1
|
Wang K, Liu Y, Li S, Zhao N, Qin F, Tao Y, Song Z. Unveiling the therapeutic potential and mechanisms of stanniocalcin-1 in retinal degeneration. Surv Ophthalmol 2025; 70:106-120. [PMID: 39270826 DOI: 10.1016/j.survophthal.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 07/30/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024]
Abstract
Retinal degeneration (RD) is a group of ocular diseases characterized by progressive photoreceptor apoptosis and visual impairment. Mitochondrial malfunction, excessive oxidative stress, and chronic activation of neuroglia collectively contribute to the development of RD. Currently, there is a lack of efficacious therapeutic interventions for RD. Stanniocalcin-1 (STC-1) is a promising candidate molecule to decelerate photoreceptor cell death. STC-1 is a secreted calcium/phosphorus regulatory protein that exerts diverse protective effects. Accumulating evidence suggests that STC-1 protects retinal cells from ischemic injury, oxidative stress, and excessive apoptosis through enhancing the expression of uncoupling protein-2 (UCP-2). Furthermore, STC-1 exerts its antiinflammatory effects by inhibiting the activation of microglia and macrophages, as well as the synthesis and secretion of proinflammatory cytokines, such as TNF-α, IL-1, and IL-6. By employing these mechanisms, STC-1 effectively shields the retinal photoreceptors and optic nerve, thereby slowing down the progression of RD. We summarize the STC-1-mediated therapeutic effects on the degenerating retina, with a particular focus on its underlying mechanisms. These findings highlight that STC-1 may act as a versatile molecule to treat degenerative retinopathy. Further research on STC-1 is imperative to establish optimal protocols for its clinical use.
Collapse
Affiliation(s)
- Kexin Wang
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Yashuang Liu
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Siyu Li
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China
| | - Na Zhao
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China
| | - Fangyuan Qin
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Ye Tao
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China.
| | - Zongming Song
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China.
| |
Collapse
|
2
|
Elashry MI, Speer J, De Marco I, Klymiuk MC, Wenisch S, Arnhold S. Extracellular Vesicles: A Novel Diagnostic Tool and Potential Therapeutic Approach for Equine Osteoarthritis. Curr Issues Mol Biol 2024; 46:13078-13104. [PMID: 39590374 PMCID: PMC11593097 DOI: 10.3390/cimb46110780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Osteoarthritis (OA) is a chronic progressive degenerative joint disease that affects a significant portion of the equine population and humans worldwide. Current treatment options for equine OA are limited and incompletely curative. Horses provide an excellent large-animal model for studying human OA. Recent advances in the field of regenerative medicine have led to the exploration of extracellular vesicles (EVs)-cargoes of microRNA, proteins, lipids, and nucleic acids-to evaluate their diagnostic value in terms of disease progression and severity, as well as a potential cell-free therapeutic approach for equine OA. EVs transmit molecular signals that influence various biological processes, including the inflammatory response, apoptosis, proliferation, and cell communication. In the present review, we summarize recent advances in the isolation and identification of EVs, the use of their biologically active components as biomarkers, and the distribution of the gap junction protein connexin 43. Moreover, we highlight the role of mesenchymal stem cell-derived EVs as a potential therapeutic tool for equine musculoskeletal disorders. This review aims to provide a comprehensive overview of the current understanding of the pathogenesis, diagnosis, and treatment strategies for OA. In particular, the roles of EVs as biomarkers in synovial fluid, chondrocytes, and plasma for the early detection of equine OA are discussed.
Collapse
Affiliation(s)
- Mohamed I. Elashry
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (J.S.); (M.C.K.); (S.A.)
| | - Julia Speer
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (J.S.); (M.C.K.); (S.A.)
| | - Isabelle De Marco
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (I.D.M.); (S.W.)
| | - Michele C. Klymiuk
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (J.S.); (M.C.K.); (S.A.)
| | - Sabine Wenisch
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (I.D.M.); (S.W.)
| | - Stefan Arnhold
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (J.S.); (M.C.K.); (S.A.)
| |
Collapse
|
3
|
Wang J, Li J, Yin L, Wang X, Dong Y, Zhao G, Shen S, Hou Y. MSCs promote the efferocytosis of large peritoneal macrophages to eliminate ferroptotic monocytes/macrophages in the injured endometria. Stem Cell Res Ther 2024; 15:127. [PMID: 38693589 PMCID: PMC11064342 DOI: 10.1186/s13287-024-03742-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND Endometria are one of the important components of the uterus, which is located in the peritoneal cavity. Endometrial injury usually leads to intrauterine adhesions (IUA), accompanied by inflammation and cell death. We previously reported that both the endometrial ferroptosis was increased and monocytes/macrophages were involved in endometrial injury of IUA. Large peritoneal macrophages (LPMs) are recently reported to migrate into the injured tissues and phagocytose dead cells to repair the tissues. We previously demonstrated that mesenchymal stromal cells (MSCs) had made excellent progress in the repair of endometrial injury. However, it is unclear whether MSCs regulate the LPM efferocytosis against ferroptotic monocytes/macrophages in the injured endometria. METHODS Here, endometrial injury in IUA mouse model was conducted by uterine curettage and LPS injection surgery and the samples were collected at different times to detect the changes of LPMs and ferroptotic monocytes/macrophages. We conducted LPMs depletion assay in vivo and LPMs and Erastin-induced ferroptotic THP-1 cells coculture systems in vitro to detect the LPM efferocytosis against ferroptotic monocytes/macrophages. The IUA model was treated with MSCs, and their effects on LPMs and endometrial repair were analyzed. Flow cytometry, western blotting, quantitative real-time PCR, immunohistochemical analysis, ELISA, and RNA-sequencing were performed. RESULTS We found that LPMs migrated to the injured uteri in response to the damage in early phase (3 h), and sustained to a later stage (7 days). Astonishingly, we found that ferroptotic monocytes/macrophages were significantly increased in the injured uteri since 12 h after injury. Moreover, LPMs cocultured with Erastin-induced ferroptotic THP-1 cells in vitro, efferocytosis of LPMs against ferroptotic monocytes/macrophages was emerged. The mRNA expression profiles revealed that LPM efferocytosis against ferroptotic monocytes/macrophages was an induction of glycolysis program and depended on the PPARγ-HK2 pathway. Importantly, we validated that MSCs promoted the efferocytic capability and migration of LPMs to the injured uteri via secreting stanniocalcin-1 (STC-1). CONCLUSION The data collectively demonstrated first the roles of LPMs via removal of ferroptotic monocytes/macrophages and provided a novel mechanism of MSCs in repairing the endometrial injury.
Collapse
Affiliation(s)
- Jiali Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, No. 22 Hankou Rd., Gulou District, Nanjing, Jiangsu, 210093, People's Republic of China
| | - Jingman Li
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, No. 22 Hankou Rd., Gulou District, Nanjing, Jiangsu, 210093, People's Republic of China
| | - Lijie Yin
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, No. 22 Hankou Rd., Gulou District, Nanjing, Jiangsu, 210093, People's Republic of China
| | - Xiuzhu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, No. 22 Hankou Rd., Gulou District, Nanjing, Jiangsu, 210093, People's Republic of China
| | - Yue Dong
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, No. 22 Hankou Rd., Gulou District, Nanjing, Jiangsu, 210093, People's Republic of China
| | - Guangfeng Zhao
- Department of Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Sunan Shen
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, No. 22 Hankou Rd., Gulou District, Nanjing, Jiangsu, 210093, People's Republic of China.
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, No. 22 Hankou Rd., Gulou District, Nanjing, Jiangsu, 210093, People's Republic of China.
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
4
|
Rendra E, Crigna AT, Daniele C, Sticht C, Cueppers M, Kluth MA, Ganss C, Frank MH, Gretz N, Bieback K. Clinical-grade human skin-derived ABCB5+ mesenchymal stromal cells exert anti-apoptotic and anti-inflammatory effects in vitro and modulate mRNA expression in a cisplatin-induced kidney injury murine model. Front Immunol 2024; 14:1228928. [PMID: 38274791 PMCID: PMC10808769 DOI: 10.3389/fimmu.2023.1228928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 12/22/2023] [Indexed: 01/27/2024] Open
Abstract
Acute kidney injury (AKI) is characterized by a rapid reduction in renal function and glomerular filtration rate (GFR). The broadly used anti-cancer chemotherapeutic agent cisplatin often induces AKI as an adverse drug side effect. Therapies targeted at the reversal of AKI and its potential progression to chronic kidney disease or end-stage renal disease are currently insufficiently effective. Mesenchymal stromal cells (MSCs) possess diverse immunomodulatory properties that confer upon them significant therapeutic potential for the treatment of diverse inflammatory disorders. Human dermal MSCs expressing ATP-Binding Cassette member B5 (ABCB5) have shown therapeutic efficacy in clinical trials in chronic skin wounds or recessive dystrophic epidermolysis bullosa. In preclinical studies, ABCB5+ MSCs have also shown to reverse metabolic reprogramming in polycystic kidney cells, suggesting a capacity for this cell subset to improve also organ function in kidney diseases. Here, we aimed to explore the therapeutic capacity of ABCB5+ MSCs to improve renal function in a preclinical rat model of cisplatin-induced AKI. First, the anti-apoptotic and immunomodulatory capacity was compared against research-grade adipose stromal cells (ASCs). Then, cross-species immunomodulatory capacity was checked, testing first inhibition of mitogen-driven peripheral blood mononuclear cells and then modulation of macrophage function. Finally, therapeutic efficacy was evaluated in a cisplatin AKI model. First, ABCB5+ MSCs suppressed cisplatin-induced apoptosis of human conditionally-immortalized proximal tubular epithelial cells in vitro, most likely by reducing oxidative stress. Second, ABCB5+ MSCs inhibited the proliferation of either human or rat peripheral blood mononuclear cells, in the human system via the Indoleamine/kynurenine axis and in the murine context via nitric oxide/nitrite. Third, ABCB5+ MSCs decreased TNF-α secretion after lipopolysaccharide stimulation and modulated phagocytosis and in both human and rat macrophages, involving prostaglandin E2 and TGF-β1, respectively. Fourth, clinical-grade ABCB5+ MSCs grafted intravenously and intraperitoneally to a cisplatin-induced AKI murine model exerted modulatory effects on mRNA expression patterns toward an anti-inflammatory and pro-regenerative state despite an apparent lack of amelioration of renal damage at physiologic, metabolic, and histologic levels. Our results demonstrate anti-inflammatory and pro-regenerative effects of clinical grade ABCB5+ MSCs in vitro and in vivo and suggest potential therapeutic utility of this cell population for treatment or prevention of cisplatin chemotherapy-induced tissue toxicity.
Collapse
Affiliation(s)
- Erika Rendra
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| | - Adriana Torres Crigna
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| | - Cristina Daniele
- Medical Faculty Mannheim, Medical Research Centre, Heidelberg University, Mannheim, Germany
| | - Carsten Sticht
- Medical Faculty Mannheim, Medical Research Centre, Heidelberg University, Mannheim, Germany
| | - Maike Cueppers
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| | | | | | - Markus H. Frank
- Transplant Research Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States
- Harvard Skin Disease Research Center, Department of Dermatology, Brigham and Women’s Hospital, Boston, MA, United States
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | - Norbert Gretz
- Medical Faculty Mannheim, Medical Research Centre, Heidelberg University, Mannheim, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
5
|
Rendra E, Uhlig S, Moskal I, Thielemann C, Klüter H, Bieback K. Adipose Stromal Cell-Derived Secretome Attenuates Cisplatin-Induced Injury In Vitro Surpassing the Intricate Interplay between Proximal Tubular Epithelial Cells and Macrophages. Cells 2024; 13:121. [PMID: 38247813 PMCID: PMC10814170 DOI: 10.3390/cells13020121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
(1) Background: The chemotherapeutic drug cisplatin exerts toxic side effects causing acute kidney injury. Mesenchymal stromal cells can ameliorate cisplatin-induced kidney injury. We hypothesize that the MSC secretome orchestrates the vicious cycle of injury and inflammation by acting on proximal tubule epithelial cells (PTECs) and macrophages individually, but further by counteracting their cellular crosstalk. (2) Methods: Conditioned medium (CM) from adipose stromal cells was used, first assessing its effect on cisplatin injury in PTECs. Second, the effects of cisplatin and the CM on macrophages were measured. Lastly, in an indirect co-culture system, the interplay between the two cell types was assessed. (3) Results: First, the CM rescued PTECs from cisplatin-induced apoptosis by reducing oxidative stress and expression of nephrotoxicity genes. Second, while cisplatin exerted only minor effects on macrophages, the CM skewed macrophage phenotypes to the anti-inflammatory M2-like phenotype and increased phagocytosis. Finally, in the co-culture system, the CM suppressed PTEC death by inhibiting apoptosis and nuclei fragmentation. The CM lowered TNF-α release, while cisplatin inhibited macrophage phagocytosis, PTECs, and the CM to a greater extent, thus enhancing it. The CM strongly dampened the inflammatory macrophage cytokine secretion triggered by PTECs. (4) Conclusions: ASC-CM surpasses the PTEC-macrophage crosstalk in cisplatin injury. The positive effects on reducing cisplatin cytotoxicity, on polarizing macrophages, and on fine-tuning cytokine secretion underscore MSCs' CM benefit to prevent kidney injury progression.
Collapse
Affiliation(s)
- Erika Rendra
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany; (E.R.); (H.K.)
| | - Stefanie Uhlig
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany; (E.R.); (H.K.)
- Flow Core Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Isabell Moskal
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany; (E.R.); (H.K.)
| | - Corinna Thielemann
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany; (E.R.); (H.K.)
| | - Harald Klüter
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany; (E.R.); (H.K.)
- Mannheim Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany; (E.R.); (H.K.)
- Flow Core Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Mannheim Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| |
Collapse
|
6
|
Choi S, An HJ, Yeo HJ, Sung MJ, Oh J, Lee K, Lee SA, Kim SK, Kim J, Kim I, Lee S. MicroRNA‑606 inhibits the growth and metastasis of triple‑negative breast cancer by targeting Stanniocalcin 1. Oncol Rep 2024; 51:2. [PMID: 37975233 PMCID: PMC10688449 DOI: 10.3892/or.2023.8661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/26/2023] [Indexed: 11/19/2023] Open
Abstract
Triple‑negative breast cancer (TNBC) is associated with a poor prognosis; however, treatments for TNBC are limited, with poor outcomes. MicroRNAs (miRNAs/miRs) are small non‑coding RNA molecules that are able to regulate gene expression. The present study aimed to identify differentially expressed miRNAs in patients with breast cancer, and to investigate the functional role of the identified miRNA targets and their effects in vitro and in vivo. Transfection with miR‑606 suppressed TNBC cell proliferation, migration, invasion and tumor sphere‑forming ability, as determined using trypan blue, Transwell and sphere formation assays. Moreover, miR‑606 induced the apoptosis of TNBC cells, as determined by flow cytometric analysis. Furthermore, intratumoral injections of miR‑606 mimics suppressed tumor growth in MDA‑MB‑231 xenografts. In addition, MDA‑MB‑231 cells transfected with miR‑606 mimics exhibited decreased lung metastatic nodules in a mouse tail vein injection model. Notably, miR‑606 and STC1 expression had opposing effects on the overall survival of patients with TNBC. The results of the present study suggested a novel tumor suppressor function for miR‑606 in TNBC, thus indicating its potential application in the development of anticancer miRNA therapeutics.
Collapse
Affiliation(s)
- Sujin Choi
- Department of Orthopedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Hyun-Ju An
- Department of Orthopedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi 13488, Republic of Korea
- SL Bio, Inc., Pocheon, Gyeonggi 11160, Republic of Korea
| | - Hyun Jeong Yeo
- Department of Orthopedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Min-Ji Sung
- Department of Orthopedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Jisu Oh
- Division of Hemato-Oncology, Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Gyeonggi 16995, Republic of Korea
| | - Kwanbum Lee
- Department of General Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Seung Ah Lee
- Department of General Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Seung Ki Kim
- Department of General Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Junhan Kim
- Department of Orthopedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Isaac Kim
- Department of General Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Soonchul Lee
- Department of Orthopedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi 13488, Republic of Korea
- SL Bio, Inc., Pocheon, Gyeonggi 11160, Republic of Korea
| |
Collapse
|
7
|
Sionov RV, Ahdut-HaCohen R. A Supportive Role of Mesenchymal Stem Cells on Insulin-Producing Langerhans Islets with a Specific Emphasis on The Secretome. Biomedicines 2023; 11:2558. [PMID: 37761001 PMCID: PMC10527322 DOI: 10.3390/biomedicines11092558] [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: 08/15/2023] [Revised: 09/06/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Type 1 Diabetes (T1D) is a chronic autoimmune disease characterized by a gradual destruction of insulin-producing β-cells in the endocrine pancreas due to innate and specific immune responses, leading to impaired glucose homeostasis. T1D patients usually require regular insulin injections after meals to maintain normal serum glucose levels. In severe cases, pancreas or Langerhans islet transplantation can assist in reaching a sufficient β-mass to normalize glucose homeostasis. The latter procedure is limited because of low donor availability, high islet loss, and immune rejection. There is still a need to develop new technologies to improve islet survival and implantation and to keep the islets functional. Mesenchymal stem cells (MSCs) are multipotent non-hematopoietic progenitor cells with high plasticity that can support human pancreatic islet function both in vitro and in vivo and islet co-transplantation with MSCs is more effective than islet transplantation alone in attenuating diabetes progression. The beneficial effect of MSCs on islet function is due to a combined effect on angiogenesis, suppression of immune responses, and secretion of growth factors essential for islet survival and function. In this review, various aspects of MSCs related to islet function and diabetes are described.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Institute of Biomedical and Oral Research (IBOR), Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Ronit Ahdut-HaCohen
- Department of Medical Neurobiology, Institute of Medical Research, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel;
- Department of Science, The David Yellin Academic College of Education, Jerusalem 9103501, Israel
| |
Collapse
|
8
|
Zhuang X, Jiang Y, Yang X, Fu L, Luo L, Dong Z, Zhao J, Hei F. Advances of mesenchymal stem cells and their derived extracellular vesicles as a promising therapy for acute respiratory distress syndrome: from bench to clinic. Front Immunol 2023; 14:1244930. [PMID: 37711624 PMCID: PMC10497773 DOI: 10.3389/fimmu.2023.1244930] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is an acute inflammatory lung injury characterized by diffuse alveolar damage. The period prevalence of ARDS was 10.4% of ICU admissions in 50 countries. Although great progress has been made in supportive care, the hospital mortality rate of severe ARDS is still up to 46.1%. Moreover, up to now, there is no effective pharmacotherapy for ARDS and most clinical trials focusing on consistently effective drugs have met disappointing results. Mesenchymal stem cells (MSCs) and their derived extracellular vesicles (EVs) have spawned intense interest of a wide range of researchers and clinicians due to their robust anti-inflammatory, anti-apoptotic and tissue regeneration properties. A growing body of evidence from preclinical studies confirmed the promising therapeutic potential of MSCs and their EVs in the treatment of ARDS. Based on the inspiring experimental results, clinical trials have been designed to evaluate safety and efficacy of MSCs and their EVs in ARDS patients. Moreover, trials exploring their optimal time window and regimen of drug administration are ongoing. Therefore, this review aims to present an overview of the characteristics of mesenchymal stem cells and their derived EVs, therapeutic mechanisms for ARDS and research progress that has been made over the past 5 years.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Feilong Hei
- Department of Cardiopulmonary Bypass, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
9
|
Mohamad-Fauzi N, Shaw C, Foutouhi SH, Hess M, Kong N, Kol A, Storey DB, Desai PT, Shah J, Borjesson D, Murray JD, Weimer BC. Salmonella enhances osteogenic differentiation in adipose-derived mesenchymal stem cells. Front Cell Dev Biol 2023; 11:1077350. [PMID: 37009487 PMCID: PMC10055666 DOI: 10.3389/fcell.2023.1077350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 02/17/2023] [Indexed: 03/17/2023] Open
Abstract
The potential of mesenchymal stem cells (MSCs) for tissue repair and regeneration has garnered great attention. While MSCs are likely to interact with microbes at sites of tissue damage and inflammation, like in the gastrointestinal system, the consequences of pathogenic association on MSC activities have yet to be elucidated. This study investigated the effects of pathogenic interaction on MSC trilineage differentiation paths and mechanisms using model intracellular pathogen Salmonella enterica ssp enterica serotype Typhimurium. The examination of key markers of differentiation, apoptosis, and immunomodulation demonstrated that Salmonella altered osteogenic and chondrogenic differentiation pathways in human and goat adipose-derived MSCs. Anti-apoptotic and pro-proliferative responses were also significantly upregulated (p < 0.05) in MSCs during Salmonella challenge. These results together indicate that Salmonella, and potentially other pathogenic bacteria, can induce pathways that influence both apoptotic response and functional differentiation trajectories in MSCs, highlighting that microbes have a potentially significant role as influencers of MSC physiology and immune activity.
Collapse
Affiliation(s)
- Nuradilla Mohamad-Fauzi
- Department of Animal Science, College of Agricultural and Environmental Sciences, University of California, Davis, Davis, CA, United States
| | - Claire Shaw
- Department of Animal Science, College of Agricultural and Environmental Sciences, University of California, Davis, Davis, CA, United States
| | - Soraya H. Foutouhi
- Department of Population Health and Reproduction, 100K Pathogen Genome Project, Davis, CA, United States
| | - Matthias Hess
- Department of Animal Science, College of Agricultural and Environmental Sciences, University of California, Davis, Davis, CA, United States
| | - Nguyet Kong
- Department of Population Health and Reproduction, 100K Pathogen Genome Project, Davis, CA, United States
| | - Amir Kol
- Department of Pathology, Microbiology and Immunology, University of California, Davis, Davis, CA, United States
| | - Dylan Bobby Storey
- Department of Population Health and Reproduction, 100K Pathogen Genome Project, Davis, CA, United States
| | - Prerak T. Desai
- Department of Population Health and Reproduction, 100K Pathogen Genome Project, Davis, CA, United States
| | - Jigna Shah
- Department of Population Health and Reproduction, 100K Pathogen Genome Project, Davis, CA, United States
| | - Dori Borjesson
- Department of Pathology, Microbiology and Immunology, University of California, Davis, Davis, CA, United States
| | - James D. Murray
- Department of Animal Science, College of Agricultural and Environmental Sciences, University of California, Davis, Davis, CA, United States
- Department of Population Health and Reproduction, 100K Pathogen Genome Project, Davis, CA, United States
- *Correspondence: James D. Murray, ; Bart C. Weimer,
| | - Bart C. Weimer
- Department of Population Health and Reproduction, 100K Pathogen Genome Project, Davis, CA, United States
- *Correspondence: James D. Murray, ; Bart C. Weimer,
| |
Collapse
|
10
|
Erkılınç G, Yüceer RO, Erdemoğlu E, Kaymak ZA, Darbaş ŞM, Bozkurt KK, Çiriş İM. The expression of stanniocalcin-1, estrogen receptor and progesterone receptor in endometrioid endometrial cancer. Turk J Obstet Gynecol 2023; 20:8-15. [PMID: 36908008 PMCID: PMC10013080 DOI: 10.4274/tjod.galenos.2023.93646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 02/19/2023] [Indexed: 03/12/2023] Open
Abstract
Objective To evaluate the expression of stanniocalcin-1 (STC-1) and to investigate the correlation of STC-1 with expression of estrogen receptor (ER), progesterone receptor (PR) and clinical parameters, histopathological findings and prognostic factors in endometrioid endometrial cancer (EEC). Materials and Methods In this retrospective study, STC-1 (cytoplasmic), ER (nuclear), and PR (nuclear) stainings were applied to tissue microarray sections of 89 EEC, 27 endometrial intraepithelial neoplasia (EIN), and 21 normal endometrium (NE). Prognostic factors such as age, tumor size, depth of myometrial invasion, lymphovascular invasion, perineural invasion, and lymph node metastasis were compared with the expression of these markers. Results ER showed significantly higher positivity in grade 1 EEC. PR expression was also higher in grade 1 EEC, but these findings were not statistically significant. Strong expression of STC-1 was observed in EIN and EECs compared with NE. STC-1 showed low staining in the NE, and high staining was also noted in the EIN foci adjacent to the NE. STC-1 expression was positively correlated with grade 1 EECs. Conclusion STC-1 expression was positively correlated with low histologic grade in EECs. STC-1 can be used for distinguishing low-grade endometrioid tumors and high -grade endometrioid tumors in curretage specimens. Since STC-1 is related to well differentiated tumors, it can also be regarded as a good prognostic factor in EECs.
Collapse
Affiliation(s)
- Gamze Erkılınç
- İzmir Urla State Hospital, Medical Pathology Laboratory, İzmir, Turkey
| | | | - Evrim Erdemoğlu
- Süleyman Demirel University Faculty of Medicine, Department of Obstetrics and Gynecology/Gynecological Oncology, Isparta, Turkey
| | - Zümrüt Arda Kaymak
- Süleyman Demirel University Faculty of Medicine, Department of Radiation Oncology, Isparta, Turkey
| | - Şerife Mehtap Darbaş
- Bartın State Hospital, Clinic of Internal Medicine/Department Endocrinology and Metabolic Disease, Bartın, Turkey
| | - Kemal Kürşat Bozkurt
- Süleyman Demirel University Faculty of Medicine, Department of Medical Pathology, Isparta, Turkey
| | - İbrahim Metin Çiriş
- Süleyman Demirel University Faculty of Medicine, Department of Medical Pathology, Isparta, Turkey
| |
Collapse
|
11
|
Ghasemi Darestani N, Gilmanova AI, Al-Gazally ME, Zekiy AO, Ansari MJ, Zabibah RS, Jawad MA, Al-Shalah SAJ, Rizaev JA, Alnassar YS, Mohammed NM, Mustafa YF, Darvishi M, Akhavan-Sigari R. Mesenchymal stem cell-released oncolytic virus: an innovative strategy for cancer treatment. Cell Commun Signal 2023; 21:43. [PMID: 36829187 PMCID: PMC9960453 DOI: 10.1186/s12964-022-01012-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/10/2022] [Indexed: 02/26/2023] Open
Abstract
Oncolytic viruses (OVs) infect, multiply, and finally remove tumor cells selectively, causing no damage to normal cells in the process. Because of their specific features, such as, the ability to induce immunogenic cell death and to contain curative transgenes in their genomes, OVs have attracted attention as candidates to be utilized in cooperation with immunotherapies for cancer treatment. This treatment takes advantage of most tumor cells' inherent tendency to be infected by certain OVs and both innate and adaptive immune responses are elicited by OV infection and oncolysis. OVs can also modulate tumor microenvironment and boost anti-tumor immune responses. Mesenchymal stem cells (MSC) are gathering interest as promising anti-cancer treatments with the ability to address a wide range of cancers. MSCs exhibit tumor-trophic migration characteristics, allowing them to be used as delivery vehicles for successful, targeted treatment of isolated tumors and metastatic malignancies. Preclinical and clinical research were reviewed in this study to discuss using MSC-released OVs as a novel method for the treatment of cancer. Video Abstract.
Collapse
Affiliation(s)
| | - Anna I Gilmanova
- Department of Prosthetic Dentistry of the I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | | | - Angelina O Zekiy
- Department of Prosthetic Dentistry of the I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | | | - Saif A J Al-Shalah
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Iraq
| | - Jasur Alimdjanovich Rizaev
- Department of Public Health and Healthcare Management, Rector, Samarkand State Medical University, Samarkand, Uzbekistan
| | | | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Mohammad Darvishi
- Department of Aerospace and Subaquatic Medicine, Infectious Diseases and Tropical Medicine Research Center (IDTMRC), AJA University of Medical Sciences, Tehran, Iran.
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center, Tuebingen, Germany.,Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
12
|
Ding W, Zhang K, Li Q, Xu L, Ma Y, Han F, Zhu L, Sun X. Advances in Understanding the Roles of Mesenchymal Stem Cells in Lung Cancer. Cell Reprogram 2023; 25:20-31. [PMID: 36594933 DOI: 10.1089/cell.2022.0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Lung cancer is the most common and deadliest type of cancer worldwide. Research concerning lung cancer has made considerable progress in recent decades, but lung cancer remains the leading cause of malignancy-related mortality rate. Mesenchymal stem cells (MSCs) mainly exist in fat, umbilical cord blood, bone marrow, bone, and muscle. MSCs are a primary component of the tumor microenvironment (TME). Recent studies have shown that MSCs have roles in lung cancer-related proliferation, invasion, migration, and angiogenesis, but the underlying mechanisms are poorly understood. Because MSCs can migrate to the TME, there is increasing attention toward the use of MSCs in drugs or gene vectors for cancer treatment. This review summarizes the roles and effects of MSCs in lung cancer, while addressing clinical applications of MSCs in lung cancer treatment.
Collapse
Affiliation(s)
- Wenli Ding
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China.,Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China.,Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China.,Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Qinying Li
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China.,Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Linfei Xu
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yanhui Ma
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China.,Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China.,Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Liang Zhu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China.,Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China.,Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| |
Collapse
|
13
|
Crippa S, Conti A, Vavassori V, Ferrari S, Beretta S, Rivis S, Bosotti R, Scala S, Pirroni S, Jofra-Hernandez R, Santi L, Basso-Ricci L, Merelli I, Genovese P, Aiuti A, Naldini L, Di Micco R, Bernardo ME. Mesenchymal stromal cells improve the transplantation outcome of CRISPR-Cas9 gene-edited human HSPCs. Mol Ther 2023; 31:230-248. [PMID: 35982622 PMCID: PMC9840125 DOI: 10.1016/j.ymthe.2022.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/29/2022] [Accepted: 08/12/2022] [Indexed: 01/26/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) have been employed in vitro to support hematopoietic stem and progenitor cell (HSPC) expansion and in vivo to promote HSPC engraftment. Based on these studies, we developed an MSC-based co-culture system to optimize the transplantation outcome of clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 gene-edited (GE) human HSPCs. We show that bone marrow (BM)-MSCs produce several hematopoietic supportive and anti-inflammatory factors capable of alleviating the proliferation arrest and mitigating the apoptotic and inflammatory programs activated in GE-HSPCs, improving their expansion and clonogenic potential in vitro. The use of BM-MSCs resulted in superior human engraftment and increased clonal output of GE-HSPCs contributing to the early phase of hematological reconstitution in the peripheral blood of transplanted mice. In conclusion, our work poses the biological bases for a novel clinical use of BM-MSCs to promote engraftment of GE-HSPCs and improve their transplantation outcome.
Collapse
Affiliation(s)
- Stefania Crippa
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.
| | - Anastasia Conti
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Valentina Vavassori
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Samuele Ferrari
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Stefano Beretta
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Silvia Rivis
- Laboratory of Tumor Inflammation and Angiogenesis, VIB-KULeuven, 3000 Leuven, Belgium
| | - Roberto Bosotti
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Serena Scala
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | - Raisa Jofra-Hernandez
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ludovica Santi
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Luca Basso-Ricci
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ivan Merelli
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; National Research Council, Institute for Biomedical Technologies, 20132 Milan, Italy
| | - Pietro Genovese
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Department of Pediatric Oncology, Harvard Medical School, Boston, MA 02115, USA
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Pediatric Immunohematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy; (")Vita Salute" San Raffaele University, 20132 Milan, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (")Vita Salute" San Raffaele University, 20132 Milan, Italy
| | - Raffaella Di Micco
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Maria Ester Bernardo
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Pediatric Immunohematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy; (")Vita Salute" San Raffaele University, 20132 Milan, Italy.
| |
Collapse
|
14
|
Regenerative Potential of Granulation Tissue in Periodontitis: A Systematic Review and Meta-analysis. Stem Cells Int 2023; 2023:8789852. [PMID: 36926181 PMCID: PMC10014158 DOI: 10.1155/2023/8789852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/27/2022] [Accepted: 02/15/2023] [Indexed: 03/12/2023] Open
Abstract
Methods Electronic searches were conducted in five databases including CENTRAL, MEDLINE, EMBASE, Web of Science, and Dentistry & Oral Sciences Source using a combination of MeSH terms and keywords up to 21 June 2022. Human studies including patients aged over 18 years with all forms of periodontitis were included. Following the risk of bias assessment, both qualitative and quantitative analyses were performed. Results A total of twelve studies were included in qualitative analysis and six of them in quantitative analyses. The evidence suggested that cells derived from periodontitis granulation tissue have osteogenic, adipogenic, chondrogenic, neurogenic, and angiogenic differentiation abilities as well as immunoregulatory properties. In particular, CD44+, CD73+, CD90+, CD105+, and CD146+ cells were found widely in granulation tissue whilst the only meta-analysis confirmed that CD90+ cells were present in lower numbers within the granulation tissue when compared with healthy periodontal tissue (WMD = -23.43%, 95% CI -30.43 to -16.44, p < 0.00001). Conclusions This review provided further evidence that granulation tissue from patients with periodontitis can be a potential stem cell source for regenerative therapy.
Collapse
|
15
|
Analysis on the desert adaptability of indigenous sheep in the southern edge of Taklimakan Desert. Sci Rep 2022; 12:12264. [PMID: 35851076 PMCID: PMC9293982 DOI: 10.1038/s41598-022-15986-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 07/04/2022] [Indexed: 11/25/2022] Open
Abstract
The southern margin of the Taklimakan Desert is characterized by low rainfall, heavy sandstorms, sparse vegetation and harsh ecological environment. The indigenous sheep in this area are rich in resources, with the advantages of perennial estrus and good resistance to stress in most sheep. Exploring the molecular markers of livestock adaptability in this environment will provide the molecular basis for breeding research to cope with extreme future changes in the desert environment. In this study, we analyzed the population genetic structure and linkage imbalance of five sheep breeds with three different agricultural geographic characteristics using four complementary genomic selection signals: fixation index (FST), cross-population extended haplotype homozygosity (xp-EHH), Rsb (extended haplotype homozygosity between-populations) and iHS (integrated haplotype homozygosity score). We used Illumina Ovine SNP 50K Genotyping BeadChip Array, and gene annotation and enrichment analysis were performed on selected regions of the obtained genome. The ovary of Qira Black sheep (Follicular phase, Luteal phase, 30th day of pregnancy, 45th day of pregnancy) was collected, and the differentially expressed genes were screened by transcriptomic sequencing. Genome-wide selective sweep results and transcriptome data were combined for association analysis to obtain candidate genes associated with perennial estrus and stable reproduction. In order to verify the significance of the results, 15 resulting genes were randomly selected for fluorescence quantitative analysis. The results showed that Dolang sheep and Qira Black sheep evolved from Kazak sheep. Linkage disequilibrium analysis showed that the decay rate of sheep breeds in the Taklimakan Desert was higher than that in Yili grassland. The signals of FST, xp-EHH, Rsb and iHS detected 526, 332, 308 and 408 genes, respectively, under the threshold of 1% and 17 overlapping genes under the threshold of 5%. A total of 29 genes were detected in association analysis of whole-genome and transcriptome data. This study reveals the genetic mechanism of perennial estrus and environmental adaptability of indigenous sheep breeds in the Taklimakan Desert. It provides a theoretical basis for the conservation and exploitation of genetic resources of indigenous sheep breeds in extreme desert environment. This provides a new perspective for the quick adaptation of sheep and other mammals to extreme environments and future climate changes.
Collapse
|
16
|
Rosa RH, Xie W, Zhao M, Tsai SH, Roddy GW, Su MG, Potts LB, Hein TW, Kuo L. Intravitreal Administration of Stanniocalcin-1 Rescues Photoreceptor Degeneration with Reduced Oxidative Stress and Inflammation in a Porcine Model of Retinitis Pigmentosa. Am J Ophthalmol 2022; 239:230-243. [PMID: 35307380 DOI: 10.1016/j.ajo.2022.03.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/09/2022] [Accepted: 03/12/2022] [Indexed: 12/01/2022]
Abstract
PURPOSE To investigate the effect of stanniocalcin-1 (STC-1), a secreted polypeptide exhibiting multiple functions in cell survival and death, on photoreceptor degeneration in a porcine model of retinitis pigmentosa (RP). METHODS P23H transgenic pigs (TG P23H) and wild-type hybrid littermates were obtained from the National Swine Resource and Research Center. Human recombinant STC-1 was injected intravitreally every 2 weeks from postnatal day 15 (P15) to P75. The contralateral eye was injected with balanced salt solution as a control. Electroretinography (ERG) and spectral domain optical coherence tomography (SD-OCT) were performed to evaluate retinal function and morphology in vivo at P90. Retinal tissue was collected for histologic analysis and molecular assays to evaluate the antioxidative and anti-inflammatory mechanisms by which STC-1 may rescue photoreceptor degeneration. RESULTS Intravitreal injection of STC-1 improved retinal function in TG P23H pigs with increased photopic and flicker ERG a- and b-wave amplitudes. Greater integrity of the ellipsoid zone (EZ) band on SD-OCT and morphologic rescue with preservation of cone photoreceptors were observed in STC-1-treated TG P23H pigs. STC-1 altered gene expression in TG P23H pig retina on microarray analysis and increased photoreceptor specific gene expression by reverse transcription-polymerase chain reaction analysis. STC-1 significantly decreased oxidative stress and the expressions of NLRP3 inflammasome, cleaved caspase-1, and IL-1β in TG P23H pig retina. CONCLUSIONS Intravitreal administration of STC-1 enhances cone photoreceptor function, improves EZ integrity, and reduces retinal degeneration through antioxidative and anti-inflammatory effects in a large animal (pig) model of the most common form of autosomal dominant RP in the United States.
Collapse
Affiliation(s)
- Robert H Rosa
- Department of Medical Physiology, Texas A&M University Health Science Center, Bryan, TX, USA (RR, WX, MZ, ST, LP, TH, LK); Department of Ophthalmology, Baylor Scott & White Eye Institute, Temple, TX, USA (RR, WX, MZ, MS, LP, TH, LK).
| | - Wankun Xie
- Department of Medical Physiology, Texas A&M University Health Science Center, Bryan, TX, USA (RR, WX, MZ, ST, LP, TH, LK); Department of Ophthalmology, Baylor Scott & White Eye Institute, Temple, TX, USA (RR, WX, MZ, MS, LP, TH, LK)
| | - Min Zhao
- Department of Medical Physiology, Texas A&M University Health Science Center, Bryan, TX, USA (RR, WX, MZ, ST, LP, TH, LK); Department of Ophthalmology, Baylor Scott & White Eye Institute, Temple, TX, USA (RR, WX, MZ, MS, LP, TH, LK)
| | - Shu-Huai Tsai
- Department of Medical Physiology, Texas A&M University Health Science Center, Bryan, TX, USA (RR, WX, MZ, ST, LP, TH, LK)
| | - Gavin W Roddy
- Department of Ophthalmology, Mayo Clinic, Rochester, MN (GR)
| | - Maxwell G Su
- Department of Ophthalmology, Baylor Scott & White Eye Institute, Temple, TX, USA (RR, WX, MZ, MS, LP, TH, LK)
| | - Luke B Potts
- Department of Medical Physiology, Texas A&M University Health Science Center, Bryan, TX, USA (RR, WX, MZ, ST, LP, TH, LK); Department of Ophthalmology, Baylor Scott & White Eye Institute, Temple, TX, USA (RR, WX, MZ, MS, LP, TH, LK)
| | - Travis W Hein
- Department of Medical Physiology, Texas A&M University Health Science Center, Bryan, TX, USA (RR, WX, MZ, ST, LP, TH, LK); Department of Ophthalmology, Baylor Scott & White Eye Institute, Temple, TX, USA (RR, WX, MZ, MS, LP, TH, LK)
| | - Lih Kuo
- Department of Medical Physiology, Texas A&M University Health Science Center, Bryan, TX, USA (RR, WX, MZ, ST, LP, TH, LK); Department of Ophthalmology, Baylor Scott & White Eye Institute, Temple, TX, USA (RR, WX, MZ, MS, LP, TH, LK)
| |
Collapse
|
17
|
Cai Y, Zhang MM, Wang M, Jiang ZH, Tan ZG. Bone Marrow-Derived Mesenchymal Stem Cell-Derived Exosomes Containing Gli1 Alleviate Microglial Activation and Neuronal Apoptosis In Vitro and in a Mouse Parkinson Disease Model by Direct Inhibition of Sp1 Signaling. J Neuropathol Exp Neurol 2022; 81:522-534. [PMID: 35609560 DOI: 10.1093/jnen/nlac037] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
This study investigated possible therapeutic effect mechanisms of exosomes from bone marrow-derived mesenchymal stem cells (BMSC) in neuronal and microglial cells and in a Parkinson disease (PD) model. Neuronal SH-SY5Y cells and microglial HMC3 cells were subjected to 1-methyl-4-phenylpyridinium (MPP+) or LPS, respectively. The mRNA and protein expression was assessed using qRT-PCR, Western blotting, and enzyme-linked immunosorbent assay. Cell viability and apoptosis of SH-SY5Y cells were examined using the MTT assay and flow cytometry. Chromatin immunoprecipitation assays were performed to assess the binding relationship between glioma-associated oncogene homolog 1 (Gli1) and the Sp1 transcription factor promoter. BMSC-derived exosomes promoted cell proliferation and inhibited apoptosis in MPP+-treated SH-SY5Y cells and suppressed inflammatory markers in LPS-treated HMC3 cells. Sp1 knockdown decreased SH-SY5Y cell damage and HMC3 immune activation. Gli1 carried by BMSC exosomes directly bound with Sp1 to inhibit Sp1-mediated LRRK2 activation whereas exosomes secreted by Gli1-knockdown in BMSC did not. In a PD mouse model induced with MPTP, BMSC exosomes decreased neuron loss injury and the inflammatory response by inhibiting Sp1 signaling. Thus, BMSC-derived exosomal Gli1 alleviates inflammatory damage and neuronal apoptosis by inhibiting Sp1 in vitro and in vivo. These findings provide the basis for the potential clinical use of BMSC-derived exosomes in PD.
Collapse
Affiliation(s)
- Yang Cai
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province 410011, P.R. China
| | - Ming-Ming Zhang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province 410011, P.R. China
| | - Ming Wang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province 410011, P.R. China
| | - Zhuo-Hang Jiang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province 410011, P.R. China
| | - Zhi-Gang Tan
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province 410011, P.R. China
| |
Collapse
|
18
|
Ohkouchi S, Kanehira M, Saigusa D, Ono M, Tazawa R, Terunuma H, Hirano T, Numakura T, Notsuda H, Inoue C, Saito-Koyama R, Tabata M, Irokawa T, Ogawa H, Kurosawa H, Okada Y. Metabolic and Epigenetic Regulation of SMAD7 by Stanniocalcin-1 (STC1) Ameliorates Lung Fibrosis. Am J Respir Cell Mol Biol 2022; 67:320-333. [PMID: 35696344 DOI: 10.1165/rcmb.2021-0445oc] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
As shown in our previous studies, the intratracheal-administration of stanniocalcin-1 (STC1) ameliorates pulmonary fibrosis by reducing oxidative and endoplasmic reticulum stress through the uncoupling of respiration in a bleomycin (BLM)-treated mouse model. However, the overall effect of STC1 on metabolism was not examined. Therefore, we first conducted a comprehensive metabolomics analysis to screen the overall metabolic changes induced by STC1 in an alveolar epithelial cell line using capillary electrophoresis time-of-flight mass spectrometry (CE-TOFMS). The results were subsequently validated in multiple alveolar epithelial and fibroblast cell lines by performing precise analyses of each substance. STC1 stimulated glycolysis, acetyl-CoA synthesis, and the methionine and cysteine-glutathione pathways, which are closely related to the uncoupling of respiration, modulation of epigenetics and reduction in oxidative stress. These results are consistent with our previous study. Subsequently, we focused on the inhibitory factor SMAD7, which exerts an antifibrotic effect and is susceptible to epigenetic regulation. STC1 upregulates SMAD7 in an uncoupling protein 2-dependent manner, induces demethylation of the SMAD7 promoter region and acetylation of the SMAD7 protein in human alveolar epithelial and fibroblast cell lines and a BLM-treated mouse model, and subsequently attenuates fibrosis. The antifibrotic effects of STC1 may partially depend on the regulation of SMAD7. In the evaluation using lung tissue from idiopathic pulmonary fibrosis patients, SMAD7 expression and acetylation were high in the alveolar structure-preserving region and low in the fibrotic region. The intratracheal-administration of STC1 may prevent the development of pulmonary fibrosis by regulating the metabolism-mediated epigenetic modification of SMAD7 in patients.
Collapse
Affiliation(s)
- Shinya Ohkouchi
- Tohoku University Graduate School of Medicine, Occupational Health, Sendai, Japan.,Institute of Devepolment, Aging and Cancer, Tohoku University, Department of Thoracic surgery, Sendai, Japan;
| | - Masahiko Kanehira
- University of Yamanashi, 38146, Center for Life Science Research, Kofu, Japan.,Institute of Devepolment, Aging and Cancer, Tohoku University, Department of Thoracic surgery, Sendai, Japan
| | - Daisuke Saigusa
- Teikyo University School of Medicine Graduate School of Medicine, 36906, Faculty of Pharma-Science, Itabashi-ku, Japan
| | - Manabu Ono
- Tohoku University Graduate School of Medicine, Department of Respiratory Medicine, Sendai, Japan
| | - Ryushi Tazawa
- Tokyo Medical and Dental University, 13100, Student Support and Health Administration Organization, Tokyo, Japan.,Niigata University, 12978, Medical and Dental Hospital, Niigata, Japan
| | | | - Taizou Hirano
- Tohoku University Graduate School of Medicine, Department of Respiratory Medicine, Sensai, Japan
| | - Tadahisa Numakura
- Tohoku University Graduate School of Medicine, Department of Respiratory Medicine, Sendai, Japan
| | - Hirotsugu Notsuda
- Institute of Devepolment, Aging and Cancer, Tohoku University, Department of Thoracic surgery, Sendai, Japan
| | - Chihiro Inoue
- Tohoku University Graduate School of Medicine, Department of Anatomic Pathology, Sendai, Japan
| | - Ryoko Saito-Koyama
- Tohoku University Graduate School of Medicine, Department of Anatomic Pathology, Sendai, Japan
| | - Masao Tabata
- Tohoku University Graduate School of Medicine, Occupational Health, Sendai, Japan
| | - Toshiya Irokawa
- Tohoku University Graduate School of Medicine, Occupational Health, Sendai, Japan
| | - Hiromasa Ogawa
- Tohoku University Graduate School of Medicine, Occupational Health, Sendai, Japan
| | - Hajime Kurosawa
- Tohoku University Graduate School of Medicine, Occupational Health, Sendai, Japan
| | - Yoshinori Okada
- Institute of Devepolment, Aging and Cancer, Tohoku University, Department of Thoracic surgery, Sendai, Japan
| |
Collapse
|
19
|
Cai JH, Sun YT, Bao S. HucMSCs-exosomes containing miR-21 promoted estrogen production in ovarian granulosa cells via LATS1-mediated phosphorylation of LOXL2 and YAP. Gen Comp Endocrinol 2022; 321-322:114015. [PMID: 35271888 DOI: 10.1016/j.ygcen.2022.114015] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/23/2022] [Accepted: 03/03/2022] [Indexed: 12/22/2022]
Abstract
BACKGROUND Premature ovarian failure (POF) is one of the common disorders found in women leading to 1% female infertility. Clinical features of POF are hypoestrogenism or estrogen deficiency. With the development of regenerative medicine, human mesenchymal stem cells (hMSCs) therapy brings new prospects for POF. This research aims to reveal the therapeutic effects and potential mechanisms of human umbilical cord mesenchymal stem cells (hucMSCs)-derived exosomes on POF. METHODS The mRNA and protein expressions in hucMSCs and ovarian granulosa cells (KGN and SVOG cells) were assessed using qRT-PCR and western blot. ELISA assay was performed to evaluate estradiol (E2) secretion in granulosa cells. The binding relationship between miR-21 and LATS1 was verified by dual-luciferase reporter assay and RNA binding protein immunoprecipitation assay (RIP) assay. Additionally, Immunoprecipitation assay was carried out to confirm Lysyl oxidase like 2 (LOXL2) was phosphorylated by large tumor suppressor 1 (LATS1). Finally, the binding relationships between Yes-associated protein (YAP), StAR and LOXL2 were verified by dual-luciferase reporter assay and/or chromatin immunoprecipitation assay (ChIP) assay. RESULTS Here our results displayed that miR-21 was overexpressed in hucMSCs and hucMSCs-derived exosomes, compared with that ovarian granulosa cells. hucMSC-exo with overexpressing miR-21 could markedly promote the secretion of estrogen in ovarian granulosa cells. LATS1 overexpression in ovarian granulosa cells reduced the secretion of estrogen. We subsequently confirmed that LATS1 was the target of miR-21. In addition, LATS1 could regulate StAR expression by phosphorylating LOXL2 and YAP. CONCLUSION miR-21 carried by hucMSCs-derived exosomes could downregulate LATS1, thereby reducing phosphorylated LOXL2 and YAP, and ultimately promoting estrogen secretion in ovarian granulosa cells.
Collapse
Affiliation(s)
- Jun-Hong Cai
- Central Laboratory, Hainan General Hospital/Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province 570311, People's Republic of China
| | - Yu-Ting Sun
- Hainan Medical University, Haikou, Hainan Province 571199, People's Republic of China
| | - Shan Bao
- Department of Gynaecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University/Hainan General Hospital, Haikou, Hainan Province 570311, People's Republic of China.
| |
Collapse
|
20
|
Bone marrow mesenchymal stem cells-derived exosomes containing miR-539-5p inhibit pyroptosis through NLRP3/caspase-1 signalling to alleviate inflammatory bowel disease. Inflamm Res 2022; 71:833-846. [PMID: 35637388 DOI: 10.1007/s00011-022-01577-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/22/2022] [Accepted: 04/14/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Exosomes derived from bone mesenchymal stem cells (BMSCs) are potential candidates for inflammatory bowel disease (IBD) treatment. The present study investigated the therapeutic effect and potential mechanism of BMSCs-derived exosomes on pyroptosis in IBD. METHODS We induced IBD in mice and cell models through dextran sulfate sodium (DSS) and LPS, respectively. The mRNA and protein expression levels were assessed by qRT-PCR, Western blotting, IF and IHC. The concentrations of IL-1β, IL-18 and TNFα were assessed using ELISA. ROS levels were determined using DCFH-DA staining. Cell proliferation of mIECs was analysed using an MTT assay. In addition, a flow cytometry assay was performed to detect pyroptosis. Finally, the binding relationship between miR-539-5p and NLRP3 was verified by a dual luciferase reporter gene assay. RESULTS Our results revealed that intraperitoneal injection of BMSCs-derived exosomes inhibited DSS-induced pyroptosis as well as IBD symptoms in mice. In addition, BMSCs-derived exosome treatment suppressed pyroptosis, ROS levels and the concentrations of proinflammatory cytokines (IL-1β, IL-18 and TNFα) in LPS-treated mIECs in a miR-539-5p-dependent manner. Further research found that miR-539-5p suppressed NLRP3 expression in mIECs by directly targeting NLRP3. As expected, pyroptosis in LPS-treated mIECs was significantly reduced by NLRP3 knockdown. In addition, NLRP3 silencing restored the inhibitory effect of exosomes derived from BMSCs transfected with miR-539-5p inhibitor on pyroptosis in LPS-treated mIECs. CONCLUSION The present study demonstrated that BMSCs-derived exosomal miR-539-5p suppresses pyroptosis through NLRP3/caspase-1 signalling to inhibit IBD progression.
Collapse
|
21
|
Molnar V, Pavelić E, Vrdoljak K, Čemerin M, Klarić E, Matišić V, Bjelica R, Brlek P, Kovačić I, Tremolada C, Primorac D. Mesenchymal Stem Cell Mechanisms of Action and Clinical Effects in Osteoarthritis: A Narrative Review. Genes (Basel) 2022; 13:genes13060949. [PMID: 35741711 PMCID: PMC9222975 DOI: 10.3390/genes13060949] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
With the insufficient satisfaction rates and high cost of operative treatment for osteoarthritis (OA), alternatives have been sought. Furthermore, the inability of current medications to arrest disease progression has led to rapidly growing clinical research relating to mesenchymal stem cells (MSCs). The availability and function of MSCs vary according to tissue source. The three primary sources include the placenta, bone marrow, and adipose tissue, all of which offer excellent safety profiles. The primary mechanisms of action are trophic and immunomodulatory effects, which prevent the further degradation of joints. However, the function and degree to which benefits are observed vary significantly based on the exosomes secreted by MSCs. Paracrine and autocrine mechanisms prevent cell apoptosis and tissue fibrosis, initiate angiogenesis, and stimulate mitosis via growth factors. MSCs have even been shown to exhibit antimicrobial effects. Clinical results incorporating clinical scores and objective radiological imaging have been promising, but a lack of standardization in isolating MSCs prevents their incorporation in current guidelines.
Collapse
Affiliation(s)
- Vilim Molnar
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (E.P.); (E.K.); (V.M.); (P.B.)
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Eduard Pavelić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (E.P.); (E.K.); (V.M.); (P.B.)
| | - Kristijan Vrdoljak
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (K.V.); (M.Č.)
| | - Martin Čemerin
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (K.V.); (M.Č.)
| | - Emil Klarić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (E.P.); (E.K.); (V.M.); (P.B.)
| | - Vid Matišić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (E.P.); (E.K.); (V.M.); (P.B.)
| | - Roko Bjelica
- Department of Oral Surgery, School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Petar Brlek
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (E.P.); (E.K.); (V.M.); (P.B.)
| | | | | | - Dragan Primorac
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (E.P.); (E.K.); (V.M.); (P.B.)
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Medical School, University of Split, 21000 Split, Croatia
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
- Medical School REGIOMED, 96450 Coburg, Germany
- Eberly College of Science, The Pennsylvania State University, University Park, PA 16802, USA
- The Henry C. Lee College of Criminal Justice and Forensic Sciences, University of New Haven, West Haven, CT 06516, USA
- Correspondence:
| |
Collapse
|
22
|
Velarde F, Ezquerra S, Delbruyere X, Caicedo A, Hidalgo Y, Khoury M. Mesenchymal stem cell-mediated transfer of mitochondria: mechanisms and functional impact. Cell Mol Life Sci 2022; 79:177. [PMID: 35247083 PMCID: PMC11073024 DOI: 10.1007/s00018-022-04207-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 01/27/2022] [Accepted: 02/11/2022] [Indexed: 12/13/2022]
Abstract
There is a steadily growing interest in the use of mitochondria as therapeutic agents. The use of mitochondria derived from mesenchymal stem/stromal cells (MSCs) for therapeutic purposes represents an innovative approach to treat many diseases (immune deregulation, inflammation-related disorders, wound healing, ischemic events, and aging) with an increasing amount of promising evidence, ranging from preclinical to clinical research. Furthermore, the eventual reversal, induced by the intercellular mitochondrial transfer, of the metabolic and pro-inflammatory profile, opens new avenues to the understanding of diseases' etiology, their relation to both systemic and local risk factors, and also leads to new therapeutic tools for the control of inflammatory and degenerative diseases. To this end, we illustrate in this review, the triggers and mechanisms behind the transfer of mitochondria employed by MSCs and the underlying benefits as well as the possible adverse effects of MSCs mitochondrial exchange. We relay the rationale and opportunities for the use of these organelles in the clinic as cell-based product.
Collapse
Affiliation(s)
- Francesca Velarde
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Sarah Ezquerra
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Xavier Delbruyere
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Andres Caicedo
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
- Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina iBioMed, Quito, Ecuador
- Mito-Act Research Consortium, Quito, Ecuador
- Sistemas Médicos SIME, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Yessia Hidalgo
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile.
| | - Maroun Khoury
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile.
| |
Collapse
|
23
|
Urtatiz O, Haage A, Tanentzapf G, Van Raamsdonk CD. Crosstalk with keratinocytes causes GNAQ oncogene specificity in melanoma. eLife 2021; 10:71825. [PMID: 34939927 PMCID: PMC8747508 DOI: 10.7554/elife.71825] [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: 06/30/2021] [Accepted: 12/21/2021] [Indexed: 11/13/2022] Open
Abstract
Different melanoma subtypes exhibit specific and non-overlapping sets of oncogene and tumor suppressor mutations, despite a common cell of origin in melanocytes. For example, activation of the Gαq/11 signaling pathway is a characteristic initiating event in primary melanomas that arise in the dermis, uveal tract, or central nervous system. It is rare in melanomas arising in the epidermis. The mechanism for this specificity is unknown. Here, we present evidence that in the mouse, crosstalk with the epidermal microenvironment actively impairs the survival of melanocytes expressing the GNAQQ209L oncogene. We found that GNAQQ209L, in combination with signaling from the interfollicular epidermis (IFE), stimulates dendrite extension, leads to actin cytoskeleton disorganization, inhibits proliferation, and promotes apoptosis in melanocytes. The effect was reversible and paracrine. In contrast, the epidermal environment increased the survival of wildtype and BrafV600E expressing melanocytes. Hence, our studies reveal the flip side of Gαq/11 signaling, which was hitherto unsuspected. In the future, the identification of the epidermal signals that restrain the GNAQQ209L oncogene could suggest novel therapies for GNAQ and GNA11 mutant melanomas.
Collapse
Affiliation(s)
- Oscar Urtatiz
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Amanda Haage
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
24
|
Dunbar H, Weiss DJ, Rolandsson Enes S, Laffey JG, English K. The Inflammatory Lung Microenvironment; a Key Mediator in MSC Licensing. Cells 2021; 10:cells10112982. [PMID: 34831203 PMCID: PMC8616504 DOI: 10.3390/cells10112982] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022] Open
Abstract
Recent clinical trials of mesenchymal stromal cell (MSC) therapy for various inflammatory conditions have highlighted the significant benefit to patients who respond to MSC administration. Thus, there is strong interest in investigating MSC therapy in acute inflammatory lung conditions, such as acute respiratory distress syndrome (ARDS). Unfortunately, not all patients respond, and evidence now suggests that the differential disease microenvironment present across patients and sub-phenotypes of disease or across disease severities influences MSC licensing, function and therapeutic efficacy. Here, we discuss the importance of licensing MSCs and the need to better understand how the disease microenvironment influences MSC activation and therapeutic actions, in addition to the need for a patient-stratification approach.
Collapse
Affiliation(s)
- Hazel Dunbar
- Department of Biology, Maynooth University, W23 F2H6 Maynooth, Ireland;
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Ireland
| | - Daniel J Weiss
- Department of Medicine, 226 Health Science Research Facility, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA;
| | - Sara Rolandsson Enes
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22100 Lund, Sweden;
| | - John G Laffey
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, H91 W2TY Galway, Ireland;
- Department of Anaesthesia, Galway University Hospitals, SAOLTA University Health Group, H91 YR71 Galway, Ireland
| | - Karen English
- Department of Biology, Maynooth University, W23 F2H6 Maynooth, Ireland;
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Ireland
- Correspondence: ; Tel.: +353-1-7086290
| |
Collapse
|
25
|
Park JM, Han YM, Hwang SJ, Kim SJ, Hahm KB. Therapeutic effects of placenta derived-, umbilical cord derived-, and adipose tissue derived-mesenchymal stem cells in chronic Helicobacter pylori infection: comparison and novel mechanisms. J Clin Biochem Nutr 2021; 69:188-202. [PMID: 34616110 PMCID: PMC8482378 DOI: 10.3164/jcbn.20-151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/13/2020] [Indexed: 12/13/2022] Open
Abstract
Supported with significant rejuvenating and regenerating actions of mesenchymal stem cells (MSCs) in various gastrointestinal diseases including Helicobacter pylori (H. pylori)-associated gastric diseases, we have compared these actions among placenta derived-MSCs (PD-MSCs), umbilical cord derived-MSCs (UC-MSCs), and adipose tissue derived-MSCs (AD-MSCs) and explored contributing genes implicated in rejuvenation of H. pylori-chronic atrophic gastritis (CAG) and tumorigenesis. In this study adopting H. pylori-initiated, high salt diet-promoted gastric carcinogenesis model, we have administered three kinds of MSCs around 15-18 weeks in H. pylori infected C57BL/6 mice and sacrificed at 24 and 48 weeks, respectively, in order to either assess the rejuvenating capability or anti-tumorigenesis. At 24 weeks, MSCs all led to significantly mitigated atrophic gastritis, for which significant inductions of autophagy, preservation of tumor suppressive 15-PGDH, attenuated apoptosis, and efficient efferocytosis was imposed with MSCs administration during atrophic gastritis. At 48 weeks, MSCs administered during H. pylori-associated atrophic gastritis afforded significant blocking the progression of CAG, as evidenced with statistically significant reduction in H. pylori-associated gastric tumor (p<0.05) accompanied with significant decreases in IL-1β, COX-2, STAT3, and NF-κB. Combined together with the changes of stanniocalcin-1 (STC-1), thrombospondin-1 (TSP-1), and IL-10 known as biomarkers reflecting stem cell activities at 48 weeks after H. pylori, PD-MSCs among MSCs afforded the best rejuvenating action against H. pylori-associated CAG via additional actions of efferocytosis, autophagy, and anti-apoptosis at 24 weeks. In conclusion, MSCs, especially PD-MSCs, exerted rejuvenating actions against H. pylori-associated CAG via anti-mutagenesis of IL-10, CD-36, ATG5 and cancer suppressive influences of STC-1, TSP-1, and 15-PGDH.
Collapse
Affiliation(s)
- Jong Min Park
- College of Oriental Medicine, Daejeon University, Daehak-ro 62, Dong-gu, Daejeon 34520, Korea
| | - Young Min Han
- Western Seoul Center, Korea Basic Science Institute, University-Industry Cooperate Building, 150 Bugahyeon-ro, Seodaemun-gu, Seoul 03759, Korea
| | - Sun Jin Hwang
- Medpacto Research Institute, Medpacto, Myungdal-ro 92, Seocho-gu, Seoul 06668, Korea
| | - Seong Jin Kim
- Medpacto Research Institute, Medpacto, Myungdal-ro 92, Seocho-gu, Seoul 06668, Korea
| | - Ki Baik Hahm
- Medpacto Research Institute, Medpacto, Myungdal-ro 92, Seocho-gu, Seoul 06668, Korea.,CHA Cancer Preventive Research Center, CHA Bio Complex, CHA University, 330 Pangyo-dong, Bundang-gu, Seongnam 13497, Korea
| |
Collapse
|
26
|
Xiong WP, Yao WQ, Wang B, Liu K. BMSCs-exosomes containing GDF-15 alleviated SH-SY5Y cell injury model of Alzheimer's disease via AKT/GSK-3β/β-catenin. Brain Res Bull 2021; 177:92-102. [PMID: 34536520 DOI: 10.1016/j.brainresbull.2021.09.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 08/27/2021] [Accepted: 09/12/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) therapy has great potential for Alzheimer's disease (AD) treatment. Here, we investigated the roles of BMSCs-exosomes containing growth differentiation factor-15 (GDF-15) in regulating SH-SY5Y cell injury in AD. METHODS The SH-SY5Y cell injury model was constructed by treating SH-SY5Y cells with 10 μM Aβ42. GDF-15 expression was assessed using qRT-PCR and western blot. CCK8 assay and flow cytometry assay were employed to elevate cell proliferation and apoptosis, respectively. The expression levels of inflammatory factors (IL-6, IL-1β, TNFα and IL-8) and Aβ42 were detected using ELISA. Besides, the levels of apoptosis-related proteins and AKT pathway-related proteins were determined using western blot. RESULTS Our results displayed that BMSCs-EVs treatment elevated cell viability, while suppressed cell apoptosis and inflammation in Aβ42-treated SH-SY5Y cells. Exosomes secreted by BMSCs after GDF-15 silence lost the ability to restore Aβ42-induced SH-SY5Y cell damage. GDF-15 treatment restored Aβ42-induced SH-SY5Y cell damage, while it was eliminated by AKT pathway inhibition. BMSCs-exosomes containing GDF-15 upregulated NEP and IDE via activation of AKT/GSK-3β/β-catenin pathway, thereby degrading Aβ42 protein to relieve SH-SY5Y cell damage. CONCLUSION BMSCs-exosomes containing GDF-15 alleviated SH-SY5Y cell damage via AKT/GSK-3β/β-catenin. Our work confers a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Wen-Ping Xiong
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.169, East Lake Road, Wuhan 430071, Hubei Province, PR China
| | - Wei-Qi Yao
- Department of Hematology, Union Hospital, Huazhong University of Science and Technology, Hubei Engineering Research Center for Human Stem Cell Preparation and Application and Resource Conservation, Wuhan 430071, Hubei Province, PR China
| | - Bei Wang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.169, East Lake Road, Wuhan 430071, Hubei Province, PR China
| | - Kui Liu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.169, East Lake Road, Wuhan 430071, Hubei Province, PR China.
| |
Collapse
|
27
|
The secretome of mesenchymal stem cells and oxidative stress: challenges and opportunities in cell-free regenerative medicine. Mol Biol Rep 2021; 48:5607-5619. [PMID: 34191238 DOI: 10.1007/s11033-021-06360-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/16/2021] [Indexed: 12/15/2022]
Abstract
Over the last decade, mesenchymal stem cells (MSCs) have been considered a suitable source for cell-based therapy, especially in regenerative medicine. First, the efficacy and functions of MSCs in clinical applications have been attributed to their differentiation ability, called homing and differentiation. However, it has recently been confirmed that MSCs mostly exert their therapeutic effects through soluble paracrine bioactive factors and extracellular vesicles, especially secretome. These secreted components play critical roles in modulating immune responses, improving the survival, and increasing the regeneration of damaged tissues. The secretome content of MSCs is variable under different conditions. Oxidative stress (OS) is one of these conditions that is highly important in MSC therapy and regenerative medicine. High levels of reactive oxygen species (ROS) are produced during isolation, cell culture, and transplantation lead to OS, which induces cell death and apoptosis and limits the efficacy of their regeneration capability. In turn, the preconditioning of MSCs in OS conditions contributes to the secretion of several proteins, cytokines, growth factors, and exosomes, which can improve the antioxidant potential of MSCs against OS. This potential of MSC secretome has turned it into a new promising cell-free tissue regeneration strategy.This review provides a view of MSC secretome under OS conditions, focusing on different secretome contents of MSCs and thier possible therapeutic potential against cell therapy.
Collapse
|
28
|
Abusharkh HA, Mallah AH, Amr MM, Mendenhall J, Gozen BA, Tingstad EM, Abu-Lail NI, Van Wie BJ. Enhanced matrix production by cocultivated human stem cells and chondrocytes under concurrent mechanical strain. In Vitro Cell Dev Biol Anim 2021; 57:631-640. [PMID: 34129185 DOI: 10.1007/s11626-021-00592-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 05/10/2021] [Indexed: 11/28/2022]
Abstract
Conventional treatments of osteoarthritis have failed to re-build functional articular cartilage. Tissue engineering clinical treatments for osteoarthritis, including autologous chondrocyte implantation, provides an alternative approach by injecting a cell suspension to fill lesions within the cartilage in osteoarthritic knees. The success of chondrocyte implantation relies on the availability of chondrogenic cell lines, and their resilience to high mechanical loading. We hypothesize we can reduce the numbers of human articular chondrocytes necessary for a treatment by supplementing cultures with human adipose-derived stem cells, in which stem cells will have protective and stimulatory effects on mixed cultures when exposed to high mechanical loads, and in which coculture will enhance production of requisite extracellular matrix proteins over those produced by stretched chondrocytes alone. In this work, adipose-derived stem cells and articular chondrocytes were cultured separately or cocultivated at ratios of 3:1, 1:1, and 1:3 in static plates or under excessive cyclic tensile strain of 10% and results were compared to culturing of both cell types alone with and without cyclic strain. Results indicate 75% of chondrocytes in engineered articular cartilage can be replaced with stem cells with enhanced collagen over all culture conditions and glycosaminoglycan content over stretched cultures of chondrocytes. This can be done without observing adverse effects on cell viability. Collagen and glycosaminoglycan secretion, when compared to chondrocyte alone under 10% strain, was enhanced 6.1- and 2-fold, respectively, by chondrocytes cocultivated with stem cells at a ratio of 1:3.
Collapse
Affiliation(s)
- Haneen A Abusharkh
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, 1505 NE Stadium Way, Pullman, WA, 99164-6515, USA
| | - Alia H Mallah
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Mahmoud M Amr
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Juana Mendenhall
- Department of Chemistry, Morehouse College, Atlanta, GA, 30314, USA
| | - Bulent A Gozen
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA, 99164-2920, USA
| | - Edwin M Tingstad
- Inland Orthopedic Surgery and Sports Medicine Clinic, Pullman, WA, 99163, USA
| | - Nehal I Abu-Lail
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Bernard J Van Wie
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, 1505 NE Stadium Way, Pullman, WA, 99164-6515, USA.
| |
Collapse
|
29
|
Wang P, Li XL, Cao ZH. STC1 ameliorates cognitive impairment and neuroinflammation of Alzheimer's disease mice via inhibition of ERK1/2 pathway. Immunobiology 2021; 226:152092. [PMID: 34004549 DOI: 10.1016/j.imbio.2021.152092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 02/04/2023]
Abstract
OBJECTIVE To investigate the regulatory role of STC1 (Stanniocalcin-1) mediated ERK1/2 pathway in cognitive impairment and neuroinflammation of Alzheimer's disease (AD). METHODS WT mice and STC1 Tg mice (transgenic overexpression of STC1) were used to establish AD models to perform behavioral test by Morris water maze. Hippocampal cell apoptosis was quantified by TUNEL staining, the levels of inflammatory cytokines in serum and hippocampal tissues determined by ELISA, as well as oxidative stress-related factors detected by corresponding testing kits, and protein expression of STC1 and ERK1/2 pathway measured by Western blotting. RESULTS Compared with WT Sham group, WT AD mice had prolonged escape latency, decreased crossing platform times, increased hippocampal cell apoptosis with up-regulated inflammatory cytokines and oxidative stress-related factors, as well as increased STC1 and ERK1/2 pathway-related molecules. By contrast, STC1 Tg AD mice showed shortened escape latency, increased crossing platform times than WT AD mice, and they also exhibited the decreased apoptosis index and inflammatory cytokines, alleviated oxidative stress-injury, down-regulated protein expression of ERK1/2 pathway, and up-regulated the protein expression of STC1 and UCP2. CONCLUSION STC1 overexpression could alleviate oxidative stress-induced injury, reduce neuroinflammation, improve cognitive function to play a neuro-protective role by inhibiting ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Pu Wang
- Department of Neurology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Xiao-Long Li
- Department of Neurology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Zhi-Hua Cao
- Department of Neurology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China.
| |
Collapse
|
30
|
Mesenchymal stromal cells for the treatment of ocular autoimmune diseases. Prog Retin Eye Res 2021; 85:100967. [PMID: 33775824 DOI: 10.1016/j.preteyeres.2021.100967] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/17/2021] [Accepted: 03/20/2021] [Indexed: 12/22/2022]
Abstract
Mesenchymal stromal cells, commonly referred to as MSCs, have emerged as a promising cell-based therapy for a range of autoimmune diseases thanks to several therapeutic advantages. Key among these are: 1) the ability to modulate innate and adaptive immune responses and to promote tissue regeneration, 2) the ease of their isolation from readily accessible tissues and expansion at scale in culture, 3) their low immunogenicity enabling use as an allogeneic "off-the-shelf" product, and 4) MSC therapy's safety and feasibility in humans, as demonstrated in more than one thousand clinical trials. Evidence from preclinical studies and early clinical trials indicate the therapeutic potential of MSCs and their derivatives for efficacy in ocular autoimmune diseases such as autoimmune uveoretinitis and Sjögren's syndrome-related dry eye disease. In this review, we provide an overview of the current understanding of the therapeutic mechanisms of MSCs, and summarize the results from preclinical and clinical studies that have used MSCs or their derivatives for the treatment of ocular autoimmune diseases. We also discuss the challenges to the successful clinical application of MSC therapy, and suggest strategies for overcoming them.
Collapse
|
31
|
Liang W, Chen X, Zhang S, Fang J, Chen M, Xu Y, Chen X. Mesenchymal stem cells as a double-edged sword in tumor growth: focusing on MSC-derived cytokines. Cell Mol Biol Lett 2021; 26:3. [PMID: 33472580 PMCID: PMC7818947 DOI: 10.1186/s11658-020-00246-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 12/27/2020] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) show homing capacity towards tumor sites. Numerous reports indicate that they are involved in multiple tumor-promoting processes through several mechanisms, including immunosuppression; stimulation of angiogenesis; transition to cancer-associated fibroblasts; inhibition of cancer cell apoptosis; induction of epithelial-mesenchymal transition (EMT); and increase metastasis and chemoresistance. However, other studies have shown that MSCs suppress tumor growth by suppressing angiogenesis, incrementing inflammatory infiltration, apoptosis and cell cycle arrest, and inhibiting the AKT and Wnt signaling pathways. In this review, we discuss the supportive and suppressive impacts of MSCs on tumor progression and metastasis. We also discuss MSC-based therapeutic strategies for cancer based on their potential for homing to tumor sites.
Collapse
Affiliation(s)
- Wenqing Liang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, 355 Xinqiao Road, Dinghai District, Zhoushan, 316000, Zhejiang, People's Republic of China.
| | - Xiaozhen Chen
- College of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang, People's Republic of China
| | - Songou Zhang
- College of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang, People's Republic of China
| | - Jian Fang
- College of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang, People's Republic of China
| | - Meikai Chen
- Department of Orthopaedics, Shaoxing People's Hospital, The First Affiliated Hospital of Shaoxing University, Shaoxing, 312000, Zhejiang, People's Republic of China
| | - Yifan Xu
- Department of Orthopaedics, Shaoxing People's Hospital, The First Affiliated Hospital of Shaoxing University, Shaoxing, 312000, Zhejiang, People's Republic of China
| | - Xuerong Chen
- Department of Orthopaedics, Shaoxing People's Hospital, The First Affiliated Hospital of Shaoxing University, Shaoxing, 312000, Zhejiang, People's Republic of China
| |
Collapse
|
32
|
Stevens HY, Bowles AC, Yeago C, Roy K. Molecular Crosstalk Between Macrophages and Mesenchymal Stromal Cells. Front Cell Dev Biol 2020; 8:600160. [PMID: 33363157 PMCID: PMC7755599 DOI: 10.3389/fcell.2020.600160] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/05/2020] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have been widely investigated for regenerative medicine applications, from treating various inflammatory diseases as a cell therapy to generating engineered tissue constructs. Numerous studies have evaluated the potential effects of MSCs following therapeutic administration. By responding to their surrounding microenvironment, MSCs may mediate immunomodulatory effects through various mechanisms that directly (i.e., contact-dependent) or indirectly (i.e., paracrine activity) alter the physiology of endogenous cells in various disease pathologies. More specifically, a pivotal crosstalk between MSCs and tissue-resident macrophages and monocytes (TMφ) has been elucidated using in vitro and in vivo preclinical studies. An improved understanding of this crosstalk could help elucidate potential mechanisms of action (MOAs) of therapeutically administered MSCs. TMφ, by nature of their remarkable functional plasticity and prevalence within the body, are uniquely positioned as critical modulators of the immune system - not only in maintaining homeostasis but also during pathogenesis. This has prompted further exploration into the cellular and molecular alterations to TMφ mediated by MSCs. In vitro assays and in vivo preclinical trials have identified key interactions mediated by MSCs that polarize the responses of TMφ from a pro-inflammatory (i.e., classical activation) to a more anti-inflammatory/reparative (i.e., alternative activation) phenotype and function. In this review, we describe physiological and pathological TMφ functions in response to various stimuli and discuss the evidence that suggest specific mechanisms through which MSCs may modulate TMφ phenotypes and functions, including paracrine interactions (e.g., secretome and extracellular vesicles), nanotube-mediated intercellular exchange, bioenergetics, and engulfment by macrophages. Continued efforts to elucidate this pivotal crosstalk may offer an improved understanding of the immunomodulatory capacity of MSCs and inform the development and testing of potential MOAs to support the therapeutic use of MSCs and MSC-derived products in various diseases.
Collapse
Affiliation(s)
- Hazel Y. Stevens
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Annie C. Bowles
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Carolyn Yeago
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
- NSF Engineering Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Georgia Institute of Technology, Atlanta, GA, United States
| | - Krishnendu Roy
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
- NSF Engineering Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Georgia Institute of Technology, Atlanta, GA, United States
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
- Center for ImmunoEngineering, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
33
|
Zheng X, Liu D. Adiponectin alleviates the symptoms of ischemic renal disease by inhibiting renal cell apoptosis. Life Sci 2020; 265:118825. [PMID: 33275989 DOI: 10.1016/j.lfs.2020.118825] [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: 08/31/2020] [Revised: 11/06/2020] [Accepted: 11/21/2020] [Indexed: 11/15/2022]
Abstract
AIMS Ischemic renal disease (IRD) can cause kidney damage and eventually lead to end-stage renal disease. Adiponectin (APN), a recently discovered collagen-like protein secreted by adipose tissues, plays an important role in regulating energy metabolism and inflammation. This study aimed to explore the specific mechanism by which APN affects IRD. MAIN METHODS We cultured human renal tubular epithelial cells (HK-2) and created a mouse model of IRD to detect apoptosis-related indicators in vitro and in vivo. KEY FINDINGS Compared with those in the control group, the apoptosis rate and expression levels of Bax and Fas increased in the CoCl2-induced hypoxia model group. However, the expression of Bcl-2 decreased, and after the combined treatment with APN, the phenomenon mentioned above was reversed. Moreover, studies have found that stanniocalcin-1 (STC-1) and uncoupling protein3 (UCP3) are also involved in the protective effect of APN. Additionally, we found that the glomeruli of the mice were significantly enlarged after the APN gene was knocked out; furthermore, the number of collagen fibers in the renal tubules, as well as the expression of the corresponding fibrogenic factors, increased significantly. More importantly, after the knockout of the APN gene, the expression of the hypoxia-inducible factors HIF-1α and HIF-1β and the apoptotic rate of renal tissue cells also increased. SIGNIFICANCE These results indicate that APN can alleviate the symptoms of IRD by inhibiting renal cell apoptosis. Thus, in the future, APN may be a new target for the treatment of IRD. CHEMICAL COMPOUNDS Cobalt chloride (PubChem CID: 24643).
Collapse
Affiliation(s)
- Xiaotong Zheng
- Department of Nephrology, Shengjing Hospital of China Medical University, NO.39 Huaxiang Road, Tiexi District, Shenyang 110022, Liaoning, PR China
| | - Dajun Liu
- Department of Nephrology, Shengjing Hospital of China Medical University, NO.39 Huaxiang Road, Tiexi District, Shenyang 110022, Liaoning, PR China.
| |
Collapse
|
34
|
Kumar A, Ghosh SB. Emerging Treatment Options of Regenerative Medicine in Severe Corona Virus/COVID 19 Infections. Int J Stem Cells 2020; 13:305-311. [PMID: 32840231 PMCID: PMC7691858 DOI: 10.15283/ijsc20076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/19/2020] [Accepted: 06/29/2020] [Indexed: 12/18/2022] Open
Abstract
COVID-19 pandemic has brought the whole world stand still, locked down in their homes, infecting more than 8 million people, and many thousands (449,182) -have lost their lives across the globe. Due to lack of any definitive medicine or vaccine, treatment options are supportive of oxygenation, antiviral, antiretroviral drugs, antibiotics, fluid/ electrolyte, mechanical ventilation with ICU (Intensive Care Unit) support, and chloroquine/hydroxychloroquine have been tried to fight this infection. However, mortality due to severe pneumonia, ARDS (Acute Respiratory Distress Syndrome), and multiorgan failure arising from the overactive immune response (storm) mediated by cytokines remains a treatment challenge in elderly and patients with severe medical comorbidities. Recently, anti-inflammatory, angiogenic, immune-modular, and healing properties of intravenous injections of culture derived stem cells have been proposed and shown to benefits in a small number of patients with severe COVID-19 infections. Based on previous experience with other viral infections, convalescent plasma, and serum transfusion are being used as a source of neutralizing antibody/factors to minimize the effects of inflammatory cytokines in this infection. Immunotherapy with purified monoclonal antibodies and conditioned serum with a mixture of unique cytokines are also being developed. Regenerative Medicine has emerged as a crucial adjuvant tool in promoting healing and early recovery in severe COVID-19 infections and other supportive treatments.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Orthopedics, My Doc Specialist Medical Centre DMCC, Dubai, UAE.,Department of Orthopedics, Saudi German Hospital, Dubai, UAE
| | | |
Collapse
|
35
|
Kumar A, Ghosh Kadamb A, Ghosh Kadamb K. Mesenchymal or Maintenance Stem Cell & Understanding Their Role in Osteoarthritis of the Knee Joint: A Review Article. THE ARCHIVES OF BONE AND JOINT SURGERY 2020; 8:560-569. [PMID: 33088856 DOI: 10.22038/abjs.2020.42536.2155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Mesenchymal Stem Cell (MSC) therapy in osteoarthritis has been hailed as a promising treatment for osteoarthritis due to their unlimited potential of healing and regeneration. Existing literature regarding their proper name, optimal sources, mechanisms of action, dosage, and route of administration, efficacy, and safety is debatable. This index review article has tried to connect these puzzling pieces of available information and brought clarity on some of these crucial issues. The author believes that Maintenance Stem Cells (MSC) may be a more suitable term than mesenchymal stem cell or medicinal signaling cells as their origin might not be limited to mesodermal tissue. Also, they have been shown capable of self-renewal, differentiation, and maintaining a cascade of healing & possibly regeneration at the implanted site. Only a small percentage of implanted MSC survive and rest undergo apoptosis after releasing growth factors, cytokines, and extracellular vesicles. These surviving MSC become active due to conformational changes induced by anti-environment stimuli and undergo limited self-renewal, proliferation, and differentiation, but only a few of them might incorporate into the host tissues. These cells generate & maintain a momentum of series of regenerative activities to improve the function of joint, stabilize or possibly enhance the cartilage quality. More randomized studies with long term follow-up are required to bring clarity on their ideal source, expansion, culture technique, optimum dosage, and route of administration and long-term safety issues.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Orthopaedics, Saudi German Hospital, Dubai, UAE
| | | | | |
Collapse
|
36
|
Bachmann J, Ehlert E, Becker M, Otto C, Radeloff K, Blunk T, Bauer-Kreisel P. Ischemia-Like Stress Conditions Stimulate Trophic Activities of Adipose-Derived Stromal/Stem Cells. Cells 2020; 9:cells9091935. [PMID: 32825678 PMCID: PMC7566001 DOI: 10.3390/cells9091935] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/31/2020] [Accepted: 08/19/2020] [Indexed: 01/02/2023] Open
Abstract
Adipose-derived stromal/stem cells (ASCs) have been shown to exert regenerative functions, which are mainly attributed to the secretion of trophic factors. Upon transplantation, ASCs are facing an ischemic environment characterized by oxygen and nutrient deprivation. However, current knowledge on the secretion capacity of ASCs under such conditions is limited. Thus, the present study focused on the secretory function of ASCs under glucose and oxygen deprivation as major components of ischemia. After exposure to glucose/oxygen deprivation, ASCs maintained distinct viability, but the metabolic activity was greatly reduced by glucose limitation. ASCs were able to secrete a broad panel of factors under glucose/oxygen deprivation as revealed by a cytokine antibody array. Quantification of selected factors by ELISA demonstrated that glucose deprivation in combination with hypoxia led to markedly higher secretion levels of the angiogenic and anti-apoptotic factors IL-6, VEGF, and stanniocalcin-1 as compared to the hypoxic condition alone. A conditioned medium of glucose/oxygen-deprived ASCs promoted the viability and tube formation of endothelial cells, and the proliferation and migration of fibroblasts. These findings indicate that ASCs are stimulated by ischemia-like stress conditions to secrete trophic factors and would be able to exert their beneficial function in an ischemic environment.
Collapse
Affiliation(s)
- Julia Bachmann
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Wuerzburg, 97080 Wuerzburg, Germany; (J.B.); (E.E.); (T.B.)
| | - Elias Ehlert
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Wuerzburg, 97080 Wuerzburg, Germany; (J.B.); (E.E.); (T.B.)
| | - Matthias Becker
- Institute for Medical Radiation and Cell Research, University of Wuerzburg, 97078 Wuerzburg, Germany;
| | - Christoph Otto
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University of Wuerzburg, 97080 Wuerzburg, Germany;
| | - Katrin Radeloff
- Department of Otorhinolaryngology, Head and Neck Surgery, Carl von Ossietzky-University of Oldenburg, 26133 Oldenburg, Germany;
| | - Torsten Blunk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Wuerzburg, 97080 Wuerzburg, Germany; (J.B.); (E.E.); (T.B.)
| | - Petra Bauer-Kreisel
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Wuerzburg, 97080 Wuerzburg, Germany; (J.B.); (E.E.); (T.B.)
- Correspondence: ; Tel.: +49-931-201-37115
| |
Collapse
|
37
|
Zonderland J, Gomes DB, Pallada Y, Moldero IL, Camarero‐Espinosa S, Moroni L. Mechanosensitive regulation of stanniocalcin-1 by zyxin and actin-myosin in human mesenchymal stromal cells. Stem Cells 2020; 38:948-959. [PMID: 32379914 PMCID: PMC7497098 DOI: 10.1002/stem.3198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/29/2020] [Accepted: 04/16/2020] [Indexed: 12/16/2022]
Abstract
Stanniocalcin-1 (STC1) secreted by mesenchymal stromal cells (MSCs) has anti-inflammatory functions, reduces apoptosis, and aids in angiogenesis, both in vitro and in vivo. However, little is known about the molecular mechanisms of its regulation. Here, we show that STC1 secretion is increased only under specific cell-stress conditions. We find that this is due to a change in actin stress fibers and actin-myosin tension. Abolishment of stress fibers by blebbistatin and knockdown of the focal adhesion protein zyxin leads to an increase in STC1 secretion. To also study this connection in 3D, where few focal adhesions and actin stress fibers are present, STC1 expression was analyzed in 3D alginate hydrogels and 3D electrospun scaffolds. Indeed, STC1 secretion was increased in these low cellular tension 3D environments. Together, our data show that STC1 does not directly respond to cell stress, but that it is regulated through mechanotransduction. This research takes a step forward in the fundamental understanding of STC1 regulation and can have implications for cell-based regenerative medicine, where cell survival, anti-inflammatory factors, and angiogenesis are critical.
Collapse
Affiliation(s)
- Jip Zonderland
- Complex Tissue Regeneration Department, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - David B. Gomes
- Complex Tissue Regeneration Department, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Yves Pallada
- Complex Tissue Regeneration Department, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Ivan L. Moldero
- Complex Tissue Regeneration Department, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Sandra Camarero‐Espinosa
- Complex Tissue Regeneration Department, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Lorenzo Moroni
- Complex Tissue Regeneration Department, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| |
Collapse
|
38
|
Shin JY, Lee PH. Mesenchymal stem cells modulate misfolded α-synuclein in parkinsonian disorders: A multitarget disease-modifying strategy. Stem Cell Res 2020; 47:101908. [PMID: 32683319 DOI: 10.1016/j.scr.2020.101908] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/22/2020] [Accepted: 07/05/2020] [Indexed: 02/07/2023] Open
Abstract
α-Synuclein (α-Syn) aggregates, the major toxic component of Lewy bodies, are proteinaceous fibrillar cytoplasmic inclusions observed in α-synucleinopathies, such as Parkinson's disease (PD), multiple system atrophy, and dementia with Lewy bodies. Overexpression of α-syn induce neuronal loss and α-syn aggregation in PD animals. Recent studies show that α-syn is released by exocytosis and can be transmitted between brain areas through cell-to-cell propagation. Moreover, aggregates of extracellular α-syn can induce neuroinflammation-mediated neurotoxic signaling through microglial activation and release of pro-inflammatory factors. Thus, modulation of α-syn might be a potential therapeutic strategy for modifying disease progression of α-synucleinopathies. Our previous studies have revealed that MSCs have potent neuroprotective effects in PD animal through modulation of neuroinflammation, inhibition of cell death, and promotion of neurogenesis. Here, we provide further evidence that MSCs have the potential to modulate α-syn-related microenvironments via enhancement of autophagy, proteolysis of α-syn aggregates, inhibition of cell-to-cell transmission of α-syn, stabilization of axonal transport, and phagocytic clearance of α-syn by microglial M2 polarization. With advantages in clinical applications, these data suggests that the use of MSCs as pharmacological modulators of α-syn propagation would be an effective therapeutic approach in PD.
Collapse
Affiliation(s)
- Jin Young Shin
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea; Severance Biomedical Science Institute, Yonsei University, Seoul, South Korea
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea; Severance Biomedical Science Institute, Yonsei University, Seoul, South Korea.
| |
Collapse
|
39
|
Challenges of Engineering Biomimetic Dental and Paradental Tissues. Tissue Eng Regen Med 2020; 17:403-421. [PMID: 32621282 DOI: 10.1007/s13770-020-00269-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/07/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Loss of the dental and paradental tissues resulting from trauma, caries or from systemic diseases considered as one of the most significant and frequent clinical problem to the healthcare professionals. Great attempts have been implemented to recreate functionally, healthy dental and paradental tissues in order to substitute dead and diseased tissues resulting from secondary trauma of car accidents, congenital malformations of cleft lip and palate or due to acquired diseases such as cancer and periodontal involvements. METHOD An extensive literature search has been done on PubMed database from 2010 to 2019 about the challenges of engineering a biomimetic tooth (BioTooth) regarding basic biology of the tooth and its supporting structures, strategies, and different techniques of obtaining biological substitutes for dental tissue engineering. RESULTS It has been found that great challenges need to be considered before engineering biomimetic individual parts of the tooth such as enamel, dentin-pulp complex and periodontium. In addition, two approaches have been adopted to engineer a BioTooth. The first one was to engineer a BioTooth as an individual unit and the other was to engineer a BioTooth with its supporting structures. CONCLUSION Engineering of BioTooth with its supporting structures thought to be in the future will replace the traditional and conventional treatment modalities in the field of dentistry. To accomplish this goal, different cell lines and growth factors with a variety of scaffolds at the nano-scale level are now in use. Recent researches in this area of interest are dedicated for this objective, both in vivo and in vitro. Despite progress in this field, there are still many challenges ahead and need to be overcome, many of which related to the basic tooth biology and its supporting structures and some others related to the sophisticated techniques isolating cells, fabricating the needed scaffolds and obtaining the signaling molecules.
Collapse
|
40
|
Sathiyanathan P, Samsonraj RM, Tan CLL, Ling L, Lezhava A, Nurcombe V, Stanton LW, Cool SM. A genomic biomarker that identifies human bone marrow-derived mesenchymal stem cells with high scalability. Stem Cells 2020; 38:1124-1136. [PMID: 32510174 DOI: 10.1002/stem.3203] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 03/29/2020] [Indexed: 12/12/2022]
Abstract
Although the application of human mesenchymal stem cells (hMSCs) to repair damaged or diseased tissues has proven relatively effective, both the donor-to-donor variability in ex vivo expansion rates and the maintenance of stemness remain a bottleneck to widespread translation. Previous work from this laboratory stratified donors into those yielding hMSCs with high- or low-growth capacity; global transcriptomic analysis revealed that high-growth-capacity hMSCs were characterized by a loss of the gene encoding glutathione S-transferase theta 1 (GSTT1). These GSTT1-null hMSCs demonstrated increased proliferative rates, clonogenic potential, and longer telomeres compared with low-growth capacity hMSCs that were GSTT1-positive. Thus, this study identifies GSTT1 as a novel genomic DNA biomarker for hMSC scalability.
Collapse
Affiliation(s)
- Padmapriya Sathiyanathan
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore.,Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Rebekah M Samsonraj
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Clarissa L L Tan
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Ling Ling
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Alexander Lezhava
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Victor Nurcombe
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Lawrence W Stanton
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Simon M Cool
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore.,Department of Orthopaedic Surgery, National University of Singapore, Singapore
| |
Collapse
|
41
|
Waclawiczek A, Hamilton A, Rouault-Pierre K, Abarrategi A, Albornoz MG, Miraki-Moud F, Bah N, Gribben J, Fitzgibbon J, Taussig D, Bonnet D. Mesenchymal niche remodeling impairs hematopoiesis via stanniocalcin 1 in acute myeloid leukemia. J Clin Invest 2020; 130:3038-3050. [PMID: 32364536 PMCID: PMC7260026 DOI: 10.1172/jci133187] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML) disrupts the generation of normal blood cells, predisposing patients to hemorrhage, anemia, and infections. Differentiation and proliferation of residual normal hematopoietic stem and progenitor cells (HSPCs) are impeded in AML-infiltrated bone marrow (BM). The underlying mechanisms and interactions of residual hematopoietic stem cells (HSCs) within the leukemic niche are poorly understood, especially in the human context. To mimic AML infiltration and dissect the cellular crosstalk in human BM, we established humanized ex vivo and in vivo niche models comprising AML cells, normal HSPCs, and mesenchymal stromal cells (MSCs). Both models replicated the suppression of phenotypically defined HSPC differentiation without affecting their viability. As occurs in AML patients, the majority of HSPCs were quiescent and showed enrichment of functional HSCs. HSPC suppression was largely dependent on secreted factors produced by transcriptionally remodeled MSCs. Secretome analysis and functional validation revealed MSC-derived stanniocalcin 1 (STC1) and its transcriptional regulator HIF-1α as limiting factors for HSPC proliferation. Abrogation of either STC1 or HIF-1α alleviated HSPC suppression by AML. This study provides a humanized model to study the crosstalk among HSPCs, leukemia, and their MSC niche, and a molecular mechanism whereby AML impairs normal hematopoiesis by remodeling the mesenchymal niche.
Collapse
MESH Headings
- Animals
- Female
- Glycoproteins/genetics
- Glycoproteins/metabolism
- HL-60 Cells
- Hematopoiesis
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Male
- Mesenchymal Stem Cells/metabolism
- Mesenchymal Stem Cells/pathology
- Mice
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- U937 Cells
Collapse
Affiliation(s)
- Alexander Waclawiczek
- Haematopoietic Stem Cell Laboratory, Francis Crick Institute, London, United Kingdom
| | - Ashley Hamilton
- Haematopoietic Stem Cell Laboratory, Francis Crick Institute, London, United Kingdom
| | - Kevin Rouault-Pierre
- Haematopoietic Stem Cell Laboratory, Francis Crick Institute, London, United Kingdom
| | - Ander Abarrategi
- Haematopoietic Stem Cell Laboratory, Francis Crick Institute, London, United Kingdom
| | | | - Farideh Miraki-Moud
- Haemato-Oncology Unit, Royal Marsden Hospital, Institute of Cancer Research, London, United Kingdom
| | - Nourdine Bah
- Bioinformatic Core Facility, Francis Crick Institute, London, United Kingdom
| | - John Gribben
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jude Fitzgibbon
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - David Taussig
- Haemato-Oncology Unit, Royal Marsden Hospital, Institute of Cancer Research, London, United Kingdom
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, Francis Crick Institute, London, United Kingdom
| |
Collapse
|
42
|
Zhao F, Yang G, Feng M, Cao Z, Liu Y, Qiu J, You L, Zheng L, Zhang T, Zhao Y. Expression, function and clinical application of stanniocalcin-1 in cancer. J Cell Mol Med 2020; 24:7686-7696. [PMID: 32468698 PMCID: PMC7348177 DOI: 10.1111/jcmm.15348] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/10/2019] [Accepted: 10/19/2019] [Indexed: 12/13/2022] Open
Abstract
The glycoprotein stanniocalcin-1 functions as a regulatory endocrine hormone that maintains the balance of calcium and phosphorus in bony fish and as a paracrine/autocrine factor involved in many physiological/pathological processes in humans, including carcinogenesis. In this review, we provide an overview of (a) the possible mechanisms through which STC1 affects the malignant properties of cancer, (b) transcriptional and post-transcriptional regulation pathways of STC1 and (c) the potential clinical relevance of STC1 as a cancer biomarker and even a therapeutic target in the future. Exploring the role of STC1 in cancer development may provide a better understanding of the tumorigenesis process in humans and may facilitate finding an effective therapeutic method against cancer.
Collapse
Affiliation(s)
- Fangyu Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengyu Feng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yueze Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
43
|
Sveiven SN, Nordgren TM. Lung-resident mesenchymal stromal cells are tissue-specific regulators of lung homeostasis. Am J Physiol Lung Cell Mol Physiol 2020; 319:L197-L210. [PMID: 32401672 DOI: 10.1152/ajplung.00049.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Until recently, data supporting the tissue-resident status of mesenchymal stromal cells (MSC) has been ambiguous since their discovery in the 1950-60s. These progenitor cells were first discovered as bone marrow-derived adult multipotent cells and believed to migrate to sites of injury, opposing the notion that they are residents of all tissue types. In recent years, however, it has been demonstrated that MSC can be found in all tissues and MSC from different tissues represent distinct populations with differential protein expression unique to each tissue type. Importantly, these cells are efficient mediators of tissue repair, regeneration, and prove to be targets for therapeutics, demonstrated by clinical trials (phase 1-4) for MSC-derived therapies for diseases like graft-versus-host-disease, multiple sclerosis, rheumatoid arthritis, and Crohn's disease. The tissue-resident status of MSC found in the lung is a key feature of their importance in the context of disease and injuries of the respiratory system, since these cells could be instrumental to providing more specific and targeted therapies. Currently, bone marrow-derived MSC have been established in the treatment of disease, including diseases of the lung. However, with lung-resident MSC representing a unique population with a different phenotypic and gene expression pattern than MSC derived from other tissues, their role in remediating lung inflammation and injury could provide enhanced efficacy over bone marrow-derived MSC methods. Through this review, lung-resident MSC will be characterized, using previously published data, by surface markers, gene expression patterns, and compared with bone-marrow MSC to highlight similarities and, importantly, differences in these cell types.
Collapse
Affiliation(s)
- Stefanie Noel Sveiven
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, California
| | - Tara M Nordgren
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, California
| |
Collapse
|
44
|
Gladkova N, Umezu T, Imanishi S, Kawana C, Ohyashiki JH, Ohyashiki K. Effect of the extracellular component of bone marrow mesenchymal stromal cells from healthy donors on hematologic neoplasms and their angiogenesis. Hum Cell 2020; 33:599-609. [PMID: 32281045 PMCID: PMC7324432 DOI: 10.1007/s13577-020-00332-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/11/2020] [Indexed: 11/05/2022]
Abstract
Bone marrow mesenchymal stromal cells (BM-MSCs) from healthy donors are a promising source of cell therapy. However, their effectiveness in cancer remains less known. This study is the first to evaluate the quality of BM-MSCs obtained from young and elderly healthy volunteers (KNT cells). The KNT cells had normal karyotypes and were positive for MSC markers (CD90, CD73, CD105). When cultured under appropriate conditions, they showed adipogenic or osteogenic potential. Hence, the anti-neoplastic effects of secretory factors [supernatant or extracellular vesicles (EV)] from KNT cells were verified using several neoplastic cells (three multiple myeloma, three myeloid leukemia, and three lymphoma cell lines). The conditioned medium (CM), but not EV, of KNT cells derived from young healthy donors significantly inhibited myeloma and lymphoma cell proliferation, but enhanced myeloid leukemia proliferation. Anti-angiogenesis effect of CM and EV derived from young KNT against hematologic neoplasia-induced angiogenesis was evident and more prominent in CM than in EV but not evident in elderly KNT-derived EV. These findings indicate that the anti-tumor effect of KNT cells depends on the types of hematologic neoplasia, with elements existing in the supernatant and not in EVs. Therefore, BM-MSC may produce soluble factors that affect cell proliferation of neoplasia, causing cell-to-cell communication. The anti-angiogenesis effect of KNT cells depends on the age of BM-MSC donors.
Collapse
Affiliation(s)
- Nina Gladkova
- Kintaro Cells Power Corporation, Tokyo, Japan.,Department of Advanced Cellular Therapy, Tokyo Medical University, Tokyo, Japan
| | - Tomohiro Umezu
- Department of Hematology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku, Tokyo, 160-0023, Japan.,Institute of Medical Science, Tokyo Medical University, Tokyo, Japan.,Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| | - Satoshi Imanishi
- Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Chiaki Kawana
- Department of Hematology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku, Tokyo, 160-0023, Japan
| | - Junko H Ohyashiki
- Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Kazuma Ohyashiki
- Department of Hematology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku, Tokyo, 160-0023, Japan.
| |
Collapse
|
45
|
Stanniocalcin-1 Alleviates Contrast-Induced Acute Kidney Injury by Regulating Mitochondrial Quality Control via the Nrf2 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1898213. [PMID: 32318235 PMCID: PMC7153002 DOI: 10.1155/2020/1898213] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/27/2020] [Accepted: 03/10/2020] [Indexed: 12/30/2022]
Abstract
Contrast-induced acute kidney injury (CI-AKI) is the third common cause of acute kidney injury (AKI), which is associated with poor short- and long-term outcomes. Currently, effective therapy strategy for CI-AKI remains lacking. Stanniocalcin-1 (STC1) is a conserved glycoprotein with antiapoptosis and anti-inflammatory functions, but the role of STC1 in controlling CI-AKI is unknown. Here, we demonstrated a protective role of STC1 in contrast-induced injury in cultured renal tubular epithelial cells and CI-AKI rat models. Recombinant human STC1 (rhSTC1) regulated mitochondrial quality control, thus suppressing contrast-induced mitochondrial damage, oxidative stress, inflammatory response, and apoptotic injury. Mechanistically, activation of the Nrf2 signaling pathway contributes critically to the renoprotective effect of STC1. Together, this study demonstrates a novel role of STC1 in preventing CI-AKI and reveals Nrf2 as a molecular target of STC1. Therefore, this study provides a promising preventive target for the treatment of CI-AKI.
Collapse
|
46
|
Seo Y, Shin TH, Ahn JS, Oh SJ, Shin YY, Yang JW, Park HY, Shin SC, Kwon HK, Kim JM, Sung ES, Park GC, Lee BJ, Kim HS. Human Tonsil-Derived Mesenchymal Stromal Cells Maintain Proliferating and ROS-Regulatory Properties via Stanniocalcin-1. Cells 2020; 9:cells9030636. [PMID: 32155780 PMCID: PMC7140534 DOI: 10.3390/cells9030636] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 02/29/2020] [Accepted: 03/04/2020] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) from various sources exhibit different potential for stemness and therapeutic abilities. Recently, we reported a unique MSCs from human palatine tonsil (TMSCs) and their superior proliferation capacity compared to MSCs from other sources. However, unique characteristics of each MSC are not yet precisely elucidated. We investigated the role of stanniocalcin-1 (STC1), an anti-oxidative hormone, in the functions of TMSCs. We found that STC1 was highly expressed in TMSC compared with MSCs from bone marrow or adipose tissue. The proliferation, senescence and differentiation of TMSCs were assessed after the inhibition of STC1 expression. STC1 inhibition resulted in a significant decrease in the proliferation of TMSCs and did not affect the differentiation potential. To reveal the anti-oxidative ability of STC1 in TMSCs themselves or against other cell types, the generation of mitochondrial reactive oxygen species (ROS) in TMSC or ROS-mediated production of interleukin (IL)-1β from macrophage-like cells were detected. Interestingly, the basal level of ROS generation in TMSCs was significantly elevated after STC1 inhibition. Moreover, down-regulation of STC1 impaired the inhibitory effect of TMSCs on IL-1β production in macrophages. Taken together, these findings indicate that STC1 is highly expressed in TMSCs and plays a critical role in proliferating and ROS-regulatory abilities.
Collapse
Affiliation(s)
- Yoojin Seo
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea; (Y.S.); (J.-S.A.); (S.-J.O.); (Y.Y.S.); (J.W.Y.)
- Dental and Life Science Institute, Pusan National University, Yangsan 50612, Korea
| | - Tae-Hoon Shin
- Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Korea; (T.-H.S.); (H.Y.P.)
- Translational Stem Cell Biology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ji-Su Ahn
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea; (Y.S.); (J.-S.A.); (S.-J.O.); (Y.Y.S.); (J.W.Y.)
- Dental and Life Science Institute, Pusan National University, Yangsan 50612, Korea
| | - Su-Jeong Oh
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea; (Y.S.); (J.-S.A.); (S.-J.O.); (Y.Y.S.); (J.W.Y.)
- Dental and Life Science Institute, Pusan National University, Yangsan 50612, Korea
| | - Ye Young Shin
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea; (Y.S.); (J.-S.A.); (S.-J.O.); (Y.Y.S.); (J.W.Y.)
- Dental and Life Science Institute, Pusan National University, Yangsan 50612, Korea
| | - Ji Won Yang
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea; (Y.S.); (J.-S.A.); (S.-J.O.); (Y.Y.S.); (J.W.Y.)
- Dental and Life Science Institute, Pusan National University, Yangsan 50612, Korea
| | - Hee Young Park
- Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Korea; (T.-H.S.); (H.Y.P.)
| | - Sung-Chan Shin
- Department of Otorhinolaryngology, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Korea; (S.-C.S.); (H.-K.K.); (J.M.K.)
| | - Hyun-Keun Kwon
- Department of Otorhinolaryngology, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Korea; (S.-C.S.); (H.-K.K.); (J.M.K.)
| | - Ji Min Kim
- Department of Otorhinolaryngology, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Korea; (S.-C.S.); (H.-K.K.); (J.M.K.)
| | - Eui-Suk Sung
- Department of Otorhinolaryngology-Head and Neck Surgery, Biomedical Research Institute, Pusan National University School of Medicine, Yangsan Pusan National University Hospital, Yangsan 50612, Korea;
| | - Gi Cheol Park
- Department of Otolaryngology – Head and Neck Surgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Korea;
| | - Byung-Joo Lee
- Department of Otorhinolaryngology, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Korea; (S.-C.S.); (H.-K.K.); (J.M.K.)
- Correspondence: (B.-J.L.); (H.-S.K.); Tel.: +82-51-240-7675 (B.-J.L.); +82-51-510-8231 (H.-S.K.)
| | - Hyung-Sik Kim
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea; (Y.S.); (J.-S.A.); (S.-J.O.); (Y.Y.S.); (J.W.Y.)
- Dental and Life Science Institute, Pusan National University, Yangsan 50612, Korea
- Correspondence: (B.-J.L.); (H.-S.K.); Tel.: +82-51-240-7675 (B.-J.L.); +82-51-510-8231 (H.-S.K.)
| |
Collapse
|
47
|
Hingert D, Nawilaijaroen P, Aldridge J, Baranto A, Brisby H. Investigation of the Effect of Secreted Factors from Mesenchymal Stem Cells on Disc Cells from Degenerated Discs. Cells Tissues Organs 2020; 208:76-88. [PMID: 32092752 DOI: 10.1159/000506350] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/02/2020] [Indexed: 11/19/2022] Open
Abstract
Low back pain is experienced by a large number of people in western countries and may be caused and influenced by many different pathologies and psychosocial factors including disc degeneration. Disc degeneration involves the increased expression of proinflammatory cytokines and matrix metalloproteinases (MMPs) in the disc environment, which leads to the loss of extracellular matrix (ECM) and the viability of the native disc cells (DCs). Treatment approaches using growth factors and cell therapy have been proposed due to the compelling results that growth factors and mesenchymal stem cells (MSCs) can influence the degenerated discs. The aim of this study was to investigate the effects of conditioned media (CM) from human MSCs (hMSCs) and connective tissue growth factor (CTGF) and TGF-β on disc cells, and hMSCs isolated from patients with degenerative discs and severe low back pain. The aim was also to examine the constituents of CM in order to study the peptides that could bring about intervertebral disc (IVD) regeneration. DCs and hMSC pellets (approx.. 200,000 cells) were cultured and stimulated with hMSC-derived CM or CTGF and TGF-β over 28 days. The effects of CM and CTGF on DCs and hMSCs were assessed via cell viability, proteoglycan production, the expression of ECM proteins, and chondrogenesis in 3D pellet culture. To identify the constituents of CM, CM was analyzed with tandem mass spectrometry. The findings indicate that CM enhanced the cellular viability and ECM production of DCs while CTGF and the control exhibited nonsignificant differences. The same was observed in the hMSC group. Mass spectrometry analysis of CM identified >700 peptides, 129 of which showed a relative abundance of ≥2 (CTGF among them). The results suggest that CM holds potential to counter the progression of disc degeneration, likely resulting from the combination of all the substances released by the hMSCs. The soluble factors released belong to different peptide families. The precise mechanism underlying the regenerative effect needs to be investigated further, prior to incorporating peptides in the development of new treatment strategies for low back pain that is potentially caused by IVD degeneration.
Collapse
Affiliation(s)
- Daphne Hingert
- Department of Orthopedics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,
| | | | - Jonathan Aldridge
- Department of Physics, Chalmers University of Technology, Gothenburg, Sweden
| | - Adad Baranto
- Department of Orthopedics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Orthopedics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Helena Brisby
- Department of Orthopedics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Orthopedics, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
48
|
Chu DT, Phuong TNT, Tien NLB, Tran DK, Thanh VV, Quang TL, Truong DT, Pham VH, Ngoc VTN, Chu-Dinh T, Kushekhar K. An Update on the Progress of Isolation, Culture, Storage, and Clinical Application of Human Bone Marrow Mesenchymal Stem/Stromal Cells. Int J Mol Sci 2020; 21:E708. [PMID: 31973182 PMCID: PMC7037097 DOI: 10.3390/ijms21030708] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 12/13/2022] Open
Abstract
Bone marrow mesenchymal stem/stromal cells (BMSCs), which are known as multipotent cells, are widely used in the treatment of various diseases via their self-renewable, differentiation, and immunomodulatory properties. In-vitro and in-vivo studies have supported the understanding mechanisms, safety, and efficacy of BMSCs therapy in clinical applications. The number of clinical trials in phase I/II is accelerating; however, they are limited in the size of subjects, regulations, and standards for the preparation and transportation and administration of BMSCs, leading to inconsistency in the input and outcome of the therapy. Based on the International Society for Cellular Therapy guidelines, the characterization, isolation, cultivation, differentiation, and applications can be optimized and standardized, which are compliant with good manufacturing practice requirements to produce clinical-grade preparation of BMSCs. This review highlights and updates on the progress of production, as well as provides further challenges in the studies of BMSCs, for the approval of BMSCs widely in clinical application.
Collapse
Affiliation(s)
- Dinh-Toi Chu
- Faculty of Biology, Hanoi National University of Education, Hanoi 100000, Vietnam
- School of Odonto Stomatology, Hanoi Medical University, Hanoi 100000, Vietnam;
| | - Thuy Nguyen Thi Phuong
- Department of Animal Science, College of Agriculture and Life Science, Chonnam National University, Gwangju 61186, Korea
| | - Nguyen Le Bao Tien
- Institute of Orthopaedics and Trauma Surgery, Viet Duc Hospital, Hanoi 100000, Vietnam; (N.L.B.T.); (V.V.T.)
| | - Dang Khoa Tran
- Department of Anatomy, University of Medicine Pham Ngoc Thach, Ho Chi Minh City 700000, Vietnam;
| | - Vo Van Thanh
- Institute of Orthopaedics and Trauma Surgery, Viet Duc Hospital, Hanoi 100000, Vietnam; (N.L.B.T.); (V.V.T.)
- Department of Surgery, Hanoi Medical University, Hanoi 100000, Vietnam
| | - Thuy Luu Quang
- Center for Anesthesia and Surgical Intensive Care, Viet Duc Hospital, Hanoi 100000, Vietnam;
| | | | - Van Huy Pham
- AI Lab, Faculty of Information Technology, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam
| | - Vo Truong Nhu Ngoc
- School of Odonto Stomatology, Hanoi Medical University, Hanoi 100000, Vietnam;
| | - Thien Chu-Dinh
- Institute for Research and Development, Duy Tan University, Danang 550000, Vietnam
| | - Kushi Kushekhar
- Institute of Cancer Research, Oslo University Hospital, 0310 Oslo, Norway;
| |
Collapse
|
49
|
Takeuchi S, Hirasaki E, Kumakura H. Muscle Spindle Density of Lateral Rotators of the Thigh in Japanese Macaques and a Gibbon. Cells Tissues Organs 2020; 208:1-12. [PMID: 31927538 PMCID: PMC7212700 DOI: 10.1159/000504958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 02/02/2020] [Accepted: 11/24/2019] [Indexed: 01/07/2023] Open
Abstract
We examined the six small lateral rotators of the hip joint, which is one of the most flexible joints and allows kinematically complex motions of the hindlimb, to elucidate the functional differentiation among these muscles and to test the hypothesis that species-specific characteristics in hindlimb use during locomotion are reflected in the muscle spindle density and in other parameters of the deep small hip joint rotators. For these purposes, we estimated the number of muscle spindles of the superior gemellus muscle (SG), inferior gemellus muscle, quadratus femoris muscle, obturator internus muscle (OI), obturator externus muscle, and piriformis muscle in three Japanese macaques and a gibbon, using 30-µm-thick serial sections throughout each muscle length after azan staining. The numbers of muscle spindles per 10,000 muscle fibers were determined to compare inter-muscle variation. The spindle density was highest in the SG and lowest in the OI in the Japanese macaques, suggesting that the SG, which is attached to the tendon of the OI, functions as a kinesiological monitor of the OI. On the other hand, SG the was missing in the gibbon, and the OI in the gibbon contained more spindles than that in the Japanese macaques. This suggests that the SG and the OI fused into one muscle in the gibbon. We postulate that the relative importance of the deep small hip rotator muscles differs between the Japanese macaques and gibbon and that the gibbon's muscles are less differentiated in terms of the spindle density, probably because this brachiating species uses its hindlimbs less frequently.
Collapse
Affiliation(s)
- Sawa Takeuchi
- Department of Biological Anthropology, Graduate School of Human Sciences, Osaka University, Suita, Japan
| | - Eishi Hirasaki
- Section of Evolutionary Morphology, Primate Research Institute, Kyoto University, Inuyama, Japan,
| | - Hiroo Kumakura
- Department of Biological Anthropology, Graduate School of Human Sciences, Osaka University, Suita, Japan
| |
Collapse
|
50
|
Adipose Stem Cell-Based Clinical Strategy for Neural Regeneration: A Review of Current Opinion. Stem Cells Int 2019; 2019:8502370. [PMID: 31827536 PMCID: PMC6885831 DOI: 10.1155/2019/8502370] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/02/2019] [Accepted: 11/11/2019] [Indexed: 02/05/2023] Open
Abstract
Nerve injury is a critical problem in the clinic. Nerve injury causes serious clinic issues including pain and dysfunctions for patients. The disconnection between damaged neural fibers and muscles will result in muscle atrophy in a few weeks if no treatment is applied. Moreover, scientists have discovered that nerve injury can affect the osteogenic differentiation of skeletal stem cells (SSCs) and the fracture repairing. In plastic surgery, muscle atrophy and bone fracture after nerve injury have plagued clinicians for many years. How to promote neural regeneration is the core issue of research in the recent years. Without obvious effects of traditional neurosurgical treatments, research on stem cells in the past 10 years has provided a new therapeutic strategy for us to address this problem. Adipose stem cells (ASCs) are a kind of mesenchymal stem cells that have differentiation potential in adipose tissue. In the recent years, ASCs have become the focus of regenerative medicine. They play a pivotal role in tissue regeneration engineering. As a type of stem cell, ASCs are becoming popular for neuroregenerative medicine due to their advantages and characteristics. In the various diseases of the nervous system, ASCs are gradually applied to treat the related diseases. This review article focuses on the mechanism and clinical application of ASCs in nerve regeneration as well as the related research on ASCs over the past decades.
Collapse
|