1
|
Berni P, Andreoli V, Conti V, Ramoni R, Basini G, Scattini G, Pascucci L, Pellegrini M, Del Bue M, Squassino GP, Paino F, Pessina A, Alessandri G, Pirazzoli P, Bosetto A, Grolli S. Evaluation of a Novel Mechanical Device for the Production of Microfragmented Adipose Tissue for Veterinary Regenerative Medicine: A Proof-of-Concept. Int J Mol Sci 2024; 25:11854. [PMID: 39519405 PMCID: PMC11546731 DOI: 10.3390/ijms252111854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/14/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Therapies based on mesenchymal stromal cells (MSCs) have become one of the most significant advancements in veterinary regenerative medicine. The isolation of MSCs is usually performed by enzymatic digestion and requires variable times for cell expansion. In addition, these procedures need to be performed in specialized laboratory facilities. An alternative approach to in vitro-expanded MSC therapy is the use of microfragmented adipose tissue (microfat), which is a rich source of cells and growth factors from the stromal vascular fraction. Recent clinical studies support its safety and efficacy in the treatment of musculoskeletal disorders and wound healing. The aim of the present work was to characterize the microfragmented adipose tissue obtained by a new mechanical device, which provides sterile tissue that is ready for use in the clinic by the veterinarian, avoiding the need for specialized laboratory facilities. Microfat-derived MSCs were compared with enzymatically isolated MSCs in terms of their phenotypic characterization, growth rate and differentiation potential. Conditioned medium derived from microfat culture was evaluated for its ability to promote MSC vitality. No differences were observed between MSCs obtained through mechanical fragmentation and those derived from collagenase digestion of adipose tissue, suggesting that the device could serve as a practical source of microfragmented adipose tissue for use in veterinary clinics.
Collapse
Affiliation(s)
- Priscilla Berni
- Department of Veterinary Medical Science, University of Parma, 43121 Parma, Italy; (V.A.); (V.C.); (R.R.); (G.B.)
| | - Valentina Andreoli
- Department of Veterinary Medical Science, University of Parma, 43121 Parma, Italy; (V.A.); (V.C.); (R.R.); (G.B.)
| | - Virna Conti
- Department of Veterinary Medical Science, University of Parma, 43121 Parma, Italy; (V.A.); (V.C.); (R.R.); (G.B.)
| | - Roberto Ramoni
- Department of Veterinary Medical Science, University of Parma, 43121 Parma, Italy; (V.A.); (V.C.); (R.R.); (G.B.)
| | - Giuseppina Basini
- Department of Veterinary Medical Science, University of Parma, 43121 Parma, Italy; (V.A.); (V.C.); (R.R.); (G.B.)
| | - Gabriele Scattini
- Department of Veterinary Medicine, University of Perugia, 06123 Perugia, Italy; (G.S.); (L.P.)
| | - Luisa Pascucci
- Department of Veterinary Medicine, University of Perugia, 06123 Perugia, Italy; (G.S.); (L.P.)
| | - Martina Pellegrini
- Istituto Zooprofilattico Sperimentale dell’Umbria e delle Marche “Togo Rosati”, 06126 Perugia, Italy;
| | | | | | - Francesca Paino
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (F.P.); (A.P.); (G.A.)
| | - Augusto Pessina
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (F.P.); (A.P.); (G.A.)
| | - Giulio Alessandri
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (F.P.); (A.P.); (G.A.)
| | | | | | - Stefano Grolli
- Department of Veterinary Medical Science, University of Parma, 43121 Parma, Italy; (V.A.); (V.C.); (R.R.); (G.B.)
| |
Collapse
|
2
|
Wu S, Zhang Y, Hou Y, Zhu J, Yang H, Cui Y. Research on the role of exosomes secreted by immortalized adipose-derived mesenchymal stem cells differentiated into pericytes in the repair of high glucose-induced retinal vascular endothelial cell damage. Exp Eye Res 2024; 247:110046. [PMID: 39147191 DOI: 10.1016/j.exer.2024.110046] [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: 06/20/2024] [Revised: 07/27/2024] [Accepted: 08/13/2024] [Indexed: 08/17/2024]
Abstract
Diabetic retinopathy, a leading cause of vision impairment, is marked by microvascular complications in the retina, including pericyte loss, a key indicator of early-stage disease. This study explores the therapeutic potential of exosomes derived from immortalized adipose-mesenchymal stem cells differentiated into pericyte-like cells in restoring the function of mouse retinal microvascular endothelial cells damaged by high glucose conditions, thereby contributing to the understanding of early diabetic retinopathy intervention strategies. To induce immortalized adipose-mesenchymal stem cells differentiation into pericyte-like cells, the study employed pericyte growth supplement. And confirmed the success of cell differentiation through the detection of α-smooth muscle actin and neural/glial antigen 2 expression by Western blot and immunofluorescence. Exosomes were isolated from the culture supernatant of immortalized adipose-mesenchymal stem cells using ultracentrifugation and characterized through Western blot for exosomal markers (CD9, CD81, and TSG101), transmission electron microscopy, and nanoparticle tracking analysis. Their influence on mouse retinal microvascular endothelial cells under high glucose stress was assessed through various functional assays. Findings revealed that exosomes, especially those from pericyte-like immortalized adipose-mesenchymal stem cells, were efficiently internalized by retinal microvascular endothelial cells and effectively counteracted high glucose-induced apoptosis. These exosomes also mitigated the rise in reactive oxygen species levels and suppressed the migratory and angiogenic properties of retinal microvascular endothelial cells, as demonstrated by Transwell and tube formation assays, respectively. Furthermore, they preserved endothelial barrier function, reducing hyperglycemia-induced permeability. At the molecular level, qRT-PCR analysis showed that exosome treatment modulated the expression of critical genes involved in angiogenesis (VEGF-A, ANG2, MMP9), inflammation (IL-1β, TNF-α), gap junction communication (CX43), and cytoskeletal regulation (ROCK1), with the most prominent effects seen with exosomes from pericyte-like immortalized adipose-mesenchymal stem cells. High glucose increased the expression of pro-angiogenic and pro-inflammatory markers, which were effectively normalized post-exosome treatment. In conclusion, this research highlights the reparative capacity of exosomes secreted by pericyte-like differentiated immortalized adipose-mesenchymal stem cells in reversing the detrimental effects of high glucose on retinal microvascular endothelial cells. By reducing apoptosis, oxidative stress, inflammation, and abnormal angiogenic behavior, these exosomes present a promising avenue for therapeutic intervention in early diabetic retinopathy. Future studies can focus on elucidating the precise molecular mechanisms and exploring their translational potential in vivo.
Collapse
Affiliation(s)
- Sihui Wu
- Department of Ophthalmology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong Province, China; School of Medicine, Shandong University, Jinan, Shandong Province, China; NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China; Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yunnan Zhang
- Department of Ophthalmology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong Province, China; School of Medicine, Shandong University, Jinan, Shandong Province, China; NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China; Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yaru Hou
- Department of Ophthalmology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong Province, China; School of Medicine, Shandong University, Jinan, Shandong Province, China; NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China; Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Jing Zhu
- Department of Ophthalmology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong Province, China
| | - Hongling Yang
- Department of Ophthalmology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong Province, China
| | - Yan Cui
- Department of Ophthalmology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
3
|
Wang C, Liu X, Zhou J, Zhang X, Zhou Z, Zhang Q. Sensory nerves drive migration of dental pulp stem cells via the CGRP-Ramp1 axis in pulp repair. Cell Mol Life Sci 2024; 81:373. [PMID: 39196292 PMCID: PMC11358583 DOI: 10.1007/s00018-024-05400-2] [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: 09/01/2023] [Revised: 07/17/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024]
Abstract
Dental pulp stem cells (DPSCs) are responsible for maintaining pulp structure and function after pulp injury. DPSCs migrate directionally to the injury site before differentiating into odontoblast-like cells, which is a prerequisite and a determinant in pulp repair. Increasing evidence suggests that sensory neuron-stem cell crosstalk is critical for maintaining normal physiological functions, and sensory nerves influence stem cells mainly by neuropeptides. However, the role of sensory nerves on DPSC behaviors after pulp injury is largely unexplored. Here, we find that sensory nerves released significant amounts of calcitonin gene-related peptide (CGRP) near the injury site, acting directly on DPSCs via receptor activity modifying protein 1 (RAMP1) to promote collective migration of DPSCs to the injury site, and ultimately promoting pulp repair. Specifically, sensory denervation leads to poor pulp repair and ectopic mineralization, in parallel with that DPSCs failed to be recruited to the injury site. Furthermore, in vitro evidence shows that sensory nerve-deficient microenvironment suppressed DPSC migration prominently among all related behaviors. Mechanistically, the CGRP-Ramp1 axis between sensory neurons and DPSCs was screened by single-cell RNA-seq analysis and immunohistochemical studies confirmed that the expression of CGRP rather than Ramp1 increases substantially near the damaged site. We further demonstrated that CGRP released by sensory nerves binds the receptor Ramp1 on DPSCs to facilitate cell collective migration by an indirect co-culture system using conditioned medium from trigeminal neurons, CGRP recombinant protein and antagonists BIBN4096. The treatment with exogenous CGRP promoted the recruitment of DPSCs, and ultimately enhanced the quality of pulp repair. Targeting the sensory nerve could therefore provide a new strategy for stem cell-based pulp repair and regeneration.
Collapse
Affiliation(s)
- Chunmeng Wang
- Department of Endodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Yanchang Middle Road, Jing'an District, Shanghai, 200072, China
| | - Xiaochen Liu
- Department of Endodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Yanchang Middle Road, Jing'an District, Shanghai, 200072, China
| | - Jiani Zhou
- Department of Endodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Yanchang Middle Road, Jing'an District, Shanghai, 200072, China
| | - Xiaoyi Zhang
- Department of Endodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Yanchang Middle Road, Jing'an District, Shanghai, 200072, China
| | - Zihao Zhou
- Department of Endodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Yanchang Middle Road, Jing'an District, Shanghai, 200072, China
| | - Qi Zhang
- Department of Endodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Yanchang Middle Road, Jing'an District, Shanghai, 200072, China.
| |
Collapse
|
4
|
Roberts EL, Lepage SIM, Koch TG, Kallos MS. Bioprocess development for cord blood mesenchymal stromal cells on microcarriers in Vertical-Wheel bioreactors. Biotechnol Bioeng 2024; 121:192-205. [PMID: 37772415 DOI: 10.1002/bit.28557] [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: 01/29/2023] [Revised: 08/30/2023] [Accepted: 09/07/2023] [Indexed: 09/30/2023]
Abstract
Equine mesenchymal stromal cells (MSCs) have been found to be beneficial for the treatment of many ailments, including orthopedic injuries, due to their superior differentiation potential and immunomodulating properties. Cell therapies require large cell numbers, which are not efficiently generated using conventional static expansion methods. Expansion of equine cord blood-derived MSCs (eCB-MSCs) in bioreactors, using microcarriers as an attachment surface, has the potential to generate large numbers of cells with increased reproducibility and homogeneity compared with static T-flask expansion. This study investigated the development of an expansion process using Vertical-Wheel (VW) bioreactors, a single-use bioreactor technology that incorporates a wheel instead of an impeller. Initially, microcarriers were screened at small scale to assess eCB-MSC attachment and growth and then in bioreactors to assess cell expansion and harvesting. The effect of different donors, serial passaging, and batch versus fed batch were all examined in 0.1 L VW bioreactors. The use of VW bioreactors with an appropriate microcarrier was shown to be able to produce cell densities of up to 1E6 cells/mL, while maintaining cell phenotype and functionality, thus demonstrating great potential for the use of these bioreactors to produce large cell numbers for cell therapies.
Collapse
Affiliation(s)
- Erin L Roberts
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - Sarah I M Lepage
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Thomas G Koch
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Michael S Kallos
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
5
|
Wu L, Zhang X, Yi C, Ren H. CD146-positive adipose-derived stem cells subpopulation enriched by albumin magnetic sphere ameliorates knee osteoarthritis pain and promotes cartilage repair. J Orthop Surg Res 2023; 18:969. [PMID: 38102700 PMCID: PMC10724978 DOI: 10.1186/s13018-023-04434-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 12/01/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND The use of adipose stem cell (ADSCs) subpopulations in cartilage repair remains poorly characterized. In this study, we constructed an albumin magnetic sphere with specific targeting of CD146 (CD146-AMs) for sorting a subpopulation of CD146-positive ADSCs (CD146 + ADSCs) and explored the role of CD146 + ADSCs on joint pain and cartilage repair in rats with knee osteoarthritis (KOA). METHODS CD146-AMs were prepared and analyzed in materialistic characterization tests. Subpopulations of CD146 + ADSCs were sorted using CD146-AMs. Surface labeling, viability, and proliferation of a subpopulation of CD146 + ADSCs were evaluated in vitro. Molecular characterization of mRNA and protein expression profiles was analyzed by microarray. A rat KOA pain model was established by the iodoacetic acid method, and KOA pain and the promotion of cartilage repair were assessed after treatment with bilateral joint cavity injections of CD146 + ADSCs. RESULTS The CD146-AMs prepared in this study had an average particle size of 242.63 ± 6.74 nm, an average potential of 33.82 ± 3.53 mv, and high CD146 targeting and low cytotoxicity. The positive rate of enriched CD146 + ADSCs was 98.21% and showed a high level of stem cell marker expression and good cell viability. Gene and protein expression profiles showed that CD146 + ADSCs have different cellular functions, especially in regulating inflammation. In the KOA model, low, medium and high concentrations of CD146 + ADSCs were able to improve KOA pain and promote cartilage repair in a concentration-dependent trend. CONCLUSIONS The CD146-AMs prepared in this study were able to safely and efficiently sort out the CD146 + ADSCs subpopulation. The subpopulation of CD146 + ADSCs has a unique molecular profile that ameliorates KOA pain and repairs cartilage damage in rats, providing a new idea for KOA treatment.
Collapse
Affiliation(s)
- Lianghao Wu
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, 201399, China
| | - Xu Zhang
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, 201399, China
| | - Chengqing Yi
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, 201399, China.
| | - Hanru Ren
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, 201399, China.
| |
Collapse
|
6
|
Roberts EL, Abraham BD, Dang T, Gysel E, Mehrpouyan S, Alizadeh AH, Koch TG, Kallos MS. Computer controlled expansion of equine cord blood mesenchymal stromal cells on microcarriers in 3 L vertical-wheel ® bioreactors. Front Bioeng Biotechnol 2023; 11:1250077. [PMID: 37929186 PMCID: PMC10622666 DOI: 10.3389/fbioe.2023.1250077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/11/2023] [Indexed: 11/07/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are an ideal cell source for allogenic cell therapy due to their immunomodulatory and differentiation properties. Equine MSCs (eMSCs) have been found to be a promising treatment for equine joint injuries including meniscal injuries, cartilage degradation, and osteoarthritis. Although the use of eMSCs has shown efficacy in preliminary studies, challenges associated with biomanufacturing remain. To achieve the required cell numbers for clinical application, bioreactor-based processes are required. Initial studies have shown that eMSCs can be cultivated in microcarrier-based, stirred suspension bioreactor culture at the laboratory 0.1 L scale using a Vertical-Wheel® (VW) bioreactor. However, investigations regarding scale up of these processes to the required biomanufacturing scales are required. This study investigated the scale-up of a equine cord blood MSC (eCB-MSC) bioprocess in VW bioreactors at three scales. This included scale-up from the 0.1-0.5 L bioreactor, scale-up from static culture to the 3 L computer-controlled bioreactor, and scale-up into the 3 L computer-controlled bioreactor using a mock clinical trial process. Results from the various scale-up experiments demonstrated similar cell expansion at the various tested scales. The 3 L computer-controlled system resulted in a final cell densities of 1.5 × 105 cells/cm2 on average, achieving 1.5 × 109 harvested cells. Biological testing of the cells showed that cell phenotype and functionality were maintained after scale-up. These findings demonstrate the scalability of an eCB-MSC bioprocess using microcarriers in VW bioreactors to achieve clinically relevant cell numbers, a critical step to translate MSC treatments from research to clinical applications. This study also represents the first known published study expanding any cell type in the 3 L VW bioreactor.
Collapse
Affiliation(s)
- E. L. Roberts
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - B. D. Abraham
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - T. Dang
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - E. Gysel
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - S. Mehrpouyan
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - A. H. Alizadeh
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - T. G. Koch
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
- eQcell Inc, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - M. S. Kallos
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
7
|
Li X, Qi Q, Li Y, Miao Q, Yin W, Pan J, Zhao Z, Chen X, Yang F, Zhou X, Huang M, Wang C, Deng L, Huang D, Qi M, Fan S, Zhang Y, Qiu S, Deng W, Liu T, Chen M, Ye W, Zhang D. TCAF2 in Pericytes Promotes Colorectal Cancer Liver Metastasis via Inhibiting Cold-Sensing TRPM8 Channel. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302717. [PMID: 37635201 PMCID: PMC10602580 DOI: 10.1002/advs.202302717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/04/2023] [Indexed: 08/29/2023]
Abstract
Hematogenous metastasis is the main approach for colorectal cancer liver metastasis (CRCLM). However, as the gatekeepers in the tumor vessels, the role of TPCs in hematogenous metastasis remains largely unknown, which may be attributed to the lack of specific biomarkers for TPC isolation. Here, microdissection combined with a pericyte medium-based approach is developed to obtain TPCs from CRC patients. Proteomic analysis reveals that TRP channel-associated factor 2 (TCAF2), a partner protein of the transient receptor potential cation channel subfamily M member 8 (TRPM8), is overexpressed in TPCs from patients with CRCLM. TCAF2 in TPCs is correlated with liver metastasis, short overall survival, and disease-free survival in CRC patients. Gain- and loss-of-function experiments validate that TCAF2 in TPCs promotes tumor cell motility, epithelial-mesenchymal transition (EMT), and CRCLM, which is attenuated in pericyte-conditional Tcaf2-knockout mice. Mechanistically, TCAF2 inhibits the expression and activity of TRPM8, leading to Wnt5a secretion in TPCs, which facilitates EMT via the activation of the STAT3 signaling pathway in tumor cells. Menthol, a TRPM8 agonist, significantly suppresses Wnt5a secretion in TPCs and CRCLM. This study reveals the previously unidentified pro-metastatic effects of TPCs from the perspective of cold-sensory receptors, providing a promising diagnostic biomarker and therapeutic target for CRCLM.
Collapse
Affiliation(s)
- Xiaobo Li
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentJinan UniversityGuangzhou510632China
- College of PharmacyJinan UniversityGuangzhou510632China
| | - Qi Qi
- MOE Key Laboratory of Tumor Molecular BiologyClinical Translational Center for Targeted DrugDepartment of PharmacologySchool of MedicineJinan UniversityGuangzhou510632China
| | - Yong Li
- College of PharmacyJinan UniversityGuangzhou510632China
- School of PharmacyNorth Sichuan Medical CollegeNanchong637100China
| | - Qun Miao
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentJinan UniversityGuangzhou510632China
- College of PharmacyJinan UniversityGuangzhou510632China
| | - Wenqian Yin
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentJinan UniversityGuangzhou510632China
- College of PharmacyJinan UniversityGuangzhou510632China
| | - Jinghua Pan
- Department of General SurgeryThe First Affiliated Hospital of Jinan UniversityGuangzhou510632China
| | - Zhan Zhao
- Department of General SurgeryThe First Affiliated Hospital of Jinan UniversityGuangzhou510632China
| | - Xiaoying Chen
- Department of BiophysicsKidney Disease Center of First Affiliated HospitalZhejiang University School of MedicineHangzhou310058China
| | - Fan Yang
- Department of BiophysicsKidney Disease Center of First Affiliated HospitalZhejiang University School of MedicineHangzhou310058China
| | - Xiaofeng Zhou
- MOE Key Laboratory of Tumor Molecular BiologyClinical Translational Center for Targeted DrugDepartment of PharmacologySchool of MedicineJinan UniversityGuangzhou510632China
| | - Maohua Huang
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentJinan UniversityGuangzhou510632China
- College of PharmacyJinan UniversityGuangzhou510632China
| | - Chenran Wang
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentJinan UniversityGuangzhou510632China
- College of PharmacyJinan UniversityGuangzhou510632China
| | - Lijuan Deng
- Formula‐Pattern Research CenterSchool of Traditional Chinese MedicineJinan UniversityGuangzhou510632China
| | - Dandan Huang
- The Sixth Affiliated Hospital of Sun Yet‐Sen UniversityGuangzhou510655China
| | - Ming Qi
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentJinan UniversityGuangzhou510632China
- College of PharmacyJinan UniversityGuangzhou510632China
| | - Shuran Fan
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentJinan UniversityGuangzhou510632China
- College of PharmacyJinan UniversityGuangzhou510632China
| | - Yiran Zhang
- Department of General SurgeryThe First Affiliated Hospital of Jinan UniversityGuangzhou510632China
| | - Shenghui Qiu
- Department of General SurgeryThe First Affiliated Hospital of Jinan UniversityGuangzhou510632China
| | - Weiqing Deng
- College of PharmacyJinan UniversityGuangzhou510632China
| | - Tongzheng Liu
- College of PharmacyJinan UniversityGuangzhou510632China
| | - Minfeng Chen
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentJinan UniversityGuangzhou510632China
- College of PharmacyJinan UniversityGuangzhou510632China
| | - Wencai Ye
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentJinan UniversityGuangzhou510632China
- College of PharmacyJinan UniversityGuangzhou510632China
| | - Dongmei Zhang
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentJinan UniversityGuangzhou510632China
- College of PharmacyJinan UniversityGuangzhou510632China
| |
Collapse
|
8
|
Phyo H, Aburza A, Mellanby K, Esteves CL. Characterization of canine adipose- and endometrium-derived Mesenchymal Stem/Stromal Cells and response to lipopolysaccharide. Front Vet Sci 2023; 10:1180760. [PMID: 37275605 PMCID: PMC10237321 DOI: 10.3389/fvets.2023.1180760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/02/2023] [Indexed: 06/07/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are used for regenerative therapy in companion animals. Their potential was initially attributed to multipotency, but subsequent studies in rodents, humans and veterinary species evidenced that MSCs produce factors that are key mediators of immune, anti-infective and angiogenic responses, which are essential in tissue repair. MSCs preparations have been classically obtained from bone marrow and adipose tissue (AT) in live animals, what requires the use of surgical procedures. In contrast, the uterus, which is naturally exposed to external insult and infection, can be accessed nonsurgically to obtain samples, or tissues can be taken after neutering. In this study, we explored the endometrium (EM) as an alternative source of MSCs, which we compared with AT obtained from canine paired samples. Canine AT- and EM-MSCs, formed CFUs when seeded at low density, underwent tri-lineage differentiation into adipocytes, osteocytes and chondrocytes, and expressed the CD markers CD73, CD90 and CD105, at equivalent levels. The immune genes IL8, CCL2 and CCL5 were equally expressed at basal levels by both cell types. However, in the presence of the inflammatory stimulus lipopolysaccharide (LPS), expression of IL8 was higher in EM- than in AT-MSCs (p < 0.04) while the other genes were equally elevated in both cell types (p < 0.03). This contrasted with the results for CD markers, where the expression was unaltered by exposing the MSCs to LPS. Overall, the results indicate that canine EM-MSCs could serve as an alternative cell source to AT-MSCs in therapeutic applications.
Collapse
|
9
|
Zhang Y, Yi Y, Xiao X, Hu L, Xu J, Zheng D, Koc HC, Chan UI, Meng Y, Lu L, Liu W, Xu X, Shao N, Cheung ECW, Xu RH, Chen G. Definitive Endodermal Cells Supply an in vitro Source of Mesenchymal Stem/Stromal Cells. Commun Biol 2023; 6:476. [PMID: 37127734 PMCID: PMC10151361 DOI: 10.1038/s42003-023-04810-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/05/2023] [Indexed: 05/03/2023] Open
Abstract
Mesenchymal stem/Stromal cells (MSCs) have great therapeutic potentials, and they have been isolated from various tissues and organs including definitive endoderm (DE) organs, such as the lung, liver and intestine. MSCs have been induced from human pluripotent stem cells (hPSCs) through multiple embryonic lineages, including the mesoderm, neural crest, and extraembryonic cells. However, it remains unclear whether hPSCs could give rise to MSCs in vitro through the endodermal lineage. Here, we report that hPSC-derived, SOX17+ definitive endoderm progenitors can further differentiate to cells expressing classic MSC markers, which we name definitive endoderm-derived MSCs (DE-MSCs). Single cell RNA sequencing demonstrates the stepwise emergence of DE-MSCs, while endoderm-specific gene expression can be elevated by signaling modulation. DE-MSCs display multipotency and immunomodulatory activity in vitro and possess therapeutic effects in a mouse ulcerative colitis model. This study reveals that, in addition to the other germ layers, the definitive endoderm can also contribute to MSCs and DE-MSCs could be a cell source for regenerative medicine.
Collapse
Affiliation(s)
- Yumeng Zhang
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Ye Yi
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Xia Xiao
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Lingling Hu
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Jiaqi Xu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Dejin Zheng
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Ho Cheng Koc
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Un In Chan
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Ya Meng
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Zhuhai Precision Medical Center, Zhuhai People's Hospital, Jinan University, Zhuhai, Guangdong, China
| | - Ligong Lu
- Zhuhai Precision Medical Center, Zhuhai People's Hospital, Jinan University, Zhuhai, Guangdong, China
| | - Weiwei Liu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Biological Imaging and Stem Cell Core Facility, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
| | - Xiaoling Xu
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
| | - Ningyi Shao
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
| | - Edwin Chong Wing Cheung
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
| | - Ren-He Xu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China.
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China.
| | - Guokai Chen
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China.
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China.
| |
Collapse
|
10
|
Heilen LB, Roßgardt J, Dern-Wieloch J, Vogelsberg J, Staszyk C. Isolation and cultivation as well as in situ identification of MSCs from equine dental pulp and periodontal ligament. Front Vet Sci 2023; 10:1116671. [PMID: 36968463 PMCID: PMC10036573 DOI: 10.3389/fvets.2023.1116671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/10/2023] [Indexed: 03/12/2023] Open
Abstract
IntroductionThe lifelong eruption places a great demand on the dental pulp and periodontal ligament (PDL) of horse teeth. Cells within the pulp and PDL seem to play a key role during this remodeling.MethodsIn this study, we isolated and cultivated MSCs (medicinal signaling cells) from dental pulp, PDL and retrobulbar fat of four horses. Subsequently, we analyzed them by flow cytometry and immunohistochemistry to determine and compare their characteristics. In addition, we localized these cells within the tissue structure via immunohistochemistry of histological sections. For these analyses, several surface markers were applied.ResultsThe described method illustrates a feasible approach to isolate and cultivate MSCs from equine dental pulp and PDL. In the flow cytometry a vast majority of cultivated cells were positive for CD90 and CD40 and negative for CD11a/18, CD45, CD105 and MHCII suggesting that these cells feature characteristics of MSCs. Immunohistochemistry of histological pulp and PDL sections showed the localization of CD90 positive cells especially in the perivascular region and the subodontoblastic layer.DiscussionOur findings indicate that the isolation and cultivation of MSCs from equine dental pulp and PDL is feasible although an elaborate and complicated harvesting protocol is required. MSCs isolated from dental pulp and PDL are regarded as candidates for new therapeutical approaches in equine dental medicine like regeneration of periodontal lesions, enhancement of periodontal re-attachment after dental replantation and stimulation of pulp-obliteration and apexification in combination with endodontic therapies.
Collapse
|
11
|
Zamith Cunha R, Zannoni A, Salamanca G, De Silva M, Rinnovati R, Gramenzi A, Forni M, Chiocchetti R. Expression of cannabinoid (CB1 and CB2) and cannabinoid-related receptors (TRPV1, GPR55, and PPARα) in the synovial membrane of the horse metacarpophalangeal joint. Front Vet Sci 2023; 10:1045030. [PMID: 36937015 PMCID: PMC10020506 DOI: 10.3389/fvets.2023.1045030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/31/2023] [Indexed: 03/06/2023] Open
Abstract
Background The metacarpophalangeal joint undergoes enormous loading during locomotion and can therefore often become inflamed, potentially resulting in osteoarthritis (OA). There are studies indicating that the endocannabinoid system (ECS) modulates synovium homeostasis, and could be a promising target for OA therapy. Some cannabinoid receptors, which modulate proliferative and secretory responses in joint inflammation, have been functionally identified in human and animal synovial cells. Objective To characterize the cellular distribution of the cannabinoid receptors 1 (CB1R) and 2 (CB2R), and the cannabinoid-related receptors transient receptor potential vanilloid type 1 (TRPV1), G protein-related receptor 55 (GPR55) and peroxisome proliferator-activated receptor alpha (PPARα) in the synovial membrane of the metacarpophalangeal joint of the horse. Animals The dorsal synovial membranes of 14 equine metacarpophalangeal joints were collected post-mortem from an abattoir. Materials and methods The dorsal synovial membranes of 14 equine metacarpophalangeal joints were collected post-mortem from an abattoir. The expression of the CB1R, CB2R, TRPV1, GPR55, and PPARα in synovial tissues was studied using qualitative and quantitative immunofluorescence, and quantitative real-time reverse transcriptase PCR (qRT-PCR). Macrophage-like (MLS) and fibroblast-like (FLS) synoviocytes were identified by means of antibodies directed against IBA1 and vimentin, respectively. Results Both the mRNA and protein expression of the CB2R, TRPV1, GPR55, and PPARα were found in the synoviocytes and blood vessels of the metacarpophalangeal joints. The synoviocytes expressed the mRNA and protein of the CB1R in some of the horses investigated, but not in all. Conclusions and clinical importance Given the expression of the CB1R, CB2R, TRPV1, GPR55, and PPARα in the synovial elements of the metacarpophalangeal joint, these findings encouraged the development of new studies supporting the use of molecules acting on these receptors to reduce the inflammation during joint inflammation in the horse.
Collapse
Affiliation(s)
- Rodrigo Zamith Cunha
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Bologna, Italy
| | - Augusta Zannoni
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Bologna, Italy
| | - Giulia Salamanca
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Bologna, Italy
| | - Margherita De Silva
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Bologna, Italy
| | - Riccardo Rinnovati
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Bologna, Italy
| | - Alessandro Gramenzi
- Faculty of Veterinary Medicine, Università degli Studi di Teramo, Teramo, Italy
| | - Monica Forni
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Bologna, Italy
| | - Roberto Chiocchetti
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Bologna, Italy
- *Correspondence: Roberto Chiocchetti
| |
Collapse
|
12
|
Hagen A, Niebert S, Brandt VP, Holland H, Melzer M, Wehrend A, Burk J. Functional properties of equine adipose-derived mesenchymal stromal cells cultured with equine platelet lysate. Front Vet Sci 2022; 9:890302. [PMID: 36016806 PMCID: PMC9395693 DOI: 10.3389/fvets.2022.890302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 07/11/2022] [Indexed: 12/03/2022] Open
Abstract
Successful translation of multipotent mesenchymal stromal cell (MSC)-based therapies into clinical reality relies on adequate cell production procedures. These should be available not only for human MSC, but also for MSC from animal species relevant to preclinical research and veterinary medicine. The cell culture medium supplementation is one of the critical aspects in MSC production. Therefore, we previously established a scalable protocol for the production of buffy-coat based equine platelet lysate (ePL). This ePL proved to be a suitable alternative to fetal bovine serum (FBS) for equine adipose-derived (AD-) MSC culture so far, as it supported AD-MSC proliferation and basic characteristics. The aim of the current study was to further analyze the functional properties of equine AD-MSC cultured with the same ePL, focusing on cell fitness, genetic stability and pro-angiogenic potency. All experiments were performed with AD-MSC from n = 5 horses, which were cultured either in medium supplemented with 10% FBS, 10% ePL or 2.5% ePL. AD-MSC cultured with 2.5% ePL, which previously showed decreased proliferation potential, displayed higher apoptosis but lower senescence levels as compared to 10% ePL medium (p < 0.05). Non-clonal chromosomal aberrations occurred in 8% of equine AD-MSC cultivated with FBS and only in 4.8% of equine AD-MSC cultivated with 10% ePL. Clonal aberrations in the AD-MSC were neither observed in FBS nor in 10% ePL medium. Analysis of AD-MSC and endothelial cells in an indirect co-culture revealed that the ePL supported the pro-angiogenic effects of AD-MSC. In the 10% ePL group, more vascular endothelial growth factor (VEGF-A) was released and highest VEGF-A concentrations were reached in the presence of ePL and co-cultured cells (p < 0.05). Correspondingly, AD-MSC expressed the VEGF receptor-2 at higher levels in the presence of ePL (p < 0.05). Finally, AD-MSC and 10% ePL together promoted the growth of endothelial cells and induced the formation of vessel-like structures in two of the samples. These data further substantiate that buffy-coat-based ePL is a valuable supplement for equine AD-MSC culture media. The ePL does not only support stable equine AD-MSC characteristics as demonstrated before, but it also enhances their functional properties.
Collapse
Affiliation(s)
- Alina Hagen
- Equine Clinic (Surgery, Orthopedics), Justus-Liebig-University Giessen, Giessen, Germany
| | - Sabine Niebert
- Equine Clinic (Surgery, Orthopedics), Justus-Liebig-University Giessen, Giessen, Germany
| | - Vivian-Pascal Brandt
- Saxon Incubator for Clinical Translation (SIKT), University of Leipzig, Leipzig, Germany
| | - Heidrun Holland
- Saxon Incubator for Clinical Translation (SIKT), University of Leipzig, Leipzig, Germany
| | - Michaela Melzer
- Equine Clinic (Surgery, Orthopedics), Justus-Liebig-University Giessen, Giessen, Germany
| | - Axel Wehrend
- Clinic for Obstetrics, Gynecology and Andrology of Large and Small Animals, Justus-Liebig-University Giessen, Giessen, Germany
| | - Janina Burk
- Equine Clinic (Surgery, Orthopedics), Justus-Liebig-University Giessen, Giessen, Germany
- *Correspondence: Janina Burk
| |
Collapse
|
13
|
Jones OY, Yeralan S. Is Long COVID a State of Systemic Pericyte Disarray? J Clin Med 2022; 11:jcm11030572. [PMID: 35160024 PMCID: PMC8836446 DOI: 10.3390/jcm11030572] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 01/03/2023] Open
Abstract
The most challenging aspect of Post-Acute Sequelae of SARS-CoV-2 Infection (PASC) or Long COVID remains for the discordance between the viral damage from acute infection in the recent past and susceptibility of Long COVID without clear evidence of post infectious inflammation or autoimmune reactions. In this communication we propose that disarray of pericytes plays a central role in emerge of Long COVID. We assume pericytes are agents with “Triple-A” qualities, i.e., analyze-adapt and advance, necessary for sustainability of host homeostasis. Based on this view, we further suggest Long COVID may provide a model system to integrate system theory and complex adaptive systems to explore a new class of maladies those are currently not well defined and with no remedies.
Collapse
Affiliation(s)
- Olcay Y. Jones
- Pediatric Rheumatology, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
- Correspondence: (O.Y.J.); (S.Y.)
| | - Sencer Yeralan
- School of IT and Engineering, ADA University, Baku AZ1008, Azerbaijan
- Correspondence: (O.Y.J.); (S.Y.)
| |
Collapse
|
14
|
Bone marrow-derived mesenchymal stem cells transplantation attenuates renal fibrosis following acute kidney injury by repairing the peritubular capillaries. Exp Cell Res 2021; 411:112983. [PMID: 34921827 DOI: 10.1016/j.yexcr.2021.112983] [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: 07/31/2021] [Revised: 12/09/2021] [Accepted: 12/14/2021] [Indexed: 11/23/2022]
Abstract
After the severe initial insults of acute kidney injury, progressive kidney tubulointerstitial fibrosis may occur, the peritubular capillary (PTC) rarefaction plays a key role in the disease progression. However, the mechanisms of PTC damage were not fully understood and potential therapeutic interventions were not explored. Previous studies of our research team and others in this field suggested that bone marrow-derived mesenchymal stem cells (BMSCs) transplanted into the AKI rat model may preserve the kidney function and pathological changes. In the current study, with the ischemia/reperfusion AKI rat model, we revealed that BMSCs transplantation attenuated the renal function decrease in the AKI model through preserving the peritubular capillaries (PTCs) function. The density of PTCs is maintained by BMSCs transplantation in the AKI model, detachment and relocation of pericytes in the PTCs diminished. Then we established that BMSCs transplantation may attenuate the renal fibrosis and preserve the kidney function after AKI by repairing the PTCs. Improving the vitality of pericytes, suppressing the detachment and trans-differentiation of pericytes, directly differentiation of BMSCs into pericytes by BMSCs transplantation all participate in the PTC repair. Through these processes, BMSCs rescued the microvascular damage and improved the density of PTCs. As a result, a preliminary conclusion can be reached that BMSCs transplantation can be an effective therapy for delaying renal fibrosis after AKI.
Collapse
|
15
|
Wright A, Arthaud-Day ML, Weiss ML. Therapeutic Use of Mesenchymal Stromal Cells: The Need for Inclusive Characterization Guidelines to Accommodate All Tissue Sources and Species. Front Cell Dev Biol 2021; 9:632717. [PMID: 33665190 PMCID: PMC7921162 DOI: 10.3389/fcell.2021.632717] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
Following their discovery over 50 years ago, mesenchymal stromal cells (MSCs) have become one of the most studied cellular therapeutic products by both academia and industry due to their regenerative potential and immunomodulatory properties. The promise of MSCs as a therapeutic modality has been demonstrated by preclinical data yet has not translated to consistent, successful clinical trial results in humans. Despite the disparities across the field, MSC shareholders are unified under one common goal-to use MSCs as a therapeutic modality to improve the quality of life for those suffering from a malady in which the standard of care is suboptimal or no longer effective. Currently, there is no Food and Drug Administration (FDA)-approved MSC therapy on the market in the United States although several MSC products have been granted regulatory approval in other countries. In this review, we intend to identify hurdles that are impeding therapeutic progress and discuss strategies that may aid in accomplishing this universal goal of widespread therapeutic use.
Collapse
Affiliation(s)
- Adrienne Wright
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, United States
| | - Marne L Arthaud-Day
- Department of Management, Kansas State University, Manhattan, KS, United States
| | - Mark L Weiss
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, United States.,Midwest Institute of Comparative Stem Cell Biotechnology, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
16
|
Zha K, Tian G, Yang Z, Sun Z, Liu S, Guo Q. [The role of CD146 in mesenchymal stem cells]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2021; 35:227-233. [PMID: 33624479 DOI: 10.7507/1002-1892.202005110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective To summarize the expression and role of CD146 in mesenchymal stem cells (MSCs). Methods The literature related to CD146 at home and abroad were extensively consulted, and the CD146 expression in MSCs and its function were summarized and analyzed. Results CD146 is a transmembrane protein that mediates the adhesion of cells to cells and extracellular matrix, and is expressed on the surface of various MSCs. More and more studies have shown that CD146 + MSCs have superior cell properties such as greater proliferation, differentiation, migration, and immune regulation abilities than CD146 - or unsorted MSCs, and the application of CD146 + MSCs in the treatment of specific diseases has also achieved better results. CD146 is also involved in mediating a variety of cellular signaling pathways, but whether it plays the same role in MSCs remains to be demonstrated by further experiments. Conclusion The utilization of CD146 + MSCs for tissue regeneration will be conducive to improving the therapeutic effect of MSCs.
Collapse
Affiliation(s)
- Kangkang Zha
- Medical School of Chinese PLA, Beijing, 100853, P.R.China;Institute of Orthopaedics, the First Medical Centre, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Beijing, 100853, P.R.China;School of Medicine, Nankai University, Tianjin, 300071, P.R.China
| | - Guangzhao Tian
- Medical School of Chinese PLA, Beijing, 100853, P.R.China;Institute of Orthopaedics, the First Medical Centre, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Beijing, 100853, P.R.China;School of Medicine, Nankai University, Tianjin, 300071, P.R.China
| | - Zhen Yang
- Medical School of Chinese PLA, Beijing, 100853, P.R.China;Institute of Orthopaedics, the First Medical Centre, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Beijing, 100853, P.R.China;School of Medicine, Nankai University, Tianjin, 300071, P.R.China
| | - Zhiqiang Sun
- Medical School of Chinese PLA, Beijing, 100853, P.R.China;Institute of Orthopaedics, the First Medical Centre, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Beijing, 100853, P.R.China;School of Medicine, Nankai University, Tianjin, 300071, P.R.China
| | - Shuyun Liu
- Institute of Orthopaedics, the First Medical Centre, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Beijing, 100853, P.R.China
| | - Quanyi Guo
- Institute of Orthopaedics, the First Medical Centre, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Beijing, 100853, P.R.China
| |
Collapse
|
17
|
Weatherall EL, Avilkina V, Cortes-Araya Y, Dan-Jumbo S, Stenhouse C, Donadeu FX, Esteves CL. Differentiation Potential of Mesenchymal Stem/Stromal Cells Is Altered by Intrauterine Growth Restriction. Front Vet Sci 2020; 7:558905. [PMID: 33251256 PMCID: PMC7676910 DOI: 10.3389/fvets.2020.558905] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/16/2020] [Indexed: 12/17/2022] Open
Abstract
Consistency in clinical outcomes is key to the success of therapeutic Mesenchymal Stem/Stromal cells (MSCs) in regenerative medicine. MSCs are used to treat both humans and companion animals (horses, dogs, and cats). The properties of MSC preparations can vary significantly with factors including tissue of origin, donor age or health status. We studied the effects of developmental programming associated with intrauterine growth restriction (IUGR) on MSC properties, particularly related to multipotency. IUGR results from inadequate uterine capacity and placental insufficiency of multifactorial origin. Both companion animals (horses, dogs, cats) and livestock (pigs, sheep, cattle) can be affected by IUGR resulting in decreased body size and other associated changes that can include, alterations in musculoskeletal development and composition, and increased adiposity. Therefore, we hypothesized that this dysregulation occurs at the level of MSCs, with the cells from IUGR animals being more prone to differentiate into adipocytes and less to other lineages such as chondrocytes and osteocytes compared to those obtained from normal animals. IUGR has consequences on health and performance in adult life and in the case of farm animals, on meat quality. In humans, IUGR is linked to increased risk of metabolic (type 2 diabetes) and other diseases (cardiovascular), later in life. Here, we studied porcine MSCs where IUGR occurs spontaneously, and shows features that recapitulate human IUGR. We compared the properties of adipose-derived MSCs from IUGR (IUGR-MSCs) and Normal (Normal-MSCs) new-born pig littermates. Both MSC types grew clonally and expressed typical MSC markers (CD105, CD90, CD44) at similar levels. Importantly, tri-lineage differentiation capacity was significantly altered by IUGR. IUGR-MSCs had higher adipogenic capacity than Normal-MSCs as evidenced by higher adipocyte content and expression of the adipogenic transcripts, PPARγ and FABP4 (P < 0.05). A similar trend was observed for fibrogenesis, where, upon differentiation, IUGR-MSCs expressed significantly higher levels of COL1A1 (P < 0.03) than Normal-MSCs. In contrast, chondrogenic and osteogenic potential were decreased in IUGR-MSCs as shown by a smaller chondrocyte pellet and osteocyte staining, and lower expression of SOX9 (P < 0.05) and RUNX2 (P < 0.02), respectively. In conclusion, the regenerative potential of MSCs appears to be determined prenatally in IUGR and this should be taken into account when selecting cell donors in regenerative therapy programmes both in humans and companion animals.
Collapse
Affiliation(s)
- Emma L Weatherall
- The Roslin Institute and The Royal (DICK) School of Veterinary Studies (R(D)SVS), The University of Edinburgh, Edinburgh, United Kingdom
| | - Viktorija Avilkina
- The Roslin Institute and The Royal (DICK) School of Veterinary Studies (R(D)SVS), The University of Edinburgh, Edinburgh, United Kingdom
| | - Yennifer Cortes-Araya
- The Roslin Institute and The Royal (DICK) School of Veterinary Studies (R(D)SVS), The University of Edinburgh, Edinburgh, United Kingdom
| | - Susan Dan-Jumbo
- The Roslin Institute and The Royal (DICK) School of Veterinary Studies (R(D)SVS), The University of Edinburgh, Edinburgh, United Kingdom
| | - Claire Stenhouse
- The Roslin Institute and The Royal (DICK) School of Veterinary Studies (R(D)SVS), The University of Edinburgh, Edinburgh, United Kingdom
| | - Francesc X Donadeu
- The Roslin Institute and The Royal (DICK) School of Veterinary Studies (R(D)SVS), The University of Edinburgh, Edinburgh, United Kingdom.,The Euan Macdonald Centre, The University of Edinburgh, Edinburgh, United Kingdom
| | - Cristina L Esteves
- The Roslin Institute and The Royal (DICK) School of Veterinary Studies (R(D)SVS), The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
18
|
Mannino G, Gennuso F, Giurdanella G, Conti F, Drago F, Salomone S, Furno DL, Bucolo C, Giuffrida R. Pericyte-like differentiation of human adipose-derived mesenchymal stem cells: An in vitro study. World J Stem Cells 2020; 12:1152-1170. [PMID: 33178398 PMCID: PMC7596446 DOI: 10.4252/wjsc.v12.i10.1152] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/18/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Adipose-derived mesenchymal stem cells (ASCs) are characterized by long-term self-renewal and a high proliferation rate. Under adequate conditions, they may differentiate into cells belonging to mesodermal, endodermal or ectodermal lineages. Pericytes support endothelial cells and play an important role in stabilizing the vessel wall at the microcirculation level. The loss of pericytes, as occurs in diabetic retinopathy, results in a breakdown of the blood-retina barrier (BRB) and infiltration of inflammatory cells. In this context, the use of pericyte-like differentiated ASCs may represent a valuable therapeutic strategy for restoring BRB damage. AIM To test in vitro strategies to obtain pericyte-like differentiation of human ASCs (hASCs). METHODS Different culture conditions were tested: hASCs cultured in a basal medium supplemented with transforming growth factor β1; and hASCs cultured in a specific pericyte medium (PM-hASCs). In a further sample, pericyte growth supplement was omitted from the PM. In addition, cultures of human retinal pericytes (hRPCs) were used for comparison. Pericyte-like differentiation of hASCs was tested by immunocytochemical staining and western blotting to evaluate the expression of α-smooth muscle actin (α-SMA) and neural/glial antigen 2 (NG2). Interactions between human retinal endothelial cells (hRECs) and different groups of hASCs were investigated in co-culture experiments. In these cases, the expression of typical junctional proteins such as vascular endothelial-Cadherin, zonula occludens-1 and Occludin were assessed in hRECs. In an in vitro model of the BRB, values of trans-endothelial electrical resistance were measured when hRECs were co-cultured with various groups of pretreated hASCs. The values observed were compared with co-cultures of hRECs and hRPCs as well as with cultures of hRECs alone. Three-dimensional co-cultures of hRECs and hRPCs or pericyte-like hASCs in Matrigel were designed to assess their reciprocal localization. RESULTS After 3-6 d of culture, α-SMA and NG2 immunocytochemistry showed that the closest pericyte-like phenotype was observed when hASCs were cultured in Pericyte Medium (PM-hASCs). In particular, α-SMA immunoreactivity, already visible at the basal level in pericytes and ASCs, was strongly increased only when transforming growth factor was added to the culture medium. NG2 expression, almost undetectable in most conditions, was substantially increased only in PM-hASCs. Immunocytochemical results were confirmed by western blot analysis. The presence of pericyte growth supplement seems to increase NG2 expression rather than α-SMA, in agreement with its role in maintaining pericytes in the proliferative state. In co-culture experiments, immunoreactivity of vascular endothelial-Cadherin, zonula occludens-1 and Occludin was considerably increased in hRECs when hRPCs or PM-hASCs were also present. Supporting results were found by trans-endothelial electrical resistance measurements, gathered at 3 and 6 d of co-culture. The highest resistance values were obtained when hRECs were co-cultured with hRPCs or PM-hASCs. The pericyte-like phenotype of PM-hASCs was also confirmed in three-dimensional co-cultures in Matrigel, where PM-hASCs and hRPCs similarly localized around the tubular formations made by hRECs. CONCLUSION PM-hASCs seem able to strengthen the intercellular junctions between hRECs, likely reinforcing the BRB; thus, hASC-based therapeutic approaches may be developed to restore the integrity of retinal microcirculation.
Collapse
Affiliation(s)
- Giuliana Mannino
- Physiology Section, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Florinda Gennuso
- Pharmacology Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania 95123, Italy
| | - Giovanni Giurdanella
- Biochemistry Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania 95123, Italy
| | - Federica Conti
- Pharmacology Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania 95123, Italy
| | - Filippo Drago
- Pharmacology Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania 95123, Italy
| | - Salvatore Salomone
- Pharmacology Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania 95123, Italy
| | - Debora Lo Furno
- Physiology Section, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy.
| | - Claudio Bucolo
- Pharmacology Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania 95123, Italy
| | - Rosario Giuffrida
- Physiology Section, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| |
Collapse
|
19
|
Amniotic fluid mesenchymal stromal cells from early stages of embryonic development have higher self-renewal potential. In Vitro Cell Dev Biol Anim 2020; 56:701-714. [PMID: 33029689 DOI: 10.1007/s11626-020-00511-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/11/2020] [Indexed: 12/21/2022]
Abstract
Amniotic fluid (AF) is a rich source of mesenchymal stromal cells (MSCs) that have the ability to differentiate into multiple lineages rendering them a promising and powerful tool for regenerative medicine. However, information regarding the differences among AFMSCs derived from different gestational stages is limited. In the present study, AFMSCs derived from 125 pregnant rats at four embryonic day (E) stages (E12, E15, E18, and E21) were isolated and cultured. The primary E15 cells were the smallest in size and the easiest to culture and usually grew in a spherical shape that resembled the growth morphology of embryonic stem cells (ESCs). Once adhered, the E12 and E15 AFMSCs grew faster and could be passaged more than 60 times while still maintaining a continuous proliferative state; however, AFMSCs derived from E18 and E21 could normally be maintained for only 10 passages. To identify the possible reasons for this difference, RT-qPCR was used to examine several genes associated with self-renewal ability and cell origin. The Sox2 expression levels indicated that AFMSCs from E12 and E15 possessed stronger self-renewal capability. The K19, Col2A1, FGF5, AFP, and SPC expression levels indicated there were mixed-population cells co-existing in the AFMSC culture. In conclusion, E15 cells were easier to culture than E12 cells, could be passaged more often, and had a higher Sox2 expression than E18 or E21 cells. The E15-derived AFMSCs had higher viability and proliferative capacity than cells from the later stages. Therefore, AF cells from the early stages could be a good choice for exploring potential treatments involving AFMSCs.
Collapse
|
20
|
Kamm JL, Parlane NA, Riley CB, Gee EK, Dittmer KE, McIlwraith CW. Blood type and breed-associated differences in cell marker expression on equine bone marrow-derived mesenchymal stem cells including major histocompatibility complex class II antigen expression. PLoS One 2019; 14:e0225161. [PMID: 31747418 PMCID: PMC6867698 DOI: 10.1371/journal.pone.0225161] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 10/28/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND As the search for an immune privileged allogeneic donor mesenchymal stem cell (MSC) line continues in equine medicine, the characterization of the cells between different sources becomes important. Our research seeks to more clearly define the MSC marker expression of different equine MSC donors. METHODS The bone marrow-derived MSCs from two equine breeds and different blood donor-types were compared over successive culture passages to determine the differential expression of important antigens. Eighteen Thoroughbreds and 18 Standardbreds, including 8 blood donor (erythrocyte Aa, Ca, and Qa antigen negative) horses, were evaluated. Bone marrow was taken from each horse for isolation and culture of MSCs. Samples from passages 2, 4, 6, and 8 were labelled and evaluated by flow cytometry. The cell surface expression of CD11a/18, CD44, CD90 and MHC class II antigens were assessed. Trilineage assays for differentiation into adipogenic, chondrogenic and osteogenic lines were performed to verify characterization of the cells as MSCs. FINDINGS There were significant differences in mesenchymal stem cell marker expression between breeds and blood antigen-type groups over time. Standardbred horses showed a significantly lower expression of MHC class II than did Thoroughbred horses at passages 2, 4 and 6. CD90 was significantly higher in universal blood donor Standardbreds as compared to non-blood donor Standardbreds over all time points. All MSC samples showed high expression of CD44 and low expression of CD11a/18. CONCLUSIONS Universal blood donor- type Standardbred MSCs from passages 2-4 show the most ideal antigen expression pattern of the horses and passages that we characterized for use as a single treatment of donor bone marrow-derived MSCs. Further work is needed to determine the significance of this differential expression along with the effect of the expression of MHC I on equine bone marrow-derived MSCs.
Collapse
Affiliation(s)
- J. Lacy Kamm
- Massey University, School of Veterinary Science, Massey University, Palmerston North, New Zealand
- Veterinary Associates, Karaka, Auckland, New Zealand
- * E-mail:
| | - Natalie A. Parlane
- AgResearch, Hopkirk Research Institute, Massey University, Palmerston North, New Zealand
| | - Christopher B. Riley
- Massey University, School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | - Erica K. Gee
- Massey University, School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | - Keren E. Dittmer
- Massey University, School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | - C. Wayne McIlwraith
- Massey University, School of Veterinary Science, Massey University, Palmerston North, New Zealand
- Colorado State University, Orthopaedic Research Center, Fort Collins, Colorado, United States of America
| |
Collapse
|
21
|
Stephens CJ, Spector JA, Butcher JT. Biofabrication of thick vascularized neo-pedicle flaps for reconstructive surgery. Transl Res 2019; 211:84-122. [PMID: 31170376 PMCID: PMC6702068 DOI: 10.1016/j.trsl.2019.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 05/06/2019] [Accepted: 05/14/2019] [Indexed: 01/01/2023]
Abstract
Wound chronicity due to intrinsic and extrinsic factors perturbs adequate lesion closure and reestablishment of the protective skin barrier. Immediate and proper care of chronic wounds is necessary for a swift recovery and a reduction of patient vulnerability to infection. Advanced therapies supplemented with standard wound care procedures have been clinically implemented to restore aberrant tissue; however, these treatments are ineffective if local vasculature is too compromised to support minimally-invasive strategies. Autologous "flaps", which are tissues equipped with their own hierarchical vascular supply, can be harvested from one region of the patient and transplanted to the wound where it is reperfused upon microsurgical anastomosis to appropriate recipient vessels. Despite the success of autologous flap transfer, these procedures are extremely invasive, incur obligatory donor-site morbidity, and require sufficient donor-tissue availability, microsurgical expertise, and specialized equipment. 3D-bioprinting modalities, such as extrusion-based bioprinting, can be used to address the clinical constraints of autologous flap transfer, primarily addressing donor-site morbidity and tissue availability. This advancement in regenerative medicine allows the biofabrication of heterogeneous tissue structures with high shape fidelity and spatial resolution to generate biomimetic constructs with the anatomically-precise geometries of native tissue to ensure tissue-specific function. Yet, meaningful progress toward this clinical application has been limited by the lack of vascularization required to meet the nutrient and oxygen demands of clinically relevant tissue volumes. Thus, various criteria for the fabrication of functional tissues with hierarchical, patent vasculature must be considered when implementing 3D-bioprinting technologies for deep, chronic wounds.
Collapse
Affiliation(s)
- Chelsea J Stephens
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Jason A Spector
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York; Division of Plastic Surgery, Weill Cornell Medical College, New York, New York
| | - Jonathan T Butcher
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York.
| |
Collapse
|
22
|
Nakagomi T, Takagi T, Beppu M, Yoshimura S, Matsuyama T. Neural regeneration by regionally induced stem cells within post-stroke brains: Novel therapy perspectives for stroke patients. World J Stem Cells 2019; 11:452-463. [PMID: 31523366 PMCID: PMC6716084 DOI: 10.4252/wjsc.v11.i8.452] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/04/2019] [Accepted: 07/16/2019] [Indexed: 02/06/2023] Open
Abstract
Ischemic stroke is a critical disease which causes serious neurological functional loss such as paresis. Hope for novel therapies is based on the increasing evidence of the presence of stem cell populations in the central nervous system (CNS) and the development of stem-cell-based therapies for stroke patients. Although mesenchymal stem cells (MSCs) represented initially a promising cell source, only a few transplanted MSCs were present near the injured areas of the CNS. Thus, regional stem cells that are present and/or induced in the CNS may be ideal when considering a treatment following ischemic stroke. In this context, we have recently showed that injury/ischemia-induced neural stem/progenitor cells (iNSPCs) and injury/ischemia-induced multipotent stem cells (iSCs) are present within post-stroke human brains and post-stroke mouse brains. This indicates that iNSPCs/iSCs could be developed for clinical applications treating patients with stroke. The present study introduces the traits of mouse and human iNSPCs, with a focus on the future perspective for CNS regenerative therapies using novel iNSPCs/iSCs.
Collapse
Affiliation(s)
- Takayuki Nakagomi
- Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
- Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Toshinori Takagi
- Department of Neurosurgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Mikiya Beppu
- Department of Neurosurgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Shinichi Yoshimura
- Department of Neurosurgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Tomohiro Matsuyama
- Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| |
Collapse
|
23
|
Abstract
Besides seminal functions in angiogenesis and blood pressure regulation, microvascular pericytes possess a latent tissue regenerative potential that can be revealed in culture following transition into mesenchymal stem cells. Endowed with robust osteogenic potential, pericytes and other related perivascular cells extracted from adipose tissue represent a potent and abundant cell source for refined bone tissue engineering and improved cell therapies of fractures and other bone defects. The use of diverse bone formation assays in vivo, which include mouse muscle pocket osteogenesis and calvaria replenishment, rat and dog spine fusion, and rat non-union fracture healing, has confirmed the superiority of purified perivascular cells for skeletal (re)generation. As a surprising observation though, despite strong endogenous bone-forming potential, perivascular cells drive bone regeneration essentially indirectly, via recruitment by secreted factors of local osteo-progenitors.
Collapse
|
24
|
Beppu M, Nakagomi T, Takagi T, Nakano-Doi A, Sakuma R, Kuramoto Y, Tatebayashi K, Matsuyama T, Yoshimura S. Isolation and Characterization of Cerebellum-Derived Stem Cells in Poststroke Human Brain. Stem Cells Dev 2019; 28:528-542. [PMID: 30767605 DOI: 10.1089/scd.2018.0232] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
There is compelling evidence that the mature central nervous system (CNS) harbors stem cell populations outside conventional neurogenic regions. We previously demonstrated that brain pericytes (PCs) in both mouse and human exhibit multipotency to differentiate into various neural lineages following cerebral ischemia. PCs are found throughout the CNS, including cerebellum, but it remains unclear whether cerebellar PCs also form ischemia-induced multipotent stem cells (iSCs). In this study, we demonstrate that putative iSCs can be isolated from poststroke human cerebellum (cerebellar iSCs [cl-iSCs]). These cl-iSCs exhibited multipotency and differentiated into electrophysiologically active neurons. Neurogenic potential was also confirmed in single-cell suspensions. DNA microarray analysis revealed highly similar gene expression patterns between PCs and cl-iSCs, suggesting PC origin. Global gene expression comparison with cerebral iSCs revealed general similarity, but cl-iSCs differentially expressed certain cerebellum-specific genes. Thus, putative iSCs are present in poststroke cerebellum and possess region-specific traits, suggesting potential capacity to regenerate functional cerebellar neurons following ischemic stroke.
Collapse
Affiliation(s)
- Mikiya Beppu
- 1 Department of Neurosurgery, Hyogo College of Medicine, Nishinomiya, Japan
| | - Takayuki Nakagomi
- 2 Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Japan.,3 Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine, Nishinomiya, Japan
| | - Toshinori Takagi
- 1 Department of Neurosurgery, Hyogo College of Medicine, Nishinomiya, Japan
| | - Akiko Nakano-Doi
- 2 Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Japan.,3 Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine, Nishinomiya, Japan
| | - Rika Sakuma
- 2 Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Japan
| | - Yoji Kuramoto
- 1 Department of Neurosurgery, Hyogo College of Medicine, Nishinomiya, Japan
| | - Kotaro Tatebayashi
- 1 Department of Neurosurgery, Hyogo College of Medicine, Nishinomiya, Japan
| | - Tomohiro Matsuyama
- 3 Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine, Nishinomiya, Japan
| | - Shinichi Yoshimura
- 1 Department of Neurosurgery, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
25
|
Lepage SIM, Lee OJ, Koch TG. Equine Cord Blood Mesenchymal Stromal Cells Have Greater Differentiation and Similar Immunosuppressive Potential to Cord Tissue Mesenchymal Stromal Cells. Stem Cells Dev 2019; 28:227-237. [PMID: 30484372 DOI: 10.1089/scd.2018.0135] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are the most common cell population studied for therapeutic use in veterinary medicine. MSCs obtained from neonatal sources such as umbilical cord tissue (CT-MSCs) or cord blood (CB-MSCs) are appealing due to the non-invasive nature of procurement and the time allowed for characterization of cells before use. However, it remains unclear as to whether CB- or CT-MSCs have equivalent progenitor and non-progenitor functions. CB-MSCs have been shown to have superior chondrogenic potential to MSCs from other sources, whereas their immunomodulatory capacity does not seem to vary significantly. Using equine CB-MSCs and CT-MSCs from the same donors, we hypothesized that MSCs from both sources would have a similar immunophenotype, that CB-MSCs would be more amenable to differentiation, and that they can equally suppress lymphocyte proliferation. We evaluated cells from both sources for "classic" equine MSC markers CD90, CD105, CD29, and CD44, as well as pericyte markers CD146, NG2, and α-SMA. Contrary to our hypothesis, CB-MSCs showed mid- to high expression of pericyte surface markers CD146 and NG2, whereas expression in CT-MSCs was absent. On trilineage differentiation, CB-MSCs were more osteogenic and chondrogenic based on alkaline phosphatase activity and glycosaminoglycan content, respectively. Finally, using a mononuclear cell (MNC) suppression assay, we determined that both CB-MSCs and CT-MSCs are capable of suppressing stimulated MNC proliferation to a similar degree. We have determined that the choice of MSC tissue source should be made with the intended application in mind. This appears to be particularly relevant if pursuing a progenitor-based treatment strategy.
Collapse
Affiliation(s)
- Sarah I M Lepage
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Canada
| | - Olivia J Lee
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Canada
| | - Thomas G Koch
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Canada
| |
Collapse
|
26
|
Cortés-Araya Y, Amilon K, Rink BE, Black G, Lisowski Z, Donadeu FX, Esteves CL. Comparison of Antibacterial and Immunological Properties of Mesenchymal Stem/Stromal Cells from Equine Bone Marrow, Endometrium, and Adipose Tissue. Stem Cells Dev 2018; 27:1518-1525. [PMID: 30044182 PMCID: PMC6209426 DOI: 10.1089/scd.2017.0241] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Equine mesenchymal stem/stromal cells (MSCs) are multipotent cells that are widely used for treatment of musculoskeletal injuries, and there is significant interest in expanding their application to nonorthopedic conditions. MSCs possess antibacterial and immunomodulatory properties that may be relevant for combating infection; however, comparative studies using MSCs from different origins have not been carried out in the horse, and this was the focus of this study. Our results showed that MSC-conditioned media attenuated the growth of Escherichia coli, and that this effect was, on average, more pronounced for endometrium (EM)-derived and adipose tissue (AT)-derived MSCs than for bone marrow (BM)-derived MSCs. In addition, the antimicrobial lipocalin-2 was expressed at mean higher levels in EM-MSCs than in AT-MSCs and BM-MSCs, and the bacterial component lipopolysaccharide (LPS) stimulated its production by all three MSC types. We also showed that MSCs express interleukin-6 (IL-6), IL-8, monocyte chemoattractant protein-1, chemokine ligand-5, and Toll-like receptor 4, and that, in general, these cytokines were induced in all cell types by LPS. Low expression levels of the macrophage marker colony-stimulating factor 1 receptor were detected in BM-MSCs and EM-MSCs but not in AT-MSCs. Altogether, these findings suggest that equine MSCs from EM, AT, and BM have both direct and indirect antimicrobial properties that may vary between MSCs from different origins and could be exploited toward improvement of regenerative therapies for horses.
Collapse
Affiliation(s)
- Yennifer Cortés-Araya
- 1 The Roslin Institute and R(D)SVS, University of Edinburgh , Edinburgh, United Kingdom
| | - Karin Amilon
- 1 The Roslin Institute and R(D)SVS, University of Edinburgh , Edinburgh, United Kingdom
| | | | - Georgina Black
- 1 The Roslin Institute and R(D)SVS, University of Edinburgh , Edinburgh, United Kingdom
| | - Zofia Lisowski
- 1 The Roslin Institute and R(D)SVS, University of Edinburgh , Edinburgh, United Kingdom
| | - Francesc Xavier Donadeu
- 1 The Roslin Institute and R(D)SVS, University of Edinburgh , Edinburgh, United Kingdom .,2 The Euan Macdonald Centre for Motor Neurone Disease Research, University of Edinburgh , Edinburgh, United Kingdom
| | - Cristina L Esteves
- 1 The Roslin Institute and R(D)SVS, University of Edinburgh , Edinburgh, United Kingdom
| |
Collapse
|
27
|
Sakuma R, Takahashi A, Nakano-Doi A, Sawada R, Kamachi S, Beppu M, Takagi T, Yoshimura S, Matsuyama T, Nakagomi T. Comparative Characterization of Ischemia-Induced Brain Multipotent Stem Cells with Mesenchymal Stem Cells: Similarities and Differences. Stem Cells Dev 2018; 27:1322-1338. [PMID: 29999479 DOI: 10.1089/scd.2018.0075] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells localized to the perivascular regions of various organs, including bone marrow (BM). While MSC transplantation represents a promising stem cell-based therapy for ischemic stroke, increasing evidence indicates that exogenously administered MSCs rarely accumulate in the injured central nervous system (CNS). Therefore, compared with MSCs, regionally derived brain multipotent stem cells may be a superior source to elicit regeneration of the CNS following ischemic injury. We previously identified ischemia-induced multipotent stem cells (iSCs) as likely originating from brain pericytes/perivascular cells (PCs) within poststroke regions. However, detailed characteristics of iSCs and their comparison with MSCs remains to be investigated. In the present study, we compared iSCs with BM-derived MSCs, with a focus on the stemness and neuron-generating activity of each cell type. From our results, stem and undifferentiated cell markers, including c-myc and Klf4, were found to be expressed in iSCs and BM-MSCs. In addition, both cell types exhibited the ability to differentiate into mesoderm lineages, including as osteoblasts, adipocytes, and chondrocytes. However, compared with BM-MSCs, high expression of neural stem cell markers, including nestin and Sox2, were found in iSCs. In addition, iSCs, but not BM-MSCs, formed neurosphere-like cell clusters that differentiated into functional neurons. These results demonstrate that iSCs are likely multipotent stem cells with the ability to differentiate into not only mesoderm, but also neural, lineages. Collectively, our novel findings suggest that locally induced iSCs may contribute to CNS repair by producing neuronal cells following ischemic stroke.
Collapse
Affiliation(s)
- Rika Sakuma
- 1 Institute for Advanced Medical Sciences , Hyogo College of Medicine, Nishinomiya, Japan
| | - Ai Takahashi
- 1 Institute for Advanced Medical Sciences , Hyogo College of Medicine, Nishinomiya, Japan .,2 Graduate School of Science and Technology, Kwansei Gakuin University , Sanda, Japan
| | - Akiko Nakano-Doi
- 1 Institute for Advanced Medical Sciences , Hyogo College of Medicine, Nishinomiya, Japan .,3 Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine , Nishinomiya, Japan
| | - Rikako Sawada
- 1 Institute for Advanced Medical Sciences , Hyogo College of Medicine, Nishinomiya, Japan .,2 Graduate School of Science and Technology, Kwansei Gakuin University , Sanda, Japan
| | - Saeko Kamachi
- 3 Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine , Nishinomiya, Japan
| | - Mikiya Beppu
- 4 Department of Neurosurgery, Hyogo College of Medicine , Nishinomiya, Japan
| | - Toshinori Takagi
- 4 Department of Neurosurgery, Hyogo College of Medicine , Nishinomiya, Japan
| | - Shinichi Yoshimura
- 4 Department of Neurosurgery, Hyogo College of Medicine , Nishinomiya, Japan
| | - Tomohiro Matsuyama
- 3 Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine , Nishinomiya, Japan
| | - Takayuki Nakagomi
- 1 Institute for Advanced Medical Sciences , Hyogo College of Medicine, Nishinomiya, Japan .,3 Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine , Nishinomiya, Japan
| |
Collapse
|
28
|
Rink BE, Kuhl J, Esteves CL, French HM, Watson E, Aurich C, Donadeu FX. Reproductive stage and sex steroid hormone levels influence the expression of mesenchymal stromal cell (MSC) markers in the equine endometrium. Theriogenology 2018; 116:34-40. [PMID: 29775846 DOI: 10.1016/j.theriogenology.2018.04.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/27/2018] [Accepted: 04/27/2018] [Indexed: 01/25/2023]
Abstract
Mesenchymal stem or stromal cells (MSCs) play key roles in tissue homeostasis. In the cyclic equine endometrium, this may be regulated by changes in serum concentrations of sex steroid hormones. This study was designed to investigate the changes in endometrial expression of MSC markers during reproductive cycles in mares and the influence of sex steroid hormones on endometrial MSC proliferation in vitro. Endometrial biopsies were collected from pony mares at different reproductive stages (estrus; day 5 and 13 after ovulation; seasonal anestrus; 20 h and 7days post-partum; n = 5 per stage) and were analyzed by RT-qPCR. MSC (CD29, CD44, CD73, CD90, CD105) and perivascular (CD146, NG2) markers were present in all samples irrespective of reproductive stage. Transcript levels of most markers were present at lowest levels on day 5 after ovulation and at 20 h post-partum. MSCs isolated from endometrial tissue (n = 6 mares) were cultured in the presence of progesterone (0.01-100 μM) and estradiol 17β (0.1-1 μM), and cell proliferation was analyzed using alamarBlue® assay. Relative to cells incubated in steroid-depleted media, both progesterone and estradiol 17β moderately increased cell proliferation (1.1- and 1.2-fold, respectively) independently of the concentration used. In conclusion, our results suggest that levels of MSC markers in equine endometrium dynamically change across reproductive cycles and that MSC populations are in part regulated by sex steroids.
Collapse
Affiliation(s)
- B Elisabeth Rink
- Ross University School of Veterinary Medicine, Basseterre, St. Kitts, West Indies; Artificial Inseminaton and Embryo Transfer, Department for Companion Animals and Horses, Vetmeduni Vienna, Vienna, Austria; The Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Juliane Kuhl
- Artificial Inseminaton and Embryo Transfer, Department for Companion Animals and Horses, Vetmeduni Vienna, Vienna, Austria
| | | | - Hilari M French
- Ross University School of Veterinary Medicine, Basseterre, St. Kitts, West Indies
| | - Elaine Watson
- Ross University School of Veterinary Medicine, Basseterre, St. Kitts, West Indies
| | - Christine Aurich
- Artificial Inseminaton and Embryo Transfer, Department for Companion Animals and Horses, Vetmeduni Vienna, Vienna, Austria.
| | | |
Collapse
|
29
|
Pericytes in Veterinary Species: Prospective Isolation, Characterization and Tissue Regeneration Potential. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1109:67-77. [DOI: 10.1007/978-3-030-02601-1_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
30
|
Esteves CL, Donadeu FX. Pericytes and their potential in regenerative medicine across species. Cytometry A 2017; 93:50-59. [PMID: 28941046 DOI: 10.1002/cyto.a.23243] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 08/15/2017] [Accepted: 08/25/2017] [Indexed: 12/17/2022]
Abstract
The discovery that pericytes are in vivo counterparts of Mesenchymal Stem/Stromal Cells (MSCs) has placed these perivascular cells in the research spotlight, bringing up hope for a well-characterized cell source for clinical applications, alternative to poorly defined, heterogeneous MSCs preparations currently in use. Native pericytes express typical MSC markers and, after isolation by fluorescence-activated cell sorting, display an MSC phenotype in culture. These features have been demonstrated in different species, including humans and horses, the main targets of regenerative treatments. Significant clinical potential of pericytes has been shown by transplantation of human cells into rodent models of tissue injury, and it is hoped that future studies will demonstrate clinical potential in veterinary species. Here, we provide an overview of the current knowledge on pericytes across different species including humans, companion and large animal models, in relation to their identification in different body tissues, methodology for prospective isolation, characterization, and potential for tissue regeneration. © 2017 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- C L Esteves
- Division of Developmental Biology, The Roslin Institute, University of Edinburgh, Edinburgh EH25 9RG, United Kingdom
| | - F X Donadeu
- Division of Developmental Biology, The Roslin Institute, University of Edinburgh, Edinburgh EH25 9RG, United Kingdom
| |
Collapse
|
31
|
Novel Regenerative Therapies Based on Regionally Induced Multipotent Stem Cells in Post-Stroke Brains: Their Origin, Characterization, and Perspective. Transl Stroke Res 2017; 8:515-528. [PMID: 28744717 DOI: 10.1007/s12975-017-0556-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/07/2017] [Accepted: 07/18/2017] [Indexed: 12/16/2022]
Abstract
Brain injuries such as ischemic stroke cause severe neural loss. Until recently, it was believed that post-ischemic areas mainly contain necrotic tissue and inflammatory cells. However, using a mouse model of cerebral infarction, we demonstrated that stem cells develop within ischemic areas. Ischemia-induced stem cells can function as neural progenitors; thus, we initially named them injury/ischemia-induced neural stem/progenitor cells (iNSPCs). However, because they differentiate into more than neural lineages, we now refer to them as ischemia-induced multipotent stem cells (iSCs). Very recently, we showed that putative iNSPCs/iSCs are present within post-stroke areas in human brains. Because iNSPCs/iSCs isolated from mouse and human ischemic tissues can differentiate into neuronal lineages in vitro, it is possible that a clearer understanding of iNSPC/iSC profiles and the molecules that regulate iNSPC/iSC fate (e.g., proliferation, differentiation, and survival) would make it possible to perform neural regeneration/repair in patients following stroke. In this article, we introduce the origin and traits of iNSPCs/iSCs based on our reports and recent viewpoints. We also discuss their possible contribution to neurogenesis through endogenous and exogenous iNSPC/iSC therapies following ischemic stroke.
Collapse
|