1
|
Li YR, Fang Y, Niu S, Chen Y, Lyu Z, Yang L. Managing allorejection in off-the-shelf CAR-engineered cell therapies. Mol Ther 2024:S1525-0016(24)00762-7. [PMID: 39600090 DOI: 10.1016/j.ymthe.2024.11.035] [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: 09/25/2024] [Revised: 10/25/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
Chimeric antigen receptor (CAR)-engineered T (CAR-T) cell therapy has revolutionized the treatment of various diseases, including cancers and autoimmune disorders. However, all US Food and Drug Administration (FDA)-approved CAR-T cell therapies are autologous, and their widespread clinical application is limited by several challenges, such as complex individualized manufacturing, high costs, and the need for patient-specific selection. Allogeneic off-the-shelf CAR-engineered cell therapy offers promising potential due to its immediate availability, consistent quality, potency, and scalability in manufacturing. Nonetheless, significant challenges, including the risks of graft-versus-host disease (GvHD) and host-cell-mediated allorejection, must be addressed. Strategies such as knocking out endogenous T cell receptors (TCRs) or using alternative therapeutic cells with low GvHD risk have shown promise in clinical trials aimed at reducing GvHD. However, mitigating allorejection remains critical for ensuring the long-term sustainability and efficacy of off-the-shelf cell products. In this review, we discuss the immunological basis of allorejection in CAR-engineered therapies and explore various strategies to overcome this challenge. We also highlight key insights from recent clinical trials, particularly related to the sustainability and immunogenicity of allogeneic CAR-engineered cell products, and address manufacturing considerations aimed at minimizing allorejection and optimizing the efficacy of this emerging therapeutic approach.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Ying Fang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Siyue Niu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yuning Chen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zibai Lyu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
2
|
Haider KH. Priming mesenchymal stem cells to develop "super stem cells". World J Stem Cells 2024; 16:623-640. [PMID: 38948094 PMCID: PMC11212549 DOI: 10.4252/wjsc.v16.i6.623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/04/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
The stem cell pre-treatment approaches at cellular and sub-cellular levels encompass physical manipulation of stem cells to growth factor treatment, genetic manipulation, and chemical and pharmacological treatment, each strategy having advantages and limitations. Most of these pre-treatment protocols are non-combinative. This editorial is a continuum of Li et al's published article and Wan et al's editorial focusing on the significance of pre-treatment strategies to enhance their stemness, immunoregulatory, and immunosuppressive properties. They have elaborated on the intricacies of the combinative pre-treatment protocol using pro-inflammatory cytokines and hypoxia. Applying a well-defined multi-pronged combinatorial strategy of mesenchymal stem cells (MSCs), pre-treatment based on the mechanistic understanding is expected to develop "Super MSCs", which will create a transformative shift in MSC-based therapies in clinical settings, potentially revolutionizing the field. Once optimized, the standardized protocols may be used with slight modifications to pre-treat different stem cells to develop "super stem cells" with augmented stemness, functionality, and reparability for diverse clinical applications with better outcomes.
Collapse
Affiliation(s)
- Khawaja Husnain Haider
- Department of Basic Sciences, Sulaiman AlRajhi University, AlQaseem 52736, Saudi Arabia.
| |
Collapse
|
3
|
Katzerke C, Schaffrath J, Lützkendorf J, Janssen M, Merbach AK, Nerger K, Binder M, Baum C, Lauer K, Rohde C, Willscher E, Müller-Tidow C, Müller LP. Reduced proliferation of bone marrow MSC after allogeneic stem cell transplantation is associated with clinical outcome. Blood Adv 2023; 7:2811-2824. [PMID: 36763527 PMCID: PMC10279553 DOI: 10.1182/bloodadvances.2022008510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/30/2022] [Accepted: 01/02/2023] [Indexed: 02/11/2023] Open
Abstract
Engraftment and differentiation of donor hematopoietic stem cells is decisive for the clinical success of allogeneic stem cell transplantation (alloSCT) and depends on the recipient's bone marrow (BM) niche. A damaged niche contributes to poor graft function after alloSCT; however, the underlying mechanisms and the role of BM multipotent mesenchymal stromal cells (MSC) are ill-defined. Upon multivariate analysis in 732 individuals, we observed a reduced presence of proliferation-capable MSC in BM aspirates from patients (N = 196) who had undergone alloSCT. This was confirmed by paired analysis in 30 patients showing a higher frequency of samples with a lack of MSC presence post-alloSCT compared with pre-alloSCT. This reduced MSC presence was associated with reduced survival of patients after alloSCT and specifically with impaired graft function. Post-alloSCT MSC showed diminished in vitro proliferation along with a transcriptional antiproliferative signature, upregulation of epithelial-mesenchymal transition and extracellular matrix pathways, and altered impact on cytokine release upon contact with hematopoietic cells. To avoid in vitro culture bias, we isolated the CD146+/CD45-/HLA-DR- BM cell fraction, which comprised the entire MSC population. The post-alloSCT isolated native CD146+MSC showed a similar reduction in proliferation capacity and shared the same antiproliferative transcriptomic signature as for post-alloSCT colony-forming unit fibroblast-derived MSC. Taken together, our data show that alloSCT confers damage to the proliferative capacity of native MSC, which is associated with reduced patient survival after alloSCT and impaired engraftment of allogeneic hematopoiesis. These data represent the basis to elucidate mechanisms of BM niche reconstitution after alloSCT and its therapeutic manipulation.
Collapse
Affiliation(s)
- Christiane Katzerke
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| | - Judith Schaffrath
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| | - Jana Lützkendorf
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| | - Maike Janssen
- Klinik für Innere Medizin V, Universitätsklinikum Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory-Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Anne-Kathrin Merbach
- Klinik für Innere Medizin V, Universitätsklinikum Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory-Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Katrin Nerger
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| | - Mascha Binder
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| | - Cornelia Baum
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| | - Kirstin Lauer
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| | - Christian Rohde
- Klinik für Innere Medizin V, Universitätsklinikum Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory-Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Edith Willscher
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| | - Carsten Müller-Tidow
- Klinik für Innere Medizin V, Universitätsklinikum Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory-Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Lutz P. Müller
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| |
Collapse
|
4
|
Gaber A, Ahmed OM, Khadrawy YA, Zoheir KMA, Abo-ELeneen RE, Alblihed MA, Elbakry AM. Mesenchymal Stem Cells and Begacestat Mitigate Amyloid-β 25-35-Induced Cognitive Decline in Rat Dams and Hippocampal Deteriorations in Offspring. BIOLOGY 2023; 12:905. [PMID: 37508337 PMCID: PMC10376406 DOI: 10.3390/biology12070905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/11/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of age-related neurodegeneration and cognitive decline. AD more commonly occurs in females than in males, so it is necessary to consider new treatments specifically targeting this population. The present study investigated the protective effects of Begacestat (γ-secretase inhibitor-953, GSI-953) and bone marrow-derived mesenchymal stem cells (BM-MSCs) during pregnancy on cognitive impairment in rat dams and neurodegeneration in offspring caused by the intracerebroventricular injection of Aβ 25-35 before pregnancy. The performances of dams injected with amyloid-β 25-35 (Aβ 25-35) during behavioral tests were significantly impaired. The offspring of Aβ 25-35-injected dams treated with BM-MSCs or GSI-953 showed a dramatically reduced number and size of activated microglial cells, enhancement in the processes length, and a decrease in the proinflammatory cytokine levels. Additionally, BM-MSC or GSI-953 therapy reduced Aβ 25-35-induced increases in tau phosphorylation and amyloid precursor protein levels in the neonates' hippocampus and elevated the lower levels of glycogen synthase kinase-3 and brain-derived neurotrophic factor; moreover, reversed Aβ 25-35-induced alterations in gene expression in the neonatal hippocampus. Finally, the treatments with BM-MSC or GSI-953 are globally beneficial against Aβ 25-35-induced brain alterations, particularly by suppressing neural inflammation, inhibiting microglial cell activation, restoring developmental plasticity, and increasing neurotrophic signaling.
Collapse
Affiliation(s)
- Asmaa Gaber
- Comparative Anatomy and Embryology Division, Department of Zoology, Faculty of Science, Beni-Suef University, Beni-Suef P.O. Box 62521, Egypt
| | - Osama M Ahmed
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, Beni-Suef P.O. Box 62521, Egypt
| | - Yasser A Khadrawy
- Medical Physiology Department, National Research Center, Giza P.O. Box 12622, Egypt
| | - Khairy M A Zoheir
- Cell Biology Department, National Research Center, Giza P.O. Box 12622, Egypt
| | - Rasha E Abo-ELeneen
- Comparative Anatomy and Embryology Division, Department of Zoology, Faculty of Science, Beni-Suef University, Beni-Suef P.O. Box 62521, Egypt
| | - Mohamed A Alblihed
- Department of Medical Microbiology, college of medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ahlam M Elbakry
- Comparative Anatomy and Embryology Division, Department of Zoology, Faculty of Science, Beni-Suef University, Beni-Suef P.O. Box 62521, Egypt
| |
Collapse
|
5
|
Gaber A, Elbakry AM, Aljarari RM, Jaber FA, Khadrawy YA, Sabry D, Abo-ELeneen RE, Ahmed OM. Bone Marrow-Derived Mesenchymal Stem Cells and γ-Secretase Inhibitor Treatments Suppress Amyloid- β25-35-Induced Cognitive Impairment in Rat Dams and Cortical Degeneration in Offspring. Stem Cells Int 2023; 2023:2690949. [PMID: 37274020 PMCID: PMC10234728 DOI: 10.1155/2023/2690949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 04/02/2023] [Accepted: 05/02/2023] [Indexed: 06/06/2023] Open
Abstract
Alzheimer's disease (AD) is the most frequent cause of age-related neurodegeneration and ensuing cognitive impairment. Progressive deposition of extracellular amyloid beta (Aβ) aggregates (plaques) and intracellular hyperphosphorylated Tau protein (p-Tau) are the core pathological markers of AD but may precede clinical symptoms by many years, presenting a therapeutic window of opportunity. Females are more frequently afflicted by AD than males, necessitating evaluation of novel treatments for the female population. The current study examined the protective efficacies of intravenous bone marrow-derived mesenchymal stem cells (BM-MSCs) and oral gamma-secretase inhibitor-953 (GSI-953) during pregnancy on cognitive impairment in rat dams and neurodegeneration in offspring induced by intracerebroventricular injection of Aβ25-35 prior to pregnancy. The Aβ25-35 (AD) group exhibited significant (P < 0.001) impairments in the Y-maze and novel object recognition test performance prior to conception. Histological analysis of the offspring cortex revealed substantial dendritic shrinkage and activation of microglial cells, while neurochemical analysis demonstrated significant increases in the proinflammatory cytokine interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α). In contrast, BM-MSC or GSI-953 treatment of dams following Aβ25-35 injection significantly (P < 0.001) reduced the number and size of activated microglial cells, markedly increased dendrite length, and reversed proinflammatory cytokine elevations in offspring. Moreover, BM-MSC or GSI-953 treatment reversed the Aβ25-35-induced amyloid precursor protein and p-Tau elevations in the offspring brain; these changes were accompanied by upregulation of the brain-derived neurotrophic factor and downregulation of glycogen synthase kinase-3β in the serum and brain. Treatment with BM-MSCs or GSI-953 also reversed Aβ25-35-induced elevations in different gene expressions in the neonatal cortex. Finally, treatment of dams with BM-MSCs or GSI-953 prevented the Aβ25-35-induced disruption of newborn brain development. Thus, BM-MSC and GSI-953 treatments have broad-spectrum effects against Aβ25-35-induced brain pathology, including the suppression of neural inflammation, restoration of developmental plasticity, and promotion of neurotrophic signaling.
Collapse
Affiliation(s)
- Asmaa Gaber
- Comparative Anatomy and Embryology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni Suef, Egypt
| | - Ahlam M. Elbakry
- Comparative Anatomy and Embryology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni Suef, Egypt
| | - Rabab M. Aljarari
- Department of Biology, College of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Fatima A. Jaber
- Department of Biology, College of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Yasser A. Khadrawy
- Medical Physiology Department, Medical Branch Department, National Research Center, Giza, Egypt
| | - Dina Sabry
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Badr University in Cairo, Cairo 11829, Egypt
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - Rasha E. Abo-ELeneen
- Comparative Anatomy and Embryology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni Suef, Egypt
| | - Osama M. Ahmed
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni Suef, Egypt
| |
Collapse
|
6
|
Jafarzadeh A, Bazargan N, Chatrabnous N, Jafarzadeh S, Nemati M. Contribution of survivin to the immune system, allergies and autoimmune diseases. Hum Immunol 2023; 84:301-310. [PMID: 36754653 DOI: 10.1016/j.humimm.2023.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 12/30/2022] [Accepted: 01/30/2023] [Indexed: 02/09/2023]
Abstract
In addition to malignancies, survivin (a member of the apoptosis inhibitor family) has been implicated in the pathogenesis of inflammatory disorders, including autoimmune and allergic diseases. Survivin is constantly expressed in the proliferating hematopoietic progenitor cells, and it is re-expressed in the mature cells of the innate and adaptive immunity, upon activation. Survivin enhances the expression of co-stimulatory molecules and MHC class II molecules in dendritic cells, and promotes the lifespan of macrophages, neutrophils, and eosinophils, while suppressing natural killer (NK) cell activity. Survivin has been implicated in T cell maturation, T cell expansion, effector CD4+ T cell differentiation, maintenance of memory CD4+ T and CD8+ T cells, as well as antibody production. Upregulated expression of survivin was indicated in the T cells as well as various samples collected from allergic patients. Survivin can contribute to the pathogenesis of allergic diseases via the promotion of the Th2 polarization, promoting IL-4 expression, compromising activation-induced cell death (AICD) in Th2 cells, and preventing apoptosis of eosinophils, as well as, amplification of eosinophilia. Moreover, survivin can interfere with clonal deletion of autoreactive T and B cells, as well as suppress Treg cell development and activity supporting the development of autoimmune diseases. This review discusses the role of survivin in immunity, allergy and autoimmunity as well as provides evidence that survivin may be considered as a novel therapeutic target for the treatment of allergic and autoimmune diseases.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Nasrin Bazargan
- Department of Internal Medicine, Medical School, Kerman University of Medical Sciences, Kerman, Iran
| | - Nazanin Chatrabnous
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Jafarzadeh
- Student Research Committee, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Nemati
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Haematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
7
|
Singh P. MSC and HSPC Coculture: Mimicking Ex Vivo Bone Marrow Niche. Methods Mol Biol 2023; 2567:181-189. [PMID: 36255702 DOI: 10.1007/978-1-0716-2679-5_12] [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] [Indexed: 06/16/2023]
Abstract
Mesenchymal stromal cells (MSCs) are the crucial component of the hematopoietic stem and progenitor cell (HSPC) niche in the bone marrow. Therefore, an ex vivo culture system that recapitulates the marrow microenvironment is important to understanding the niche's regulatory role on HSPC function and improving ex vivo HSPC expansion for clinical transplantation. Herein, a procedure for ex vivo expansion of MSCs from human bone marrow cells and their identification and characterization is described. In addition, a protocol for MSC and HSPC coculture assay is presented. This MSC-HSPC coculture assay can be used for ex vivo expansion of HSPC. Furthermore, this assay is also useful for qualitative analysis of MSCs capable of supporting hematopoiesis.
Collapse
Affiliation(s)
- Pratibha Singh
- Department of Medicine/Hematology Oncology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
8
|
The functional mechanism of bone marrow-derived mesenchymal stem cells in the treatment of animal models with Alzheimer's disease: crosstalk between autophagy and apoptosis. Stem Cell Res Ther 2022; 13:90. [PMID: 35241159 PMCID: PMC8895531 DOI: 10.1186/s13287-022-02765-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/24/2021] [Indexed: 12/25/2022] Open
Abstract
The transplantation of bone marrow-derived mesenchymal stem cells (BMMSCs) alleviates neuropathology and improves cognitive deficits in animal models with Alzheimer's disease. However, the underlying mechanism remains undefined. Based on meta-analysis and comprehensive review, high-profile studies support the theory that transplanted BMMSCs activate autophagy, as evidenced by the expression levels of signal molecules such as Beclin-1, Atg5, LC3-II, and mTOR. Functional autophagy mitigates neuronal apoptosis, which is reflected by the alterations of IAPs, Bcl-2, caspase-3, and so forth. Moreover, the transplantation of BMMSCs can decrease aberrant amyloid-beta peptides as well as tau aggregates, inhibit neuroinflammation, and stimulate synaptogenesis. There is a signal crosstalk between autophagy and apoptosis, which may be regulated to produce synergistic effect on the preconditioning of stem cells. Forasmuch, the therapeutic effect of transplanted BMMSCs can be enhanced by autophagy and/or apoptosis modulators.
Collapse
|
9
|
Pereira AR, Trivanović D, Stahlhut P, Rudert M, Groll J, Herrmann M. Preservation of the naïve features of mesenchymal stromal cells in vitro: Comparison of cell- and bone-derived decellularized extracellular matrix. J Tissue Eng 2022; 13:20417314221074453. [PMID: 35154631 PMCID: PMC8829705 DOI: 10.1177/20417314221074453] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/04/2022] [Indexed: 12/14/2022] Open
Abstract
The fate and behavior of bone marrow mesenchymal stem/stromal cells (BM-MSC) is bidirectionally influenced by their microenvironment, the stem cell niche, where a magnitude of biochemical and physical cues communicate in an extremely orchestrated way. It is known that simplified 2D in vitro systems for BM-MSC culture do not represent their naïve physiological environment. Here, we developed four different 2D cell-based decellularized matrices (dECM) and a 3D decellularized human trabecular-bone scaffold (dBone) to evaluate BM-MSC behavior. The obtained cell-derived matrices provided a reliable tool for cell shape-based analyses of typical features associated with osteogenic differentiation at high-throughput level. On the other hand, exploratory proteomics analysis identified native bone-specific proteins selectively expressed in dBone but not in dECM models. Together with its architectural complexity, the physico-chemical properties of dBone triggered the upregulation of stemness associated genes and niche-related protein expression, proving in vitro conservation of the naïve features of BM-MSC.
Collapse
Affiliation(s)
- Ana Rita Pereira
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, Wuerzburg, Germany
- Bernhard-Heine-Centrum for Locomotion Research, University of Wuerzburg, Wuerzburg, Germany
| | - Drenka Trivanović
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, Wuerzburg, Germany
- Bernhard-Heine-Centrum for Locomotion Research, University of Wuerzburg, Wuerzburg, Germany
| | - Philipp Stahlhut
- Chair for Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Wuerzburg, Wuerzburg, Germany
| | - Maximilian Rudert
- Department of Orthopedic Surgery, Koenig-Ludwig-Haus, University of Wuerzburg, Wuerzburg, Germany
| | - Jürgen Groll
- Chair for Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Wuerzburg, Wuerzburg, Germany
| | - Marietta Herrmann
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, Wuerzburg, Germany
- Bernhard-Heine-Centrum for Locomotion Research, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
10
|
Wang X, Huang Z, Zeng L, Jin X, Yan A, Zhang Y, Tan W. The Role of Survivin and Transcription Factor FOXP1 in Scarring After Glaucoma Surgery. Transl Vis Sci Technol 2022; 11:19. [PMID: 35142784 PMCID: PMC8842717 DOI: 10.1167/tvst.11.2.19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Purpose This study aims to elucidate the role and mechanism of survivin and FOXP1 in scarring after glaucoma surgery and to evaluate the prevention and treatment of excessive wound healing and scar formation in an in vitro model of glaucoma filtration surgery. Methods Human Tenon's capsule fibroblasts (HTFs) were used with TGF-β to establish an in vitro cell model after glaucoma, observe survivin expression in the cell model, and observe HTFs proliferation after treatment with survivin inhibitor YM155 and the expression of α-SMA and collagen type I. In addition, the effects of survivin and cell proliferation in HTFs after knockdown of FOXP1 were observed by Western blot analysis. Results Survivin was upregulated in HTFs after glaucoma surgery, and it could promote the cell proliferation of HTFs. After treatment with its inhibitor YM155, the cell proliferation of HTFs was inhibited, and the expression of α-SMA and collagen type I were decreased. The results showed that in knockdown of FOXP1, the expression of survivin was downregulated, and the cell proliferation of HTFs was significantly reduced. Conclusions This study demonstrates that targeting survivin with an inhibitory YM155 reduced fibrosis and the extracellular matrix (ECM), and it was regulated by the FOXP1 transcription factor. These results suggest that survivin could be a potential target for treating scar formation after glaucoma surgery. Translational Relevance Together with the results from previous survivin and FOXP1 preclinical studies, these data support the evaluation of this gene therapy candidate in clinical trials as a potential durable treatment for antiscarring of glaucoma surgery.
Collapse
Affiliation(s)
- Xiaocong Wang
- Soochow University, Suzhou, Jiangsu, China.,Medical College of Soochow University, Suzhou, Jiangsu, China.,Graduate School of Zunyi Medical University, Zunyi, Guizhou,China.,Department of Ophthalmology, The Third Affiliated Hospital of Zunyi, Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Zhihua Huang
- Graduate School of Zunyi Medical University, Zunyi, Guizhou,China.,The Ninth People's Hospital of Chongqing, Chongqing, China
| | - Lan Zeng
- Graduate School of Zunyi Medical University, Zunyi, Guizhou,China.,Department of Ophthalmology, The Third Affiliated Hospital of Zunyi, Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Xin Jin
- Department of Ophthalmology, The Third Affiliated Hospital of Zunyi, Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Ai Yan
- Department of Ophthalmology, The Third Affiliated Hospital of Zunyi, Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Ying Zhang
- Department of Ophthalmology, The Third Affiliated Hospital of Zunyi, Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Wei Tan
- Soochow University, Suzhou, Jiangsu, China.,Medical College of Soochow University, Suzhou, Jiangsu, China.,Graduate School of Zunyi Medical University, Zunyi, Guizhou,China.,Department of Ophthalmology, The Third Affiliated Hospital of Zunyi, Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| |
Collapse
|
11
|
Novel Balance Mechanism Participates in Stem Cell Therapy to Alleviate Neuropathology and Cognitive Impairment in Animal Models with Alzheimer's Disease. Cells 2021; 10:cells10102757. [PMID: 34685737 PMCID: PMC8534506 DOI: 10.3390/cells10102757] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/23/2021] [Accepted: 08/27/2021] [Indexed: 12/12/2022] Open
Abstract
Stem cell therapy improves memory loss and cognitive deficits in animal models with Alzheimer's disease. The underlying mechanism remains to be determined, but it may involve the interaction of stem cells with hippocampal cells. The transplantation of stem cells alters the pathological state and establishes a novel balance based on multiple signaling pathways. The new balance mechanism is regulated by various autocrine and paracrine cytokines, including signal molecules that target (a) cell growth and death. Stem cell treatment stimulates neurogenesis and inhibits apoptosis, which is regulated by the crosstalk between apoptosis and autophagy-(b) Aβ and tau pathology. Aberrant Aβ plaques and neurofibrillary tau tangles are mitigated subsequent to stem cell intervention-(c) inflammation. Neuroinflammation in the lesion is relieved, which may be related to the microglial M1/M2 polarization-(d) immunoregulation. The transplanted stem cells modulate immune cells and shape the pathophysiological roles of immune-related genes such as TREM2, CR1, and CD33-(e) synaptogenesis. The functional reconstruction of synaptic connections can be promoted by stem cell therapy through multi-level signaling, such as autophagy, microglial activity, and remyelination. The regulation of new balance mechanism provides perspective and challenge for the treatment of Alzheimer's disease.
Collapse
|
12
|
Abstract
Survivin is one of the rare proteins that is differentially expressed in normal and cancer cells and is directly or indirectly involved in numerous pathways required for tumor maintenance. It is expressed in almost all cancers and its expression has been detected at early stages of cancer. These traits make survivin an exceptionally attractive target for cancer therapeutics. Even with these promising features to be an oncotherapeutic target, there has been limited success in the clinical trials targeting survivin. Only recently it has emerged that survivin was not being specifically targeted which could have resulted in the negative clinical outcome. Also, focus of research has now shifted from survivin expression in the overall heterogeneous tumor cell populations to survivin expression in cancer stem cells as these cells have proved to be the major drivers of tumors. Therefore, in this review we have analyzed the expression of survivin in normal and cancer cells with a particular focus on its expression in cancer stem cell compartment. We have discussed the major signaling pathways involved in regulation of survivin. We have explored the current development status of various types of interventions for inhibition of survivin. Furthermore, we have discussed the challenges involving the development of potent and specific survivin inhibitors for cancer therapeutics. Finally we have given insights for some of the promising future anticancer treatments.
Collapse
|
13
|
Singh P, Mohammad KS, Pelus LM. CXCR4 expression in the bone marrow microenvironment is required for hematopoietic stem and progenitor cell maintenance and early hematopoietic regeneration after myeloablation. Stem Cells 2020; 38:849-859. [PMID: 32159901 DOI: 10.1002/stem.3174] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 02/14/2020] [Accepted: 02/28/2020] [Indexed: 11/10/2022]
Abstract
The bone marrow (BM) microenvironment/niche plays a key role in regulating hematopoietic stem and progenitor cell (HSPC) activities; however, mechanisms regulating niche cell function are not well understood. In this study, we show that niche intrinsic expression of the CXCR4 chemokine receptor critically regulates HSPC maintenance during steady state, and promotes early hematopoietic regeneration after myeloablative irradiation. At steady state, chimeric mice with wild-type (WT) HSPC and marrow stroma that lack CXCR4 show decreased HSPC quiescence, and their repopulation capacity was markedly reduced. Mesenchymal stromal cells (MSC) were significantly reduced in the BM of CXCR4 deficient mice, which was accompanied by decreased levels of the HSPC supporting factors stromal cell-derived factor-1 (SDF-1) and stem cell factor (SCF). CXCR4 also plays a crucial role in survival and restoration of BM stromal cells after myeloablative irradiation, where the loss of BM stromal cells was more severe in CXCR4-deficient mice compared to WT mice. In addition, transplantation of WT donor HSPC into CXCR4-deficient recipient mice demonstrated reduced HSPC homing and early hematopoietic reconstitution. We found that CXCR4 signaling attenuates irradiation-induced BM stromal cell loss by upregulating the expression of the antiapoptotic protein Survivin via the PI3K pathway. Our study suggests that SDF-1-CXCR4 signaling in the stromal microenvironment cells plays a crucial role in maintenance of HSPCs during homeostasis, and promotes niche regeneration and early hematopoietic reconstitution after transplantation. Modulation of CXCR4 signaling in the HSPC microenvironment could be a means to enhance hematopoietic recovery after clinical hematopoietic cell transplantation.
Collapse
Affiliation(s)
- Pratibha Singh
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Khalid S Mohammad
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Louis M Pelus
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
14
|
BIRC5 Expression is Regulated in Uterine Epithelium During the Estrous Cycle. Genes (Basel) 2020; 11:genes11030282. [PMID: 32155884 PMCID: PMC7140846 DOI: 10.3390/genes11030282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/28/2020] [Accepted: 03/03/2020] [Indexed: 12/12/2022] Open
Abstract
Baculoviral inhibitor of apoptosis repeat-containing 5 (Birc5), also known as survivin, is a member of the inhibitor of apoptosis (IAP) family of proteins and regulates the size of tissues through cell division control. The uterus is the most dynamically sized organ among tissues during the estrous cycle. Although Birc5 is expressed in some terminally differentiated cells, the regulation of its expression in the uterus remains unknown. We investigated the regulation of Birc5 expression in the mouse uterus. RT-PCR analysis showed that Birc5 was expressed in various tissues, including the uterus; the expression level of Birc5 was significantly higher at the diestrus stage. Immunohistochemistry and Western blotting analysis revealed that Birc5 was more active in luminal and glandular epithelium than in endometrial stroma. In ovariectomized mice, Birc5 expression in the uterus was gradually increased by estrogen treatment; however, progesterone injection decreased its expression. Estrogen-induced Birc5 expression was blocked by treatment with estrogen receptor antagonist, ICI 182, 780 and progesterone-reduced Birc5 expression was inhibited by the progesterone receptor antagonist RU486. These results suggest that Birc5 expression is dynamically regulated by a combination of estrogen and progesterone via their receptor-mediated signaling.
Collapse
|
15
|
Naji A, Favier B, Deschaseaux F, Rouas-Freiss N, Eitoku M, Suganuma N. Mesenchymal stem/stromal cell function in modulating cell death. Stem Cell Res Ther 2019; 10:56. [PMID: 30760307 PMCID: PMC6374902 DOI: 10.1186/s13287-019-1158-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) delivered as cell therapy to individuals with degenerative and/or inflammatory disorders can help improve organ features and resolve inflammation, as demonstrated in preclinical studies and to some extent in clinical studies. MSCs have trophic, homing/migration, and immunosuppression functions, with many benefits in therapeutics. MSC functions are thought to depend on the paracrine action of soluble factors and/or the expression of membrane-bound molecules, mostly belonging to the molecular class of adhesion molecules, chemokines, enzymes, growth factors, and interleukins. Cutting-edge studies underline bioactive exchanges, including that of ions, nucleic acids, proteins, and organelles transferred from MSCs to stressed cells, thereby improving the cells' survival and function. From this aspect, MSC death modulation function appears as a decisive biological function that could carry a significant part of the therapeutic effects of MSCs. Identifying the function and modes of actions of MSCs in modulating cell death may be exploited to enhance consistency and efficiency of cell therapy that is based on MSCs as medical treatment for degenerative and/or inflammatory diseases. Here, we review the essentials of MSC functions in modulating cell death in unfit cells, and its modes of actions based on current advances and outline the clinical implications.
Collapse
Affiliation(s)
- Abderrahim Naji
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School (KMS), Kochi University, Kohasu, Oko-Cho, Nankoku City, Kochi Prefecture, 783-8505, Japan.
| | - Benoit Favier
- CEA-Université Paris Sud INSERM U1184, IDMIT Department, IBFJ, DRF, Fontenay-aux-Roses, France
| | - Frédéric Deschaseaux
- STROMALab, UMR 5273 CNRS, INSERM U1031, Etablissement Français du Sang (EFS) Occitanie, Université de Toulouse, Toulouse, France
| | - Nathalie Rouas-Freiss
- CEA, DRF-Institut Francois Jacob, Division de recherche en hématologie et immunologie (SRHI), Hôpital Saint-Louis, Paris, France
| | - Masamitsu Eitoku
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School (KMS), Kochi University, Kohasu, Oko-Cho, Nankoku City, Kochi Prefecture, 783-8505, Japan
| | - Narufumi Suganuma
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School (KMS), Kochi University, Kohasu, Oko-Cho, Nankoku City, Kochi Prefecture, 783-8505, Japan.
| |
Collapse
|
16
|
Antiapoptotic Molecule Survivin in Transplantation: Helpful or Harmful? J Transplant 2018; 2018:6492034. [PMID: 30364092 PMCID: PMC6188762 DOI: 10.1155/2018/6492034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 08/11/2018] [Accepted: 08/19/2018] [Indexed: 11/17/2022] Open
Abstract
Survivin, an antiapoptotic molecule from inhibitor of apoptosis protein (IAP) family, is most known for its implication in cancer as there are some efforts to apply it for diagnostic as well as therapeutic purposes in oncology. On the other hand, it could be a useful molecule to be positively targeted when trying to save tissue and promote cells viability. Since protecting the allograft from ischemia reperfusion injury and inflammation-induced damage is a considerable objective in transplantation, it is reasonable to take advantage from antiapoptotic agents like survivin in order to achieve this goal. However, survivin's potential ability to induce malignancies makes some concerns about its use in clinic. The other barrier is this molecule's involvement in lymphocytes development and proliferation which might increase the risk of graft rejection due to adaptive immune system overactivation. In this review we summarize the few studies carried out about survivin's effect on graft survival and probable advantages and disadvantages of its overexpression in transplantation.
Collapse
|