1
|
Neves FL, Amaral MNGA, da Silva SFD, Silva IMM, Laranjeira PMDS, Pinto CRDJ, Paiva AA, Dias ASDS, Coelho MLACV. Immunoparalysis in critically ill children. Immunology 2023; 168:597-609. [PMID: 36279244 DOI: 10.1111/imm.13595] [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: 05/12/2022] [Accepted: 10/20/2022] [Indexed: 11/30/2022] Open
Abstract
Immunoparalysis is associated with poorer outcomes in the paediatric intensive care unit (PICU) setting. We aimed to determine the group of patients with higher chances of immunoparalysis and correlate this status with increased risks of nosocomial infection and adverse clinical parameters. We conducted an exploratory study with prospective data collection in a university-affiliated tertiary medical, surgical, and cardiac PICU. Fifteen patients with multiple organ dysfunction syndrome were included over a period of 6 months. Monocyte's human leucocyte antigen (HLA)-DR expression and tumour necrosis factor (TNF)-α and interleukin (IL)-6 production were measured by flow-cytometry at three time points (T1 = 1-2 days; T2 = 3-5 days; T3 = 6-8 days). Using the paediatric logistic organ dysfunction-2 score to assess initial disease severity, we established the optimal cut-off values of the evaluated parameters to identify the subset of patients with a higher probability of immunoparalysis. A comparative analysis was performed between them. Sixty per cent were males; the median age was 4.1 years. Considering the presence of two criteria in T1 (classical monocytes mean fluorescence intensity [MFI] for HLA-DR ≤ 1758.5, area under the curve (AUC) = 0.775; and frequency of monocytes producing IL-6 ≤ 68.5%, AUC = 0.905) or in T3 (classical monocytes MFI of HLA-DR ≤ 2587.5, AUC = 0.675; and frequency of monocytes producing TNF-α ≤ 93.5%, AUC = 0.833), a variable to define immunoparalysis was obtained (100% sensitivity, 81.5% specificity). Forty per cent of patients were assigned to the immunoparalysis group. In this: a higher frequency of nosocomial infection (p = 0.011), vasoactive inotropic score (p = 0.014) and length of hospital stay (p = 0.036) was observed. In the subgroup with the diagnosis of sepsis/septic shock (n = 5), patients showed higher percentages of non-classical monocytes (p = 0.004). No mortality was recorded. A reduction in classical monocytes HLA-DR expression with lower frequencies of monocytes producing TNF-α and IL-6 during the first week of critical illness, appears to be a good marker of immunoparalysis; these findings relate to an increased risk of nosocomial infection and deleterious outcomes. The increased frequency of non-classical monocytes in patients with sepsis/septic shock is suggestive of a better prognosis.
Collapse
Affiliation(s)
- Filipa Loureiro Neves
- Pediatric Intensive Care Unit, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Children and Women Department, Centro Hospitalar do Médio Tejo, Torres Novas, Portugal
| | | | - Sandra Filomena Durães da Silva
- Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
| | - Isabel Maria Melo Silva
- Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
| | - Paula Margarida Dos Santos Laranjeira
- Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR) - Group of Environment, Genetics and Oncobiology (CIMAGO) - Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Carla Regina de Jesus Pinto
- Pediatric Intensive Care Unit, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- University Clinic of Pediatrics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Artur Augusto Paiva
- Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR) - Group of Environment, Genetics and Oncobiology (CIMAGO) - Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Instituto Politécnico de Coimbra, ESTESC-Coimbra Health School, Ciências Biomédicas Laboratoriais, Coimbra, Portugal
| | - Andrea Sofia da Silva Dias
- Pediatric Intensive Care Unit, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- University Clinic of Pediatrics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | |
Collapse
|
2
|
Satani N, Parsha K, Davis C, Gee A, Olson SD, Aronowski J, Savitz SI. Peripheral blood monocytes as a therapeutic target for marrow stromal cells in stroke patients. Front Neurol 2022; 13:958579. [PMID: 36277912 PMCID: PMC9580494 DOI: 10.3389/fneur.2022.958579] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/05/2022] [Indexed: 11/22/2022] Open
Abstract
Background Systemic administration of marrow stromal cells (MSCs) leads to the release of a broad range of factors mediating recovery in rodent stroke models. The release of these factors could depend on the various cell types within the peripheral blood as they contact systemically administered MSCs. In this study, we assessed the immunomodulatory interactions of MSCs with peripheral blood derived monocytes (Mϕ) collected from acute stroke patients. Methods Peripheral blood from stroke patients was collected at 5–7 days (N = 5) after symptom onset and from age-matched healthy controls (N = 5) using mononuclear cell preparation (CPT) tubes. After processing, plasma and other cellular fractions were removed, and Mϕ were isolated from the mononuclear fraction using CD14 microbeads. Mϕ were then either cultured alone or co-cultured with MSCs in a trans-well cell-culture system. Secretomes were analyzed after 24 h of co-cultures using a MAGPIX reader. Results Our results show that there is a higher release of IFN-γ and IL-10 from monocytes isolated from peripheral blood at day 5–7 after stroke compared with monocytes from healthy controls. In trans-well co-cultures of MSCs and monocytes isolated from stroke patients, we found statistically significant increased levels of IL-4 and MCP-1, and decreased levels of IL-6, IL-1β, and TNF-α. Addition of MSCs to monocytes increased the secretions of Fractalkine, IL-6, and MCP-1, while the secretions of TNF-α decreased, as compared to the secretions from monocytes alone. When MSCs were added to monocytes from stroke patients, they decreased the levels of IL-1β, and increased the levels of IL-10 significantly more as compared to when they were added to monocytes from control patients. Conclusion The systemic circulation of stroke patients may differentially interact with MSCs to release soluble factors integral to their paracrine mechanisms of benefit. Our study finds that the effect of MSCs on Mϕ is different on those derived from stroke patients blood as compared to healthy controls. These findings suggest immunomodulation of peripheral immune cells as a therapeutic target for MSCs in patients with acute stroke.
Collapse
Affiliation(s)
- Nikunj Satani
- Department of Neurology, McGovern Medical School, Institute for Stroke and Cerebrovascular Diseases, The University of Texas Health Science Center at Houston, Houston, TX, United States
- *Correspondence: Nikunj Satani
| | - Kaushik Parsha
- Department of Neurology, McGovern Medical School, Institute for Stroke and Cerebrovascular Diseases, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Courtney Davis
- Department of Neurology, McGovern Medical School, Institute for Stroke and Cerebrovascular Diseases, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Adrian Gee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, United States
| | - Scott D. Olson
- Department of Pediatric Surgery, McGovern Medical School at UTHealth, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jaroslaw Aronowski
- Department of Neurology, McGovern Medical School, Institute for Stroke and Cerebrovascular Diseases, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Sean I. Savitz
- Department of Neurology, McGovern Medical School, Institute for Stroke and Cerebrovascular Diseases, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
3
|
Connection between Mesenchymal Stem Cells Therapy and Osteoclasts in Osteoarthritis. Int J Mol Sci 2022; 23:ijms23094693. [PMID: 35563083 PMCID: PMC9102843 DOI: 10.3390/ijms23094693] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 12/12/2022] Open
Abstract
The use of mesenchymal stem cells constitutes a promising therapeutic approach, as it has shown beneficial effects in different pathologies. Numerous in vitro, pre-clinical, and, to a lesser extent, clinical trials have been published for osteoarthritis. Osteoarthritis is a type of arthritis that affects diarthritic joints in which the most common and studied effect is cartilage degradation. Nowadays, it is known that osteoarthritis is a disease with a very powerful inflammatory component that affects the subchondral bone and the rest of the tissues that make up the joint. This inflammatory component may induce the differentiation of osteoclasts, the bone-resorbing cells. Subchondral bone degradation has been suggested as a key process in the pathogenesis of osteoarthritis. However, very few published studies directly focus on the activity of mesenchymal stem cells on osteoclasts, contrary to what happens with other cell types of the joint, such as chondrocytes, synoviocytes, and osteoblasts. In this review, we try to gather the published bibliography in relation to the effects of mesenchymal stem cells on osteoclastogenesis. Although we find promising results, we point out the need for further studies that can support mesenchymal stem cells as a therapeutic tool for osteoclasts and their consequences on the osteoarthritic joint.
Collapse
|
4
|
Laranjeira P, Pedrosa M, Duarte C, Pedreiro S, Antunes B, Ribeiro T, dos Santos F, Martinho A, Fardilha M, Domingues MR, Abecasis M, Pereira da Silva JA, Paiva A. Human Bone Marrow Mesenchymal Stromal/Stem Cells Regulate the Proinflammatory Response of Monocytes and Myeloid Dendritic Cells from Patients with Rheumatoid Arthritis. Pharmaceutics 2022; 14:pharmaceutics14020404. [PMID: 35214136 PMCID: PMC8880255 DOI: 10.3390/pharmaceutics14020404] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 12/15/2022] Open
Abstract
Rheumatoid arthritis (RA) is a disabling autoimmune disease whose treatment is ineffective for one-third of patients. Thus, the immunomodulatory potential of mesenchymal stromal/stem cells (MSCs) makes MSC-based therapy a promising approach to RA. This study aimed to explore the immunomodulatory action of human bone marrow (BM)-MSCs on myeloid dendritic cells (mDCs) and monocytes, especially on cytokines/chemokines involved in RA physiopathology. For that, LPS plus IFNγ-stimulated peripheral blood mononuclear cells from RA patients (n = 12) and healthy individuals (n = 6) were co-cultured with allogeneic BM-MSCs. TNF-α, CD83, CCR7 and MIP-1β protein levels were assessed in mDCs, classical, intermediate, and non-classical monocytes. mRNA expression of other cytokines/chemokines was also evaluated. BM-MSCs effectively reduced TNF-α, CD83, CCR7 and MIP-1β protein levels in mDCs and all monocyte subsets, in RA patients. The inhibition of TNF-α production was mainly achieved by the reduction of the percentage of cellsproducing this cytokine. BM-MSCs exhibited a remarkable suppressive action over antigen-presenting cells from RA patients, potentially affecting their ability to stimulate the immune adaptive response at different levels, by hampering their migration to the lymph node and the production of proinflammatory cytokines and chemokines. Accordingly, MSC-based therapies can be a valuable approach for RA treatment, especially for non-responder patients.
Collapse
Affiliation(s)
- Paula Laranjeira
- Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra, Av. Bissaya Barreto, Bloco de Celas, 3000-075 Coimbra, Portugal;
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal, Quinta da Vinha Moura, São Martinho do Bispo, 3041-861 Coimbra, Portugal; (M.P.); (S.P.); (A.M.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (C.D.); (J.A.P.d.S.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Polo 1, 1.° Piso, FMUC, Rua Larga, 3004-504 Coimbra, Portugal
| | - Mónia Pedrosa
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal, Quinta da Vinha Moura, São Martinho do Bispo, 3041-861 Coimbra, Portugal; (M.P.); (S.P.); (A.M.)
- Signal Transduction Laboratory, Center of Cellular Biology, SACS and Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal;
- Enzifarma—Diagnostica e Farmacêutica, S.A., Estrada da Luz, n.° 90, 2° F, 1600-160 Lisbon, Portugal
| | - Cátia Duarte
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (C.D.); (J.A.P.d.S.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Rheumatology Department, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, Praceta Professor Mota Pinto, 3000-075 Coimbra, Portugal
| | - Susana Pedreiro
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal, Quinta da Vinha Moura, São Martinho do Bispo, 3041-861 Coimbra, Portugal; (M.P.); (S.P.); (A.M.)
| | - Brígida Antunes
- Cell2B Advanced Therapeutics, SA, Biocant Park, Núcleo 04, Lote 4A, 3060-197 Cantanhede, Portugal; ; (B.A.); (T.R.); (F.d.S.)
| | - Tânia Ribeiro
- Cell2B Advanced Therapeutics, SA, Biocant Park, Núcleo 04, Lote 4A, 3060-197 Cantanhede, Portugal; ; (B.A.); (T.R.); (F.d.S.)
| | - Francisco dos Santos
- Cell2B Advanced Therapeutics, SA, Biocant Park, Núcleo 04, Lote 4A, 3060-197 Cantanhede, Portugal; ; (B.A.); (T.R.); (F.d.S.)
- Stemlab SA, Biocant Park, Núcleo 04, Lote 2, 3060-197 Cantanhede, Portugal
| | - António Martinho
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal, Quinta da Vinha Moura, São Martinho do Bispo, 3041-861 Coimbra, Portugal; (M.P.); (S.P.); (A.M.)
| | - Margarida Fardilha
- Signal Transduction Laboratory, Center of Cellular Biology, SACS and Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal;
- Laboratory of Signal Transduction, Institute of Biomedicine—iBiMED, Department of Medical Sciences, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - M. Rosário Domingues
- Mass Spectrometry Centre, LAQV REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal;
- Centre for Environmental and Marine Studies (CESAM), Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal
| | - Manuel Abecasis
- Serviço de Transplantação de Progenitores Hematopoiéticos (UTM), Instituto Português de Oncologia de Lisboa Francisco Gentil, Rua Professor Lima Basto, 1099-023 Lisbon, Portugal;
- Instituto Português do Sangue e da Transplantação—CEDACE, Alameda das Linhas de Torres, 117, 1769-001 Lisbon, Portugal
| | - José António Pereira da Silva
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (C.D.); (J.A.P.d.S.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Rheumatology Department, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, Praceta Professor Mota Pinto, 3000-075 Coimbra, Portugal
| | - Artur Paiva
- Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra, Av. Bissaya Barreto, Bloco de Celas, 3000-075 Coimbra, Portugal;
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal, Quinta da Vinha Moura, São Martinho do Bispo, 3041-861 Coimbra, Portugal; (M.P.); (S.P.); (A.M.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (C.D.); (J.A.P.d.S.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Instituto Politécnico de Coimbra, ESTESC-Coimbra Health School, Ciências Biomédicas Laboratoriais, Rua 5 de Outubro, 3046-854 Coimbra, Portugal
- Correspondence: ; Tel.: +351-239-488-700
| |
Collapse
|
5
|
Xie Y, Liu S, Wang L, Yang H, Tai C, Ling L, Chen L, Liu S, Wang B. Individual heterogeneity screened umbilical cord-derived mesenchymal stromal cells with high Treg promotion demonstrate improved recovery of mouse liver fibrosis. Stem Cell Res Ther 2021; 12:359. [PMID: 34158112 PMCID: PMC8220795 DOI: 10.1186/s13287-021-02430-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 06/05/2021] [Indexed: 12/15/2022] Open
Abstract
Background To investigate the heterogeneities of human umbilical cord mesenchymal stromal cells (HUCMSCs) derived from different donors and their therapeutic variations when applied to mouse liver fibrosis model. Methods The characteristics of HUCMSCs derived from multiple donors were comprehensively analyzed including expressions of surface markers, viability, growth curve, karyotype analysis, tumorigenicity, differentiation potentials, and immune regulation capability. Then, the HUCMSCs with distinct immunomodulatory effects were applied to treat mouse liver fibrosis and their therapeutic effects were observed. Results The HUCMSCs derived from multiple donors kept a high consistency in surface marker expressions, viability, growth curve, and tumorigenicity in nude mice but had robust heterogeneities in differentiation potentials and immune regulations. In addition, three HUCMSC lines applied to mice liver fibrosis model had different therapeutic outcomes, in line with individual immune regulation capability. Conclusion The HUCMSCs derived from different donors have individual heterogeneity, which potentially lead to distinct therapeutic outcomes in mouse liver fibrosis, indicating we could make use of the donor-variation of MSCs to screen out guaranteed general indicators of MSCs for specific diseases in further stromal cell therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02430-6.
Collapse
Affiliation(s)
- Yuanyuan Xie
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210000, People's Republic of China
| | - Shuo Liu
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210000, People's Republic of China
| | - Liudi Wang
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210000, People's Republic of China
| | - Hui Yang
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210000, People's Republic of China
| | - Chenxu Tai
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210000, People's Republic of China
| | - Li Ling
- Department of Endocrinology, University Health Science Center, Hua Zhong University of Science and Technology Union Shenzhen Hospital and The 6th Affiliated Hospital of Shenzhen, Shenzhen, 518052, Guangdong, People's Republic of China
| | - Libo Chen
- Department of Endocrinology, University Health Science Center, Hua Zhong University of Science and Technology Union Shenzhen Hospital and The 6th Affiliated Hospital of Shenzhen, Shenzhen, 518052, Guangdong, People's Republic of China
| | - Shanshan Liu
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210000, People's Republic of China
| | - Bin Wang
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210000, People's Republic of China.
| |
Collapse
|
6
|
Corey S, Bonsack B, Heyck M, Shear A, Sadanandan N, Zhang H, Borlongan CV. Harnessing the anti-inflammatory properties of stem cells for transplant therapy in hemorrhagic stroke. BRAIN HEMORRHAGES 2020; 1:24-33. [PMID: 34056567 PMCID: PMC8158660 DOI: 10.1016/j.hest.2019.12.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hemorrhagic stroke is a global health crisis plagued by neuroinflammation in the acute and chronic phases. Neuroinflammation approximates secondary cell death, which in turn robustly contributes to stroke pathology. Both the physiological and behavioral symptoms of stroke correlate with various inflammatory responses in animal and human studies. That slowing the secondary cell death mediated by this inflammation may attenuate stroke pathology presents a novel treatment strategy. To this end, experimental therapies employing stem cell transplants support their potential for neuroprotection and neuroregeneration after hemorrhagic stroke. In this review, we evaluate experiments using different types of stem cell transplants as treatments for stroke-induced neuroinflammation. We also update this emerging area by examining recent preclinical and clinical trials that have deployed these therapies. While further investigations are warranted to solidify their therapeutic profile, the reviewed studies largely posit stem cells as safe and potent biologics for stroke, specifically owing to their mode of action for sequestering neuroinflammation and promoting neuroregenerative processes.
Collapse
Affiliation(s)
- Sydney Corey
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Brooke Bonsack
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Matt Heyck
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Alex Shear
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Nadia Sadanandan
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Henry Zhang
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| |
Collapse
|
7
|
Short and Long Term Clinical and Immunologic Follow up after Bone Marrow Mesenchymal Stromal Cell Therapy in Progressive Multiple Sclerosis-A Phase I Study. J Clin Med 2019; 8:jcm8122102. [PMID: 31810187 PMCID: PMC6947442 DOI: 10.3390/jcm8122102] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/20/2022] Open
Abstract
Bone marrow derived mesenchymal stromal cells (BM-MSCs) have emerged as a possible new therapy for Multiple Sclerosis (MS), however studies regarding efficacy and in vivo immune response have been limited and inconclusive. We conducted a phase I clinical study assessing safety and clinical and peripheral immune responses after MSC therapy in MS. Seven patients with progressive MS were intravenously infused with a single dose of autologous MSC (1–2 × 106 MSCs/kg body weight). The infusions were safe and well tolerated when given during clinical remission. Five out of seven patients completed the follow up of 48 weeks post-infusion. Brain magnetic resonance imaging (MRI) showed the absence of new T2 lesions at 12 weeks in 5/6 patients, while 3/5 had accumulated new T2 lesions at 48 weeks. Patient expanded disability status scales (EDSS) were stable in 6/6 at 12 weeks but declined in 3/5 patients at 48 weeks. Early changes of circulating microRNA levels (2 h) and increased proportion of FOXP3+ Tregs were detected at 7 days post-infusion compared to baseline levels. In conclusion, MSC therapy was safe and well tolerated and is associated with possible transient beneficial clinical and peripheral immunotolerogenic effects.
Collapse
|
8
|
Liu J, Zhang L, Liu M. Mechanisms supporting potential use of bone marrow-derived mesenchymal stem cells in psychocardiology. Am J Transl Res 2019; 11:6717-6738. [PMID: 31814884 PMCID: PMC6895510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/25/2019] [Indexed: 06/10/2023]
Abstract
Despite great efforts made in recent years, globally cardiovascular disease (CVD) remains the most common and devastating disease. Pharmacological, interventional and surgical treatments have proved to be only partly satisfactory for the majority of patients. A major underlying cause of poor prognosis is a high comorbidity rate between CVD and mental illness, which calls for the approaches of psychocardiology. As psychiatric disorders and CVD can influence each other bidirectionally, it is necessary to develop novel therapies targeting both systems simultaneously. Therefore, innovative stem cell (SC) therapy has become the most promising treatment strategy in psychocardiology. Bone marrow-derived mesenchymal stem/stromal cells (BM-MSCs), among all different types of SCs, have drawn the most attention due to unique advantages in terms of ethical considerations, low immunogenicity and simplicity of preparation. In this review, we survey recent publications and clinical trials to summarize the knowledge and progress gained so far. Moreover, we discuss the feasibility of the clinical application of BM-MSCs in the area of psychocardiology.
Collapse
Affiliation(s)
- Jianyang Liu
- Department of Cardiology, Beijing Anzhen Hospital Affiliated to Capital Medical University Beijing 100029, China
| | - Lijun Zhang
- Department of Cardiology, Beijing Anzhen Hospital Affiliated to Capital Medical University Beijing 100029, China
| | - Meiyan Liu
- Department of Cardiology, Beijing Anzhen Hospital Affiliated to Capital Medical University Beijing 100029, China
| |
Collapse
|
9
|
Pedrosa M, Gomes J, Laranjeira P, Duarte C, Pedreiro S, Antunes B, Ribeiro T, Santos F, Martinho A, Fardilha M, Domingues MR, Abecasis M, P da Silva JA, Paiva A. Immunomodulatory effect of human bone marrow-derived mesenchymal stromal/stem cells on peripheral blood T cells from rheumatoid arthritis patients. J Tissue Eng Regen Med 2019; 14:16-28. [PMID: 31502378 DOI: 10.1002/term.2958] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/04/2019] [Accepted: 09/03/2019] [Indexed: 12/18/2022]
Abstract
Rheumatoid arthritis (RA) is a Th1/Th17-mediated autoimmune disease whose current treatment, consisting in the blockage of inflammatory cytokines by disease-modifying antirheumatic drugs, is not effective for all patients. The therapeutic potential of mesenchymal stromal/stem cells' (MSCs) immunomodulatory properties is being explored in RA. Here, we investigate the effect of human bone marrow (BM)-MSCs on the expression of cytokines involved in RA physiopathology by the distinct functional compartments of CD4+ and CD8+ T cells from RA patients. Peripheral blood mononuclear cells from healthy individuals (n = 6) and RA patients (n = 12) were stimulated with phorbol myristate acetate plus ionomycin and cultured in the presence/absence of BM-MSCs. The expression of (interleukin) IL-2, tumor necrosis factor alpha (TNF-α), and interferon-gamma (IFN-γ) was evaluated in naive, central memory, effector memory, and effector CD4+ and CD8+ T cells, whereas IL-6, IL-9, and IL-17 expression was measured in total CD4+ and CD8+ T cells. mRNA expression of IL-4, IL-10, transforming growth factor beta (TGF-β), cytotoxic T-lymphocyte-associated antigen 4, and/or forkhead box P3 was quantified in fluorescence-activated cell sorting-purified CD4+ T cells, CD8+ T cells, and CD4+ Treg. BM-MSCs inhibited the production of TNF-α, IL-17, IL-6, IL-2, IFN-γ, and IL-9 by T cells from RA patients, mainly by reducing the percentage of cells producing cytokines. This inhibitory effect was transversal to all T cell subsets analyzed. At mRNA level, BM-MSCs increased expression of IL-10 and TGF-β by CD4+ and CD8+ T cells. BM-MSCs displayed a striking inhibitory action over T cells from RA patients, reducing the expression of cytokines involved in RA physiopathology. Remarkably, BM-MSC-derived immunomodulation affected either naive, effector, and memory T cells.
Collapse
Affiliation(s)
- Mónia Pedrosa
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal.,Signal Transduction Laboratory, Center of Cellular Biology, SACS and Department of Biology, University of Aveiro, Aveiro, Portugal
| | - Joana Gomes
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal.,Mass Spectrometry Center, QOPNA, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Paula Laranjeira
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Cátia Duarte
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal.,Rheumatology Department, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Susana Pedreiro
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal
| | | | - Tânia Ribeiro
- Cell2B Advanced Therapeutics, SA, Cantanhede, Portugal
| | - Francisco Santos
- Cell2B Advanced Therapeutics, SA, Cantanhede, Portugal.,Stemlab SA, Cantanhede, Portugal
| | - António Martinho
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal
| | - Margarida Fardilha
- Signal Transduction Laboratory, Center of Cellular Biology, SACS and Department of Biology, University of Aveiro, Aveiro, Portugal.,Laboratory of Signal Transduction, Institute of Biomedicine-iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - M Rosário Domingues
- Mass Spectrometry Center, QOPNA, Department of Chemistry, University of Aveiro, Aveiro, Portugal.,Departament of Biology & CESAM, University of Aveiro, Aveiro, Portugal
| | - Manuel Abecasis
- Serviço de Transplantação de Progenitores Hematopoiéticos (UTM), Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa, Portugal
| | - José António P da Silva
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal.,Rheumatology Department, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Artur Paiva
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal.,Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Instituto Politecnico de Coimbra, ESTESC-Coimbra Health School, Ciencias Biomedicas Laboratoriais, Coimbra, Portugal
| |
Collapse
|
10
|
Abdi J, Rastgoo N, Chen Y, Chen GA, Chang H. Ectopic expression of BIRC5-targeting miR-101-3p overcomes bone marrow stroma-mediated drug resistance in multiple myeloma cells. BMC Cancer 2019; 19:975. [PMID: 31638931 PMCID: PMC6805455 DOI: 10.1186/s12885-019-6151-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 09/11/2019] [Indexed: 12/27/2022] Open
Abstract
Background Multiple myeloma (MM) cells gain protection against drugs through interaction with bone marrow stromal cells (BMSCs). This form of resistance largely accounts for resistance to therapy in MM patients which warrants further exploration to identify more potential therapeutic targets. Methods We performed miRNA/mRNA qPCR arrays and western blotting to analyze transcriptional and translational changes in MM cells co-cultured with BMSCs. Drug cytotoxicity and apoptosis in MMGFP-BMSC co-cultures were measured using fluorescence plate reader and flowcytometry, respectively. miRNA was overexpressed in MM cell lines using Lentiviral transduction, miRNA-3’UTR binding was examined using luciferase assay. Results We found that BMSCs downregulated miR-101-3p and upregulated survivin (BIRC5) in MM cells. Survivin was downregulated by miR-101-3p overexpression and found to be a direct target of miR-101-3p using 3’UTR luciferase assay. Overexpression of survivin increased viability of MM cells in the presence of anti-myeloma drugs, and miR-101-3p inhibition by anti-miR against miR-101-3p upregulated survivin. Furthermore, overexpression of miR-101-3p or silencing of survivin triggered apoptosis in MM cells and sensitized them to anti-myeloma drugs in the presence of BMSCs overcoming the stroma-induced drug resistance. Conclusions Our study demonstrates that BMSC-induced resistance to drugs is associated with survivin upregulation which is a direct target of miR-101-3p. This study also identifies miR-101-3p-survivin interaction as a druggable target involved in stroma-mediated drug resistance in MM and suggests it for developing more efficient therapeutic strategies.
Collapse
Affiliation(s)
- Jahangir Abdi
- Dept. of Laboratory Hematology, Laboratory Medicine Program, Toronto General Hospital, University Health Network, 200 Elizabeth Street, 11E-413, Toronto, Ontario, M5G 2C4, Canada
| | - Nasrin Rastgoo
- Dept. of Laboratory Hematology, Laboratory Medicine Program, Toronto General Hospital, University Health Network, 200 Elizabeth Street, 11E-413, Toronto, Ontario, M5G 2C4, Canada
| | - Yan Chen
- Dept. of Laboratory Hematology, Laboratory Medicine Program, Toronto General Hospital, University Health Network, 200 Elizabeth Street, 11E-413, Toronto, Ontario, M5G 2C4, Canada
| | - Guo An Chen
- Department of Hematology/Oncology, First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Hong Chang
- Dept. of Laboratory Hematology, Laboratory Medicine Program, Toronto General Hospital, University Health Network, 200 Elizabeth Street, 11E-413, Toronto, Ontario, M5G 2C4, Canada. .,Dept. of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Canada.
| |
Collapse
|
11
|
Guan YQ, Zhao CS, Zou HQ, Yan XM, Luo LL, Wu JL, Li X, Zhang YA. Aging, rather than Parkinson's disease, affects the responsiveness of PBMCs to the immunosuppression of bone marrow mesenchymal stem cells. Mol Med Rep 2018; 19:165-176. [PMID: 30483752 PMCID: PMC6297737 DOI: 10.3892/mmr.2018.9670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 10/19/2018] [Indexed: 01/22/2023] Open
Abstract
Whether aging or Parkinson's disease (PD) affects the responses of peripheral blood mononuclear cells (PBMCs) to immunosuppression by bone marrow‑derived mesenchymal stem cell (BM‑MSCs) and which cytokines are more effective in inducing BM‑MSCs to be immunosuppressive remains to be elucidated. PBMCs were isolated from healthy young (age 26‑35), healthy middle‑aged (age 56‑60) and middle‑aged PD‑affected individuals. All the recruits were male. The mitogen‑stimulated PBMCs and proinflammatory cytokine‑pretreated BM‑MSCs were co‑cultured. The PBMC proliferation was measured using Cell Counting Kit‑8, while the cytokine secretion was assayed by cytometric bead array technology. The immunosuppressive ability of BM‑MSCs was confirmed in young healthy, middle‑aged healthy and middle‑aged PD‑affected individuals. Among the three groups, the PBMC proliferation and cytokine secretion of the young healthy group were suppressed more significantly compared with those of the middle‑aged healthy and middle‑aged PD‑affected group. No significant differences were identified in the PBMC proliferation and cytokine secretion between the patients with PD and the middle‑aged healthy subjects. Interferon (IFN)‑γ synergized with tumor necrosis factor (TNF)‑α, interleukin (IL)‑1α or IL‑1β was more effective than either one alone, and the combinations of IFN‑γ + IL‑1α and IFN‑γ + IL‑1β were more effective than IFN‑γ + TNF‑α in inducing BM‑MSCs to inhibit PBMC proliferation. The results of the present study suggested that aging, rather than PD, affects the response of PBMCs toward the suppression of BM‑MSC, at least in middle‑aged males. Patients with PD aged 56‑60 remain eligible for anti‑inflammatory BM‑MSC‑based therapy. Treatment of BM‑MSCs with IFN‑γ + IL‑1α or IFN‑γ + IL‑1β prior to transplantation may result in improved immunosuppressive effects.
Collapse
Affiliation(s)
- Yun-Qian Guan
- Department of Cell Biology, Key laboratory of Ministry of Education, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Chun-Song Zhao
- Department of Cell Biology, Key laboratory of Ministry of Education, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Hai-Qiang Zou
- Department of Neurology, The General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China
| | - Xiao-Ming Yan
- Department of Function Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Lu-Lu Luo
- Department of Neurology, Northern Jiangsu People's Hospital, Clinical Medical School of Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China
| | - Jia-Lin Wu
- Department of Neurology, Northern Jiangsu People's Hospital, Clinical Medical School of Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China
| | - Xiaobo Li
- Department of Neurology, Northern Jiangsu People's Hospital, Clinical Medical School of Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China
| | - Yu Alex Zhang
- Department of Cell Biology, Key laboratory of Ministry of Education, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| |
Collapse
|
12
|
Liu HM, Lin R, Song Y, Liu WT. Adoptive immunotherapy for autoimmune hepatitis. Shijie Huaren Xiaohua Zazhi 2018; 26:1434-1438. [DOI: 10.11569/wcjd.v26.i24.1434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Autoimmune hepatitis (AIH) is a chronic inflammatory liver disease caused by an autoimmune disorder, which has attracted more and more attention due to its unique clinical and pathological features. European epidemiological data show that the incidence of AIH is increasing year by year, but its pathogenesis is not yet clear, and the targeted treatments are limited. Immunotherapy of autoimmune hepatitis has been extensively studied in recent years, especially in the area of adoptive immunotherapy. In this paper, we summarize the functional mechanism and clinical applications of adoptive immunotherapy with different kinds of immunocompetent cells in the treatment of AIH.
Collapse
Affiliation(s)
- Hui-Min Liu
- Department of Gastroenterology, the General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Rui Lin
- Department of Gastroenterology, the General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Yan Song
- Department of Gastroenterology, the General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Wen-Tian Liu
- Department of Gastroenterology, the General Hospital of Tianjin Medical University, Tianjin 300052, China
| |
Collapse
|
13
|
Li J, Yawno T, Sutherland AE, Gurung S, Paton M, McDonald C, Tiwari A, Pham Y, Castillo-Melendez M, Jenkin G, Miller SL. Preterm umbilical cord blood derived mesenchymal stem/stromal cells protect preterm white matter brain development against hypoxia-ischemia. Exp Neurol 2018; 308:120-131. [PMID: 30012511 DOI: 10.1016/j.expneurol.2018.07.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/16/2018] [Accepted: 07/12/2018] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Preterm infants are at high risk for white matter injury and subsequent neurodevelopmental impairments. Mesenchymal stem/stromal cells (MSC) have anti-inflammatory/immunomodulatory actions and are of interest for neural repair in adults and newborns. This study examined the neuroprotective effects of allogeneic MSC, derived from preterm umbilical cord blood (UCB), in a preterm sheep model of white matter injury. METHODS Quad-lineage differentiation, clonogenicity and self-renewal ability of UCB-derived MSC were confirmed. Chronically instrumented fetal sheep (0.7 gestation) received either 25 min hypoxia-ischemia (HI) to induce preterm brain injury, or sham-HI. Ten million MSC, or saline, were administered iv to fetuses at 12 h after HI. Fetal brains were collected 10d after HI for histopathology and immunocytochemistry. RESULTS HI induced white matter injury, as indicated by a reduction in CNPase-positive myelin fiber density. HI also induced microglial activation (Iba-1) in the periventricular white matter and internal capsule (P < .05 vs control). MSC administration following HI preserved myelination (P < .05), modified microglial activation, and promoted macrophage migration (CD163) and cell proliferation (Ki-67) within cerebral white matter (P < .05). Cerebral CXCL10 concentration was increased following MSC administration (P < .05), which was likely associated with macrophage migration and cell proliferation within the preterm brain. Additionally, MSC administration reduced systemic pro-inflammatory cytokine TNFα at 3d post-HI (P < .05). CONCLUSIONS UCB-derived MSC therapy preserved white matter brain structure following preterm HI, mediated by a suppression of microglial activation, promotion of macrophage migration and acceleration of self-repair within the preterm brain. UCB-derived MSC are neuroprotective, acting via peripheral and cerebral anti-inflammatory and immunomodulatory mechanisms.
Collapse
Affiliation(s)
- Jingang Li
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Tamara Yawno
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Amy E Sutherland
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Shanti Gurung
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Madison Paton
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Courtney McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Abhilasha Tiwari
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Yen Pham
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | | | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
14
|
Scutera S, Salvi V, Lorenzi L, Piersigilli G, Lonardi S, Alotto D, Casarin S, Castagnoli C, Dander E, D'Amico G, Sozzani S, Musso T. Adaptive Regulation of Osteopontin Production by Dendritic Cells Through the Bidirectional Interaction With Mesenchymal Stromal Cells. Front Immunol 2018; 9:1207. [PMID: 29910810 PMCID: PMC5992779 DOI: 10.3389/fimmu.2018.01207] [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: 12/07/2017] [Accepted: 05/14/2018] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) exert immunosuppressive effects on immune cells including dendritic cells (DCs). However, many details of the bidirectional interaction of MSCs with DCs are still unsolved and information on key molecules by which DCs can modulate MSC functions is limited. Here, we report that osteopontin (OPN), a cytokine involved in homeostatic and pathophysiologic responses, is constitutively expressed by DCs and regulated in the DC/MSC cocultures depending on the activation state of MSCs. Resting MSCs promoted OPN production, whereas the production of OPN was suppressed when MSCs were activated by proinflammatory cytokines (i.e., TNF-α, IL-6, and IL-1β). OPN induction required cell-to-cell contact, mediated at least in part, by β1 integrin (CD29). Conversely, activated MSCs inhibited the release of OPN via the production of soluble factors with a major role played by Prostaglandin E2 (PGE2). Accordingly, pretreatment with indomethacin significantly abrogated the MSC-mediated suppression of OPN while the direct addition of exogenous PGE2 inhibited OPN production by DCs. Furthermore, DC-conditioned medium promoted osteogenic differentiation of MSCs with a concomitant inhibition of adipogenesis. These effects were paralleled by the repression of the adipogenic markers PPARγ, adiponectin, and FABP4, and induction of the osteogenic markers alkaline phosphatase, RUNX2, and of the bone-anabolic chemokine CCL5. Notably, blocking OPN activity with RGD peptides or with an antibody against CD29, one of the OPN receptors, prevented the effects of DC-conditioned medium on MSC differentiation and CCL5 induction. Because MSCs have a key role in maintenance of bone marrow (BM) hematopoietic stem cell niche through reciprocal regulation with immune cells, we investigated the possible MSC/DC interaction in human BM by immunohistochemistry. Although DCs (CD1c+) are a small percentage of BM cells, we demonstrated colocalization of CD271+ MSCs with CD1c+ DCs in normal and myelodysplastic BM. OPN reactivity was observed in occasional CD1c+ cells in the proximity of CD271+ MSCs. Altogether, these results candidate OPN as a signal modulated by MSCs according to their activation status and involved in DC regulation of MSC differentiation.
Collapse
Affiliation(s)
- Sara Scutera
- Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Valentina Salvi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luisa Lorenzi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giorgia Piersigilli
- Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Silvia Lonardi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Daniela Alotto
- Skin Bank, Department of General and Specialized Surgery, A.O.U. Citta della Salute e della Scienza di Torino, Turin, Italy
| | - Stefania Casarin
- Skin Bank, Department of General and Specialized Surgery, A.O.U. Citta della Salute e della Scienza di Torino, Turin, Italy
| | - Carlotta Castagnoli
- Skin Bank, Department of General and Specialized Surgery, A.O.U. Citta della Salute e della Scienza di Torino, Turin, Italy
| | - Erica Dander
- "M. Tettamanti" Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy
| | - Giovanna D'Amico
- "M. Tettamanti" Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy
| | - Silvano Sozzani
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Tiziana Musso
- Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| |
Collapse
|
15
|
Stem cell therapy for abrogating stroke-induced neuroinflammation and relevant secondary cell death mechanisms. Prog Neurobiol 2017; 158:94-131. [PMID: 28743464 DOI: 10.1016/j.pneurobio.2017.07.004] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/13/2022]
Abstract
Ischemic stroke is a leading cause of death worldwide. A key secondary cell death mechanism mediating neurological damage following the initial episode of ischemic stroke is the upregulation of endogenous neuroinflammatory processes to levels that destroy hypoxic tissue local to the area of insult, induce apoptosis, and initiate a feedback loop of inflammatory cascades that can expand the region of damage. Stem cell therapy has emerged as an experimental treatment for stroke, and accumulating evidence supports the therapeutic efficacy of stem cells to abrogate stroke-induced inflammation. In this review, we investigate clinically relevant stem cell types, such as hematopoietic stem cells (HSCs), mesenchymal stem cells (MSCs), endothelial progenitor cells (EPCs), very small embryonic-like stem cells (VSELs), neural stem cells (NSCs), extraembryonic stem cells, adipose tissue-derived stem cells, breast milk-derived stem cells, menstrual blood-derived stem cells, dental tissue-derived stem cells, induced pluripotent stem cells (iPSCs), teratocarcinoma-derived Ntera2/D1 neuron-like cells (NT2N), c-mycER(TAM) modified NSCs (CTX0E03), and notch-transfected mesenchymal stromal cells (SB623), comparing their potential efficacy to sequester stroke-induced neuroinflammation and their feasibility as translational clinical cell sources. To this end, we highlight that MSCs, with a proven track record of safety and efficacy as a transplantable cell for hematologic diseases, stand as an attractive cell type that confers superior anti-inflammatory effects in stroke both in vitro and in vivo. That stem cells can mount a robust anti-inflammatory action against stroke complements the regenerative processes of cell replacement and neurotrophic factor secretion conventionally ascribed to cell-based therapy in neurological disorders.
Collapse
|
16
|
Contreras-Kallens P, Terraza C, Oyarce K, Gajardo T, Campos-Mora M, Barroilhet MT, Álvarez C, Fuentes R, Figueroa F, Khoury M, Pino-Lagos K. Mesenchymal stem cells and their immunosuppressive role in transplantation tolerance. Ann N Y Acad Sci 2017; 1417:35-56. [PMID: 28700815 DOI: 10.1111/nyas.13364] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 03/13/2017] [Accepted: 03/29/2017] [Indexed: 12/23/2022]
Abstract
Since they were first described, mesenchymal stem cells (MSCs) have been shown to have important effector mechanisms and the potential for use in cell therapy. A great deal of research has been focused on unveiling how MSCs contribute to anti-inflammatory responses, including describing several cell populations involved and identifying soluble and other effector molecules. In this review, we discuss some of the contemporary evidence for use of MSCs in the field of immune tolerance, with a special emphasis on transplantation. Although considerable effort has been devoted to understanding the biological function of MSCs, additional resources are required to clarify the mechanisms of their induction of immune tolerance, which will undoubtedly lead to improved clinical outcomes for MSC-based therapies.
Collapse
Affiliation(s)
- Pamina Contreras-Kallens
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Claudia Terraza
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Karina Oyarce
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Tania Gajardo
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Mauricio Campos-Mora
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - María Teresa Barroilhet
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Carla Álvarez
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Ricardo Fuentes
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Fernando Figueroa
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Maroun Khoury
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Cells for Cells, Santiago, Chile.,Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Karina Pino-Lagos
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| |
Collapse
|
17
|
de Wolf C, van de Bovenkamp M, Hoefnagel M. Regulatory perspective on in vitro potency assays for human mesenchymal stromal cells used in immunotherapy. Cytotherapy 2017; 19:784-797. [PMID: 28457740 DOI: 10.1016/j.jcyt.2017.03.076] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/10/2017] [Accepted: 03/21/2017] [Indexed: 12/20/2022]
Abstract
Mesenchymal stromal cells (MSCs) are multipotent cells derived from various tissues that can differentiate into several cell types. MSCs are able to modulate the response of immune cells of the innate and adaptive immune system. Because of these multimodal properties, the potential use of MSCs for immunotherapies is currently explored in various clinical indications. Due to the diversity of potential MSC medicinal products at the level of cell source, manufacturing process and indication, distinct functionality tests may be needed to ensure the quality for each of the different products. In this review, we focus on in vitro potency assays proposed for characterization and release of different MSC medicinal products. We discuss the most used functional assays, as presented in scientific advices and literature, highlighting specific advantages and limitations of the various assays. Currently, the most proposed and accepted potency assay for release is based on in vitro inhibition of T cell proliferation or other functionalities. However, for some products, assays based on other MSC or responder cell properties may be more appropriate. In all cases, the biological relevance of the proposed assay for the intended clinical activity should be substantiated with appropriate product-specific (non-)clinical data. In case practical considerations prevent the use of the ideal potency assay at release, use of a surrogate marker or test could be considered if correlation with functionality has been demonstrated. Nevertheless, as the field of MSC immunology is evolving, improvements can be expected in relevant assays and consequently in guidance related to potency testing.
Collapse
|
18
|
Lee HJ, Kim SN, Jeon MS, Yi T, Song SU. ICOSL expression in human bone marrow-derived mesenchymal stem cells promotes induction of regulatory T cells. Sci Rep 2017; 7:44486. [PMID: 28290526 PMCID: PMC5349520 DOI: 10.1038/srep44486] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 02/09/2017] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can modulate lymphocyte proliferation and function. One of the immunomodulatory functions of MSCs involves CD4+CD25+FoxP3+ regulatory T cells (Tregs), which negatively regulate inflammatory responses. MSC-mediated Treg induction is supposed to be regulated by mechanisms requiring both soluble and cell contact-dependent factors. Although the involvement of soluble factors has been revealed, the contact-dependent mechanisms in MSC-mediated Treg induction remain unclear. We attempted to identify molecule(s) other than secreted factors that are responsible for MSC-mediated Treg induction and to uncover the underlying mechanisms. Under in vitro Treg-inducing conditions, ICOSL expression in MSCs coincided with Treg induction in co-cultures of MSCs with CD4+ T cells. When cultured in a transwell plate, MSCs failed to induce Tregs. Neutralization or knockdown of ICOSL significantly reduced Tregs and their IL-10 release. ICOSL overexpression in MSCs promoted induction of functional Tregs. ICOSL-ICOS signaling promoted Treg differentiation from CD4+ T cells through activation of the phosphoinositide 3-kinase-Akt pathway. MSCs primed with Interleukin-1β significantly induced Tregs through ICOSL upregulation. We demonstrated that the Treg-inducing activity of MSCs is proportionate to their basal ICOSL expression. This study provides evidence that ICOSL expression in human MSCs plays an important role in contact-dependent regulation of MSC-mediated Treg induction.
Collapse
Affiliation(s)
- Hyun-Joo Lee
- Translational Research Center, Inha University School of Medicine, Incheon, Republic of Korea.,Drug Development Program, Department of Biomedical Sciences, Inha University School of Medicine, Incheon, Republic of Korea
| | - Si-Na Kim
- Translational Research Center, Inha University School of Medicine, Incheon, Republic of Korea.,Drug Development Program, Department of Biomedical Sciences, Inha University School of Medicine, Incheon, Republic of Korea.,SCM Lifesciences Co. Ltd., Incheon, Republic of Korea
| | - Myung-Shin Jeon
- Translational Research Center, Inha University School of Medicine, Incheon, Republic of Korea
| | - TacGhee Yi
- Translational Research Center, Inha University School of Medicine, Incheon, Republic of Korea.,SunCreate Co. Ltd., Yangju, Republic of Korea
| | - Sun U Song
- Translational Research Center, Inha University School of Medicine, Incheon, Republic of Korea.,SCM Lifesciences Co. Ltd., Incheon, Republic of Korea
| |
Collapse
|
19
|
Overexpression of soluble RAGE in mesenchymal stem cells enhances their immunoregulatory potential for cellular therapy in autoimmune arthritis. Sci Rep 2016; 6:35933. [PMID: 27804999 PMCID: PMC5090969 DOI: 10.1038/srep35933] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 10/06/2016] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are attractive agents for cellular therapy in rheumatoid arthritis (RA). The receptor for advanced glycation end products (RAGE) serves as a pattern recognition receptor for endogenous inflammatory ligands. Soluble RAGE (sRAGE) is a truncated form of RAGE that functions as a decoy and acts as an anti-inflammatory molecule. The aim of this study was to determine whether sRAGE has therapeutic effects and the mechanisms active in sRAGE-overexpressing MSCs (sRAGE-MSCs) in an experimental model of RA. sRAGE-MSCs were generated by DNA transfection of human adipose tissue-derived MSCs (Ad-hMSCs). MSCs showed increased expression of VEGF, IL-1β, IL-6, and HMGB-1 under inflammatory conditions. However, sRAGE-MSCs showed significantly lower production of these proinflammatory molecules. Expression of immunomodulatory molecules such as IL-10, TGF-β, and indoleamine 2, 3-dioxygenase was higher in sRAGE-MSCs than in mock-MSCs. sRAGE-MSCs showed enhanced migration potential. Transplantation of sRAGE-MSCs into arthritic IL-1Ra-knockout mice markedly suppressed inflammatory arthritis, decreased Th17 cells, and reciprocally increased regulatory T cells. The differentiation of IFN-γ+CD4+ and IL-17+CD4+ cells was inhibited by incubation with sRAGE-MSCs compared with mock-MSCs. These findings suggest that sRAGE overexpression in Ad-hMSCs optimizes their immunoregulatory properties, which may be useful as a novel cellular therapy for RA.
Collapse
|
20
|
Najar M, Raicevic G, Fayyad-Kazan H, Bron D, Toungouz M, Lagneaux L. Mesenchymal stromal cells and immunomodulation: A gathering of regulatory immune cells. Cytotherapy 2016; 18:160-71. [PMID: 26794710 DOI: 10.1016/j.jcyt.2015.10.011] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 10/01/2015] [Accepted: 10/13/2015] [Indexed: 12/13/2022]
Abstract
Because of their well-recognized immunomodulatory properties, mesenchymal stromal cells (MSCs) represent an attractive cell population for therapeutic purposes. In particular, there is growing interest in the use of MSCs as cellular immunotherapeutics for tolerance induction in allogeneic transplantations and the treatment of autoimmune diseases. However, multiple mechanisms have been identified to mediate the immunomodulatory effects of MSCs, sometimes with several ambiguities and inconsistencies. Although published studies have mainly reported the role of soluble factors, we believe that a sizeable cellular component plays a critical role in MSC immunomodulation. We refer to these cells as regulatory immune cells, which are generated from both the innate and adaptive responses after co-culture with MSCs. In this review, we discuss the nature and role of these immune regulatory cells as well as the role of different mediators, and, in particular, regulatory immune cell induction by MSCs through interleukin-10. Once induced, immune regulatory cells accumulate and converge their regulatory pathways to create a tolerogenic environment conducive for immunomodulation. Thus, a better understanding of these regulatory immune cells, in terms of how they can be optimally manipulated and induced, would be suitable for improving MSC-based immunomodulatory therapeutic strategies.
Collapse
Affiliation(s)
- Mehdi Najar
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles, Campus Erasme, Brussels, Belgium.
| | - Gordana Raicevic
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles, Campus Erasme, Brussels, Belgium
| | - Hussein Fayyad-Kazan
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles, Campus Erasme, Brussels, Belgium
| | - Dominique Bron
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles, Campus Erasme, Brussels, Belgium
| | - Michel Toungouz
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles, Campus Erasme, Brussels, Belgium
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles, Campus Erasme, Brussels, Belgium
| |
Collapse
|
21
|
Muir P, Hans EC, Racette M, Volstad N, Sample SJ, Heaton C, Holzman G, Schaefer SL, Bloom DD, Bleedorn JA, Hao Z, Amene E, Suresh M, Hematti P. Autologous Bone Marrow-Derived Mesenchymal Stem Cells Modulate Molecular Markers of Inflammation in Dogs with Cruciate Ligament Rupture. PLoS One 2016; 11:e0159095. [PMID: 27575050 PMCID: PMC5005014 DOI: 10.1371/journal.pone.0159095] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 06/27/2016] [Indexed: 01/22/2023] Open
Abstract
Mid-substance rupture of the canine cranial cruciate ligament rupture (CR) and associated stifle osteoarthritis (OA) is an important veterinary health problem. CR causes stifle joint instability and contralateral CR often develops. The dog is an important model for human anterior cruciate ligament (ACL) rupture, where rupture of graft repair or the contralateral ACL is also common. This suggests that both genetic and environmental factors may increase ligament rupture risk. We investigated use of bone marrow-derived mesenchymal stem cells (BM-MSCs) to reduce systemic and stifle joint inflammatory responses in dogs with CR. Twelve dogs with unilateral CR and contralateral stable partial CR were enrolled prospectively. BM-MSCs were collected during surgical treatment of the unstable CR stifle and culture-expanded. BM-MSCs were subsequently injected at a dose of 2x106 BM-MSCs/kg intravenously and 5x106 BM-MSCs by intra-articular injection of the partial CR stifle. Blood (entry, 4 and 8 weeks) and stifle synovial fluid (entry and 8 weeks) were obtained after BM-MSC injection. No adverse events after BM-MSC treatment were detected. Circulating CD8+ T lymphocytes were lower after BM-MSC injection. Serum C-reactive protein (CRP) was decreased at 4 weeks and serum CXCL8 was increased at 8 weeks. Synovial CRP in the complete CR stifle was decreased at 8 weeks. Synovial IFNγ was also lower in both stifles after BM-MSC injection. Synovial/serum CRP ratio at diagnosis in the partial CR stifle was significantly correlated with development of a second CR. Systemic and intra-articular injection of autologous BM-MSCs in dogs with partial CR suppresses systemic and stifle joint inflammation, including CRP concentrations. Intra-articular injection of autologous BM-MSCs had profound effects on the correlation and conditional dependencies of cytokines using causal networks. Such treatment effects could ameliorate risk of a second CR by modifying the stifle joint inflammatory response associated with cranial cruciate ligament matrix degeneration or damage.
Collapse
Affiliation(s)
- Peter Muir
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
- * E-mail:
| | - Eric C. Hans
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Molly Racette
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Nicola Volstad
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Susannah J. Sample
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Caitlin Heaton
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Gerianne Holzman
- UW Veterinary Care Hospital, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Susan L. Schaefer
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Debra D. Bloom
- Department of Medicine, School of Medicine & Public Health, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States of America
| | - Jason A. Bleedorn
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Zhengling Hao
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Ermias Amene
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - M. Suresh
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Peiman Hematti
- Department of Medicine, School of Medicine & Public Health, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States of America
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, 53705, United States of America
| |
Collapse
|
22
|
Pleyer L, Valent P, Greil R. Mesenchymal Stem and Progenitor Cells in Normal and Dysplastic Hematopoiesis-Masters of Survival and Clonality? Int J Mol Sci 2016; 17:ijms17071009. [PMID: 27355944 PMCID: PMC4964385 DOI: 10.3390/ijms17071009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 05/20/2016] [Accepted: 06/08/2016] [Indexed: 02/07/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are malignant hematopoietic stem cell disorders that have the capacity to progress to acute myeloid leukemia (AML). Accumulating evidence suggests that the altered bone marrow (BM) microenvironment in general, and in particular the components of the stem cell niche, including mesenchymal stem cells (MSCs) and their progeny, play a pivotal role in the evolution and propagation of MDS. We here present an overview of the role of MSCs in the pathogenesis of MDS, with emphasis on cellular interactions in the BM microenvironment and related stem cell niche concepts. MSCs have potent immunomodulatory capacities and communicate with diverse immune cells, but also interact with various other cellular components of the microenvironment as well as with normal and leukemic stem and progenitor cells. Moreover, compared to normal MSCs, MSCs in MDS and AML often exhibit altered gene expression profiles, an aberrant phenotype, and abnormal functional properties. These alterations supposedly contribute to the “reprogramming” of the stem cell niche into a disease-permissive microenvironment where an altered immune system, abnormal stem cell niche interactions, and an impaired growth control lead to disease progression. The current article also reviews molecular targets that play a role in such cellular interactions and possibilities to interfere with abnormal stem cell niche interactions by using specific targeted drugs.
Collapse
Affiliation(s)
- Lisa Pleyer
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology & Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Richard Greil
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| |
Collapse
|
23
|
Ulusoy C, Zibandeh N, Yıldırım S, Trakas N, Zisimopoulou P, Küçükerden M, Tașlı H, Tzartos S, Göker K, Tüzün E, Akkoç T. Dental follicle mesenchymal stem cell administration ameliorates muscle weakness in MuSK-immunized mice. J Neuroinflammation 2015; 12:231. [PMID: 26646841 PMCID: PMC4673854 DOI: 10.1186/s12974-015-0451-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 12/04/2015] [Indexed: 01/09/2023] Open
Abstract
Background Myasthenia gravis (MG) is an antibody-mediated autoimmune disease of the neuromuscular junction (NMJ), mostly associated with acetylcholine receptor (AChR) antibodies. Around 5–10 % of MG patients show antibodies to muscle-specific tyrosine kinase (MuSK). Mesenchymal stem cell (MSC) administration has been shown to ameliorate muscle weakness in the experimental autoimmune myasthenia gravis (EAMG) model induced by AChR immunization. Methods To investigate the efficacy of stem cell treatment in MuSK-related EAMG, clinical and immunological features of MuSK-immunized mice with or without dental follicle MSC (DFMSC) treatment were compared. Results MuSK-immunized mice intravenously treated with DFMSC after second and third immunizations showed significantly lower EAMG incidence and severity and reduced serum anti-MuSK antibody, NMJ IgG, and C3 deposit levels and CD11b+ lymph node cell ratios. Moreover, lymph node cells of DFMSC-administered mice showed reduced proliferation and IL-6 and IL-12 production responses to MuSK stimulation. By contrast, proportions of B and T cell populations and production of a wide variety of cytokines were not affected from DFMSC treatment. Conclusions Our results suggest that DFMSC treatment shows its beneficial effects mostly through suppression of innate immune system, whereas other immune functions appear to be preserved. Stem cell treatment might thus constitute a specific and effective treatment method in MuSK-associated MG. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0451-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Canan Ulusoy
- Department of Neuroscience, Institute for Experimental Medical Research (DETAE), Istanbul University, Istanbul, Turkey
| | - Noushin Zibandeh
- Division of Pediatric Allergy and Immunology, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - Selin Yıldırım
- Division of Pediatric Allergy and Immunology, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - Nikolaos Trakas
- Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | | | - Melike Küçükerden
- Department of Neuroscience, Institute for Experimental Medical Research (DETAE), Istanbul University, Istanbul, Turkey
| | - Hatice Tașlı
- Department of Neuroscience, Institute for Experimental Medical Research (DETAE), Istanbul University, Istanbul, Turkey
| | - Socrates Tzartos
- Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Kamil Göker
- Department of Oral and Maxillofacial Surgery, Marmara University Faculty of Dentistry, Istanbul, Turkey
| | - Erdem Tüzün
- Department of Neuroscience, Institute for Experimental Medical Research (DETAE), Istanbul University, Istanbul, Turkey. .,Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Capa, Turkey.
| | - Tunç Akkoç
- Division of Pediatric Allergy and Immunology, Marmara University Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
24
|
Mattar P, Bieback K. Comparing the Immunomodulatory Properties of Bone Marrow, Adipose Tissue, and Birth-Associated Tissue Mesenchymal Stromal Cells. Front Immunol 2015; 6:560. [PMID: 26579133 PMCID: PMC4630659 DOI: 10.3389/fimmu.2015.00560] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 10/19/2015] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal cells (MSC) have gained immense attraction in regenerative medicine, tissue engineering, and immunotherapy. This is based on their differentiation potential and the supply of pro-regenerative and immunomodulatory signals. MSC can be isolated from a multitude of tissue sources, but mainly bone marrow, adipose tissue, and birth-associated tissues (e.g., umbilical cord, cord blood, placenta) appear to be relevant for clinical translation in immune-mediated disorders. However, only a few studies directly compared the immunomodulatory potency of MSC from different tissue sources. This review compiles the current literature regarding the similarities and differences between these three sources for MSCs with a special focus on their immunomodulatory effects on T-lymphocyte subsets and monocytes, macrophages, and dendritic cells.
Collapse
Affiliation(s)
- Philipp Mattar
- Stem Cell Laboratory, Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University , Heidelberg , Germany ; German Red Cross Blood Service Baden-Württemberg - Hessen , Mannheim , Germany
| | - Karen Bieback
- Stem Cell Laboratory, Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University , Heidelberg , Germany ; German Red Cross Blood Service Baden-Württemberg - Hessen , Mannheim , Germany
| |
Collapse
|