed University) Pune 412115 India
| | - Vaijayanti Kale
- Symbiosis Centre for Stem Cell ResearchSymbiosis International (Deemed University) Pune 412115 India
| | - Anuradha Vaidya
- Symbiosis Centre for Stem Cell ResearchSymbiosis International (Deemed University) Pune 412115 India
- Symbiosis School of Biological SciencesSymbiosis International (Deemed University) Pune 412115 India
| |
Collapse
|
191
|
A Small-Sized Population of Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells Shows High Stemness Properties and Therapeutic Benefit. Stem Cells Int 2020; 2020:5924983. [PMID: 32399043 PMCID: PMC7204153 DOI: 10.1155/2020/5924983] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 03/11/2020] [Accepted: 03/24/2020] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem cells (MSCs) represent a promising means to promote tissue regeneration. However, the heterogeneity of MSCs impedes their use for regenerative medicine. Further investigation of this phenotype is required to develop cell therapies with improved clinical efficacy. Here, a small-sized population of human umbilical cord blood-derived MSCs (UCB-MSCs) was isolated using a filter and centrifuge system to analyze its stem cell characteristics. Consequently, this population showed higher cell growth and lower senescence. Additionally, it exhibited diverse stem cell properties including differentiation, stemness, and adhesion, as compared to those of the population before isolation. Using cell surface protein array or sorting analysis, both EGFR and CD49f were identified as markers associated with the small-sized population. Accordingly, suppression of these surface proteins abolished the superior characteristics of this population. Moreover, compared to that with large or nonisolated populations, the small-sized population showed greater therapeutic efficacy by promoting the engraftment potential of infused cells and reducing lung damage in an emphysema mouse model. Therefore, the isolation of this small-sized population of UCB-MSCs could be a simple and effective way to enhance the efficacy of cell therapy.
Collapse
|
192
|
Gladkova N, Umezu T, Imanishi S, Kawana C, Ohyashiki JH, Ohyashiki K. Effect of the extracellular component of bone marrow mesenchymal stromal cells from healthy donors on hematologic neoplasms and their angiogenesis. Hum Cell 2020; 33:599-609. [PMID: 32281045 PMCID: PMC7324432 DOI: 10.1007/s13577-020-00332-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/11/2020] [Indexed: 11/05/2022]
Abstract
Bone marrow mesenchymal stromal cells (BM-MSCs) from healthy donors are a promising source of cell therapy. However, their effectiveness in cancer remains less known. This study is the first to evaluate the quality of BM-MSCs obtained from young and elderly healthy volunteers (KNT cells). The KNT cells had normal karyotypes and were positive for MSC markers (CD90, CD73, CD105). When cultured under appropriate conditions, they showed adipogenic or osteogenic potential. Hence, the anti-neoplastic effects of secretory factors [supernatant or extracellular vesicles (EV)] from KNT cells were verified using several neoplastic cells (three multiple myeloma, three myeloid leukemia, and three lymphoma cell lines). The conditioned medium (CM), but not EV, of KNT cells derived from young healthy donors significantly inhibited myeloma and lymphoma cell proliferation, but enhanced myeloid leukemia proliferation. Anti-angiogenesis effect of CM and EV derived from young KNT against hematologic neoplasia-induced angiogenesis was evident and more prominent in CM than in EV but not evident in elderly KNT-derived EV. These findings indicate that the anti-tumor effect of KNT cells depends on the types of hematologic neoplasia, with elements existing in the supernatant and not in EVs. Therefore, BM-MSC may produce soluble factors that affect cell proliferation of neoplasia, causing cell-to-cell communication. The anti-angiogenesis effect of KNT cells depends on the age of BM-MSC donors.
Collapse
Affiliation(s)
- Nina Gladkova
- Kintaro Cells Power Corporation, Tokyo, Japan.,Department of Advanced Cellular Therapy, Tokyo Medical University, Tokyo, Japan
| | - Tomohiro Umezu
- Department of Hematology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku, Tokyo, 160-0023, Japan.,Institute of Medical Science, Tokyo Medical University, Tokyo, Japan.,Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| | - Satoshi Imanishi
- Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Chiaki Kawana
- Department of Hematology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku, Tokyo, 160-0023, Japan
| | - Junko H Ohyashiki
- Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Kazuma Ohyashiki
- Department of Hematology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku, Tokyo, 160-0023, Japan.
| |
Collapse
|
193
|
Zhao M, Tao F, Venkatraman A, Li Z, Smith SE, Unruh J, Chen S, Ward C, Qian P, Perry JM, Marshall H, Wang J, He XC, Li L. N-Cadherin-Expressing Bone and Marrow Stromal Progenitor Cells Maintain Reserve Hematopoietic Stem Cells. Cell Rep 2020; 26:652-669.e6. [PMID: 30650358 DOI: 10.1016/j.celrep.2018.12.093] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 10/31/2018] [Accepted: 12/20/2018] [Indexed: 12/15/2022] Open
Abstract
Regulation of hematopoietic stem cells (HSCs) by bone marrow (BM) niches has been extensively studied; however, whether and how HSC subpopulations are distinctively regulated by BM niches remain unclear. Here, we functionally distinguished reserve HSCs (rHSCs) from primed HSCs (pHSCs) based on their response to chemotherapy and examined how they are dichotomously regulated by BM niches. Both pHSCs and rHSCs supported long-term hematopoiesis in homeostasis; however, pHSCs were sensitive but rHSCs were resistant to chemotherapy. Surviving rHSCs restored the HSC pool and supported hematopoietic regeneration after chemotherapy. The rHSCs were preferentially maintained in the endosteal region that enriches N-cadherin+ (N-cad+) bone-lining cells in homeostasis and post-chemotherapy. N-cad+ cells were functional bone and marrow stromal progenitor cells (BMSPCs), giving rise to osteoblasts, adipocytes, and chondrocytes in vitro and in vivo. Finally, ablation of N-cad+ niche cells or deletion of SCF from N-cad+ niche cells impaired rHSC maintenance during homeostasis and regeneration.
Collapse
Affiliation(s)
- Meng Zhao
- Institute of Hematology, the Third Affiliated Hospital of Sun Yat-Sen University; Key Laboratory of Stem Cells and Tissue Engineering Sun Yat-Sen University, Guangzhou 510000, China; Stowers Institute for Medical Research, Kansas City, MO 66110, USA
| | - Fang Tao
- Stowers Institute for Medical Research, Kansas City, MO 66110, USA; Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | | - Zhenrui Li
- Stowers Institute for Medical Research, Kansas City, MO 66110, USA
| | - Sarah E Smith
- Stowers Institute for Medical Research, Kansas City, MO 66110, USA
| | - Jay Unruh
- Stowers Institute for Medical Research, Kansas City, MO 66110, USA
| | - Shiyuan Chen
- Stowers Institute for Medical Research, Kansas City, MO 66110, USA
| | - Christina Ward
- Stowers Institute for Medical Research, Kansas City, MO 66110, USA
| | - Pengxu Qian
- Stowers Institute for Medical Research, Kansas City, MO 66110, USA; Center of Stem Cell and Regenerative Medicine, Institute of Hematology, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China 310058
| | - John M Perry
- Stowers Institute for Medical Research, Kansas City, MO 66110, USA; Children's Research Institute, Children's Mercy, Kansas City, MO 64108, USA
| | - Heather Marshall
- Stowers Institute for Medical Research, Kansas City, MO 66110, USA
| | - Jinxi Wang
- Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Xi C He
- Stowers Institute for Medical Research, Kansas City, MO 66110, USA
| | - Linheng Li
- Stowers Institute for Medical Research, Kansas City, MO 66110, USA; Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
194
|
Sinha S, Biswas M, Chatterjee SS, Kumar S, Sengupta A. Pbrm1 Steers Mesenchymal Stromal Cell Osteolineage Differentiation by Integrating PBAF-Dependent Chromatin Remodeling and BMP/TGF-β Signaling. Cell Rep 2020; 31:107570. [DOI: 10.1016/j.celrep.2020.107570] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 02/17/2020] [Accepted: 04/02/2020] [Indexed: 12/31/2022] Open
|
195
|
Sun JL, Yan JF, Li J, Wang WR, Yu SB, Zhang HY, Huang F, Niu LN, Jiao K. Conditional deletion of Adrb2 in mesenchymal stem cells attenuates osteoarthritis-like defects in temporomandibular joint. Bone 2020; 133:115229. [PMID: 31926929 DOI: 10.1016/j.bone.2020.115229] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/31/2019] [Accepted: 01/08/2020] [Indexed: 01/07/2023]
Abstract
β2-adrenergic signal transduction in mesenchymal stem cells (MSCs) induces subchondral bone loss in osteoarthritis (OA) of temporomandibular joints (TMJs). However, whether conditional deletion of β2-adrenergic receptor (Adrb2) in nestin+ MSCs can alleviate TMJ-OA development remains unknown. In this study, nestin-Cre mice were crossed with Adrb2 flox mice to generate mice lacking Adrb2 expression specifically in the nestin+ MSCs (Adrb2-/-), and TMJ-OA development in such mice was investigated. Adrb2 flox mice (Adrb2+/+) and Adrb2-/- mice were subjected to unilateral anterior crossbite (UAC), while mice in the control group were subjected to sham operation. Adrb2+/+ and Adrb2-/- mice in the control group showed no distinguishable phenotypic changes in body weight and length, mandibular condylar size, and other histomorphological parameters of the condylar subchondral bone. A significant increase in subchondral bone loss and cartilage degradation was observed in Adrb2+/+ UAC mice; the former was characterized by decreased bone mineral density, bone volume fraction, and trabecular plate thickness, and increased trabecular separation, osteoclast number and osteoclast surface, and pro-osteoclastic factor expression; the latter was characterized by decreased cartilage thickness, chondrocyte density, proteoglycan area, and collagen II and aggrecan expression, but increased matrix metalloproteinase and alkaline phosphatase expression and percentage area of calcified cartilage. Adrb2 deletion in nestin+ MSCs largely attenuated UAC-induced increase in condylar subchondral bone loss, cartilage degradation, and aberrant calcification at the osteochondral interface. Thus, Adrb2-expressing MSCs in the condylar subchondral bone play an important role in TMJ-OA progression and may serve as novel therapeutic targets for TMJ-OA.
Collapse
Affiliation(s)
- Jin-Long Sun
- State Key Laboratory of Military Stomatology, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China.; Department of Stomatology, Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Jian-Fei Yan
- State Key Laboratory of Military Stomatology, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Jing Li
- State Key Laboratory of Military Stomatology, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Wan-Rong Wang
- State Key Laboratory of Military Stomatology, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Shi-Bin Yu
- State Key Laboratory of Military Stomatology, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Hong-Yun Zhang
- State Key Laboratory of Military Stomatology, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Fei Huang
- Department of Stomatology, Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Li-Na Niu
- State Key Laboratory of Military Stomatology, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China..
| | - Kai Jiao
- State Key Laboratory of Military Stomatology, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China..
| |
Collapse
|
196
|
Xu J, Lian W, Chen J, Li W, Li L, Huang Z. Chemical-defined medium supporting the expansion of human mesenchymal stem cells. Stem Cell Res Ther 2020; 11:125. [PMID: 32192530 PMCID: PMC7083066 DOI: 10.1186/s13287-020-01641-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/03/2020] [Accepted: 03/10/2020] [Indexed: 12/04/2022] Open
Abstract
Objectives Mesenchymal stem cells (MSCs) have been intensively investigated as to their therapeutic potentials. However, the full chemical-defined medium supporting the isolation and expansion of human MSCs has not been developed yet. Materials and methods Here, we developed the full chemical-defined medium, NBVbe medium, via RNA sequencing, bioinformatic analysis, and growth factor screening. Results The NBVbe medium contains N2B27 medium with the BSA (bovine serum albumin) replaced by the recombinant human albumin, bFGF (basic fibroblast growth factor), vitamin C, and EGF (epidermal growth factor). The NBVbe medium could support the isolation and expansion of human MSCs from the umbilical cords. Conclusions The full chemical-defined medium supporting the isolation and expansion of human MSCs has been developed. This would be helpful for further optimization of the MSC medium, their clinical applications, and molecular characterization.
Collapse
Affiliation(s)
- Jianyong Xu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, School of Medicine, Shenzhen University, Nanhai Avenue 3688, Shenzhen, 518060, Guangdong, People's Republic of China.
| | - Wei Lian
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, School of Medicine, Shenzhen University, Nanhai Avenue 3688, Shenzhen, 518060, Guangdong, People's Republic of China
| | - Jieting Chen
- Department of Obstetrics, People's Hospital of Baoan, Shenzhen, 518055, People's Republic of China
| | - Wenlei Li
- Department of Obstetrics, Women and Children Health Institute of Futian, Shenzhen, 518055, People's Republic of China
| | - Lingyun Li
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, School of Medicine, Shenzhen University, Nanhai Avenue 3688, Shenzhen, 518060, Guangdong, People's Republic of China
| | - Zhong Huang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, School of Medicine, Shenzhen University, Nanhai Avenue 3688, Shenzhen, 518060, Guangdong, People's Republic of China
| |
Collapse
|
197
|
Munir S, Basu A, Maity P, Krug L, Haas P, Jiang D, Strauss G, Wlaschek M, Geiger H, Singh K, Scharffetter-Kochanek K. TLR4-dependent shaping of the wound site by MSCs accelerates wound healing. EMBO Rep 2020; 21:e48777. [PMID: 32162777 PMCID: PMC7202058 DOI: 10.15252/embr.201948777] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 02/14/2020] [Accepted: 02/18/2020] [Indexed: 12/13/2022] Open
Abstract
We here address the question whether the unique capacity of mesenchymal stem cells to re‐establish tissue homeostasis depends on their potential to sense pathogen‐associated molecular pattern and, in consequence, mount an adaptive response in the interest of tissue repair. After injection of MSCs primed with the bacterial wall component LPS into murine wounds, an unexpected acceleration of healing occurs, clearly exceeding that of non‐primed MSCs. This correlates with a fundamental reprogramming of the transcriptome in LPS‐treated MSCs as deduced from RNAseq analysis and its validation. A network of genes mediating the adaptive response through the Toll‐like receptor 4 (TLR4) pathway responsible for neutrophil and macrophage recruitment and their activation profoundly contributes to enhanced wound healing. In fact, injection of LPS‐primed MSCs silenced for TLR4 fails to accelerate wound healing. These unprecedented findings hold substantial promise to refine current MSC‐based therapies for difficult‐to‐treat wounds.
Collapse
Affiliation(s)
- Saira Munir
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany
| | - Abhijit Basu
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany
| | - Pallab Maity
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany.,Aging Research Center (ARC), Ulm, Germany
| | - Linda Krug
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany.,Aging Research Center (ARC), Ulm, Germany
| | - Philipp Haas
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany
| | - Dongsheng Jiang
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany
| | - Gudrun Strauss
- Department of Pediatrics and Adolescent Medicine, Ulm University, Ulm, Germany
| | - Meinhard Wlaschek
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany
| | - Hartmut Geiger
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany.,Aging Research Center (ARC), Ulm, Germany.,Institute of Molecular Medicine and Stem Cell Aging, Ulm University, Ulm, Germany.,Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Karmveer Singh
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany.,Aging Research Center (ARC), Ulm, Germany
| | - Karin Scharffetter-Kochanek
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany.,Aging Research Center (ARC), Ulm, Germany
| |
Collapse
|
198
|
Zhang J, Liu Y, Yin W, Hu X. Adipose-derived stromal cells in regulation of hematopoiesis. Cell Mol Biol Lett 2020; 25:16. [PMID: 32161623 PMCID: PMC7059705 DOI: 10.1186/s11658-020-00209-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/25/2020] [Indexed: 12/21/2022] Open
Abstract
Over the past decade, mesenchymal stromal cells (MSCs) found in the bone marrow microenvironment have been considered to be important candidates in cellular therapy. However, the application of MSCs in clinical settings is limited by the difficulty and low efficiency associated with the separation of MSCs from the bone marrow. Therefore, distinct sources of MSCs have been extensively explored. Adipose-derived stromal cells (ASCs), a cell line similar to MSCs, have been identified as a promising source. ASCs have become increasingly popular in many fields, as they can be conveniently extracted from fat tissue. This review focuses on the properties of ASCs in hematopoietic regulation and the underlying mechanisms, as well as the current applications and future perspectives in ASC-based therapy.
Collapse
Affiliation(s)
- Jing Zhang
- 1Department of Transfusion Medicine, Xijing Hospital, Xi'an, 710032 China
| | - Yunsheng Liu
- 2Department of Rocket Force Medicine, Third Military Medical University, Chongqing, 400038 China
| | - Wen Yin
- 1Department of Transfusion Medicine, Xijing Hospital, Xi'an, 710032 China
| | - Xingbin Hu
- 1Department of Transfusion Medicine, Xijing Hospital, Xi'an, 710032 China
| |
Collapse
|
199
|
Derecka M, Herman JS, Cauchy P, Ramamoorthy S, Lupar E, Grün D, Grosschedl R. EBF1-deficient bone marrow stroma elicits persistent changes in HSC potential. Nat Immunol 2020; 21:261-273. [PMID: 32066955 DOI: 10.1038/s41590-020-0595-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 01/09/2020] [Indexed: 12/11/2022]
Abstract
Crosstalk between mesenchymal stromal cells (MSCs) and hematopoietic stem cells (HSCs) is essential for hematopoietic homeostasis and lineage output. Here, we investigate how transcriptional changes in bone marrow (BM) MSCs result in long-lasting effects on HSCs. Single-cell analysis of Cxcl12-abundant reticular (CAR) cells and PDGFRα+Sca1+ (PαS) cells revealed an extensive cellular heterogeneity but uniform expression of the transcription factor gene Ebf1. Conditional deletion of Ebf1 in these MSCs altered their cellular composition, chromatin structure and gene expression profiles, including the reduced expression of adhesion-related genes. Functionally, the stromal-specific Ebf1 inactivation results in impaired adhesion of HSCs, leading to reduced quiescence and diminished myeloid output. Most notably, HSCs residing in the Ebf1-deficient niche underwent changes in their cellular composition and chromatin structure that persist in serial transplantations. Thus, genetic alterations in the BM niche lead to long-term functional changes of HSCs.
Collapse
Affiliation(s)
- Marta Derecka
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.
| | - Josip Stefan Herman
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany.,International Max Planck Research School for Molecular and Cellular Biology, Freiburg, Germany
| | - Pierre Cauchy
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | | | - Ekaterina Lupar
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Dominic Grün
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.,Centre for Integrative Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Rudolf Grosschedl
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.
| |
Collapse
|
200
|
Huo J, Zhang L, Ren X, Li C, Li X, Dong P, Zheng X, Huang J, Shao Y, Ge M, Zhang J, Wang M, Nie N, Jin P, Zheng Y. Multifaceted characterization of the signatures and efficacy of mesenchymal stem/stromal cells in acquired aplastic anemia. Stem Cell Res Ther 2020; 11:59. [PMID: 32054519 PMCID: PMC7020384 DOI: 10.1186/s13287-020-1577-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 11/17/2019] [Accepted: 02/03/2020] [Indexed: 01/07/2023] Open
Abstract
Background Longitudinal studies have verified the pivotal role of mesenchymal stem/stromal cells (MSCs) in the bone marrow microenvironment for hematopoiesis and coordinate contribution to leukemia pathogenesis. However, the precise characteristics and alternation of MSCs during acquired aplastic anemia (AA) remain obscure. Methods In this study, we originally collected samples from both healthy donors (HD) and AA patients to dissect the hematological changes. To systematically evaluate the biological defects of AA-derived MSCs (AA-MSCs), we analyzed alterations in cellular morphology, immunophenotype, multi-lineage differentiation, cell migration, cellular apoptosis, and chromosome karyocyte, together with the immunosuppressive effect on the activation and differentiation of lymphocytes. With the aid of whole genome sequencing and bioinformatic analysis, we try to compare the differences between AA-MSCs and HD-derived MSCs (HD-MSCs) upon the molecular genetics, especially the immune-associated gene expression pattern. In addition, the efficacy of umbilical cord-derived MSC (UC-MSC) transplantation on AA mice was evaluated by utilizing survivorship curve, histologic sections, and blood cell analyses. Results In coincidence with the current reports, AA patients showed abnormal subsets of lymphocytes and higher contents of proinflammatory cytokines. Although with similar immunophenotype and chromosome karyotype to HD-MSCs, AA-MSCs showed distinguishable morphology and multiple distinct characteristics including genetic properties. In addition, the immunosuppressive effect on lymphocytes was significantly impaired in AA-MSCs. What is more, the cardinal symptoms of AA mice were largely rescued by systemic transplantation of UC-MSCs. Conclusions Herein, we systematically investigated the signatures and efficacy of MSCs to dissect the alterations occurred in AA both at the cellular and molecular levels. Different from HD-MSCs, AA-MSCs exhibited multifaceted defects in biological characteristics and alterative molecular genetics in the whole genome. Our findings have provided systematic and overwhelming new evidence for the defects of AA-MSCs, together with effectiveness assessments of UC-MSCs on AA as well.
Collapse
Affiliation(s)
- Jiali Huo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Leisheng Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China. .,The Postdoctoral Research Station, School of Medicine, Nankai University, Tianjin, 300071, China.
| | - Xiang Ren
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Chengwen Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Xingxin Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Peiyuan Dong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Xuan Zheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Jinbo Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Yingqi Shao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Meili Ge
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Jing Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Min Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Neng Nie
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Peng Jin
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Yizhou Zheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China.
| |
Collapse
|