1
|
Yao J, Sun L, Gao F, Zhu W. Mesenchymal stem/stromal cells: dedicator to maintain tumor homeostasis. Hum Cell 2024; 38:21. [PMID: 39607530 DOI: 10.1007/s13577-024-01154-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Mesenchymal stem/stromal cells (MSCs) act as a factor in tumor recurrence after drug treatment with their involvement observed in various cancer types. As a constituent of the tumor microenvironment (TME), MSCs not only provide support to tumor growth but also establish connections with diverse cell populations within the TME, serving as mediators linking different tumor-associated components. MSCs play an important role in maintaining tumor progression due to their stem cell properties and remarkable differentiation capacity. Given the intensification of tumor research and the encouraging results achieved in recent years,the aim of this article is to investigate the supportive role of MSCs in tumor cells as well as in various cellular and non-cellular components of the tumor microenvironment. Furthermore, the article shows that MSCs do not have a specific anatomical ecological niche and describes the contribution of MSCs to the maintenance of tumor homeostasis on the basis of homing, plasticity and tumor-forming properties. By elucidating the critical roles of different components of TME, this study provides a comprehensive understanding of tumor therapy and may offer new insights into defeating cancer.
Collapse
Affiliation(s)
- Juncun Yao
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Li Sun
- Department of Clinical Laboratory, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu Province, People's Republic of China
| | - Feng Gao
- Department of Surgery, Jingjiang People's Hospital, Jingjiang, 214500, People's Republic of China.
| | - Wei Zhu
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China.
| |
Collapse
|
2
|
Rui X, Calderon FA, Wobma H, Gerdemann U, Albanese A, Cagnin L, McGuckin C, Michaelis KA, Naqvi K, Blazar BR, Tkachev V, Kean LS. Human OX40L-CAR-T regs target activated antigen-presenting cells and control T cell alloreactivity. Sci Transl Med 2024; 16:eadj9331. [PMID: 39413160 PMCID: PMC11789419 DOI: 10.1126/scitranslmed.adj9331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 07/09/2024] [Accepted: 09/23/2024] [Indexed: 10/18/2024]
Abstract
Regulatory T cells (Tregs) make major contributions to immune homeostasis. Because Treg dysfunction can lead to both allo- and autoimmunity, there is interest in correcting these disorders through Treg adoptive transfer. Two of the central challenges in clinically deploying Treg cellular therapies are ensuring phenotypic stability and maximizing potency. Here, we describe an approach to address both issues through the creation of OX40 ligand (OX40L)-specific chimeric antigen receptor (CAR)-Tregs under the control of a synthetic forkhead box P3 (FOXP3) promoter. The creation of these CAR-Tregs enabled selective Treg stimulation by engagement of OX40L, a key activation antigen in alloimmunity, including both graft-versus-host disease and solid organ transplant rejection, and autoimmunity, including rheumatoid arthritis, systemic sclerosis, and systemic lupus erythematosus. We demonstrated that OX40L-CAR-Tregs were robustly activated in the presence of OX40L-expressing cells, leading to up-regulation of Treg suppressive proteins without induction of proinflammatory cytokine production. Compared with control Tregs, OX40L-CAR-Tregs more potently suppressed alloreactive T cell proliferation in vitro and were directly inhibitory toward activated monocyte-derived dendritic cells (DCs). We identified trogocytosis as one of the central mechanisms by which these CAR-Tregs effectively decrease extracellular display of OX40L, resulting in decreased DC stimulatory capacity. OX40L-CAR-Tregs demonstrated an enhanced ability to control xenogeneic graft-versus-host disease compared with control Tregs without abolishing the graft-versus-leukemia effect. These results suggest that OX40L-CAR-Tregs may have wide applicability as a potent cellular therapy to control both allo- and autoimmune diseases.
Collapse
Affiliation(s)
- Xianliang Rui
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Francesca Alvarez Calderon
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Holly Wobma
- Harvard Medical School, Boston, MA 02115, USA
- Division of Immunology, Boston Children’s Hospital, Boston, MA 02215, USA
| | - Ulrike Gerdemann
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Alexandre Albanese
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Lorenzo Cagnin
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Connor McGuckin
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
| | | | - Kisa Naqvi
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Bruce R. Blazar
- Division of Pediatric Blood and Marrow Transplant and Cellular Therapy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Victor Tkachev
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Leslie S. Kean
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
3
|
Larey AM, Spoerer TM, Daga KR, Morfin MG, Hynds HM, Carpenter J, Hines KM, Marklein RA. High throughput screening of mesenchymal stromal cell morphological response to inflammatory signals for bioreactor-based manufacturing of extracellular vesicles that modulate microglia. Bioact Mater 2024; 37:153-171. [PMID: 38549769 PMCID: PMC10972802 DOI: 10.1016/j.bioactmat.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/14/2024] [Accepted: 03/07/2024] [Indexed: 04/09/2024] Open
Abstract
Due to their immunomodulatory function, mesenchymal stromal cells (MSCs) are a promising therapeutic with the potential to treat neuroinflammation associated with neurodegenerative diseases. This function is mediated by secreted extracellular vesicles (MSC-EVs). Despite established safety, MSC clinical translation has been unsuccessful due to inconsistent clinical outcomes resulting from functional heterogeneity. Current approaches to mitigate functional heterogeneity include 'priming' MSCs with inflammatory signals to enhance function. However, comprehensive evaluation of priming and its effects on MSC-EV function has not been performed. Furthermore, clinical translation of MSC-EV therapies requires significant manufacturing scale-up, yet few studies have investigated the effects of priming in bioreactors. As MSC morphology has been shown to predict their immunomodulatory function, we screened MSC morphological response to an array of priming signals and evaluated MSC-EV identity and potency in response to priming in flasks and bioreactors. We identified unique priming conditions corresponding to distinct morphologies. These conditions demonstrated a range of MSC-EV preparation quality and lipidome, allowing us to discover a novel MSC-EV manufacturing condition, as well as gain insight into potential mechanisms of MSC-EV microglia modulation. Our novel screening approach and application of priming to MSC-EV bioreactor manufacturing informs refinement of larger-scale manufacturing and enhancement of MSC-EV function.
Collapse
Affiliation(s)
- Andrew M. Larey
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Thomas M. Spoerer
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Kanupriya R. Daga
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Maria G. Morfin
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Hannah M. Hynds
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Jana Carpenter
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Kelly M. Hines
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Ross A. Marklein
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| |
Collapse
|
4
|
Kuang YH, Zhu W, Lin G, Cheng LM, Qin Q, Huang ZJ, Shi YL, Zhang CL, Xu JH, Yan KX, Lv CZ, Li W, Han Q, Stambler I, Lim LW, Chakrabarti S, Ulfhake B, Min KJ, Ellison-Hughes G, Cho WC, Jin K, Yao D, Lu C, Zhao RC, Chen X. Expert Consensus on the Application of Stem Cells in Psoriasis Research and Clinical Trials. Aging Dis 2024:AD.2024.0012. [PMID: 39012666 DOI: 10.14336/ad.2024.0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/04/2024] [Indexed: 07/17/2024] Open
Abstract
Psoriasis is an immune-mediated, chronic, relapsing, inflammatory, systemic disease induced by individual-environmental interactions, and is often lifelong because of the difficulty of treatment. In recent years, a variety of targeted therapies, including biologics, have improved the lesions and quality of life of most psoriasis patients, but they still do not address the problem of relapse and may be associated with decreased efficacy or adverse events such as infections over time. Therefore, there is an urgent need for breakthroughs in psoriasis treatment and in relapse-delaying and non-pharmacologic strategies, and stem cell therapy for psoriasis has emerged. In recent years, research on stem cell therapy for psoriasis has received a lot of attention, however, there is no reference standard as well as consensus in this field of research. Therefore, according to the latest consensus and guidelines, combined with relevant literature reports, clinical practice experience and the results of discussions with experts, this consensus specifies the types of stem cells commonly used in the treatment of psoriasis, the methods, dosages, and routes of stem cell therapy for psoriasis, as well as the clinical evaluations (efficacy and safety) of stem cell therapy for psoriasis. In addition, this consensus also provides normative standards for the processes of collection, preparation, preservation and quality control of stem cells and their related products, as well as recommendations for the management of stem cells during infusion for the treatment of psoriasis. This consensus provides the latest specific reference standards and practice guidelines for the field of stem cell therapy for psoriasis.
Collapse
Affiliation(s)
- Ye-Hong Kuang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
| | - Wu Zhu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- National Engineering Research Center of Human Stem Cell, Changsha, China
| | - La-Mei Cheng
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- National Engineering Research Center of Human Stem Cell, Changsha, China
| | - Qun Qin
- The Office of Drug Clinical Trials Institution, Xiangya Hospital, Central South University, Changsha, China
| | - Zhi-Jun Huang
- Center for Clinical Pharmacology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yu-Ling Shi
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
- Department of Dermatology, Shanghai Skin Disease Hospital, Institute of Psoriasis, School of Medicine, Tongji University, Shanghai, China
| | - Chun-Lei Zhang
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| | - Jin-Hua Xu
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ke-Xiang Yan
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Cheng-Zhi Lv
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
- Department of Dermatology, Dalian Dermatosis Hospital, Dalian, China
| | - Wei Li
- Department of Dermatology, Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
| | - Qin Han
- International Society on Aging and Disease, Fort Worth, TX, USA
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
- Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing, China
| | - Ilia Stambler
- International Society on Aging and Disease, Fort Worth, TX, USA
- Department of Science, Technology and Society, Bar Ilan University, Ramat Gan, Israel
| | - Lee Wei Lim
- International Society on Aging and Disease, Fort Worth, TX, USA
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Sasanka Chakrabarti
- International Society on Aging and Disease, Fort Worth, TX, USA
- Maharishi Markandeshwar Deemed University, Mullana-Ambala, India
| | - Brun Ulfhake
- International Society on Aging and Disease, Fort Worth, TX, USA
- Karolinska University Hospital, Stockholm, Sweden
| | - Kyung-Jin Min
- International Society on Aging and Disease, Fort Worth, TX, USA
- Department of Biological Sciences, Inha University, Incheon, Republic of Korea
| | - Georgina Ellison-Hughes
- International Society on Aging and Disease, Fort Worth, TX, USA
- School of Basic and Medical Biosciences, Faculty of Life Sciences &;amp Medicine, King's College London, London, UK
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Kunlin Jin
- International Society on Aging and Disease, Fort Worth, TX, USA
- University of North Texas Health Science Center, Bryan, TX, USA
| | - Danni Yao
- Department of Dermatology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Chuanjian Lu
- Department of Dermatology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Robert Chunhua Zhao
- International Society on Aging and Disease, Fort Worth, TX, USA
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
- Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
| |
Collapse
|
5
|
Herger N, Heggli I, Mengis T, Devan J, Arpesella L, Brunner F, Distler O, Dudli S. Impacts of priming on distinct immunosuppressive mechanisms of mesenchymal stromal cells under translationally relevant conditions. Stem Cell Res Ther 2024; 15:65. [PMID: 38443999 PMCID: PMC10916130 DOI: 10.1186/s13287-024-03677-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 02/21/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND The multimodal properties of mesenchymal stromal cells (MSCs), particularly their ability to modulate immune responses is of high interest in translational research. Pro-inflammatory, hypoxic, and 3D culture priming are promising and often used strategies to improve the immunosuppressive potency of MSCs, but the underlying mechanisms are not well understood. Therefore, the aims of this study were (i) to compare the effects of pro-inflammatory, hypoxic, and 3D culture priming on the in vitro immunosuppressive potential of MSCs, (ii) to assess if immunosuppressive priming effects are temporally preserved under standard and translationally relevant culture conditions, and (iii) to investigate if the three priming strategies engage the same immunosuppressive mechanisms. METHODS Functional in vitro T cell suppressive potency measurements were conducted to assess the impact of pro-inflammatory, hypoxic, and 3D culture priming on the immunosuppressive potential of human bone marrow-derived MSCs. Primed MSCs were either cultured under standard cell culture conditions or translationally relevant culture conditions, and their transcriptomic adaptations were monitored over time. Next-generation sequencing was performed to assess if different priming strategies activate distinct immunosuppressive mechanisms. RESULTS (i) Pro-inflammatory, hypoxic, and 3D culture priming induced profound transcriptomic changes in MSCs resulting in a significantly enhanced T cell suppressive potential of pro-inflammatory and 3D culture primed MSCs. (ii) Priming effects rapidly faded under standard cell culture conditions but were partially preserved under translationally relevant conditions. Interestingly, continuous 3D culture priming of MSCs maintained the immunosuppressive potency of MSCs. (iii) Next-generation sequencing revealed that priming strategy-specific differentially expressed genes are involved in the T cell suppressive capacity of MSCs, indicating that different priming strategies engage distinct immunosuppressive mechanisms. CONCLUSION Priming can be a useful approach to improve the immunosuppressive potency of MSCs. However, future studies involving primed MSCs should carefully consider the significant impact of translationally relevant conditions on the preservation of priming effects. Continuous 3D culture could act as a functionalized formulation, supporting the administration of MSC spheroids for a sustainably improved immunosuppressive potency.
Collapse
Affiliation(s)
- Nick Herger
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Balgrist Campus, Zurich, Switzerland.
| | - Irina Heggli
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Balgrist Campus, Zurich, Switzerland
| | - Tamara Mengis
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Balgrist Campus, Zurich, Switzerland
| | - Jan Devan
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Balgrist Campus, Zurich, Switzerland
| | - Leonardo Arpesella
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Balgrist Campus, Zurich, Switzerland
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia
| | - Florian Brunner
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Balgrist Campus, Zurich, Switzerland
| | - Oliver Distler
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Stefan Dudli
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Balgrist Campus, Zurich, Switzerland
| |
Collapse
|
6
|
Galera MR, Svalgaard J, Woetmann A. Therapeutic potential of adipose derived stromal cells for major skin inflammatory diseases. Front Med (Lausanne) 2024; 11:1298229. [PMID: 38463491 PMCID: PMC10921940 DOI: 10.3389/fmed.2024.1298229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/31/2024] [Indexed: 03/12/2024] Open
Abstract
Inflammatory skin diseases like psoriasis and atopic dermatitis are chronic inflammatory skin conditions continuously under investigation due to increased prevalence and lack of cure. Moreover, long-term treatments available are often associated with adverse effects and drug resistance. Consequently, there is a clear unmet need for new therapeutic approaches. One promising and cutting-edge treatment option is the use of adipose-derived mesenchymal stromal cells (AD-MSCs) due to its immunomodulatory and anti-inflammatory properties. Therefore, this mini review aims to highlight why adipose-derived mesenchymal stromal cells are a potential new treatment for these diseases by summarizing the pre-clinical and clinical studies investigated up to date and addressing current limitations and unresolved clinical questions from a dermatological and immunomodulatory point of view.
Collapse
Affiliation(s)
- Marina Ramírez Galera
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Anders Woetmann
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Mahmoud M, Abdel-Rasheed M, Galal ER, El-Awady RR. Factors Defining Human Adipose Stem/Stromal Cell Immunomodulation in Vitro. Stem Cell Rev Rep 2024; 20:175-205. [PMID: 37962697 PMCID: PMC10799834 DOI: 10.1007/s12015-023-10654-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2023] [Indexed: 11/15/2023]
Abstract
Human adipose tissue-derived stem/stromal cells (hASCs) are adult multipotent mesenchymal stem/stromal cells with immunomodulatory capacities. Here, we present up-to-date knowledge on the impact of different experimental and donor-related factors on hASC immunoregulatory functions in vitro. The experimental determinants include the immunological status of hASCs relative to target immune cells, contact vs. contactless interaction, and oxygen tension. Factors such as the ratio of hASCs to immune cells, the cellular context, the immune cell activation status, and coculture duration are also discussed. Conditioning of hASCs with different approaches before interaction with immune cells, hASC culture in xenogenic or xenofree culture medium, hASC culture in two-dimension vs. three-dimension with biomaterials, and the hASC passage number are among the experimental parameters that greatly may impact the hASC immunosuppressive potential in vitro, thus, they are also considered. Moreover, the influence of donor-related characteristics such as age, sex, and health status on hASC immunomodulation in vitro is reviewed. By analysis of the literature studies, most of the indicated determinants have been investigated in broad non-standardized ranges, so the results are not univocal. Clear conclusions cannot be drawn for the fine-tuned scenarios of many important factors to set a standard hASC immunopotency assay. Such variability needs to be carefully considered in further standardized research. Importantly, field experts' opinions may help to make it clearer.
Collapse
Affiliation(s)
- Marwa Mahmoud
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, 12622, Cairo Governorate, Egypt.
- Department of Medical Molecular Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt.
| | - Mazen Abdel-Rasheed
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, 12622, Cairo Governorate, Egypt
- Department of Reproductive Health Research, National Research Centre, Cairo, Egypt
| | - Eman Reda Galal
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Rehab R El-Awady
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
8
|
Larey AM, Spoerer TM, Daga KR, Morfin MG, Hynds HM, Carpenter J, Hines KM, Marklein RA. High throughput screening of mesenchymal stromal cell morphological response to inflammatory signals for bioreactor-based manufacturing of extracellular vesicles that modulate microglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.19.567730. [PMID: 38014258 PMCID: PMC10680807 DOI: 10.1101/2023.11.19.567730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Due to their immunomodulatory function, mesenchymal stromal cells (MSCs) are a promising therapeutic with the potential to treat neuroinflammation associated with neurodegenerative diseases. This function can be mediated by secreted extracellular vesicles (MSC-EVs). Despite established safety, MSC clinical translation has been unsuccessful due to inconsistent clinical outcomes resulting from functional heterogeneity. Current approaches to mitigate functional heterogeneity include 'priming' MSCs with inflammatory signals to enhance function. However, comprehensive evaluation of priming and its effects on MSC-EV function has not been performed. Clinical translation of MSC-EV therapies requires significant manufacturing scale-up, yet few studies have investigated the effects of priming in bioreactors. As MSC morphology has been shown to predict their immunomodulatory function, we screened MSC morphological response to an array of priming signals and evaluated MSC-EV identity and potency in response to priming in flasks and bioreactors. We identified unique priming conditions corresponding to distinct morphologies. These conditions demonstrated a range of MSC-EV preparation quality and lipidome, allowing us to discover a novel MSC-EV manufacturing condition, as well as gain insight into potential mechanisms of MSC-EV microglia modulation. Our novel screening approach and application of priming to MSC-EV bioreactor manufacturing informs refinement of larger-scale manufacturing and enhancement of MSC-EV function.
Collapse
Affiliation(s)
- Andrew M. Larey
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Thomas M. Spoerer
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Kanupriya R. Daga
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Maria G. Morfin
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Hannah M. Hynds
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Jana Carpenter
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Kelly M. Hines
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Ross A. Marklein
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| |
Collapse
|
9
|
Neo SH, Her Z, Othman R, Tee CA, Ong LC, Wang Y, Tan I, Tan J, Yang Y, Yang Z, Chen Q, Boyer LA. Expansion of human bone marrow-derived mesenchymal stromal cells with enhanced immunomodulatory properties. Stem Cell Res Ther 2023; 14:259. [PMID: 37726837 PMCID: PMC10510228 DOI: 10.1186/s13287-023-03481-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 08/29/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) have broad potential as a cell therapy including for the treatment of drug-resistant inflammatory conditions with abnormal T cell proliferation such as graft-versus-host disease (GVHD). Clinical success, however, has been complicated by the heterogeneity of culture-expanded MSCs as well as donor variability. Here, we devise culture conditions that promote expansion of MSCs with enhanced immunomodulatory functions both in vitro and in animal models of GVHD. METHODS Human bone marrow-derived MSCs were expanded at high-confluency (MSCHC) and low-confluency state (MSCLC). Their immunomodulatory properties were evaluated with in vitro co-culture assays based on suppression of activated T cell proliferation and secretion of pro-inflammatory cytokines from activated T cells. Metabolic state of these cells was determined, while RNA sequencing was performed to explore transcriptome of these MSCs. Ex vivo expanded MSCHC or MSCLC was injected into human peripheral blood mononuclear cells (PBMC)-induced GVHD mouse model to determine their in vivo therapeutic efficacy based on clinical grade scoring, human CD45+ blood count and histopathological examination. RESULTS As compared to MSCLC, MSCHC significantly reduced both the proliferation of anti-CD3/CD28-activated T cells and secretion of pro-inflammatory cytokines upon MSCHC co-culture across several donors even in the absence of cytokine priming. Mechanistically, metabolic analysis of MSCHC prior to co-culture with activated T cells showed increased glycolytic metabolism and lactate secretion compared to MSCLC, consistent with their ability to inhibit T cell proliferation. Transcriptome analysis further revealed differential expression of immunomodulatory genes including TRIM29, BPIFB4, MMP3 and SPP1 in MSCHC as well as enriched pathways including cytokine-cytokine receptor interactions, cell adhesion and PI3K-AKT signalling. Lastly, we demonstrate in a human PBMC-induced GVHD mouse model that delivery of MSCHC showed greater suppression of inflammation and improved outcomes compared to MSCLC and saline controls. CONCLUSION Our study provides evidence that ex vivo expansion of MSCs at high confluency alters the metabolic and transcriptomic states of these cells. Importantly, this approach maximizes the production of MSCs with enhanced immunomodulatory functions without priming, thus providing a non-invasive and generalizable strategy for improving the use of MSCs for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Shu Hui Neo
- Critical Analytics for Manufacturing of Personalized Medicine (CAMP), Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
| | - Zhisheng Her
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
- Invivocue Pte Ltd, 51 Science Park Road, #01-11/13 The Aries, Singapore Science Park II, Singapore, 117586, Republic of Singapore
| | - Rashidah Othman
- Critical Analytics for Manufacturing of Personalized Medicine (CAMP), Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
| | - Ching Ann Tee
- Critical Analytics for Manufacturing of Personalized Medicine (CAMP), Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
| | - Li Ching Ong
- Invivocue Pte Ltd, 51 Science Park Road, #01-11/13 The Aries, Singapore Science Park II, Singapore, 117586, Republic of Singapore
| | - Yuehua Wang
- Invivocue Pte Ltd, 51 Science Park Road, #01-11/13 The Aries, Singapore Science Park II, Singapore, 117586, Republic of Singapore
| | - Irwin Tan
- Invivocue Pte Ltd, 51 Science Park Road, #01-11/13 The Aries, Singapore Science Park II, Singapore, 117586, Republic of Singapore
| | - Jaylen Tan
- Critical Analytics for Manufacturing of Personalized Medicine (CAMP), Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
| | - Yanmeng Yang
- Critical Analytics for Manufacturing of Personalized Medicine (CAMP), Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
| | - Zheng Yang
- Critical Analytics for Manufacturing of Personalized Medicine (CAMP), Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
- Department of Orthopaedic Surgery, National University of Singapore, NUHS, 1E Kent Ridge RoadTower Block 11, Singapore, 119288, Republic of Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore, 117510, Republic of Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore.
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Republic of Singapore.
| | - Laurie A Boyer
- Critical Analytics for Manufacturing of Personalized Medicine (CAMP), Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore.
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA.
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA.
| |
Collapse
|
10
|
Al-Akashi Z, Zujur D, Kamiya D, Kato T, Kondo T, Ikeya M. Selective vulnerability of human-induced pluripotent stem cells to dihydroorotate dehydrogenase inhibition during mesenchymal stem/stromal cell purification. Front Cell Dev Biol 2023; 11:1089945. [PMID: 36814599 PMCID: PMC9939518 DOI: 10.3389/fcell.2023.1089945] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/24/2023] [Indexed: 02/08/2023] Open
Abstract
The use of induced mesenchymal stem/stromal cells (iMSCs) derived from human induced pluripotent stem cells (hiPSCs) in regenerative medicine involves the risk of teratoma formation due to hiPSCs contamination in iMSCs. Therefore, eradicating the remaining undifferentiated hiPSCs is crucial for the effectiveness of the strategy. The present study demonstrates the Brequinar (BRQ)-induced inhibition of dihydroorotate dehydrogenase (DHODH), a key enzyme in de novo pyrimidine biosynthesis, selectively induces apoptosis, cell cycle arrest, and differentiation; furthermore, it promotes transcriptional changes and prevents the growth of 3-dimensional hiPSC aggregates. Contrastingly, BRQ-treated iMSCs showed no changes in survival, differentiation potential, or gene expression. The results suggest that BRQ is a potential agent for the effective purification of iMSCs from a mixed population of iMSCs and hiPSCs, which is a crucial step in successful iMSC-based therapy.
Collapse
Affiliation(s)
- Ziadoon Al-Akashi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Denise Zujur
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Daisuke Kamiya
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan,Takeda-CiRA Joint Program, Fujisawa, Kanagawa, Japan
| | - Tomohisa Kato
- Medical Research Institute, Kanazawa Medical University, Kanazawa, Japan
| | - Toru Kondo
- Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Makoto Ikeya
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan,Takeda-CiRA Joint Program, Fujisawa, Kanagawa, Japan,*Correspondence: Makoto Ikeya,
| |
Collapse
|
11
|
Gornostaeva AN, Buravkova LB. Changes Induced by Inflammatory-Activated Immune Cell Microenvironment in the Paracrine Profile of MSC. Bull Exp Biol Med 2023; 174:544-548. [PMID: 36894814 DOI: 10.1007/s10517-023-05745-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Indexed: 03/11/2023]
Abstract
We studied the influence of activated innate and adaptive immune cells on the production of growth factors by human adipose tissue multipotent mesenchymal stromal cells (MSC). MSC showed immunosuppressive properties in vitro: decreased activation and proliferation of stimulated immune cells. T-cell interaction with MSC resulted with increased secretion of EGF, PDGF-AB/BB, FGF-2, and VEGF growth factors. Co-culturing with natural killer cells also stimulated TGFα production. The intensity of the effect varied depending on the type of immune cells. Natural killer caused a more significant increase in PDGF-AB/BB and FGF-2 secretion, while VEGF secretion increased stronger after co-culturing with T cells. The obtained data indicate the possibility of increasing reparative potential of MSC under the influence of inflammatory microenvironment.
Collapse
Affiliation(s)
- A N Gornostaeva
- Institute of Biomedical Problems, State Research Center, Russian Academy of Sciences, Moscow, Russia.
| | - L B Buravkova
- Institute of Biomedical Problems, State Research Center, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
12
|
Hackel A, Vollmer S, Bruderek K, Lang S, Brandau S. Immunological priming of mesenchymal stromal/stem cells and their extracellular vesicles augments their therapeutic benefits in experimental graft-versus-host disease via engagement of PD-1 ligands. Front Immunol 2023; 14:1078551. [PMID: 36875112 PMCID: PMC9978482 DOI: 10.3389/fimmu.2023.1078551] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/09/2023] [Indexed: 02/18/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) and their extracellular vesicles (EVs) exert profound anti-inflammatory and regenerative effects in inflammation and tissue damage, which makes them an attractive tool for cellular therapies. In this study we have assessed the inducible immunoregulatory properties of MSCs and their EVs upon stimulation with different combinations of cytokines. First, we found that MSCs primed with IFN-γ, TNF-α and IL-1β, upregulate the expression of PD-1 ligands, as crucial mediators of their immunomodulatory activity. Further, primed MSCs and MSC-EVs, compared to unstimulated MSCs and MSC-EVs, had increased immunosuppressive effects on activated T cells and mediated an enhanced induction of regulatory T cells, in a PD-1 dependent manner. Importantly, EVs derived from primed MSCs reduced the clinical score and prolonged the survival of mice in a model of graft-versus-host disease. These effects could be reversed in vitro and in vivo by adding neutralizing antibodies directed against PD-L1 and PD-L2 to both, MSCs and their EVs. In conclusion, our data reveal a priming strategy that potentiates the immunoregulatory function of MSCs and their EVs. This concept also provides new opportunities to improve the clinical applicability and efficiency of cellular or EV-based therapeutic MSC products.
Collapse
Affiliation(s)
- Alexander Hackel
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sebastian Vollmer
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Kirsten Bruderek
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stephan Lang
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sven Brandau
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
13
|
Robb KP, Audet J, Gandhi R, Viswanathan S. Putative critical quality attribute matrix identifies mesenchymal stromal cells with potent immunomodulatory and angiogenic "fitness" ranges in response to culture process parameters. Front Immunol 2022; 13:972095. [PMID: 36532069 PMCID: PMC9747767 DOI: 10.3389/fimmu.2022.972095] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/27/2022] [Indexed: 12/05/2022] Open
Abstract
Adipose-derived mesenchymal stromal cells (MSC(AT)) display immunomodulatory and angiogenic properties, but an improved understanding of quantitative critical quality attributes (CQAs) that inform basal MSC(AT) fitness ranges for immunomodulatory and/or angiogenic applications is urgently needed for effective clinical translation. We constructed an in vitro matrix of multivariate readouts to identify putative CQAs that were sensitive enough to discriminate between specific critical processing parameters (CPPs) chosen for their ability to enhance MSC immunomodulatory and angiogenic potencies, with consideration for donor heterogeneity. We compared 3D aggregate culture conditions (3D normoxic, 3D-N) and 2D hypoxic (2D-H) culture as non-genetic CPP conditions that augment immunomodulatory and angiogenic fitness of MSC(AT). We measured multivariate panels of curated genes, soluble factors, and morphometric features for MSC(AT) cultured under varying CPP and licensing conditions, and we benchmarked these against two functional and therapeutically relevant anchor assays - in vitro monocyte/macrophage (MΦ) polarization and in vitro angiogenesis. Our results showed that varying CPP conditions was the primary driver of MSC(AT) immunomodulatory fitness; 3D-N conditions induced greater MSC(AT)-mediated MΦ polarization toward inflammation-resolving subtypes. In contrast, donor heterogeneity was the primary driver of MSC(AT) angiogenic fitness. Our analysis further revealed panels of putative CQAs with minimum and maximum values that consisted of twenty MSC(AT) characteristics that informed immunomodulatory fitness ranges, and ten MSC(AT) characteristics that informed angiogenic fitness ranges. Interestingly, many of the putative CQAs consisted of angiogenic genes or soluble factors that were inversely correlated with immunomodulatory functions (THBS1, CCN2, EDN1, PDGFA, VEGFA, EDIL3, ANGPT1, and ANG genes), and positively correlated to angiogenic functions (VEGF protein), respectively. We applied desirability analysis to empirically rank the putative CQAs for MSC(AT) under varying CPP conditions and donors to numerically identify the desirable CPP conditions or donors with maximal MSC(AT) immunomodulatory and/or angiogenic fitness. Taken together, our approach enabled combinatorial analysis of the matrix of multivariate readouts to provide putative quantitative CQAs that were sensitive to variations in select CPPs that enhance MSC immunomodulatory/angiogenic potency, and donor heterogeneity. These putative CQAs may be used to prospectively screen potent MSC(AT) donors or cell culture conditions to optimize for desired basal MSC(AT) immunomodulatory or angiogenic fitness.
Collapse
Affiliation(s)
- Kevin P. Robb
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada,Krembil Research Institute, University Health Network, Toronto, ON, Canada,Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Julie Audet
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Rajiv Gandhi
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada,Department of Surgery, Division of Orthopedic Surgery, University of Toronto, Toronto, ON, Canada
| | - Sowmya Viswanathan
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada,Krembil Research Institute, University Health Network, Toronto, ON, Canada,Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada,Department of Medicine, Division of Hematology, University of Toronto, Toronto, ON, Canada,*Correspondence: Sowmya Viswanathan,
| |
Collapse
|
14
|
Doglio M, Crossland RE, Alho AC, Penack O, Dickinson AM, Stary G, Lacerda JF, Eissner G, Inngjerdingen M. Cell-based therapy in prophylaxis and treatment of chronic graft-versus-host disease. Front Immunol 2022; 13:1045168. [PMID: 36466922 PMCID: PMC9714556 DOI: 10.3389/fimmu.2022.1045168] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/31/2022] [Indexed: 08/31/2023] Open
Abstract
Hematopoietic allogeneic stem cell transplantation (allo-SCT) is a curative option for patients with hematological malignancies. However, due to disparities in major and minor histocompatibility antigens between donor and recipient, severe inflammatory complications can occur, among which chronic graft-versus-host disease (cGVHD) can be life-threatening. A classical therapeutic approach to the prevention and treatment of cGVHD has been broad immunosuppression, but more recently adjuvant immunotherapies have been tested. This review summarizes and discusses immunomodulatory approaches with T cells, including chimeric antigen receptor (CAR) and regulatory T cells, with natural killer (NK) cells and innate lymphoid cells (ILCs), and finally with mesenchymal stromal cells (MSC) and extracellular vesicles thereof. Clinical studies and pre-clinical research results are presented likewise.
Collapse
Affiliation(s)
- Matteo Doglio
- Experimental Haematology Unit, Division of Immunology Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, Milan, Italy
| | - Rachel E. Crossland
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ana C. Alho
- JLacerda Lab, Hematology and Transplantation Immunology, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
- Serviço de Hematologia e Transplantação de Medula, Hospital de Santa Maria, Centro Hospitalar Universitário de Lisboa Norte, Lisbon, Portugal
| | - Olaf Penack
- Department of Hematology, Oncology, and Cancer Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Anne M. Dickinson
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- Alcyomics Ltd, Newcastle upon Tyne, United Kingdom
| | - Georg Stary
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - João F. Lacerda
- JLacerda Lab, Hematology and Transplantation Immunology, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
- Serviço de Hematologia e Transplantação de Medula, Hospital de Santa Maria, Centro Hospitalar Universitário de Lisboa Norte, Lisbon, Portugal
| | - Günther Eissner
- Systems Biology Ireland, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Marit Inngjerdingen
- Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway
| |
Collapse
|
15
|
Sarsenova M, Kim Y, Raziyeva K, Kazybay B, Ogay V, Saparov A. Recent advances to enhance the immunomodulatory potential of mesenchymal stem cells. Front Immunol 2022; 13:1010399. [PMID: 36211399 PMCID: PMC9537745 DOI: 10.3389/fimmu.2022.1010399] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/07/2022] [Indexed: 11/19/2022] Open
Abstract
Considering the unique therapeutic potential of mesenchymal stem cells (MSCs), including their immunosuppressive and immunomodulatory properties as well as their ability to improve tissue regeneration, these cells have attracted the attention of scientists and clinicians for the treatment of different inflammatory and immune system mediated disorders. However, various clinical trials using MSCs for the therapeutic purpose are conflicting and differ from the results of promising preclinical studies. This inconsistency is caused by several factors such as poor migration and homing capacities, low survival rate, low level of proliferation and differentiation, and donor-dependent variation of the cells. Enhancement and retention of persistent therapeutic effects of the cells remain a challenge to overcome in MSC-based therapy. In this review, we summarized various approaches to enhance the clinical outcomes of MSC-based therapy as well as revised current and future perspectives for the creation of cellular products with improved potential for diverse clinical applications.
Collapse
Affiliation(s)
- Madina Sarsenova
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Yevgeniy Kim
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Kamila Raziyeva
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Bexultan Kazybay
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Vyacheslav Ogay
- Laboratory of Stem Cells, National Center for Biotechnology, Nur-Sultan, Kazakhstan
| | - Arman Saparov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
- *Correspondence: Arman Saparov,
| |
Collapse
|
16
|
Wiese DM, Wood CA, Ford BN, Braid LR. Cytokine Activation Reveals Tissue-Imprinted Gene Profiles of Mesenchymal Stromal Cells. Front Immunol 2022; 13:917790. [PMID: 35924240 PMCID: PMC9341285 DOI: 10.3389/fimmu.2022.917790] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/10/2022] [Indexed: 11/30/2022] Open
Abstract
Development of standardized metrics to support manufacturing and regulatory approval of mesenchymal stromal cell (MSC) products is confounded by heterogeneity of MSC populations. Many reports describe fundamental differences between MSCs from various tissues and compare unstimulated and activated counterparts. However, molecular information comparing biological profiles of activated MSCs across different origins and donors is limited. To better understand common and source-specific mechanisms of action, we compared the responses of 3 donor populations each of human umbilical cord (UC) and bone marrow (BM) MSCs to TNF-α, IL-1β or IFN-γ. Transcriptome profiles were analysed by microarray and select secretome profiles were assessed by multiplex immunoassay. Unstimulated (resting) UC and BM-MSCs differentially expressed (DE) 174 genes. Signatures of TNF-α-stimulated BM and UC-MSCs included 45 and 14 new DE genes, respectively, while all but 7 of the initial 174 DE genes were expressed at comparable levels after licensing. After IL-1β activation, only 5 of the 174 DE genes remained significantly different, while 6 new DE genes were identified. IFN-γ elicited a robust transcriptome response from both cell types, yet nearly all differences (171/174) between resting populations were attenuated. Nine DE genes predominantly corresponding to immunogenic cell surface proteins emerged as a BM-MSC signature of IFN-γ activation. Changes in protein synthesis of select analytes correlated modestly with transcript levels. The dynamic responses of licensed MSCs documented herein, which attenuated heterogeneity between unstimulated populations, provide new insight into common and source-imprinted responses to cytokine activation and can inform strategic development of meaningful, standardized assays.
Collapse
Affiliation(s)
| | | | - Barry N. Ford
- Defence Research and Development Canada Suffield Research Centre, Casualty Management Section, Medicine Hat, AB, Canada
| | - Lorena R. Braid
- Aurora BioSolutions Inc., Medicine Hat, AB, Canada
- Simon Fraser University, Department of Molecular Biology and Biochemistry, Burnaby, BC, Canada
- *Correspondence: Lorena R. Braid, ;
| |
Collapse
|
17
|
Saheli M, Khoramipour K, Vosough M, Piryaei A, Rahmati M, Suzuki K. Athletes' Mesenchymal Stem Cells Could Be the Best Choice for Cell Therapy in Omicron-Infected Patients. Cells 2022; 11:1926. [PMID: 35741055 PMCID: PMC9221912 DOI: 10.3390/cells11121926] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/08/2022] [Accepted: 06/12/2022] [Indexed: 02/05/2023] Open
Abstract
New severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variant, Omicron, contains 32 mutations that have caused a high incidence of breakthrough infections or re-infections. These mutations have reduced vaccine protection against Omicron and other new emerging variants. This highlights the need to find effective treatment, which is suggested to be stem cell-based therapy. Stem cells could support respiratory epithelial cells and they could restore alveolar bioenergetics. In addition, they can increase the secretion of immunomodulatory cytokines. However, after transplantation, cell survival and growth rate are low because of an inappropriate microenvironment, and stem cells face ischemia, inflammation, and oxidative stress in the transplantation niche which reduces the cells' survival and growth. Exercise-training can upregulate antioxidant, anti-inflammatory, and anti-apoptotic defense mechanisms and increase growth signaling, thereby improving transplanted cells' survival and growth. Hence, using athletes' stem cells may increase stem-cell therapy outcomes in Omicron-affected patients.
Collapse
Affiliation(s)
- Mona Saheli
- Department of Anatomical Sciences, and Pathology and Stem Cell Research Centre, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman 7616914115, Iran;
| | - Kayvan Khoramipour
- Neuroscience Research Center, Institute of Neuropharmacology, and Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman 7616914115, Iran
- Student Research Committee, Kerman University of Medical Sciences, Kerman 7619813159, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran;
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, 17177 Stockholm, Sweden
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran;
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1434875451, Iran
| | - Masoud Rahmati
- Department of Physical Education and Sport Sciences, Faculty of Literature and Human Sciences, Lorestan University, Khoramabad 6815144316, Iran;
| | - Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, Tokorozawa 359-1192, Saitama, Japan
| |
Collapse
|
18
|
Kyawsoewin M, Limraksasin P, Ngaokrajang U, Pavasant P, Osathanon T. Extracellular adenosine triphosphate induces IDO and IFNγ expression of human periodontal ligament cells through P 2 X 7 receptor signaling. J Periodontal Res 2022; 57:742-753. [PMID: 35510301 DOI: 10.1111/jre.12997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/31/2022] [Accepted: 04/19/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Mechanical stimuli induce the release of adenosine triphosphate into the extracellular environment by human periodontal ligament cells (hPDLCs). Extracellular adenosine triphosphate (eATP) plays the role in both inflammation and osteogenic differentiation. eATP involves in immunosuppressive action by increasing immunosuppressive molecules IDO and IFNγ expression on immune cells. However, the role of eATP on the immunomodulation of hPDLCs remains unclear. This study aimed to examine the effects of eATP on the IDO and IFNγ expression of hPDLCs and the participation of purinergic P2 receptors in this phenomenon. METHODS hPDLCs were treated with eATP. The mRNA and protein expression of indoleamine-pyrrole 2,3-dioxygenase (IDO) and interferon-gamma (IFNγ) were determined. The role of the purinergic P2 receptor was determined using calcium chelator (EGTA) and PKC inhibitor (PKCi). Chemical inhibitors (KN62 and BBG), small interfering RNA (siRNA), and P2 X7 receptor agonist (BzATP) were used to confirm the involvement of P2 X7 receptors on IDO and IFNγ induction by hPDLCs. RESULTS eATP significantly enhanced mRNA expression of IDO and IFNγ. Moreover, eATP increased kynurenine which is the active metabolite of tryptophan breakdown catalyzed by the IDO enzyme and significantly induced IFNγ protein expression. EGTA and PKCi reduced eATP-induced IDO and IFNγ expressions by hPDLCs, confirming the role of calcium signaling. Chemical P2 X7 inhibitors (KN62 and BBG) and siRNA targeting the P2 X7 receptor significantly inhibited the eATP-induced IDO and IFNγ production. Correspondingly, BzATP markedly increased IDO and IFNγ expression. CONCLUSION eATP induced immunosuppressive function of hPDLCs by promoting IDO and IFNγ production via P2 X7 receptor signaling. eATP may become a promising target for periodontal regeneration by modulating immune response and further triggering tissue healing.
Collapse
Affiliation(s)
- Maythwe Kyawsoewin
- Center of Excellence for Regenerative Dentistry, Chulalongkorn University, Bangkok, Thailand.,Department of Oral Biological Science, University of Dental Medicine, Yangon, Myanmar
| | - Phoonsuk Limraksasin
- Center of Excellence for Regenerative Dentistry, Chulalongkorn University, Bangkok, Thailand.,Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Utapin Ngaokrajang
- Center of Excellence for Regenerative Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Prasit Pavasant
- Center of Excellence for Regenerative Dentistry, Chulalongkorn University, Bangkok, Thailand.,Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Thanaphum Osathanon
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
19
|
Zoehler B, Fracaro L, Boldrini-Leite LM, da Silva JS, Travers PJ, Brofman PRS, Bicalho MDG, Senegaglia AC. HLA-G and CD152 Expression Levels Encourage the Use of Umbilical Cord Tissue-Derived Mesenchymal Stromal Cells as an Alternative for Immunosuppressive Therapy. Cells 2022; 11:cells11081339. [PMID: 35456019 PMCID: PMC9032010 DOI: 10.3390/cells11081339] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/30/2022] [Accepted: 04/11/2022] [Indexed: 12/04/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have been used in immunosuppressive therapy due to their therapeutic effects, with the HLA-G molecule seeming to play a fundamental role. This work evaluated alternative MSC sources to bone marrow (BM), namely, umbilical cord tissue (UC), adipose tissue (AD) and dental pulp tissue (DP), and the influence of interferon-γ (IFN-γ) and hypoxia on the cultivation of these cells for use in immunosuppression therapies. Expression of costimulatory markers CD40, CD80 and CD86 and immunosuppressive molecules CD152 and HLA-G was analyzed. Lymphocyte inhibition assays were also performed. Sequencing of the HLA-G gene from exons 1 to 5 was performed using next-generation sequencing to determine the presence of alleles. UC-derived MSCs (UCMSCs) expressed higher CD152 and HLA-G1 under standard cultivation. UCMSCs and DP-derived MSCs (DPSCs) secreted similar levels of HLA-G5. All MSC sources inhibited the proliferation of peripheral blood mononuclear cells (PBMCs); growth under regular versus hypoxic conditions resulted in similar levels of inhibition. When IFN-γ was added, PBMC growth was inhibited to a lesser extent by UCMSCs. The HLA-G*01:04:01:01 allele appears to generate a more efficient MSC response in inhibiting lymphocyte proliferation. However, the strength of this conclusion was limited by the small sample size. UCMSCs are an excellent alternative to BM in immunosuppressive therapy: they express high concentrations of inhibitory molecules and can be cultivated without stimuli, which minimizes cost.
Collapse
Affiliation(s)
- Bernardo Zoehler
- Immunogenetics and Histocompatibility Laboratory, Department of Genetics, Universidade Federal do Paraná (UFPR), Curitiba 81530-001, PR, Brazil; (J.S.d.S.); (M.d.G.B.)
- Correspondence: (B.Z.); (A.C.S.)
| | - Letícia Fracaro
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80910-215, PR, Brazil; (L.F.); (L.M.B.-L.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine, INCT-REGENERA, Rio de Janeiro 21941-902, RJ, Brazil
| | - Lidiane Maria Boldrini-Leite
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80910-215, PR, Brazil; (L.F.); (L.M.B.-L.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine, INCT-REGENERA, Rio de Janeiro 21941-902, RJ, Brazil
| | - José Samuel da Silva
- Immunogenetics and Histocompatibility Laboratory, Department of Genetics, Universidade Federal do Paraná (UFPR), Curitiba 81530-001, PR, Brazil; (J.S.d.S.); (M.d.G.B.)
| | - Paul J. Travers
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK;
| | - Paulo Roberto Slud Brofman
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80910-215, PR, Brazil; (L.F.); (L.M.B.-L.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine, INCT-REGENERA, Rio de Janeiro 21941-902, RJ, Brazil
| | - Maria da Graça Bicalho
- Immunogenetics and Histocompatibility Laboratory, Department of Genetics, Universidade Federal do Paraná (UFPR), Curitiba 81530-001, PR, Brazil; (J.S.d.S.); (M.d.G.B.)
| | - Alexandra Cristina Senegaglia
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80910-215, PR, Brazil; (L.F.); (L.M.B.-L.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine, INCT-REGENERA, Rio de Janeiro 21941-902, RJ, Brazil
- Correspondence: (B.Z.); (A.C.S.)
| |
Collapse
|
20
|
Gupta S, Rawat S, Krishnakumar V, Rao EP, Mohanty S. Hypoxia preconditioning elicit differential response in tissue-specific MSCs via immunomodulation and exosomal secretion. Cell Tissue Res 2022; 388:535-548. [PMID: 35316374 DOI: 10.1007/s00441-022-03615-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 03/15/2022] [Indexed: 11/02/2022]
Abstract
Mesenchymal stromal cells (MSCs) are emerging as an ideal candidate for regenerative medicine. It is known that the culture conditions impact the cellular properties of MSCs and their therapeutic behavior. Moreover, maintenance of MSCs in low oxygen tension for a short duration has shown to be beneficial for MSCs as it is similar to that of their physiological niche. However, the precise mechanism through which hypoxia pre-conditioning affects MSCs is not clear yet. Thus, in this study, we have investigated the effect of hypoxia exposure (1% O2) on tissue-specific MSCs over a period of time under serum-free culture conditions and evaluated the changes in expression of immuno-modulatory molecules and exosome biogenesis and secretion markers. It was observed that all MSCs responded differentially towards hypoxia exposure as indicated by the expression of HIF-1α. Moreover, this short-term exposure did not induce any changes in MSCs cellular morphology, proliferation rate, and surface marker profiling. In addition, we observed an enhancement in the expression of immunomodulatory factors (HLA-G, PGE-2, and IDO) after hypoxia exposure of 12 to 24 h in all tissue-specific MSCs. Interestingly, we have also observed the upregulation in exosome secretion that was further corelated to the upregulation of expression of exosome biogenesis and secretion markers (ALIX, TSG101, RAB27a, RAB27b). Though there was a differential response of MSCs where WJ-MSCs and BM-MSCs showed upregulation of these markers at 6-12 h of hypoxia pre-conditioning, while AD-MSCs showed similar changes beyond 24 h of hypoxia exposure.
Collapse
Affiliation(s)
- Suchi Gupta
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, 1stFloor, ORBO Complex, Ansari Nagar, New Delhi, India
| | - Sonali Rawat
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, 1stFloor, ORBO Complex, Ansari Nagar, New Delhi, India
| | - Vishnu Krishnakumar
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, 1stFloor, ORBO Complex, Ansari Nagar, New Delhi, India
| | - E Pranshu Rao
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, 1stFloor, ORBO Complex, Ansari Nagar, New Delhi, India
| | - Sujata Mohanty
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, 1stFloor, ORBO Complex, Ansari Nagar, New Delhi, India.
| |
Collapse
|
21
|
Uberti B, Plaza A, Henríquez C. Pre-conditioning Strategies for Mesenchymal Stromal/Stem Cells in Inflammatory Conditions of Livestock Species. Front Vet Sci 2022; 9:806069. [PMID: 35372550 PMCID: PMC8974404 DOI: 10.3389/fvets.2022.806069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/16/2022] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) therapy has been a cornerstone of regenerative medicine in humans and animals since their identification in 1968. MSCs can interact and modulate the activity of practically all cellular components of the immune response, either through cell-cell contact or paracrine secretion of soluble mediators, which makes them an attractive alternative to conventional therapies for the treatment of chronic inflammatory and immune-mediated diseases. Many of the mechanisms described as necessary for MSCs to modulate the immune/inflammatory response appear to be dependent on the animal species and source. Although there is evidence demonstrating an in vitro immunomodulatory effect of MSCs, there are disparate results between the beneficial effect of MSCs in preclinical models and their actual use in clinical diseases. This discordance might be due to cells' limited survival or impaired function in the inflammatory environment after transplantation. This limited efficacy may be due to several factors, including the small amount of MSCs inoculated, MSC administration late in the course of the disease, low MSC survival rates in vivo, cryopreservation and thawing effects, and impaired MSC potency/biological activity. Multiple physical and chemical pre-conditioning strategies can enhance the survival rate and potency of MSCs; this paper focuses on hypoxic conditions, with inflammatory cytokines, or with different pattern recognition receptor ligands. These different pre-conditioning strategies can modify MSCs metabolism, gene expression, proliferation, and survivability after transplantation.
Collapse
Affiliation(s)
- Benjamin Uberti
- Instituto de Ciencias Clínicas, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Anita Plaza
- Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Claudio Henríquez
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
- *Correspondence: Claudio Henríquez
| |
Collapse
|
22
|
Arnaiz E, Harris AL. Role of Hypoxia in the Interferon Response. Front Immunol 2022; 13:821816. [PMID: 35251003 PMCID: PMC8895238 DOI: 10.3389/fimmu.2022.821816] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
In solid tumors, as the tumor grows and the disease progresses, hypoxic regions are often generated, but in contrast to most normal cells which cannot survive under these conditions, tumour cells adapt to hypoxia by HIF-driven mechanisms. Hypoxia can further promote cancer development by generating an immunosuppressive environment within the tumour mass, which allows tumour cells to escape the immune system recognition. This is achieved by recruiting immunosuppressive cells and by upregulating molecules which block immune cell activation. Hypoxia can also confer resistance to antitumor therapies by inducing the expression of membrane proteins that increase drug efflux or by inhibiting the apoptosis of treated cells. In addition, tumor cells require an active interferon (IFN) signalling pathway for the success of many anticancer therapies, such as radiotherapy or chemotherapy. Therefore, hypoxic effects on this pathway needs to be addressed for a successful treatment.
Collapse
Affiliation(s)
- Esther Arnaiz
- Department of Oncology, University of Oxford, Oxford, United Kingdom
- Cambridge Institute for Therapeutic Immunology & Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge, United Kingdom
| | - Adrian L. Harris
- Department of Oncology, University of Oxford, Oxford, United Kingdom
- *Correspondence: Adrian L. Harris,
| |
Collapse
|
23
|
Mesenchymal Stem Cell-Based Therapy for Rheumatoid Arthritis. Int J Mol Sci 2021; 22:ijms222111592. [PMID: 34769021 PMCID: PMC8584240 DOI: 10.3390/ijms222111592] [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: 10/11/2021] [Revised: 10/23/2021] [Accepted: 10/24/2021] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have great potential to differentiate into various types of cells, including but not limited to, adipocytes, chondrocytes and osteoblasts. In addition to their progenitor characteristics, MSCs hold unique immunomodulatory properties that provide new opportunities in the treatment of autoimmune diseases, and can serve as a promising tool in stem cell-based therapy. Rheumatoid arthritis (RA) is a chronic systemic autoimmune disorder that deteriorates quality and function of the synovium membrane, resulting in chronic inflammation, pain and progressive cartilage and bone destruction. The mechanism of RA pathogenesis is associated with dysregulation of innate and adaptive immunity. Current conventional treatments by steroid drugs, antirheumatic drugs and biological agents are being applied in clinical practice. However, long-term use of these drugs causes side effects, and some RA patients may acquire resistance to these drugs. In this regard, recently investigated MSC-based therapy is considered as a promising approach in RA treatment. In this study, we review conventional and modern treatment approaches, such as MSC-based therapy through the understanding of the link between MSCs and the innate and adaptive immune systems. Moreover, we discuss recent achievements in preclinical and clinical studies as well as various strategies for the enhancement of MSC immunoregulatory properties.
Collapse
|
24
|
Rozier P, Maumus M, Maria ATJ, Toupet K, Jorgensen C, Guilpain P, Noël D. Lung Fibrosis Is Improved by Extracellular Vesicles from IFNγ-Primed Mesenchymal Stromal Cells in Murine Systemic Sclerosis. Cells 2021; 10:2727. [PMID: 34685707 PMCID: PMC8535048 DOI: 10.3390/cells10102727] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Systemic sclerosis (SSc) is a severe autoimmune disease for which mesenchymal stromal cells (MSCs)-based therapy was reported to reduce SSc-related symptoms in pre-clinical studies. Recently, extracellular vesicles released by MSCs (MSC-EVs) were shown to mediate most of their therapeutic effect. Here, we aimed at improving their efficacy by increasing the MSC-EV dose or by IFNγ-priming of MSCs. METHODS small size (ssEVs) and large size EVs (lsEVs) were recovered from murine MSCs that were pre-activated using 1 or 20 ng/mL of IFNγ. In the HOCl-induced model of SSc, mice were treated with EVs at day 21 and sacrificed at day 42. Lung and skin samples were collected for histological and molecular analyses. RESULTS increasing the dose of MSC-EVs did not add benefit to the dose previously reported to be efficient in SSc. By contrast, IFNγ pre-activation improved MSC-EVs-based treatment, essentially in the lungs. Low doses of IFNγ decreased the expression of fibrotic markers, while high doses improved remodeling and anti-inflammatory markers. IFNγ pre-activation upregulated iNos, IL1ra and Il6 in MSCs and ssEVs and the PGE2 protein in lsEVs. CONCLUSION IFNγ-pre-activation improved the therapeutic effect of MSC-EVs preferentially in the lungs of SSc mice by modulating anti-inflammatory and anti-fibrotic markers.
Collapse
Affiliation(s)
- Pauline Rozier
- IRMB, University of Montpellier, INSERM, 34295 Montpellier, France; (P.R.); (M.M.); (A.T.J.M.); (K.T.); (C.J.); (P.G.)
| | - Marie Maumus
- IRMB, University of Montpellier, INSERM, 34295 Montpellier, France; (P.R.); (M.M.); (A.T.J.M.); (K.T.); (C.J.); (P.G.)
| | - Alexandre Thibault Jacques Maria
- IRMB, University of Montpellier, INSERM, 34295 Montpellier, France; (P.R.); (M.M.); (A.T.J.M.); (K.T.); (C.J.); (P.G.)
- Department of Internal Medicine, Multi-Organic Diseases, CHU, 34295 Montpellier, France
| | - Karine Toupet
- IRMB, University of Montpellier, INSERM, 34295 Montpellier, France; (P.R.); (M.M.); (A.T.J.M.); (K.T.); (C.J.); (P.G.)
| | - Christian Jorgensen
- IRMB, University of Montpellier, INSERM, 34295 Montpellier, France; (P.R.); (M.M.); (A.T.J.M.); (K.T.); (C.J.); (P.G.)
- Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU, 34295 Montpellier, France
| | - Philippe Guilpain
- IRMB, University of Montpellier, INSERM, 34295 Montpellier, France; (P.R.); (M.M.); (A.T.J.M.); (K.T.); (C.J.); (P.G.)
- Department of Internal Medicine, Multi-Organic Diseases, CHU, 34295 Montpellier, France
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, 34295 Montpellier, France; (P.R.); (M.M.); (A.T.J.M.); (K.T.); (C.J.); (P.G.)
- Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU, 34295 Montpellier, France
| |
Collapse
|
25
|
He H, Takahashi A, Mukai T, Hori A, Narita M, Tojo A, Yang T, Nagamura-Inoue T. The Immunomodulatory Effect of Triptolide on Mesenchymal Stromal Cells. Front Immunol 2021; 12:686356. [PMID: 34484183 PMCID: PMC8415460 DOI: 10.3389/fimmu.2021.686356] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/27/2021] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are known to have immunosuppressive ability and have been used in clinical treatment of acute graft-versus-host disease, one of severe complications of the hematopoietic stem cell transplantation. However, MSCs are activated to suppress the immune system only after encountering an inflammatory stimulation. Thus, it will be ideal if MSCs are primed to be activated and ready to suppress the immune reaction before being administered. Triptolide (TPL) is a diterpene triepoxide purified from a Chinese herb-Tripterygium wilfordii Hook.f. It has been shown to possess anti-inflammatory and immunosuppressive properties in vitro. In this study, we aimed to use TPL to prime umbilical cord-derived MSCs (TPL-primed UC-MSCs) to enter a stronger immunosuppressive status. UC-MSCs were primed with TPL, which was washed out thoroughly, and the TPL-primed UC-MSCs were resuspended in fresh medium. Although TPL inhibited the proliferation of UC-MSCs, 0.01 μM TPL for 24 h was tolerable. The surface markers of TPL-primed UC-MSCs were identical to those of non-primed UC-MSCs. TPL-primed UC-MSCs exhibited stronger anti-proliferative effect for activated CD4+ and CD8+ T cells in the allogeneic mixed lymphocyte reaction assay than the non-primed UC-MSCs. TPL-primed UC-MSCs promoted the expression of IDO-1 in the presence of IFN-γ, but TPL alone was not sufficient. Furthermore, TPL-primed UC-MSCs showed increased expression of PD-L1. Conclusively, upregulation of IDO-1 in the presence of IFN-γ and induction of PD-L1 enhances the immunosuppressive potency of TPL-primed UC-MSCs on the proliferation of activated T cells. Thus, TPL- primed MSCs may provide a novel immunosuppressive cell therapy.
Collapse
Affiliation(s)
- Haiping He
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan.,Department of Hematology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Atsuko Takahashi
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Takeo Mukai
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Akiko Hori
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Miwako Narita
- Laboratory of Hematology and Oncology, School of Health Science, Niigata University Faculty of Medicine, Niigata, Japan
| | - Arinobu Tojo
- Division of Molecular Therapy, Center for Advanced Medical Research, Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Tonghua Yang
- Department of Hematology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| |
Collapse
|
26
|
Functional heterogeneity of IFN-γ-licensed mesenchymal stromal cell immunosuppressive capacity on biomaterials. Proc Natl Acad Sci U S A 2021; 118:2105972118. [PMID: 34446555 DOI: 10.1073/pnas.2105972118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are increasingly combined with biomaterials to enhance their therapeutic properties, including their immunosuppressive function. However, clinical trials utilizing MSCs with or without biomaterials have shown limited success, potentially due to their functional heterogeneity across different donors and among different subpopulations of cells. Here, we evaluated the immunosuppressive capacity, as measured by the ability to reduce T-cell proliferation and activation, of interferon-gamma (IFN-γ)-licensed MSCs from multiple donors on fibrin and collagen hydrogels, the two most commonly utilized biomaterials in combination with MSCs in clinical trials worldwide according to ClinicalTrials.gov Variations in the immunosuppressive capacity between IFN-γ-licensed MSC donors on the biomaterials correlated with the magnitude of indoleamine-2,3-dioxygenase activity. Immunosuppressive capacity of the IFN-γ-licensed MSCs depended on the αV/α5 integrins when cultured on fibrin and on the α2/β1 integrins when cultured on collagen. While all tested MSCs were nearly 100% positive for these integrins, sorted MSCs that expressed higher levels of αV/α5 integrins demonstrated greater immunosuppressive capacity with IFN-γ licensing than MSCs that expressed lower levels of these integrins on fibrin. These findings were equivalent for MSCs sorted based on the α2/β1 integrins on collagen. These results demonstrate the importance of integrin engagement to IFN-γ licensed MSC immunosuppressive capacity and that IFN-γ-licensed MSC subpopulations of varying immunosuppressive capacity can be identified by the magnitude of integrin expression specific to each biomaterial.
Collapse
|
27
|
Infusion of Mesenchymal Stem Cells to Treat Graft Versus Host Disease: the Role of HLA-G and the Impact of its Polymorphisms. Stem Cell Rev Rep 2021; 16:459-471. [PMID: 32088839 DOI: 10.1007/s12015-020-09960-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hematopoietic stem-cell transplantation is widely performed for the treatment of hematologic diseases and is increasingly being used for the experimental treatment of various autoimmune diseases. Despite the rapid evolution of this therapy, the mortality rate of patients undergoing this procedure is still high, mainly due to the development of graft versus host disease (GvHD). Even with the administration of immunosuppressive therapy, some patients manifest the chronic form of the disease. For these cases, infusion of mesenchymal stem cells (MSCs) was proposed as a therapeutic strategy, considering the immunosuppressive potential of these cells. This review describes the main results obtained in cell therapy with MSCs for the treatment of GvHD. Despite the encouraging results found, some points differed among the studies. Although the factors that influence the different results are uncertain, some investigators have suggested that variations in immunosuppressive molecules are responsible for these divergences. We highlight the key role of the HLA-G gene in modulating the immune response, and the importance of the polymorphisms and alleles of this gene associated with the outcome of the transplants. We suggest that the HLA-G gene and its polymorphisms be analyzed as a factor in selecting the MSCs to be used in treating GvHD, given its strong immunosuppressive role.
Collapse
|
28
|
Noronha NC, Mizukami A, Orellana MD, Oliveira MC, Covas DT, Swiech K, Malmegrim KC. Hypoxia priming improves in vitro angiogenic properties of umbilical cord derived-mesenchymal stromal cells expanded in stirred-tank bioreactor. Biochem Eng J 2021. [DOI: 10.1016/j.bej.2021.107949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
29
|
Abstract
Mesenchymal stromal cells (MSC) are multipotent precursor cells that can be derived from a variety of tissue sources, with a working definition based on immunophenotyping and cell differentiation capacity. Despite historical roots in the field of tissue engineering, they have generated great interest as cell therapies for their immune regulatory function, which has led to numerous clinical trials for a range of inflammatory and autoimmune conditions. Importantly, due to the lack of traditional MHC expression and their expression of other immune regulatory proteins, they can be used from third party donors without generating a dangerous alloreactivity. After 20 years of clinical trials, they have earned themselves an excellent safety record but are currently only approved for use in Canada, New Zealand, Japan, South Korea and Europe due to a lack of consistent efficacy data. In the United States, the indication that has seen the most progress is steroid refractory acute graft-versus-host disease (SR-aGVHD). Issues with early clinical trials can be attributed to both challenges with defining optimal patient populations and trial design as well as limitations related to commercial manufacturing. Earlier this year, the encouraging data for a repeat Phase III trial in pediatric patients with SR-aGVHD was published. This review provides information on the proposed mechanism of action of MSCs, clinical utilization of MSCs with focus on SR-aGVHD and potential modalities that can improve the efficacy of MSCs.
Collapse
Affiliation(s)
- Holly Wobma
- Department of Pediatrics, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115, USA
| | - Prakash Satwani
- Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Department of Pediatrics, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
30
|
Contreras-Lopez R, Elizondo-Vega R, Luque-Campos N, Torres MJ, Pradenas C, Tejedor G, Paredes-Martínez MJ, Vega-Letter AM, Campos-Mora M, Rigual-Gonzalez Y, Oyarce K, Salgado M, Jorgensen C, Khoury M, Garcia-Robles MDLÁ, Altamirano C, Djouad F, Luz-Crawford P. The ATP synthase inhibition induces an AMPK-dependent glycolytic switch of mesenchymal stem cells that enhances their immunotherapeutic potential. Am J Cancer Res 2021; 11:445-460. [PMID: 33391485 PMCID: PMC7681096 DOI: 10.7150/thno.51631] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023] Open
Abstract
Objectives: Mesenchymal Stem/Stromal Cells (MSC) are promising therapeutic tools for inflammatory diseases due to their potent immunoregulatory capacities. Their suppressive activity mainly depends on inflammatory cues that have been recently associated with changes in MSC bioenergetic status towards a glycolytic metabolism. However, the molecular mechanisms behind this metabolic reprogramming and its impact on MSC therapeutic properties have not been investigated. Methods: Human and murine-derived MSC were metabolically reprogramed using pro-inflammatory cytokines, an inhibitor of ATP synthase (oligomycin), or 2-deoxy-D-glucose (2DG). The immunosuppressive activity of these cells was tested in vitro using co-culture experiments with pro-inflammatory T cells and in vivo with the Delayed-Type Hypersensitivity (DTH) and the Graph versus Host Disease (GVHD) murine models. Results: We found that the oligomycin-mediated pro-glycolytic switch of MSC significantly enhanced their immunosuppressive properties in vitro. Conversely, glycolysis inhibition using 2DG significantly reduced MSC immunoregulatory effects. Moreover, in vivo, MSC glycolytic reprogramming significantly increased their therapeutic benefit in the DTH and GVHD mouse models. Finally, we demonstrated that the MSC glycolytic switch effect partly depends on the activation of the AMPK signaling pathway. Conclusion: Altogether, our findings show that AMPK-dependent glycolytic reprogramming of MSC using an ATP synthase inhibitor contributes to their immunosuppressive and therapeutic functions, and suggest that pro-glycolytic drugs might be used to improve MSC-based therapy.
Collapse
|
31
|
Abreu SC, Hampton TH, Hoffman E, Dearborn J, Ashare A, Singh Sidhu K, Matthews DE, McKenna DH, Amiel E, Barua J, Krasnodembskaya A, English K, Mahon B, Dos Santos C, Cruz FF, Chambers DC, Liu KD, Matthay MA, Cramer RA, Stanton BA, Rocco PRM, Wargo MJ, Weiss DJ, Rolandsson Enes S. Differential effects of the cystic fibrosis lung inflammatory environment on mesenchymal stromal cells. Am J Physiol Lung Cell Mol Physiol 2020; 319:L908-L925. [PMID: 32901521 PMCID: PMC7792680 DOI: 10.1152/ajplung.00218.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/14/2020] [Accepted: 08/23/2020] [Indexed: 12/23/2022] Open
Abstract
Growing evidence demonstrates that human mesenchymal stromal cells (MSCs) modify their in vivo anti-inflammatory actions depending on the specific inflammatory environment encountered. Understanding this better is crucial to refine MSC-based cell therapies for lung and other diseases. Using acute exacerbations of cystic fibrosis (CF) lung disease as a model, the effects of ex vivo MSC exposure to clinical bronchoalveolar lavage fluid (BALF) samples, as a surrogate for the in vivo clinical lung environment, on MSC viability, gene expression, secreted cytokines, and mitochondrial function were compared with effects of BALF collected from healthy volunteers. CF BALF samples that cultured positive for Aspergillus sp. (Asp) induced rapid MSC death, usually within several hours of exposure. Further analyses suggested the fungal toxin gliotoxin as a potential mediator contributing to CF BALF-induced MSC death. RNA sequencing analyses of MSCs exposed to either Asp+ or Asp- CF BALF samples identified a number of differentially expressed transcripts, including those involved in interferon signaling, antimicrobial gene expression, and cell death. Toxicity did not correlate with bacterial lung infections. These results suggest that the potential use of MSC-based cell therapies for CF or other lung diseases may not be warranted in the presence of Aspergillus.
Collapse
Affiliation(s)
- Soraia C Abreu
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thomas H Hampton
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Evan Hoffman
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Jacob Dearborn
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Alix Ashare
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
- Section of Pulmonary and Critical Care Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | | | - Dwight E Matthews
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
- Department of Chemistry, University of Vermont, Burlington, Vermont
| | - David H McKenna
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Eyal Amiel
- Department of Biomedical and Health Sciences, College of Nursing and Health Sciences, University of Vermont, Burlington, Vermont
| | - Jayita Barua
- Division of Pulmonary Disease and Critical Care, University of Vermont, and The Vermont Lung Center, Burlington, Vermont
| | - Anna Krasnodembskaya
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University of Belfast, Belfast, United Kingdom
| | - Karen English
- Cellular Immunology Laboratory, Biology Department, Human Health Research Institute, Maynooth University, Maynooth, Ireland
| | - Bernard Mahon
- Immunology & Cell Biology Laboratory, Biology Department, Human Health Research Institute, Maynooth University, Maynooth, Ireland
| | - Claudia Dos Santos
- Departments of Medicine and Critical Care Medicine and the Keenan Research Center for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Daniel C Chambers
- School of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Queenland Lung Transplant Service, The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Kathleen D Liu
- Departments of Medicine and Anesthesiology and the Cardiovascular Research Institute, University of California, San Francisco, California
| | - Michael A Matthay
- Departments of Medicine and Anesthesiology and the Cardiovascular Research Institute, University of California, San Francisco, California
| | - Robert A Cramer
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Bruce A Stanton
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Matthew J Wargo
- Department of Microbiology & Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Daniel J Weiss
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Sara Rolandsson Enes
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
- Department of Experimental Medical Science, Lung Biology Unit, Lund University, Lund, Sweden
| |
Collapse
|
32
|
Contreras-Lopez RA, Elizondo-Vega R, Torres MJ, Vega-Letter AM, Luque-Campos N, Paredes-Martinez MJ, Pradenas C, Tejedor G, Oyarce K, Salgado M, Jorgensen C, Khoury M, Kronke G, Garcia-Robles MA, Altamirano C, Luz-Crawford P, Djouad F. PPARβ/δ-dependent MSC metabolism determines their immunoregulatory properties. Sci Rep 2020; 10:11423. [PMID: 32651456 PMCID: PMC7351754 DOI: 10.1038/s41598-020-68347-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 06/19/2020] [Indexed: 01/01/2023] Open
Abstract
Mesenchymal stem cell (MSC)-based therapy is being increasingly considered a powerful opportunity for several disorders based on MSC immunoregulatory properties. Nonetheless, MSC are versatile and plastic cells that require an efficient control of their features and functions for their optimal use in clinic. Recently, we have shown that PPARβ/δ is pivotal for MSC immunoregulatory and therapeutic functions. However, the role of PPARβ/δ on MSC metabolic activity and the relevance of PPARβ/δ metabolic control on MSC immunosuppressive properties have never been addressed. Here, we demonstrate that PPARβ/δ deficiency forces MSC metabolic adaptation increasing their glycolytic activity required for their immunoregulatory functions on Th1 and Th17 cells. Additionally, we show that the inhibition of the mitochondrial production of ATP in MSC expressing PPARβ/δ, promotes their metabolic switch towards aerobic glycolysis to stably enhance their immunosuppressive capacities significantly. Altogether, these data demonstrate that PPARβ/δ governs the immunoregulatory potential of MSC by dictating their metabolic reprogramming and pave the way for enhancing MSC immunoregulatory properties and counteracting their versatility.
Collapse
Affiliation(s)
- R A Contreras-Lopez
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile.,IRMB, Univ Montpellier, INSERM, CHU Montpellier, Inserm U 1183, IRMB, Hôpital Saint-Eloi, 80 Avenue Augustin Fliche, 34295, Montpellier Cedex 5, France
| | - R Elizondo-Vega
- Facultad de Ciencias Biológicas, Departamento de Biología Celular, Laboratorio de Biología Celular, Universidad de Concepción, Concepción, Chile
| | - M J Torres
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaiso, Valparaiso, Chile
| | - A M Vega-Letter
- Cells for Cells, Consorcio Regenero, Las Condes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - N Luque-Campos
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - M J Paredes-Martinez
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - C Pradenas
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - G Tejedor
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Inserm U 1183, IRMB, Hôpital Saint-Eloi, 80 Avenue Augustin Fliche, 34295, Montpellier Cedex 5, France
| | - K Oyarce
- Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile
| | - M Salgado
- Facultad de Ciencias Biológicas, Departamento de Biología Celular, Laboratorio de Biología Celular, Universidad de Concepción, Concepción, Chile
| | - C Jorgensen
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Inserm U 1183, IRMB, Hôpital Saint-Eloi, 80 Avenue Augustin Fliche, 34295, Montpellier Cedex 5, France
| | - M Khoury
- Cells for Cells, Consorcio Regenero, Las Condes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - G Kronke
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, 91054, Erlangen, Germany
| | - M A Garcia-Robles
- Facultad de Ciencias Biológicas, Departamento de Biología Celular, Laboratorio de Biología Celular, Universidad de Concepción, Concepción, Chile
| | - C Altamirano
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaiso, Valparaiso, Chile
| | - P Luz-Crawford
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile.
| | - F Djouad
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Inserm U 1183, IRMB, Hôpital Saint-Eloi, 80 Avenue Augustin Fliche, 34295, Montpellier Cedex 5, France.
| |
Collapse
|
33
|
Daltro SRT, Meira CS, Santos IP, Ribeiro dos Santos R, Soares MBP. Mesenchymal Stem Cells and Atopic Dermatitis: A Review. Front Cell Dev Biol 2020; 8:326. [PMID: 32478072 PMCID: PMC7240073 DOI: 10.3389/fcell.2020.00326] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 04/16/2020] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are stromal-derived non-hematopoietic progenitor cells that reside in and can be expanded from various tissues sources of adult and neonatal origin, such as the bone marrow, umbilical cord, umbilical cord blood, adipose tissue, amniotic fluid, placenta, dental pulp and skin. The discovery of the immunosuppressing action of MSCs on T cells has opened new perspectives for their use as a therapeutic agent for immune-mediated disorders, including allergies. Atopic dermatitis (AD), a chronic and relapsing skin disorder that affects up to 20% of children and up to 3% of adults worldwide, is characterized by pruritic eczematous lesions, impaired cutaneous barrier function, Th2 type immune hyperactivation and, frequently, elevation of serum immunoglobulin E levels. Although, in the dermatology field, the application of MSCs as a therapeutic agent was initiated using the concept of cell replacement for skin defects and wound healing, accumulating evidence have shown that MSC-mediated immunomodulation can be applicable to the treatment of inflammatory/allergic skin disorders. Here we reviewed the pre-clinical and clinical studies and possible biological mechanisms of MSCs as a therapeutic tool for the treatment of atopic dermatitis.
Collapse
Affiliation(s)
| | | | | | - Ricardo Ribeiro dos Santos
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- Health Institute of Technology, National Industrial Learning Service - Integrated Manufacturing and Technology Campus (SENAI-CIMATEC), Salvador, Brazil
- National Institute of Science and Technology for Regenerative Medicine (INCT-REGENERA), Rio de Janeiro, Brazil
| | - Milena Botelho Pereira Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- Health Institute of Technology, National Industrial Learning Service - Integrated Manufacturing and Technology Campus (SENAI-CIMATEC), Salvador, Brazil
- National Institute of Science and Technology for Regenerative Medicine (INCT-REGENERA), Rio de Janeiro, Brazil
| |
Collapse
|
34
|
Contreras-Lopez R, Elizondo-Vega R, Paredes MJ, Luque-Campos N, Torres MJ, Tejedor G, Vega-Letter AM, Figueroa-Valdés A, Pradenas C, Oyarce K, Jorgensen C, Khoury M, Garcia-Robles MDLA, Altamirano C, Djouad F, Luz-Crawford P. HIF1α-dependent metabolic reprogramming governs mesenchymal stem/stromal cell immunoregulatory functions. FASEB J 2020; 34:8250-8264. [PMID: 32333618 DOI: 10.1096/fj.201902232r] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 04/02/2020] [Accepted: 04/06/2020] [Indexed: 12/12/2022]
Abstract
Hypoxia-inducible factor 1 α (HIF1α), a regulator of metabolic change, is required for the survival and differentiation potential of mesenchymal stem/stromal cells (MSC). Its role in MSC immunoregulatory activity, however, has not been completely elucidated. In the present study, we evaluate the role of HIF1α on MSC immunosuppressive potential. We show that HIF1α silencing in MSC decreases their inhibitory potential on Th1 and Th17 cell generation and limits their capacity to generate regulatory T cells. This reduced immunosuppressive potential of MSC is associated with a metabolic switch from glycolysis to OXPHOS and a reduced capacity to express or produce some immunosuppressive mediators including Intercellular Adhesion Molecule (ICAM), IL-6, and nitric oxide (NO). Moreover, using the Delayed-Type Hypersensitivity murine model (DTH), we confirm, in vivo, the critical role of HIF1α on MSC immunosuppressive effect. Indeed, we show that HIF1α silencing impairs MSC capacity to reduce inflammation and inhibit the generation of pro-inflammatory T cells. This study reveals the pivotal role of HIF1α on MSC immunosuppressive activity through the regulation of their metabolic status and identifies HIF1α as a novel mediator of MSC immunotherapeutic potential.
Collapse
Affiliation(s)
- Rafael Contreras-Lopez
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Roberto Elizondo-Vega
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Maria Jose Paredes
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Noymar Luque-Campos
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Maria Jose Torres
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Catolica de Valparaiso, Valparaiso, Chile
| | - Gautier Tejedor
- IRMB, INSERM, Université de Montpellier, Montpellier, France
| | - Ana Maria Vega-Letter
- Cells for Cells, Consorcio Regenero, Las Condes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Aliosha Figueroa-Valdés
- Cells for Cells, Consorcio Regenero, Las Condes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Carolina Pradenas
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Karina Oyarce
- Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile
| | | | - Maroun Khoury
- Cells for Cells, Consorcio Regenero, Las Condes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Maria de Los Angeles Garcia-Robles
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Claudia Altamirano
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Catolica de Valparaiso, Valparaiso, Chile
| | - Farida Djouad
- IRMB, INSERM, Université de Montpellier, Montpellier, France
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| |
Collapse
|
35
|
Mohapatra SR, Sadik A, Tykocinski LO, Dietze J, Poschet G, Heiland I, Opitz CA. Hypoxia Inducible Factor 1α Inhibits the Expression of Immunosuppressive Tryptophan-2,3-Dioxygenase in Glioblastoma. Front Immunol 2019; 10:2762. [PMID: 31866995 PMCID: PMC6905408 DOI: 10.3389/fimmu.2019.02762] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
Abnormal circulation in solid tumors results in hypoxia, which modulates both tumor intrinsic malignant properties as well as anti-tumor immune responses. Given the importance of hypoxia in glioblastoma (GBM) biology and particularly in shaping anti-tumor immunity, we analyzed which immunomodulatory genes are differentially regulated in response to hypoxia in GBM cells. Gene expression analyses identified the immunosuppressive enzyme tryptophan-2,3-dioxygenase (TDO2) as the second most downregulated gene in GBM cells cultured under hypoxic conditions. TDO2 catalyses the oxidation of tryptophan to N-formyl kynurenine, which is the first and rate-limiting step of Trp degradation along the kynurenine pathway (KP). In multiple GBM cell lines hypoxia reduced TDO2 expression both at mRNA and protein levels. The downregulation of TDO2 through hypoxia was reversible as re-oxygenation rescued TDO2 expression. Computational modeling of tryptophan metabolism predicted reduced flux through the KP and lower intracellular concentrations of kynurenine and its downstream metabolite 3-hydroxyanthranilic acid under hypoxia. Metabolic measurements confirmed the predicted changes, thus demonstrating the ability of the mathematical model to infer intracellular tryptophan metabolite concentrations. Moreover, we identified hypoxia inducible factor 1α (HIF1α) to regulate TDO2 expression under hypoxic conditions, as the HIF1α-stabilizing agents dimethyloxalylglycine (DMOG) and cobalt chloride reduced TDO2 expression. Knockdown of HIF1α restored the expression of TDO2 upon cobalt chloride treatment, confirming that HIF1α controls TDO2 expression. To investigate the immunoregulatory effects of this novel mechanism of TDO2 regulation, we co-cultured isolated T cells with TDO2-expressing GBM cells under normoxic and hypoxic conditions. Under normoxia TDO2-expressing GBM cells suppressed T cell proliferation, while hypoxia restored the proliferation of the T cells, likely due to the reduction in kynurenine levels produced by the GBM cells. Taken together, our data suggest that the regulation of TDO2 expression by HIF1α may be involved in modulating anti-tumor immunity in GBM.
Collapse
Affiliation(s)
- Soumya R Mohapatra
- DKTK Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ahmed Sadik
- DKTK Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Bioscience, Heidelberg University, Heidelberg, Germany
| | - Lars-Oliver Tykocinski
- Division of Rheumatology, Department of Medicine V, University Hospital of Heidelberg, Heidelberg, Germany
| | - Jørn Dietze
- Department of Arctic and Marine Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Gernot Poschet
- Centre for Organismal Studies (COS), University of Heidelberg, Heidelberg, Germany
| | - Ines Heiland
- Department of Arctic and Marine Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Christiane A Opitz
- DKTK Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Neurology Clinic and National Center for Tumor Diseases, University Hospital of Heidelberg, Heidelberg, Germany
| |
Collapse
|
36
|
Palomares Cabeza V, Hoogduijn MJ, Kraaijeveld R, Franquesa M, Witte-Bouma J, Wolvius EB, Farrell E, Brama PAJ. Pediatric Mesenchymal Stem Cells Exhibit Immunomodulatory Properties Toward Allogeneic T and B Cells Under Inflammatory Conditions. Front Bioeng Biotechnol 2019; 7:142. [PMID: 31245368 PMCID: PMC6581756 DOI: 10.3389/fbioe.2019.00142] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/28/2019] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells from pediatric patients (pMSCs) are an attractive cell source in regenerative medicine, due to their higher proliferation rates and better differentiation abilities compared to adult MSCs (aMSCs). We have previously characterized the immunomodulatory abilities of pMSCs on T cells under co-culture. It has also been reported that aMSCs can inhibit B cell proliferation and maturation under inflammatory conditions. In this study, we therefore aimed to clarify the immunomodulatory effect of pMSCs toward T and B cells in an inflammatory microenvironment. Bone marrow derived pMSCs were primed to simulate inflammatory conditions by exposure with 50 ng/mL of IFN-γ for 3 days. To analyze the interaction between pMSCs and T cells, CD3/CD28 stimulated peripheral blood mononuclear cells (PBMCs) were co-cultured with primed or unprimed pMSCs. To investigate B cell responses, quiescent B cells obtained from spleens by CD43 negative selection were stimulated with anti-IgM, anti-CD40, IL-2, and co-cultured with either IFN-γ primed or unprimed pMSC. pMSC phenotype, B and T cell proliferation, and B cell functionality were analyzed. Gene expression of indoleamine 2,3-dioxygenease (IDO), as well as the expression of HLA-ABC, HLA-DR and the co-stimulatory molecules CD80 and CD86 was upregulated on pMSCs upon IFN-γ priming. IFN-γ did not alter the immunomodulatory abilities of pMSCs upon CD4+ nor CD8+ stimulated T cells compared to unprimed pMSCs. IFN-γ primed pMSCs but not unprimed pMSCs strongly inhibited naïve (CD19+CD27−), memory (CD19+CD27+), and total B cell proliferation. Antibody-producing plasmablast (CD19+CD27highCD38high) formation and IgG production were also significantly inhibited by IFN-γ primed pMSCs compared to unprimed pMSCs. Collectively, these results show that pMSCs have immunomodulatory effects upon the adaptive immune response which can be potentiated by inflammatory stimuli. This knowledge is useful in regenerative medicine and allogeneic transplantation applications toward tailoring pMSCs function to best modulate the immune response for a successful implant engraftment and avoidance of a strong immune reaction.
Collapse
Affiliation(s)
- Virginia Palomares Cabeza
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, Netherlands.,Nephrology and Transplantation, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands.,School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Martin Johannes Hoogduijn
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Rens Kraaijeveld
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Marcella Franquesa
- REMAR Group and Nephrology Service, Germans Trias i Pujol Health Science Institute and University Hospital, Badalona, Spain
| | - Janneke Witte-Bouma
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Eppo B Wolvius
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Pieter A J Brama
- School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
37
|
Noronha NDC, Mizukami A, Caliári-Oliveira C, Cominal JG, Rocha JLM, Covas DT, Swiech K, Malmegrim KCR. Priming approaches to improve the efficacy of mesenchymal stromal cell-based therapies. Stem Cell Res Ther 2019; 10:131. [PMID: 31046833 PMCID: PMC6498654 DOI: 10.1186/s13287-019-1224-y] [Citation(s) in RCA: 374] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multipotent mesenchymal stromal cells (MSC) have been widely explored for cell-based therapy of immune-mediated, inflammatory, and degenerative diseases, due to their immunosuppressive, immunomodulatory, and regenerative potentials. Preclinical studies and clinical trials have demonstrated promising therapeutic results although these have been somewhat limited. Aspects such as low in vivo MSC survival in inhospitable disease microenvironments, requirements for ex vivo cell overexpansion prior to infusions, intrinsic differences between MSC and different sources and donors, variability of culturing protocols, and potency assays to evaluate MSC products have been described as limitations in the field. In recent years, priming approaches to empower MSC have been investigated, thereby generating cellular products with improved potential for different clinical applications. Herein, we review the current priming approaches that aim to increase MSC therapeutic efficacy. Priming with cytokines and growth factors, hypoxia, pharmacological drugs, biomaterials, and different culture conditions, as well as other diverse molecules, are revised from current and future perspectives.
Collapse
Affiliation(s)
- Nádia de Cássia Noronha
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Graduate Program on Bioscience and Biotechnology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Amanda Mizukami
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Juçara Gastaldi Cominal
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Graduate Program on Bioscience and Biotechnology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - José Lucas M Rocha
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Graduate Program on Basic and Applied Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Dimas Tadeu Covas
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Kamilla Swiech
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Kelen C R Malmegrim
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil. .,Department of Clinical, Toxicological and Bromatological Analysis, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café, s/n°, Ribeirão Preto, SP, 14010-903, Brazil.
| |
Collapse
|