1
|
Wei Z, Yu Q, Xie Q, Liao D, Gou X, Chen S. A comparative analysis of stem cells derived from young rabbit knee joints: the potentially superior performance of decellularized extracellular matrix pretreated infrapatellar fat pad stem cells on nanofiber scaffolds. Front Cell Dev Biol 2025; 13:1539308. [PMID: 40206399 PMCID: PMC11979179 DOI: 10.3389/fcell.2025.1539308] [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: 12/04/2024] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Introduction Osteoarthritis (OA) remains a significant clinical challenge, necessitating improved strategies for cartilage repair. Stem cells and scaffolds have crucial roles in tissue repair and regeneration. In this study, we comprehensively investigated the proliferation and differentiation potential of infrapatellar fat pad stem cells (IFPSCs), synovium-derived stem cells (SDSCs), and bone marrow stem cells (BMSCs) from unpretreated knee joints in young rabbits, and after decellularized extracellular matrix (dECM) deposition by stem cell pretreatment in vitro. Methods We also examined adhesion and differentiation effects of poly-L-lactic acid (PLLA) and poly-D, L-lactic acid (PDLLA) scaffolds after inoculation with the three stem cell types. We conducted osteogenic, adipogenic, and chondrogenic induction studies using three unpretreated stem cell groups, nine stem cell groups cross-preconditioned with different dECM types, and six stem cell groups cultured on nanofiber PLLA and PDLLA scaffolds. Staining and PCR analyses were then performed. Results In vitro studies indicated that without pretreatment, IFPSCs exhibited the highest proliferation capacity, followed by SDSCs, while BMSCs had the lowest proliferation rate. After cross-pretreatment with dECMs from different sources, IFPSCs pretreated with IECM (decellularized extracellular matrix deposited by IFPSCs) showed the greatest proliferation. BMSCs displayed the highest osteogenic potential, while SDSCs and IFPSCs showed greater chondrogenic potential. No significant differences were observed in adipogenic potential among the three groups. BMSCs exhibited reduced osteogenic potential after pretreatment with all three dECMs, whereas IFPSCs and SDSCs showed enhanced osteogenic potential following SECM and IECM pretreatment, respectively. Additionally, all 3 cell types showed reduced lipogenic potential after pretreatment with the three dECM types. For chondrogenesis, BECM pretreatment were suitable for enhancing the chondrogenic potential of all 3 cell types. Furthermore, BMSCs and IFPSCs exhibited better adhesion and survival than SDSCs on electrospun scaffolds, which mimicked dECM structures. Besides, BMSCs and IFPSCs are more suitable for PLLA to promote osteogenic, adipogenic, and chondrogenic differentiation, whereas SDSCs are better suited for PDLLA. Discussion Overall, it is anticipated that IFPSCs can be expanded with BECM pretreatment in vitro, and when combined with degradable nanofiber PLLA scaffolds in vivo, will facilitate better OA repair.
Collapse
Affiliation(s)
- Zhixin Wei
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Qingqing Yu
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Qingyun Xie
- Department of Orthopaedics, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Dongfa Liao
- Department of Orthopaedics, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Xue Gou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Song Chen
- Department of Orthopaedics, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
2
|
Wang B, Lu Z, Gao G, Mikaeiliagah E, Wang L, Yu Q, Wang Z, Hu G, Chen S, Zhang X, Pei M. Distinct role of perlecan in mesenchymal tissue regeneration via genetic and epigenetic modification. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2025; 508:161103. [PMID: 40124846 PMCID: PMC11928145 DOI: 10.1016/j.cej.2025.161103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Perlecan (HSPG2), a key component of basement membrane proteins, plays a crucial role in tissue development and regeneration. However, its direct impact on mesenchymal tissue differentiation, mediated through both genetic modification (gain- and loss-of-function mutations) and epigenetic changes (matrix microenvironment alterations), remains underexplored. In this study, we utilized CRISPR/Cas9 to achieve knockout (KO) and overexpression (OE) of HSPG2 in human fetal nucleus pulposus stem/progenitor cells (NPSCs) and adult infrapatellar fat pad-derived stem cells (IPFSCs) to investigate perlecan's influence on mesenchymal differentiation. We also assessed the effects of decellularized extracellular matrix (dECM) derived from fetal NPSCs with modified HSPG2 expression on the proliferation and differentiation of adult NPSCs. Our findings demonstrate that HSPG2-KO enhance chondrogenic differentiation, while HSPG2-OE suppressed adipogenic differentiation in both fetal NPSCs and adult IPFSCs. Notably, dECM from HSPG2-OE fetal NPSCs significantly promoted chondrogenic differentiation in adult NPSCs, suggesting potential applications for perlecan in developing advanced biomaterials for cartilage regeneration.
Collapse
Affiliation(s)
- Bin Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Department of Orthopaedics, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, China
| | - Zhihua Lu
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Medical School, Yangzhou Polytechnic College, Yangzhou, Jiangsu 225009, China
| | - Gongming Gao
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Department of Orthopaedics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Elmira Mikaeiliagah
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
| | - Lei Wang
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
| | - Qingqing Yu
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Zhuo Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Department of Orthopaedics, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, China
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Bioinformatics Core, West Virginia University, Morgantown, WV, USA
| | - Song Chen
- Department of Orthopaedics, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Xiaobing Zhang
- Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Peking Union Medical College, Tianjin, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
3
|
Jia B, Han X, Li X, Zhang L, Ma F, Wang Y, Wang X, Yan Y, Li Y, Shen J, Chen X, Li X, Zhang Q, Hu P, Du R. Deer antler reserve mesenchyme cells modified with miR-145 promote chondrogenesis in cartilage regeneration. Front Vet Sci 2024; 11:1500969. [PMID: 39776601 PMCID: PMC11705092 DOI: 10.3389/fvets.2024.1500969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Deer antler-derived reserve mesenchyme cells (RMCs) are a promising source of cells for cartilage regeneration therapy due to their chondrogenic differentiation potential. However, the regulatory mechanism has not yet been elucidated. In this study, we analyzed the role of microRNAs (miRNAs) in regulating the differentiation of RMCs and in the post-transcriptional regulation of chondrogenesis and hypertrophic differentiation at the molecular and histological levels. The results showed that RMCs showed typical MSC differentiation potentials. During chondrogenic differentiation, we obtained the expression profile of miRNAs, among which miR- 145 was the most prominent candidate as a key microRNA involved in the balance of chondral and endochondral differentiation. Knockdown of miR-145 promoted chondrogenesis and inhibited hypertrophy differentiation in RMCs. Mechanically, by prediction through online databases combined with dual-luciferase reporter assay, SOX9 was suggested as a target of miR-145. Further validation experiments confirmed that knockdown of miR-145 contributed to the balance between endochondral versus chondral differentiation of RMCs by targeting SOX9. Additionally, RMCs transfected with the miR-145-knockdown-mediated lentiviral vector successfully promoted cartilage regeneration in vivo. In summary, our study suggested that the reciprocal negative feedback between SOX9 and miR-145 was essential for balancing between endochondral versus chondral differentiation of RMCs. Our study suggested that modification of RMCs using miRNAs transduction might be an effective treatment for cartilage defects.
Collapse
Affiliation(s)
- Boyin Jia
- College of Animal Medicine/College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun, China
| | - Xintong Han
- College of Animal Medicine/College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Xin Li
- College of Animal Medicine/College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Linlin Zhang
- College of Animal Medicine/College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Fuquan Ma
- College of Animal Medicine/College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yusu Wang
- Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun, China
| | - Xue Wang
- College of Animal Medicine/College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yaru Yan
- College of Animal Medicine/College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yaxin Li
- College of Animal Medicine/College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Junnan Shen
- College of Animal Medicine/College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Xinran Chen
- College of Animal Medicine/College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Xinyi Li
- College of Animal Medicine/College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Qianzhen Zhang
- College of Animal Medicine/College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Pengfei Hu
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, China
| | - Rui Du
- Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun, China
- Yanbian University, Yanbian, China
| |
Collapse
|
4
|
Zhang Y, He X, Ge Z, Wang B, Ni M, Cai G. Investigating the differential therapeutic efficacy and mechanisms of human umbilical cord mesenchymal stem cells at various passages in osteoarthritis treatment. Tissue Cell 2024; 90:102499. [PMID: 39126832 DOI: 10.1016/j.tice.2024.102499] [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: 06/08/2023] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024]
Abstract
This study aimed to assess the clinical efficacy of umbilical cord mesenchymal stem cells (hUC-MSCs) from different passages (P3, P8, and P13) in the treatment of knee osteoarthritis (OA) and explore the underlying mechanisms. The hUC-MSCs from each passage were characterized and evaluated for their stemness, migration, proliferation, and marker expression. Rats with OA were treated with hUC-MSCs from each passage, and the therapeutic effects were assessed based on knee swelling, discomfort, and pathological examination of the knee joint. Co-culture experiments were conducted to examine the ability of hUC-MSCs to stimulate type II collagen synthesis and inhibit MMP13 expression in chondrocytes. Telomere length and telomerase activity of hUC-MSCs from each passage were measured to investigate the reasons for the observed differences in clinical efficacy. The results revealed that P3 and P8 hUC-MSCs exhibited superior osteogenic and chondrogenic differentiation potential compared to P13, while P13 demonstrated stronger adipogenic differentiation. The wound healing rate was significantly higher in the P3 and P8 groups compared to P13. All hUC-MSC groups expressed high levels of CD90 and CD105, indicating their mesenchymal stem cell characteristics, while CD31 and CD45 were not expressed. CD105 expression was significantly reduced in the P13 group. In the treatment of rat osteoarthritis, there were no significant differences in knee swelling, discomfort, Mankin scores, and pathological findings between P3 and P8 hUC-MSC treatments. However, there was a significant difference between the 8th and 13th passages. Co-culture experiments showed that hUC-MSCs from P3 and P8 enhanced type II collagen synthesis and reduced MMP13 expression in chondrocytes. Although no significant difference was observed between the P3 and P8 groups, a significant difference was found between the P13 and P8 groups. Telomere length analysis revealed that P13 samples had significantly shorter telomeres compared to both P3 and P8. The telomerase activity was positive in P3 and P8 hUC-MSCs, indicating no significant difference between these passages, while it was negative in P13 hUC-MSCs. In conclusion, P3 and P8 hUC-MSCs exhibited superior therapeutic potential for knee osteoarthritis compared to P13, possibly due to their enhanced differentiation capacity and telomerase activity.
Collapse
Affiliation(s)
- Yingkai Zhang
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China; Department of Orthopaedic Surgery, Zhongshan Hospital Fudan University, Shanghai City 200032, PR China
| | - Xianwei He
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China
| | - Zhe Ge
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China
| | - Bingnan Wang
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China
| | - Miaozhong Ni
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China
| | - Guoping Cai
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China.
| |
Collapse
|
5
|
Daldrup-Link HE, Suryadevara V, Tanyildizi Y, Nernekli K, Tang JH, Meade TJ. Musculoskeletal imaging of senescence. Skeletal Radiol 2024; 53:1879-1887. [PMID: 38329533 PMCID: PMC11303117 DOI: 10.1007/s00256-024-04585-8] [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: 11/30/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 02/09/2024]
Abstract
Senescent cells play a vital role in the pathogenesis of musculoskeletal (MSK) diseases, such as chronic inflammatory joint disorders, rheumatoid arthritis (RA), and osteoarthritis (OA). Cellular senescence in articular joints represents a response of local cells to persistent stress that leads to cell-cycle arrest and enhanced production of inflammatory cytokines, which in turn perpetuates joint damage and leads to significant morbidities in afflicted patients. It has been recently discovered that clearance of senescent cells by novel "senolytic" therapies can attenuate the chronic inflammatory microenvironment of RA and OA, preventing further disease progression and supporting healing processes. To identify patients who might benefit from these new senolytic therapies and monitor therapy response, there is an unmet need to identify and map senescent cells in articular joints and related musculoskeletal tissues. To fill this gap, new imaging biomarkers are being developed to detect and characterize senescent cells in human joints and musculoskeletal tissues. This review article will provide an overview of these efforts. New imaging biomarkers for senescence cells are expected to significantly improve the specificity of state-of-the-art imaging technologies for diagnosing musculoskeletal disorders.
Collapse
Affiliation(s)
- Heike E Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, USA.
| | - Vidyani Suryadevara
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, USA
| | - Yasemin Tanyildizi
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, USA
| | - Kerem Nernekli
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, USA
| | - Jian-Hong Tang
- Department of Chemistry, Northwestern University, Evanston, USA
| | - Thomas J Meade
- Department of Chemistry, Northwestern University, Evanston, USA
| |
Collapse
|
6
|
Jeyaraman M, Jeyaraman N, Nallakumarasamy A, Ramasubramanian S, Yadav S. Critical Challenges and Frontiers in Cartilage Tissue Engineering. Cureus 2024; 16:e53095. [PMID: 38414693 PMCID: PMC10897756 DOI: 10.7759/cureus.53095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2024] [Indexed: 02/29/2024] Open
Abstract
Cartilage tissue engineering has witnessed considerable advancements since its establishment in 1977, evolving from rudimentary surgical interventions to more nuanced biotechnological approaches. The field has navigated various challenges encompassing cellular considerations, scaffold material selection, environmental factors, and ethical and regulatory constraints. Innovations in cell source diversification, including chondrocytes, mesenchymal stem cells, and induced pluripotent stem cells, have been instrumental but not without their limitations, such as restricted cell proliferation and ethical dilemmas. Scaffold materials offer a unique dichotomy between natural substrates, which provide biocompatibility, and synthetic matrices, which grant mechanical integrity. However, translational hurdles in clinical applicability persist. Environmental factors, such as growth factors and thermal and mechanical forces, have been recognized as influential variables in cellular behavior and tissue maturation. Despite these strides, integration with host tissue remains a significant challenge, involving mechanical and immunological complexities. Looking forward, emerging technologies such as 3D and 4D printing, nanotechnology, and molecular therapies hold the promise of refining scaffold design and enhancing tissue regeneration. As the field continues to mature, a multidisciplinary approach encompassing thorough scientific investigation and collaboration is indispensable for overcoming existing challenges and realizing its full clinical potential.
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, IND
| | - Naveen Jeyaraman
- Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, IND
| | - Arulkumar Nallakumarasamy
- Orthopaedics, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Karaikal, IND
| | | | - Sankalp Yadav
- Medicine, Shri Madan Lal Khurana Chest Clinic, New Delhi, IND
| |
Collapse
|
7
|
Pei YA, Mikaeiliagah E, Wang B, Zhang X, Pei M. The matrix microenvironment influences but does not dominate tissue-specific stem cell lineage differentiation. Mater Today Bio 2023; 23:100805. [PMID: 37766896 PMCID: PMC10519827 DOI: 10.1016/j.mtbio.2023.100805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/25/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Mesenchymal stem cells (MSCs) play a pivotal role in tissue engineering and regenerative medicine, with their clinical application often hindered by cell senescence during ex vivo expansion. Recent studies suggest that MSC-deposited decellularized extracellular matrix (dECM) offers a conducive microenvironment that fosters cell proliferation and accentuates stem cell differentiation. However, the ability of this matrix environment to govern lineage differentiation of tissue-specific stem cells remains ambiguous. This research employs human adipose-derived MSCs (ADSCs) and synovium-derived MSCs (SDSCs) as models for adipogenesis and chondrogenesis differentiation pathways, respectively. Genetically modified dECM (GMdECM), produced by SV40LT-transduced immortalized cells, was studied for its influence on cell differentiation. Both types of immortalized cells displayed a reduction in chondrogenic ability but an enhancement in adipogenic potential. ADSCs grown on ADSC-deposited dECM showed stable chondrogenic potential but increased adipogenic capacity; conversely, SDSCs expanded on SDSC-generated dECM displayed elevated chondrogenic capacity and diminished adipogenic potential. This cell-dependent response was confirmed through GMdECM expansion, with SDSCs showing enhanced chondrogenesis. However, ADSCs did not exhibit improved chondrogenic potential on GMdECM, suggesting that the matrix microenvironment does not dictate the final differentiation path of tissue-specific stem cells. Potential molecular mechanisms, such as elevated basement membrane protein expression in GMdECMs and dynamic TWIST1 expression during expansion and chondrogenic induction, may underpin the strong chondrogenic differentiation of GMdECM-expanded SDSCs.
Collapse
Affiliation(s)
- Yixuan Amy Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elmira Mikaeiliagah
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Department of Biology, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| | - Bin Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Department of Foot and Hand Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu, China
| | - Xiaobing Zhang
- Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Peking Union Medical College, Tianjin, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
8
|
Arias C, Vásquez B, Salazar LA. Propolis as a Potential Therapeutic Agent to Counteract Age-Related Changes in Cartilage: An In Vivo Study. Int J Mol Sci 2023; 24:14272. [PMID: 37762574 PMCID: PMC10532056 DOI: 10.3390/ijms241814272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Aging is intricately linked to chronic low-grade systemic inflammation, which plays a significant role in various age-related conditions, including osteoarthritis (OA). The aging process significantly influences the development of OA due to alterations in cartilage composition, reduced proteoglycan content, dysregulation of growth factor signaling, and heightened oxidative stress. Propolis, a natural product renowned for its potent antioxidant and anti-inflammatory properties, has the potential to mitigate age-induced changes in cartilage. The primary objective of this study was to rigorously assess the impact of in vivo propolis treatment on the histopathological characteristics of knee articular cartilage in senescent rats. This study involved a cohort of twenty male Sprague-Dawley rats, randomly allocated into four distinct groups for comparative analysis: YR (control group consisting of young rats), SR (senescent rats), SR-EEP (senescent rats treated with an ethanolic extract of propolis, EEP), and SR-V (senescent rats administered with a control vehicle). This study employed comprehensive histological and stereological analyses of knee articular cartilage. Propolis treatment exhibited a significant capacity to alleviate the severity of osteoarthritis, enhance the structural integrity of cartilage, and augment chondrocyte density. These promising findings underscore the potential of propolis as a compelling therapeutic agent to counteract age-related alterations in cartilage and, importantly, to potentially forestall the onset of osteoarthritis.
Collapse
Affiliation(s)
- Consuelo Arias
- Escuela de Kinesiología, Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Santiago 8380000, Chile;
| | - Bélgica Vásquez
- Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Avenida Francisco Salazar 01145, Temuco 4811230, Chile
- Centre of Excellence in Morphological and Surgical Studies, Universidad de La Frontera, Avenida Francisco Salazar 01145, Temuco 4811230, Chile
| | - Luis A. Salazar
- Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Avenida Francisco Salazar 01145, Temuco 4811230, Chile
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Avenida Francisco Salazar 01145, Temuco 4811230, Chile
| |
Collapse
|
9
|
Zhang Y, Qi G, Yan Y, Wang C, Wang Z, Jiang C, Jiang Z, Ma T, Zhang C, Yan Z. Exosomes derived from bone marrow mesenchymal stem cells pretreated with decellularized extracellular matrix enhance the alleviation of osteoarthritis through miR-3473b/phosphatase and tensin homolog axis. J Gene Med 2023; 25:e3510. [PMID: 36998238 DOI: 10.1002/jgm.3510] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 02/22/2023] [Accepted: 03/27/2023] [Indexed: 04/01/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a prevalent degenerative articular disease for which there is no effective treatment. Progress has been made in mesenchymal stem cell (MSC)-based therapy in OA, and the efficacy has been demonstrated to be a result of paracrine exosomes from MSCs. Decellularized extracellular matrix (dECM) provides an optimum microenvironment for the expansion of MSCs. In the present study, we aimed to investigate whether exosomes isolated from bone marrow mesenchymal stem cells (BMSCs) with dECM pretreatment (dECM-BMSC-Exos) enhance the amelioration of OA. METHODS Exosomes from BMSCs with or without dECM pretreatment were isolated. We measured and compared the effect of the BMSC-Exo and dECM-BMSC-Exo on interleukin (IL)-1β-induced chondrocytes by analyzing proliferation, anabolism and catabolism, migration and apoptosis in vitro. The in vivo experiment was performed by articular injection of exosomes into DMM mice, followed by histological evaluation of cartilage. MicroRNA sequencing of exosomes was performed on BMSC-Exo and dECM-BMSC-Exo to investigate the underlying mechanism. The function of miR-3473b was validated by rescue studies in vitro and in vivo using antagomir-3473b. RESULTS IL-1β-treated chondrocytes treated with dECM-BMSC-Exos showed enhanced proliferation, anabolism, migration and anti-apoptosis properties compared to BMSC-Exos. DMM mice injected with dECM-BMSC-Exo showed better cartilage regeneration than those injected with BMSC-Exo. Interestingly, miR-3473b was significantly elevated in dECM-BMSC-Exos and was found to mediate the protective effect in chondrocytes by targeting phosphatase and tensin homolog (PTEN), which activated the PTEN/AKT signaling pathway. CONCLUSIONS dECM-BMSC-Exo can enhance the alleviation of osteoarthritis via promoting migration, improving anabolism and inhibiting apoptosis of chondrocytes by upregulating miR-3473b, which targets PTEN.
Collapse
Affiliation(s)
- Yueqi Zhang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guobin Qi
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuheng Yan
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chenzhong Wang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhe Wang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chang Jiang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zengxin Jiang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai, China
| | - Tianle Ma
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chi Zhang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zuoqin Yan
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Acellular nerve grafts supplemented with induced pluripotent stem cell-derived exosomes promote peripheral nerve reconstruction and motor function recovery. Bioact Mater 2022; 15:272-287. [PMID: 35356813 PMCID: PMC8935093 DOI: 10.1016/j.bioactmat.2021.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/05/2021] [Accepted: 12/10/2021] [Indexed: 12/14/2022] Open
Abstract
Peripheral nerve injury is a great challenge in clinical work due to the restricted repair gap and weak regrowth ability. Herein, we selected induced pluripotent stem cells (iPSCs) derived exosomes to supplement acellular nerve grafts (ANGs) with the aim of restoring long-distance peripheral nerve defects. Human fibroblasts were reprogrammed into iPSCs through non-integrating transduction of Oct3/4, Sox2, Klf4, and c-Myc. The obtained iPSCs had highly active alkaline phosphatase expression and expressed Oct4, SSEA4, Nanog, Sox2, which also differentiated into all three germ layers in vivo and differentiated into mature peripheral neurons and Schwann cells (SCs) in vitro. After isolation and biological characteristics of iPSCs-derived exosomes, we found that numerous PKH26-labeled exosomes were internalized inside SCs through endocytotic pathway and exhibited a proliferative effect on SCs that were involved in the process of axonal regeneration and remyelination. After that, we prepared ANGs via optimized chemical extracted process to bridge 15 mm long-distance peripheral nerve gaps in rats. Owing to the promotion of iPSCs-derived exosomes, satisfactory regenerative outcomes were achieved including gait behavior analysis, electrophysiological assessment, and morphological analysis of regenerated nerves. Especially, motor function was restored with comparable to those achieved with nerve autografts and there were no significant differences in the fiber diameter and area of reinnervated muscle fibers. Taken together, our combined use of iPSCs-derived exosomes with ANGs demonstrates good promise to restore long-distance peripheral nerve defects, and thus represents a cell-free strategy for future clinical applications. IPSCs-derived exosomes provide a novel cell-free strategy with the regenerative power of iPSCs. ANGs supplemented with iPSCs-derived exosomes show enhanced peripheral repair and accelerated motor functional recovery. IPSCs-derived exosomes provide equivalent histological morphology to autologous nerve transplantation.
Collapse
|
11
|
Chen T, Peng Y, Hu W, Shi H, Li P, Que Y, Qiu J, Qiu X, Gao B, Zhou H, Chen Y, Zhu Y, Li S, Liang A, Gao W, Huang D. Irisin enhances chondrogenic differentiation of human mesenchymal stem cells via Rap1/PI3K/AKT axis. Stem Cell Res Ther 2022; 13:392. [PMID: 35922833 PMCID: PMC9351134 DOI: 10.1186/s13287-022-03092-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/17/2022] [Indexed: 12/11/2022] Open
Abstract
Background Human mesenchymal stem cells (hMSCs) have been proven to have inherent chondrogenic differentiation potential, which appears to be used in cartilage regeneration. Increasing evidence suggests that irisin enhances osteoblast differentiation of MSCs, but little is known about its potential on chondrogenic differentiation. Methods In the study, we investigated the effects of irisin on chondrogenic differentiation of hMSCs using a high-density pellet culture system. The cartilage pellets were evaluated by morphology, and the metabolism of cartilage matrix was detected by qPCR, western blot and immunohistochemistry. Next, RNA-seq was performed to explore the underlying mechanism. Furthermore, using the transduction of plasmid, miRNAs mimics and inhibitor, the activation of Rap1/PI3K/AKT axis, the expression level of SIPA1L2, and the functional verification of miR-125b-5p were detected on day 7 of chondrogenic differentiation of hMSCs. Results Compared with the controls, we found that irisin treatment could significantly enhance the chondrogenic differentiation of hMSCs, enlarge the induced-cartilage tissue and up-regulate the expression levels of cartilage markers. RNA-seq indicated that irisin activated the Rap1 and PI3K/AKT signaling pathway, and the lower expression level of SIPA1L2 and the higher expression level of miR-125b-5p were found in irisin-treated group. Further, we found that irisin treatment could up-regulate the expression level of miR-125b-5p, targeting SIPA1L2 and consequently activating the Rap1/PI3K/AKT axis on the process of chondrogenic differentiation of hMSCs. Conclusions Collectively, our study reveals that irisin can enhance chondrogenic differentiation of hMSCs via the Rap1/PI3K/AKT pathway, suggesting that irisin possesses prospects in cartilage regeneration. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03092-8.
Collapse
Affiliation(s)
- Taiqiu Chen
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Yan Peng
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Wenjun Hu
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Huihong Shi
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Pengfei Li
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Yichen Que
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Jincheng Qiu
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Xianjian Qiu
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Bo Gao
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Hang Zhou
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Yanbo Chen
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Yuanxin Zhu
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Shaoguang Li
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Anjing Liang
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Wenjie Gao
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China.
| | - Dongsheng Huang
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China.
| |
Collapse
|
12
|
High expression of Piezo1 induces senescence in chondrocytes through calcium ions accumulation. Biochem Biophys Res Commun 2022; 607:138-145. [DOI: 10.1016/j.bbrc.2022.03.119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 11/21/2022]
|
13
|
Biodegradable Poly(D-L-lactide-co-glycolide) (PLGA)-Infiltrated Bioactive Glass (CAR12N) Scaffolds Maintain Mesenchymal Stem Cell Chondrogenesis for Cartilage Tissue Engineering. Cells 2022; 11:cells11091577. [PMID: 35563883 PMCID: PMC9100331 DOI: 10.3390/cells11091577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/01/2022] [Accepted: 05/03/2022] [Indexed: 12/11/2022] Open
Abstract
Regeneration of articular cartilage remains challenging. The aim of this study was to increase the stability of pure bioactive glass (BG) scaffolds by means of solvent phase polymer infiltration and to maintain cell adherence on the glass struts. Therefore, BG scaffolds either pure or enhanced with three different amounts of poly(D-L-lactide-co-glycolide) (PLGA) were characterized in detail. Scaffolds were seeded with primary porcine articular chondrocytes (pACs) and human mesenchymal stem cells (hMSCs) in a dynamic long-term culture (35 days). Light microscopy evaluations showed that PLGA was detectable in every region of the scaffold. Porosity was greater than 70%. The biomechanical stability was increased by polymer infiltration. PLGA infiltration did not result in a decrease in viability of both cell types, but increased DNA and sulfated glycosaminoglycan (sGAG) contents of hMSCs-colonized scaffolds. Successful chondrogenesis of hMSC-colonized scaffolds was demonstrated by immunocytochemical staining of collagen type II, cartilage proteoglycans and the transcription factor SOX9. PLGA-infiltrated scaffolds showed a higher relative expression of cartilage related genes not only of pAC-, but also of hMSC-colonized scaffolds in comparison to the pure BG. Based on the novel data, our recommendation is BG scaffolds with single infiltrated PLGA for cartilage tissue engineering.
Collapse
|
14
|
Jelodari S, Ebrahimi Sadrabadi A, Zarei F, Jahangir S, Azami M, Sheykhhasan M, Hosseini S. New Insights into Cartilage Tissue Engineering: Improvement of Tissue-Scaffold Integration to Enhance Cartilage Regeneration. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7638245. [PMID: 35118158 PMCID: PMC8807044 DOI: 10.1155/2022/7638245] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 12/20/2021] [Accepted: 12/29/2021] [Indexed: 02/05/2023]
Abstract
Distinctive characteristics of articular cartilage such as avascularity and low chondrocyte conversion rate present numerous challenges for orthopedists. Tissue engineering is a novel approach that ameliorates the regeneration process by exploiting the potential of cells, biodegradable materials, and growth factors. However, problems exist with the use of tissue-engineered construct, the most important of which is scaffold-cartilage integration. Recently, many attempts have been made to address this challenge via manipulation of cellular, material, and biomolecular composition of engineered tissue. Hence, in this review, we highlight strategies that facilitate cartilage-scaffold integration. Recent advances in where efficient integration between a scaffold and native cartilage could be achieved are emphasized, in addition to the positive aspects and remaining problems that will drive future research.
Collapse
Affiliation(s)
- Sahar Jelodari
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amin Ebrahimi Sadrabadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fatemeh Zarei
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Mahmoud Azami
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Sheykhhasan
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research (ACECR), Qom Branch, Qom, Iran
| | - Samaneh Hosseini
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
15
|
Xiang XN, Zhu SY, He HC, Yu X, Xu Y, He CQ. Mesenchymal stromal cell-based therapy for cartilage regeneration in knee osteoarthritis. Stem Cell Res Ther 2022; 13:14. [PMID: 35012666 PMCID: PMC8751117 DOI: 10.1186/s13287-021-02689-9] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/07/2021] [Indexed: 02/08/2023] Open
Abstract
Osteoarthritis, as a degenerative disease, is a common problem and results in high socioeconomic costs and rates of disability. The most commonly affected joint is the knee and characterized by progressive destruction of articular cartilage, loss of extracellular matrix, and progressive inflammation. Mesenchymal stromal cell (MSC)-based therapy has been explored as a new regenerative treatment for knee osteoarthritis in recent years. However, the detailed functions of MSC-based therapy and related mechanism, especially of cartilage regeneration, have not been explained. Hence, this review summarized how to choose, authenticate, and culture different origins of MSCs and derived exosomes. Moreover, clinical application and the latest mechanistical findings of MSC-based therapy in cartilage regeneration were also demonstrated.
Collapse
Affiliation(s)
- Xiao-Na Xiang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Si-Yi Zhu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Hong-Chen He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xi Yu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yang Xu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Cheng-Qi He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China. .,School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China. .,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China. .,Rehabilitation Medicine Centre, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
16
|
Pei YA, Pei M. Hypoxia Modulates Regenerative Potential of Fetal Stem Cells. APPLIED SCIENCES (BASEL, SWITZERLAND) 2022; 12:363. [PMID: 36660242 PMCID: PMC9846719 DOI: 10.3390/app12010363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Adult mesenchymal stem cells (MSCs) are prone to senescence, which limits the scope of their use in tissue engineering and regeneration and increases the likelihood of post-implantation failure. As a robust alternative cell source, fetal stem cells can prevent an immune reaction and senescence. However, few studies use this cell type. In this study, we sought to characterize fetal cells' regenerative potential in hypoxic conditions. Specifically, we examined whether hypoxic exposure during the expansion and differentiation phases would affect human fetal nucleus pulposus cell (NPC) and fetal synovium-derived stem cell (SDSC) plasticity and three-lineage differentiation potential. We concluded that fetal NPCs represent the most promising cell source for chondrogenic differentiation, as they are more responsive and display stronger phenotypic stability, particularly when expanded and differentiated in hypoxic conditions. Fetal SDSCs have less potential for chondrogenic differentiation compared to their adult counterpart. This study also indicated that fetal SDSCs exhibit a discrepancy in adipogenic and osteogenic differentiation in response to hypoxia.
Collapse
Affiliation(s)
- Yixuan Amy Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA
- WVU Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
17
|
Wang Y, Pei YA, Sun Y, Zhou S, Zhang XB, Pei M. Stem cells immortalized by hTERT perform differently from those immortalized by SV40LT in proliferation, differentiation, and reconstruction of matrix microenvironment. Acta Biomater 2021; 136:184-198. [PMID: 34551328 PMCID: PMC8627502 DOI: 10.1016/j.actbio.2021.09.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 11/22/2022]
Abstract
Although matrix microenvironment has the potential to improve expanded stem cell proliferation and differentiation capacity, decellularized extracellular matrix (dECM) deposited by senescent cells does not contribute to the rejuvenation of adult stem cells, which has become a barrier to personalized stem cell therapy. Genetic modification is an effective strategy to protect cells from senescence but it carries the increased risk of malignant transformation and genetic instability. In this study, lentivirus carrying either human telomerase reverse transcriptase (hTERT) or simian virus 40 large T antigen (SV40LT) was used to transduce human infrapatellar fat pad-derived stem cells (IPFSCs). We found that virus transduction modified the proliferative, chondrogenic, and adipogenic abilities of IPFSCs. Interestingly, dECM deposited by immortalized cells significantly influenced replicative senescent IPFSCs in proliferation and differentiation preference, the effect of which is hinged on the approach of immortalization using either SV40LT or hTERT. Our findings indicate both dECM expansion and immortalization strategies can be used for replicative senescent adult stem cells' proliferation and lineage-specific differentiation, which benefits future stem cell-based tissue regeneration. This approach may also work for adult stem cells with premature senescence in elderly/aged patients, which needs further investigation. STATEMENT OF SIGNIFICANCE: Adult stem cells are a promising solution for autologous cell-based therapy. Unfortunately, cell senescence due to donor age and/or ex vivo expansion prevents clinical application. Recent progress with decellularized extracellular matrix provides a potential for the rejuvenation of senescent stem cells by improving their proliferation and differentiation capacities. Given the fact that the young matrix can provide a healthy and energetic microenvironment, in this study, two approaches using lentivirus transduction of hTERT and SV40LT were compared. The goal was to immortalize donor cells for deposition of decellularized extracellular matrix. The matrix was demonstrated to contribute diverging effects on the chondrogenic and adipogenic differentiation of expanded stem cells and exhibited proliferation benefits as well. These findings provide an invaluable asset for stem cell-based tissue regeneration.
Collapse
Affiliation(s)
- Yiming Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA; Department of Joint Surgery, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Yixuan Amy Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA; Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yuan Sun
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
| | - Sheng Zhou
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
| | - Xiao-Bing Zhang
- State Key Laboratory of Experimental Hematology, Tianjin, China; Department of Medicine, Loma Linda University, Loma Linda, CA, USA.
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA; WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
18
|
Solanki P, Ansari MD, Anjali, Khan I, Jahan RN, Nikita, Pandit J, Aqil M, Ahmad FJ, Sultana Y. Repurposing pentosan polysulfate sodium as hyaluronic acid linked polyion complex nanoparticles for the management of osteoarthritis: A potential approach. Med Hypotheses 2021; 157:110713. [PMID: 34710749 DOI: 10.1016/j.mehy.2021.110713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/10/2021] [Accepted: 10/17/2021] [Indexed: 10/20/2022]
Abstract
Osteoarthritis is still a disease burden for pharmaceutical scientists and strategy makers. It is associated with the chronic inflammation of joints especially weight-bearing joints like knee, hip, backbone, and phalanges. NSAIDs that are used for the management of inflammation associated with osteoarthritis have high side effects related to gastric upset, gastric ulcer, and long term treatment associated with liver and kidney damage. Nanotechnology has gained a huge scope for the management of arthritis as it can reach out to the deep inside the cell and alter cellular physiology as desired. The present study hypothesizes the use of polyion complex nanoparticles of hyaluronic acid linked Pentosan polysulfate sodium, a disease-modifying agent for the treatment of osteoarthritis administered through transdermal route. The hypothesis involves the use of drug repurposing as the drug was initially approved for interstitial cystitis, a condition of the urinary bladder associated with pain and swelling. Being very low oral bioavailability and gastric irritation profile, the transdermal route would be beneficial. To overcome the problem associated with the oral route, there is a need for the targeted approach that will particularly reach at inflammatory sites. Thereby transdermal delivery of hyaluronic acid linked Pentosan polysulfate sodium through polyion complex nanoparticle therapy will be a novel therapeutic approach to combat osteoarthritis.
Collapse
Affiliation(s)
- Pavitra Solanki
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062 India.
| | - Mohd Danish Ansari
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062 India
| | - Anjali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062 India
| | - Iram Khan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062 India
| | - Rao Nargis Jahan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062 India
| | - Nikita
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062 India
| | - Jayamanti Pandit
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062 India
| | - Mohd Aqil
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062 India
| | - Farhan J Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062 India
| | - Yasmin Sultana
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062 India.
| |
Collapse
|
19
|
Tudorachi NB, Totu EE, Fifere A, Ardeleanu V, Mocanu V, Mircea C, Isildak I, Smilkov K, Cărăuşu EM. The Implication of Reactive Oxygen Species and Antioxidants in Knee Osteoarthritis. Antioxidants (Basel) 2021; 10:985. [PMID: 34205576 PMCID: PMC8233827 DOI: 10.3390/antiox10060985] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/11/2021] [Accepted: 06/17/2021] [Indexed: 12/16/2022] Open
Abstract
Knee osteoarthritis (KOA) is a chronic multifactorial pathology and a current and essential challenge for public health, with a negative impact on the geriatric patient's quality of life. The pathophysiology is not fully known; therefore, no specific treatment has been found to date. The increase in the number of newly diagnosed cases of KOA is worrying, and it is essential to reduce the risk factors and detect those with a protective role in this context. The destructive effects of free radicals consist of the acceleration of chondrosenescence and apoptosis. Among other risk factors, the influence of redox imbalance on the homeostasis of the osteoarticular system is highlighted. The evolution of KOA can be correlated with oxidative stress markers or antioxidant status. These factors reveal the importance of maintaining a redox balance for the joints and the whole body's health, emphasizing the importance of an individualized therapeutic approach based on antioxidant effects. This paper aims to present an updated picture of the implications of reactive oxygen species (ROS) in KOA from pathophysiological and biochemical perspectives, focusing on antioxidant systems that could establish the premises for appropriate treatment to restore the redox balance and improve the condition of patients with KOA.
Collapse
Affiliation(s)
- Nicoleta Bianca Tudorachi
- Faculty of Medicine, “Ovidius” University of Constanța, Mamaia Boulevard 124, 900527 Constanța, Romania; (N.B.T.); (V.A.)
| | - Eugenia Eftimie Totu
- Faculty of Applied Chemistry and Material Science, University Politehnica of Bucharest, 1–5 Polizu Street, 011061 Bucharest, Romania
| | - Adrian Fifere
- Centre of Advanced Research in Bionanoconjugates and Biopolymers Department, “Petru Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania
| | - Valeriu Ardeleanu
- Faculty of Medicine, “Ovidius” University of Constanța, Mamaia Boulevard 124, 900527 Constanța, Romania; (N.B.T.); (V.A.)
| | - Veronica Mocanu
- Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania; (V.M.); (C.M.)
| | - Cornelia Mircea
- Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania; (V.M.); (C.M.)
| | - Ibrahim Isildak
- Faculty of Chemistry-Metallurgy, Department of Bioengineering, Yildiz Technical University, Istanbul 34220, Turkey;
| | - Katarina Smilkov
- Faculty of Medical Sciences, Division of Pharmacy, Department of Applied Pharmacy, Goce Delcev University, Krste Misirkov Street, No. 10-A, 2000 Stip, North Macedonia;
| | - Elena Mihaela Cărăuşu
- Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, “Nicolae Leon” Building, 13 Grigore Ghica Street, 700259 Iasi, Romania;
| |
Collapse
|
20
|
Stone RN, Frahs SM, Hardy MJ, Fujimoto A, Pu X, Keller-Peck C, Oxford JT. Decellularized Porcine Cartilage Scaffold; Validation of Decellularization and Evaluation of Biomarkers of Chondrogenesis. Int J Mol Sci 2021; 22:6241. [PMID: 34207917 PMCID: PMC8230108 DOI: 10.3390/ijms22126241] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/01/2021] [Accepted: 06/04/2021] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis is a major concern in the United States and worldwide. Current non-surgical and surgical approaches alleviate pain but show little evidence of cartilage restoration. Cell-based treatments may hold promise for the regeneration of hyaline cartilage-like tissue at the site of injury or wear. Cell-cell and cell-matrix interactions have been shown to drive cell differentiation pathways. Biomaterials for clinically relevant applications can be generated from decellularized porcine auricular cartilage. This material may represent a suitable scaffold on which to seed and grow chondrocytes to create new cartilage. In this study, we used decellularization techniques to create an extracellular matrix scaffold that supports chondrocyte cell attachment and growth in tissue culture conditions. Results presented here evaluate the decellularization process histologically and molecularly. We identified new and novel biomarker profiles that may aid future cartilage decellularization efforts. Additionally, the resulting scaffold was characterized using scanning electron microscopy, fluorescence microscopy, and proteomics. Cellular response to the decellularized scaffold was evaluated by quantitative real-time PCR for gene expression analysis.
Collapse
Affiliation(s)
- Roxanne N. Stone
- Interdisciplinary Studies Program, Boise State University, Boise, ID 83725, USA;
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
| | - Stephanie M. Frahs
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
- Biomolecular Sciences Graduate Programs, Boise State University, Boise, ID 83725, USA
| | - Makenna J. Hardy
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
- Biomolecular Sciences Graduate Programs, Boise State University, Boise, ID 83725, USA
| | - Akina Fujimoto
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
| | - Xinzhu Pu
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
- Biomolecular Sciences Graduate Programs, Boise State University, Boise, ID 83725, USA
| | - Cynthia Keller-Peck
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
| | - Julia Thom Oxford
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
- Biomolecular Sciences Graduate Programs, Boise State University, Boise, ID 83725, USA
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| |
Collapse
|
21
|
Huang Y, He Y, Makarcyzk MJ, Lin H. Senolytic Peptide FOXO4-DRI Selectively Removes Senescent Cells From in vitro Expanded Human Chondrocytes. Front Bioeng Biotechnol 2021; 9:677576. [PMID: 33996787 PMCID: PMC8116695 DOI: 10.3389/fbioe.2021.677576] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 04/12/2021] [Indexed: 12/16/2022] Open
Abstract
Autologous chondrocyte implantation (ACI) is a procedure used to treat articular cartilage injuries and prevent the onset of post-traumatic osteoarthritis. In vitro expansion of chondrocytes, a necessary step in ACI, results in the generation of senescent cells that adversely affect the quality and quantity of newly formed cartilage. Recently, a senolytic peptide, fork head box O transcription factor 4-D-Retro-Inverso (FOXO4-DRI), was reported to selectively kill the senescent fibroblasts. In this study, we hypothesized that FOXO4-DRI treatment could remove the senescent cells in the expanded chondrocytes, thus enhancing their potential in generating high-quality cartilage. To simulate the in vitro expansion for ACI, chondrocytes isolated from healthy donors were expanded to population doubling level (PDL) 9, representing chondrocytes ready for implantation. Cells at PDL3 were also used to serve as the minimally expanded control. Results showed that the treatment of FOXO4-DRI removed more than half of the cells in PDL9 but did not significantly affect the cell number of PDL3 chondrocytes. Compared to the untreated control, the senescence level in FOXO4-DRI treated PDL9 chondrocytes was significantly reduced. Based on the result from standard pellet culture, FOXO4-DRI pre-treatment did not enhance the chondrogenic potential of PDL9 chondrocytes. However, the cartilage tissue generated from FOXO4-DRI pretreated PDL9 cells displayed lower expression of senescence-relevant secretory factors than that from the untreated control group. Taken together, FOXO4-DRI is able to remove the senescent cells in PDL9 chondrocytes, but its utility in promoting cartilage formation from the in vitro expanded chondrocytes needs further investigation.
Collapse
Affiliation(s)
- Yuzhao Huang
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuchen He
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Meagan J Makarcyzk
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
22
|
You R, Wang L, Liu L, Wang Y, Han K, Lin H, Wang Y, Raftery D, Guan YQ. Probing cell metabolism on insulin like growth factor(IGF)-1/tumor necrosis factor(TNF)-α and chargeable polymers co-immobilized conjugates. J Tissue Eng Regen Med 2021; 15:256-268. [PMID: 33462987 DOI: 10.1002/term.3174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 11/08/2022]
Abstract
Cell culturing on different synthetic biomaterials would reprogram cell metabolism for adaption to their living conditions because such alterations in cell metabolism were necessary for cellular functions on them. Here we used metabolomics to uncover metabolic changes when liver cells were cultured on insulin-like growth factor (IGF)/tumor necrosis factor-α (TNF-α) and chargeable polymers co-modified biomaterials with the aim to explain their modulating effects on cell metabolism. The results showed that cell metabolism on IGF-1/TNF-α co-immobilized conjugates was significantly regulated according to their scatterings on the score plot of principal component analysis. Specifically, cell metabolisms were reprogrammed to the higher level of pyrimidine metabolism, β-alanine metabolism, and pantothenate and CoA biosynthesis, and the lower level of methionine salvage pathway in order to promote cell growth on IGF/TNF-α co-modified surface. Furthermore, cell senescence on PSt-PAAm-IGF/TNF-α surface was delayed through the regulation of branch amino acid metabolism and AMPK signal pathway. The research showed that metabolomics had great potential to uncover the molecular interaction between biomaterials and seeded cells, and provide the insights about cell metabolic reprogramming on IGF/TNF-α co-modified conjugates for cell growth.
Collapse
Affiliation(s)
- Rong You
- School of Life Science, South China Normal University, Guangzhou, China.,South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, China
| | - Lanqing Wang
- School of Life Science, South China Normal University, Guangzhou, China.,South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, China
| | - Li Liu
- School of Life Science, South China Normal University, Guangzhou, China.,South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, China
| | - Yuanjian Wang
- School of Life Science, South China Normal University, Guangzhou, China.,South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, China
| | - Kaibin Han
- School of Life Science, South China Normal University, Guangzhou, China.,South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, China
| | - Haiting Lin
- School of Life Science, South China Normal University, Guangzhou, China.,South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, China
| | - Yibei Wang
- School of Life Science, South China Normal University, Guangzhou, China.,South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, China
| | | | - Yan-Qing Guan
- School of Life Science, South China Normal University, Guangzhou, China.,South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, China.,Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
23
|
Wang Y, Hu G, Hill RC, Dzieciatkowska M, Hansen KC, Zhang XB, Yan Z, Pei M. Matrix reverses immortalization-mediated stem cell fate determination. Biomaterials 2021; 265:120387. [PMID: 32987274 PMCID: PMC7944411 DOI: 10.1016/j.biomaterials.2020.120387] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/24/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
Primary cell culture in vitro suffers from cellular senescence. We hypothesized that expansion on decellularized extracellular matrix (dECM) deposited by simian virus 40 large T antigen (SV40LT) transduced autologous infrapatellar fat pad stem cells (IPFSCs) could rejuvenate high-passage IPFSCs in both proliferation and chondrogenic differentiation. In the study, we found that SV40LT transduced IPFSCs exhibited increased proliferation and adipogenic potential but decreased chondrogenic potential. Expansion on dECMs deposited by passage 5 IPFSCs yielded IPFSCs with dramatically increased proliferation and chondrogenic differentiation capacity; however, this enhanced capacity diminished if IPFSCs were grown on dECM deposited by passage 15 IPFSCs. Interestingly, expansion on dECM deposited by SV40LT transduced IPFSCs yielded IPFSCs with enhanced proliferation and chondrogenic capacity but decreased adipogenic potential, particularly for the dECM group derived from SV40LT transduced passage 15 cells. Our immunofluorescence staining and proteomics data identify matrix components such as basement membrane proteins as top candidates for matrix mediated IPFSC rejuvenation. Both cell proliferation and differentiation were endorsed by transcripts measured by RNASeq during the process. This study provides a promising model for in-depth investigation of the matrix protein influence on surrounding stem cell differentiation.
Collapse
Affiliation(s)
- Yiming Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA; Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA; Bioinformatics Core, West Virginia University, Morgantown, WV, USA
| | - Ryan C Hill
- Department of Biochemistry & Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry & Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| | - Kirk C Hansen
- Department of Biochemistry & Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| | - Xiao-Bing Zhang
- State Key Laboratory of Experimental Hematology, Tianjin, China; Department of Medicine, Loma Linda University, Loma Linda, CA, USA.
| | - Zuoqin Yan
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA; WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
24
|
Li T, Chen S, Pei M. Contribution of neural crest-derived stem cells and nasal chondrocytes to articular cartilage regeneration. Cell Mol Life Sci 2020; 77:4847-4859. [PMID: 32504256 PMCID: PMC9150440 DOI: 10.1007/s00018-020-03567-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/06/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022]
Abstract
Due to poor self-regenerative potential of articular cartilage, stem cell-based regeneration becomes a hopeful approach for the treatment of articular cartilage defects. Recent studies indicate that neural crest-derived cells (NCDCs) have the potential for repairing articular cartilage with even greater chondrogenic capacity than mesoderm-derived cells (MDCs): a conventional stem cell source for cartilage regeneration. Given that NCDCs originate from a different germ layer in the early embryo compared with MDCs that give rise to articular cartilage, a mystery remains regarding their capacity for articular cartilage regeneration. In this review, we summarize the similarities and differences between MDCs and NCDCs including articular and nasal chondrocytes in cell origin, anatomy, and chondrogenic differentiation and propose that NCDCs might be promising cell origins for articular cartilage regeneration.
Collapse
Affiliation(s)
- Tianyou Li
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
- Department of Pediatric Orthopaedics, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Song Chen
- Department of Orthopaedics, The General Hospital of Western Theater Command, Chengdu, 610083, Sichuan, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA.
- WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
25
|
Lu Z, Zhou S, Vaida J, Gao G, Stewart A, Parenti J, Yan L, Pei M. Unfavorable Contribution of a Tissue-Engineering Cartilage Graft to Osteochondral Defect Repair in Young Rabbits. Front Cell Dev Biol 2020; 8:595518. [PMID: 33195273 PMCID: PMC7658375 DOI: 10.3389/fcell.2020.595518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 10/12/2020] [Indexed: 11/17/2022] Open
Abstract
A stem cell-based tissue-engineering approach is a promising strategy for treatment of cartilage defects. However, there are conflicting data in the feasibility of using this approach in young recipients. A young rabbit model with an average age of 7.7 months old was used to evaluate the effect of a tissue-engineering approach on the treatment of osteochondral defects. Following in vitro evaluation of proliferation and chondrogenic capacity of infrapatellar fat pad-derived stem cells (IPFSCs) after expansion on either tissue culture plastic (TCP) or decellularized extracellular matrix (dECM), a premature tissue construct engineered from pretreated IPFSCs was used to repair osteochondral defects in young rabbits. We found that dECM expanded IPFSCs exhibited higher proliferation and chondrogenic differentiation compared to TCP expanded cells in both pellet and tissue construct culture systems. Six weeks after creation of bilateral osteochondral defects in the femoral trochlear groove of rabbits, the Empty group (left untreated) had the best cartilage resurfacing with the highest score in Modified O’Driscoll Scale (MODS) than the other groups; however, this score had no significant difference compared to that of 15-week samples, indicating that young rabbits stop growing cartilage once they reach 9 months old. Interestingly, implantation of premature tissue constructs from both dECM and TCP groups exhibited significantly improved cartilage repair at 15 weeks compared to those at six weeks (about 9 months old), indicating that a tissue-engineering approach is able to repair adult cartilage defects. We also found that implanted pre-labeled cells in premature tissue constructs were undetectable in resurfaced cartilage at both time points. This study suggests that young rabbits (less than 9 months old) might respond differently to the classical tissue-engineering approach that is considered as a potential treatment for cartilage defects in adult rabbits.
Collapse
Affiliation(s)
- Zhihua Lu
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States.,Department of Orthopedics, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, China
| | - Sheng Zhou
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
| | - Justin Vaida
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
| | - Gongming Gao
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
| | - Amanda Stewart
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
| | - Joshua Parenti
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
| | - Lianqi Yan
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States.,WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
26
|
Arias C, Saavedra N, Leal K, Vásquez B, Abdalla DSP, Salazar LA. Histological Evaluation and Gene Expression Profiling of Autophagy-Related Genes for Cartilage of Young and Senescent Rats. Int J Mol Sci 2020; 21:ijms21228607. [PMID: 33203108 PMCID: PMC7697851 DOI: 10.3390/ijms21228607] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/13/2020] [Accepted: 11/13/2020] [Indexed: 01/06/2023] Open
Abstract
Autophagy is a cellular mechanism that protects cells from stress by digesting non-functional cellular components. In the cartilage, chondrocytes depend on autophagy as a principal mechanism to maintain cellular homeostasis. This protective role diminishes prior to the structural damage that normally occurs during aging. Considering that aging is the main risk factor for osteoarthritis, evaluating the expression of genes associated with autophagy in senescent cartilage might allow for the identification of potential therapeutic targets for treatment. Thus, we studied two groups of young and senescent rats. A histological analysis of cartilage and gene expression quantification for autophagy-related genes were performed. In aged cartilage, morphological changes were observed, such as an increase in cartilage degeneration as measured by the modified Mankin score, a decrease in the number of chondrocytes and collagen II (Col2a1), and an increase in matrix metalloproteinase 13 (Mmp13). Moreover, 84 genes associated with autophagy were evaluated by a PCR array analysis, and 15 of them were found to be significantly decreased with aging. Furthermore, an in silico analysis based on by two different bioinformatics software tools revealed that several processes including cellular homeostasis, autophagosome assembly, and aging—as well as several biological pathways such as autophagy, insulin-like growth factor 1 (IGF-1) signaling, PI3K (phosphoinositide 3-kinase)/AKT (serine/threonine kinase) signaling, and mammalian target of rapamycin (mTOR) signaling—were enriched. In conclusion, the analysis identified some potential targets for osteoarthritis treatment that would allow for the development of new therapeutic strategies for this chronic disease.
Collapse
Affiliation(s)
- Consuelo Arias
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Av. Francisco Salazar 01145, Temuco 4811230, Chile; (C.A.); (N.S.); (K.L.)
- Carrera de Kinesiología, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Av. Alemania 1090, Temuco 4810101, Chile
| | - Nicolás Saavedra
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Av. Francisco Salazar 01145, Temuco 4811230, Chile; (C.A.); (N.S.); (K.L.)
| | - Karla Leal
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Av. Francisco Salazar 01145, Temuco 4811230, Chile; (C.A.); (N.S.); (K.L.)
| | - Bélgica Vásquez
- Facultad de Ciencias de la Salud, Universidad de Tarapacá, Av. General Velásquez 1775, Arica 1000007, Chile;
| | - Dulcineia S. P. Abdalla
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, Universidade de São Paulo, Avenida Professor Lineu Prestes 580, São Paulo CEP 05508-000, SP, Brazil;
| | - Luis A. Salazar
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Av. Francisco Salazar 01145, Temuco 4811230, Chile; (C.A.); (N.S.); (K.L.)
- Correspondence: ; Tel.: +56-45-259-6724
| |
Collapse
|
27
|
Wang T, Hill RC, Dzieciatkowska M, Zhu L, Infante AM, Hu G, Hansen KC, Pei M. Site-Dependent Lineage Preference of Adipose Stem Cells. Front Cell Dev Biol 2020; 8:237. [PMID: 32351957 PMCID: PMC7174673 DOI: 10.3389/fcell.2020.00237] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/20/2020] [Indexed: 12/15/2022] Open
Abstract
Adult stem cells have unique properties in both proliferation and differentiation preference. In this study, we hypothesized that adipose stem cells have a depot-dependent lineage preference. Four rabbits were used to provide donor-matched adipose stem cells from either subcutaneous adipose tissue (ScAT) or infrapatellar fat pad (IPFP). Proliferation and multi-lineage differentiation were evaluated in adipose stem cells from donor-matched ScAT and IPFP. RNA sequencing (RNA-seq) and proteomics were conducted to uncover potential molecular discrepancy in adipose stem cells and their corresponding matrix microenvironments. We found that stem cells from ScAT exhibited significantly higher proliferation and adipogenic capacity compared to those from donor-matched IPFP while stem cells from IPFP displayed significantly higher chondrogenic potential compared to those from donor-matched ScAT. Our findings are strongly endorsed by supportive data from transcriptome and proteomics analyses, indicating a site-dependent lineage preference of adipose stem cells.
Collapse
Affiliation(s)
- Tingliang Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopedics, West Virginia University, Morgantown, WV, United States
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ryan C. Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, United States
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, United States
| | - Lian Zhu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aniello M. Infante
- Bioinformatics Core Facility, West Virginia University, Morgantown, WV, United States
| | - Gangqing Hu
- Bioinformatics Core Facility, West Virginia University, Morgantown, WV, United States
- Department of Microbiology, Immunology and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV, United States
| | - Kirk C. Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, United States
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopedics, West Virginia University, Morgantown, WV, United States
- WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
28
|
Mohamed NS, Wilkie WA, Remily EA, Delanois RE. Can human placental extract help patients with osteoarthritis? ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:62. [PMID: 32176220 PMCID: PMC7048971 DOI: 10.21037/atm.2019.12.135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 12/16/2019] [Indexed: 11/06/2022]
Affiliation(s)
- Nequesha S Mohamed
- Rubin Institute for Advanced Orthopedics, Center for Joint Preservation and Replacement, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Wayne A Wilkie
- Rubin Institute for Advanced Orthopedics, Center for Joint Preservation and Replacement, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Ethan A Remily
- Rubin Institute for Advanced Orthopedics, Center for Joint Preservation and Replacement, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Ronald E Delanois
- Rubin Institute for Advanced Orthopedics, Center for Joint Preservation and Replacement, Sinai Hospital of Baltimore, Baltimore, MD, USA
| |
Collapse
|
29
|
Khademi-Shirvan M, Ghorbaninejad M, Hosseini S, Baghaban Eslaminejad M. The Importance of Stem Cell Senescence in Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1288:87-102. [PMID: 32026416 DOI: 10.1007/5584_2020_489] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) are an interesting tool in regenerative medicine and a unique cell-based therapy to treat aging-associated diseases. Successful MSC therapy needs a large-scale cell culture, and requires a prolonged in vitro cell culture that subsequently leads to cell senescence. Administration of senescent MSCs results in inefficient cell differentiation in the clinical setting. Therefore, it is of utmost importance to enhance our knowledge about the aging process and methods to detect cell senescence in order to overcome this challenge. Numerous studies have addressed senescence in various aspects. Here, we review the characteristics of MSCs, how aging affects their features, mechanisms involved in aging of MSCs, and potential approaches to detect MSC senescence in vitro.
Collapse
Affiliation(s)
- Maliheh Khademi-Shirvan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mahsa Ghorbaninejad
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
30
|
Melatonin Reverses the Loss of Stemness Induced by TNF- α in Human Bone Marrow Mesenchymal Stem Cells through Upregulation of YAP Expression. Stem Cells Int 2019; 2019:6568394. [PMID: 32082385 PMCID: PMC7012241 DOI: 10.1155/2019/6568394] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are promising candidates for tissue regeneration and disease treatment. However, long-term in vitro culture results in loss of MSC stemness. The inflammation that occurs at stem cell transplant sites (such as that resulting from TNF-α) is a contributing factor for stem cell treatment failure. Currently, there is little evidence regarding the protective role of melatonin with regard to the negative effects of TNF-α on the stemness of MSCs. In this study, we report a melatonin-based method to reduce the inflammatory effects on the stemness of bone marrow mesenchymal stem cells (BMMSCs). The results of colony formation assays, Alizarin red staining, western blotting, and reverse transcription-polymerase chain reactions suggest that melatonin can reverse the inflammatory damage caused by TNF-α treatment in the third, seventh, and tenth generations of primary BMMSCs (vs. control and the TNF-α-treated group). Meanwhile, a detailed analysis of the molecular mechanisms showed that the melatonin receptor and YAP signaling pathway are closely related to the role that melatonin plays in negative inflammatory effects against BMMSCs. In addition, in vivo experiments showed that melatonin could reverse the damage caused by TNF-α on bone regeneration by BMMSCs in nude mice. Overall, our results suggest that melatonin can reverse the loss of stemness caused by inflammatory factor TNF-α in BMMSCs. Our results also provide a practical strategy for the application of BMMSCs in tissue engineering and cell therapy.
Collapse
|
31
|
Li J, Narayanan K, Zhang Y, Hill RC, He F, Hansen KC, Pei M. Role of lineage-specific matrix in stem cell chondrogenesis. Biomaterials 2019; 231:119681. [PMID: 31864016 DOI: 10.1016/j.biomaterials.2019.119681] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/06/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022]
Abstract
Cartilage repair in clinics is a challenge owing to the limited regenerative capacities of cartilage. Synovium-derived stem cells (SDSCs) are suggested as tissue-specific stem cells for chondrogenesis. In this study, we hypothesize that decellularized extracellular matrix (dECM) deposited by SDSCs could provide a superior tissue-specific matrix microenvironment for optimal rejuvenation of adult SDSCs for cartilage regeneration. dECMs were deposited by adult stem cells with varying chondrogenic capacities; SDSCs (strong) (SECM), adipose-derived stem cells (weak) (AECM) and dermal fibroblasts (weak) (DECM), and urine-derived stem cells (none) (UECM). Plastic flasks (Plastic) were used as a control substrate. Human SDSCs were expanded on the above substrates for one passage and examined for chondrogenic capacities. We found that each dECM consisted of unique matrix proteins and exhibited varied stiffnesses, which affected cell morphology and elasticity. Human SDSCs grown on dECMs displayed a significant increase in cell proliferation and unique surface phenotypes. Under induction media, dECM expanded cells yielded pellets with a dramatically increased number of chondrogenic markers. Interestingly, SECM expanded cells had less potential for hypertrophy compared to those grown on other dECMs, indicating that a tissue-specific matrix might provide a superior microenvironment for stem cell chondrogenic differentiation.
Collapse
Affiliation(s)
- Jingting Li
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA; Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Karthikeyan Narayanan
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA; Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Ying Zhang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA; Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, Shandong, 272067, China
| | - Ryan C Hill
- Department of Biochemistry & Molecular Genetics, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Fan He
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Kirk C Hansen
- Department of Biochemistry & Molecular Genetics, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA; WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
32
|
Wang Y, Fu Y, Yan Z, Zhang XB, Pei M. Impact of Fibronectin Knockout on Proliferation and Differentiation of Human Infrapatellar Fat Pad-Derived Stem Cells. Front Bioeng Biotechnol 2019; 7:321. [PMID: 31803729 PMCID: PMC6873900 DOI: 10.3389/fbioe.2019.00321] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/28/2019] [Indexed: 12/15/2022] Open
Abstract
Fibronectin plays an essential role in tissue development and regeneration. However, the effects of fibronectin knockout (FN1-KO) on stem cells' proliferation and differentiation remain unknown. In this study, CRISPR/Cas9 generated FN1-KO in human infrapatellar fat pad-derived stem cells (IPFSCs) was evaluated for proliferation ability including cell cycle and surface markers as well as stemness gene expression and for differentiation capacity including chondrogenic and adipogenic differentiation. High passage IPFSCs were also evaluated for proliferation and differentiation capacity after expansion on decellularized ECM (dECM) deposited by FN1-KO cells. Successful FN1-KO in IPFSCs was confirmed by Sanger sequencing and Inference of CRISPR Edits analysis (ICE) as well as immunostaining for fibronectin expression. Compared to the GFP control, FN1-KO cells showed an increase in cell growth, percentage of cells in the S and G2 phases, and CD105 and CD146 expression but a decrease in expression of stemness markers CD73, CD90, SSEA4, and mesenchymal condensation marker CDH2 gene. FN1-KO decreased both chondrogenic and adipogenic differentiation capacity. Interestingly, IPFSCs grown on dECMs deposited by FN1-KO cells exhibited a decrease in cell proliferation along with a decline in CDH2 expression. After induction, IPFSCs plated on dECMs deposited by FN1-KO cells also displayed decreased expression of both chondrogenic and adipogenic capacity. We concluded that FN1-KO increased human IPFSCs' proliferation capacity; however, this capacity was reversed after expansion on dECM deposited by FN1-KO cells. Significance of fibronectin in chondrogenic and adipogenic differentiation was demonstrated in both FN1-KO IPFSCs and FN(-) matrix microenvironment.
Collapse
Affiliation(s)
- Yiming Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yawen Fu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Tianjin, China
- Department of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Zuoqin Yan
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao-Bing Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Tianjin, China
- Department of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
- WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
33
|
Frahs S, Reeck JC, Yocham KM, Frederiksen A, Fujimoto K, Scott CM, Beard RS, Brown RJ, Lujan TJ, Solov’yov IA, Estrada D, Oxford JT. Prechondrogenic ATDC5 Cell Attachment and Differentiation on Graphene Foam; Modulation by Surface Functionalization with Fibronectin. ACS APPLIED MATERIALS & INTERFACES 2019; 11:41906-41924. [PMID: 31639302 PMCID: PMC6858527 DOI: 10.1021/acsami.9b14670] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 10/22/2019] [Indexed: 05/25/2023]
Abstract
Graphene foam holds promise for tissue engineering applications. In this study, graphene foam was used as a three-dimension scaffold to evaluate cell attachment, cell morphology, and molecular markers of early differentiation. The aim of this study was to determine if cell attachment and elaboration of an extracellular matrix would be modulated by functionalization of graphene foam with fibronectin, an extracellular matrix protein that cells adhere well to, prior to the establishment of three-dimensional cell culture. The molecular dynamic simulation demonstrated that the fibronectin-graphene interaction was stabilized predominantly through interaction between the graphene and arginine side chains of the protein. Quasi-static and dynamic mechanical testing indicated that fibronectin functionalization of graphene altered the mechanical properties of graphene foam. The elastic strength of the scaffold increased due to fibronectin, but the viscoelastic mechanical behavior remained unchanged. An additive effect was observed in the mechanical stiffness when the graphene foam was both coated with fibronectin and cultured with cells for 28 days. Cytoskeletal organization assessed by fluorescence microscopy demonstrated a fibronectin-dependent reorganization of the actin cytoskeleton and an increase in actin stress fibers. Gene expression assessed by quantitative real-time polymerase chain reaction of 9 genes encoding cell attachment proteins (Cd44, Ctnna1, Ctnnb1, Itga3, Itga5, Itgav, Itgb1, Ncam1, Sgce), 16 genes encoding extracellular matrix proteins (Col1a1, Col2a1, Col3a1, Col5a1, Col6a1, Ecm1, Emilin1, Fn1, Hapln1, Lamb3, Postn, Sparc, Spp1, Thbs1, Thbs2, Tnc), and 9 genes encoding modulators of remodeling (Adamts1, Adamts2, Ctgf, Mmp14, Mmp2, Tgfbi, Timp1, Timp2, Timp3) indicated that graphene foam provided a microenvironment conducive to expression of genes that are important in early chondrogenesis. Functionalization of graphene foam with fibronectin modified the cellular response to graphene foam, demonstrated by decreases in relative gene expression levels. These findings illustrate the combinatorial factors of microscale materials properties and nanoscale molecular features to consider in the design of three-dimensional graphene scaffolds for tissue engineering applications.
Collapse
Affiliation(s)
- Stephanie
M. Frahs
- Center
of Biomedical Research Excellence in Matrix Biology, Biomolecular
Research Center, Boise State University, Boise, Idaho 83725, United States
| | - Jonathon C. Reeck
- Center
of Biomedical Research Excellence in Matrix Biology, Biomolecular
Research Center, Boise State University, Boise, Idaho 83725, United States
| | - Katie M. Yocham
- Department
of Mechanical and Biomedical Engineering, Boise State University, Boise, Idaho 83725, United States
- Micron
School of Materials Science and Engineering, Boise State University, Boise, Idaho 83725, United States
| | - Anders Frederiksen
- University
of Southern Denmark, Department of Physics,
Chemistry and Pharmacy, Campusvej 55, 5230 Odense M, Denmark
| | - Kiyo Fujimoto
- Micron
School of Materials Science and Engineering, Boise State University, Boise, Idaho 83725, United States
| | - Crystal M. Scott
- Center
of Biomedical Research Excellence in Matrix Biology, Biomolecular
Research Center, Boise State University, Boise, Idaho 83725, United States
| | - Richard S. Beard
- Center
of Biomedical Research Excellence in Matrix Biology, Biomolecular
Research Center, Boise State University, Boise, Idaho 83725, United States
| | - Raquel J. Brown
- Center
of Biomedical Research Excellence in Matrix Biology, Biomolecular
Research Center, Boise State University, Boise, Idaho 83725, United States
| | - Trevor J. Lujan
- Department
of Mechanical and Biomedical Engineering, Boise State University, Boise, Idaho 83725, United States
| | - Ilia A. Solov’yov
- Department
of Physics, Carl von Ossietzky Universität
Oldenburg, Carl-von-Ossietzky-Straße
9-11, 26129 Oldenburg, Germany
| | - David Estrada
- Micron
School of Materials Science and Engineering, Boise State University, Boise, Idaho 83725, United States
| | - Julia Thom Oxford
- Center
of Biomedical Research Excellence in Matrix Biology, Biomolecular
Research Center, Boise State University, Boise, Idaho 83725, United States
- Department
of Biological Sciences, Boise State University, Boise, Idaho 83725, United States
| |
Collapse
|
34
|
De Angelis E, Cacchioli A, Ravanetti F, Bileti R, Cavalli V, Martelli P, Borghetti P. Gene expression markers in horse articular chondrocytes: Chondrogenic differentiaton IN VITRO depends on the proliferative potential and ageing. Implication for tissue engineering of cartilage. Res Vet Sci 2019; 128:107-117. [PMID: 31778851 DOI: 10.1016/j.rvsc.2019.10.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 09/05/2019] [Accepted: 10/31/2019] [Indexed: 02/06/2023]
Abstract
Chondrocyte dedifferentiation is a key limitation in therapies based on autologous chondrocyte implantation for cartilage repair. Articular chondrocytes, obtained from (metacarpophalangeal and metatarsophalangeal) joints of different aged horses, were cultured in monolayer for several passages (P0 to P8). Cumulative Populations Doublings Levels (PDL) and gene expression of relevant chondrocyte phenotypic markers were analysed during culturing. Overall data confirmed that, during proliferation in vitro, horse chondrocytes undergo marked morphological and phenotypic alterations of their differentiation status. Particularly, the dedifferentiation started early in culture (P0-P1) and was very marked at P3 subculture (PDL 4-6): proliferative phase after P3 could be critical for maintenance/loss of differentiation potential. In elderly animals, chondrocytes showed aspects of dedifferentiation shortly after their isolation, associated with reduced proliferative capacity. Regarding the gene expression of major cartilage markers (Col2, Aggrecan, SOX9) there was a very early reduction (P1) in proliferating chondrocytes independent of age. The chondrocytes from adult donors showed a more stable expression (up to P3) of some (Col6, Fibromodulin, SOX6, TGβ1) markers of mature cartilage; these markers could be tested as parameter to determine the dedifferentiation level. This study can provide parameters to identify up to which "culture step" chondrocytes for implantation with a conserved phenotypic potential can be obtained, and to test the efficiency of biomaterial scaffold or chondroinductive media/signals to maintain/recover the chondrocyte phenotype. Moreover, the determination of levels and time related expression of these markers can be useful during the chondroinduction of mesenchymal stem cells.
Collapse
Affiliation(s)
| | | | | | - Rossana Bileti
- Department of Veterinary Sciences, University of Parma, Italy
| | - Valeria Cavalli
- Department of Veterinary Sciences, University of Parma, Italy
| | - Paolo Martelli
- Department of Veterinary Sciences, University of Parma, Italy
| | - Paolo Borghetti
- Department of Veterinary Sciences, University of Parma, Italy
| |
Collapse
|
35
|
Zhang W, Yang J, Zhu Y, Sun X, Guo W, Liu X, Jing X, Guo G, Guo Q, Peng J, Zhu X. Extracellular matrix derived by human umbilical cord-deposited mesenchymal stem cells accelerates chondrocyte proliferation and differentiation potential in vitro. Cell Tissue Bank 2019; 20:351-365. [PMID: 31218457 DOI: 10.1007/s10561-019-09774-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 05/10/2019] [Indexed: 12/16/2022]
Abstract
The extracellular matrix (ECM) is a dynamic and intricate three-dimensional (3D) microenvironment with excellent biophysical, biomechanical, and biochemical properties that may directly or indirectly regulate cell behavior, including proliferation, adhesion, migration, and differentiation. Compared with tissue-derived ECM, cell-derived ECM potentially has more advantages, including less potential for pathogen transfer, fewer inflammatory or anti-host immune responses, and a closer resemblance to the native ECM microenvironment. Different types of cell-derived ECM, such as adipose stem cells, synovium-derived stem cells and bone marrow stromal cells, their effects on articular chondrocytes which have been researched. In this study, we aimed to develop a 3D cell culture substrate using decellularized ECM derived from human umbilical cord-derived mesenchymal stem cells (hUCMSCs), and evaluated the effects on articular chondrocytes. We evaluated the morphology and components of hUCMSC-derived ECM using physical and chemical methods. Morphological, histological, immunohistochemical, biochemical, and real-time PCR analyses demonstrated that proliferation and differentiation capacity of chondrocytes using the 3D hUCMSC-derived ECM culture substrate was superior to that using non-coated two-dimensional plastic culture plates. In conclusion, 3D decellularized ECM derived from hUCMSCs offers a tissue-specific microenvironment for in vitro culture of chondrocytes, which not only markedly promoted chondrocyte proliferation but also preserved the differentiation capacity of chondrocytes. Therefore, our findings suggest that a 3D cell-derived ECM microenvironment represents a promising prospect for autologous chondrocyte-based cartilage tissue engineering and regeneration. The hUCMSC-derived ECM as a biomaterial is used for the preparation of scaffold or hybrid scaffold products which need to further study in the future.
Collapse
Affiliation(s)
- Weixiang Zhang
- The People's Hospital of Lanxi, 1359th Shanxi Road, Lanxi, 321100, China.,Institute of Orthopedics, Chinese PLA General Hospital, 28th Fuxing Road, Beijing, 100853, China
| | - Jianhua Yang
- School of Medicine, Jiamusi University, 148th Xuefu Road, Xiangyang District, Jiamusi, 154007, China
| | - Yun Zhu
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, 999777, Hong Kong
| | - Xun Sun
- Institute of Orthopedics, Chinese PLA General Hospital, 28th Fuxing Road, Beijing, 100853, China.,School of Medicine, Nankai University, 94th Weijin Road, Nankai District, Tianjin, 300071, China
| | - Weimin Guo
- Institute of Orthopedics, Chinese PLA General Hospital, 28th Fuxing Road, Beijing, 100853, China
| | - Xuejian Liu
- School of Medicine, Jiamusi University, 148th Xuefu Road, Xiangyang District, Jiamusi, 154007, China.,Institute of Orthopedics, Chinese PLA General Hospital, 28th Fuxing Road, Beijing, 100853, China.,Zhengzhou Yihe Hospital Affiliated to Henan University, 69th Nongyedong Road, Zhengzhou, 450000, China
| | - Xiaoguang Jing
- School of Medicine, Jiamusi University, 148th Xuefu Road, Xiangyang District, Jiamusi, 154007, China.,Institute of Orthopedics, Chinese PLA General Hospital, 28th Fuxing Road, Beijing, 100853, China
| | - Ganggang Guo
- School of Medicine, Jiamusi University, 148th Xuefu Road, Xiangyang District, Jiamusi, 154007, China.,Institute of Orthopedics, Chinese PLA General Hospital, 28th Fuxing Road, Beijing, 100853, China
| | - Quanyi Guo
- Institute of Orthopedics, Chinese PLA General Hospital, 28th Fuxing Road, Beijing, 100853, China
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital, 28th Fuxing Road, Beijing, 100853, China
| | - Xiaofeng Zhu
- School of Medicine, Jiamusi University, 148th Xuefu Road, Xiangyang District, Jiamusi, 154007, China. .,Medical Research Center of Mudanjiang Medical School, 3th Tongxiang Road, Aimin District, Mudanjiang, 157011, China. .,Institute of Neurosciences, Jiamusi University, 148th Xuefu Road, Xiangyang District, Jiamusi, 154007, China.
| |
Collapse
|
36
|
Freitag J, Wickham J, Shah K, Tenen A. Effect of autologous adipose-derived mesenchymal stem cell therapy in the treatment of acromioclavicular joint osteoarthritis. BMJ Case Rep 2019; 12:12/2/e227865. [PMID: 30819682 PMCID: PMC6398814 DOI: 10.1136/bcr-2018-227865] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The aim of this case report is to evaluate the efficacy of mesenchymal stem cell (MSC) therapy in the treatment of small joint osteoarthritis (OA). Acromio-clavicular (AC) joint OA is an under-diagnosed and yet frequent source of shoulder pain. MSCs have shown evidence of benefit in the treatment of knee OA. This is the first report to describe the use of MSC therapy in OA of the upper limb. A 43-year-old patient presents with painful AC joint OA and undergoes MSC therapy. The patient reported pain and functional improvement as assessed by the Disability of Arm, Shoulder and Hand Score and Numeric Pain Rating Scale. Imaging at 12 months showed structural improvement with reduction in subchondral oedema, synovitis and subchondral cysts. This case is the first to show the benefit of MSC therapy in the treatment of small joint arthropathy and also of the upper limb. Trial registration number: Australian New Zealand Clinical Trials Registry (ACTRN12617000638336).
Collapse
Affiliation(s)
- Julien Freitag
- School of Biomedical Science, Charles Sturt University, Orange, New South Wales, Australia.,Melbourne Stem Cell Centre, Box Hill North, Victoria, Australia.,Magellan Stem Cells, Box Hill North, Victoria, Australia
| | - James Wickham
- School of Biomedical Science, Charles Sturt University, Orange, New South Wales, Australia
| | - Kiran Shah
- Magellan Stem Cells, Box Hill North, Victoria, Australia
| | - Abi Tenen
- Melbourne Stem Cell Centre, Box Hill North, Victoria, Australia.,Magellan Stem Cells, Box Hill North, Victoria, Australia.,Monash University School of Primary Health Care, Monash University, Notting Hill, Victoria, Australia.,Vision Eye Institute, Melbourne, Victoria, Australia
| |
Collapse
|
37
|
Park YB, Ha CW, Kim JA, Kim S, Park YG. Comparison of Undifferentiated Versus Chondrogenic Predifferentiated Mesenchymal Stem Cells Derived From Human Umbilical Cord Blood for Cartilage Repair in a Rat Model. Am J Sports Med 2019; 47:451-461. [PMID: 30640523 DOI: 10.1177/0363546518815151] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) have gained much interest as a promising cell source for regenerative medicine owing to the noninvasive collection, availability, high expansion capacity, and low immunogenicity. However, few in vivo studies have reported the use of hUCB-MSCs on cartilage repair. Moreover, little study has been conducted on the effects of chondrogenic predifferentiation of hUCB-MSCs on cartilage repair. PURPOSE To compare the effectiveness of transplanting undifferentiated versus chondrogenic predifferentiated mesenchymal stem cells (MSCs) for treating osteochondral defects. STUDY DESIGN Controlled laboratory study. METHODS Critical-sized osteochondral defects were created in the trochlear grooves of rat femurs. In 20 rats, a composite of chondrogenic predifferentiated hUCB-MSCs (chondro-MSCs) and 4% hyaluronic acid (HA) hydrogel was transplanted into defects in the right knees, whereas undifferentiated hUCB-MSCs (undiff-MSCs) and 4% HA hydrogel were transplanted into the left knees. In the control groups, 4% HA hydrogel without MSCs was transplanted into defects in the right knees, and the defects in the left knees were left untreated in 20 rats. The cartilage repair was evaluated at 8 and 16 weeks after surgery. RESULTS Transplanting undiff-MSCs resulted in overall superior cartilage repair as compared with chondro-MSCs, HA alone, or no treatment. The articular surfaces of the defect sites in the undiff-MSC group were relatively smoother than those of the other treatments. The undiff-MSC group showed cellular morphology and arrangement similar to surrounding normal articular cartilage tissue at 16 weeks, both of which were also better than those of the other groups. In addition, the undiff-MSC group showed coloration similar to surrounding normal articular cartilage tissue at 16 weeks in safranin O and type II collagen immunohistochemical staining. The histological scores also revealed that cartilage repair with undiff-MSCs was better than that with chondro-MSCs, HA alone, or no treatment ( P < .05 in all). CONCLUSION This study demonstrated that treatment with undiff-MSCs resulted in more favorable cartilage repair than that with chondro-MSCs in a rat model. These findings indicate that chondrogenic predifferentiation of MSCs before transplantation does not enhance cartilage repair. CLINICAL RELEVANCE The results of this study support the use of undifferentiated MSCs, rather than chondrogenic predifferentiated MSCs, as a stem cell therapy strategy for cartilage repair.
Collapse
Affiliation(s)
- Yong-Beom Park
- Department of Orthopedic Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Chul-Won Ha
- Department of Orthopedic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Jin-A Kim
- Department of Orthopedic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Seongchan Kim
- Department of Orthopedic Surgery, Jeju National University Hospital, Jeju National University School of Medicine, Jeju, Republic of Korea
| | - Yong-Geun Park
- Department of Orthopedic Surgery, Jeju National University Hospital, Jeju National University School of Medicine, Jeju, Republic of Korea
| |
Collapse
|
38
|
Sun Y, Chen S, Pei M. Comparative advantages of infrapatellar fat pad: an emerging stem cell source for regenerative medicine. Rheumatology (Oxford) 2018; 57:2072-2086. [PMID: 29373763 PMCID: PMC6256334 DOI: 10.1093/rheumatology/kex487] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 11/13/2017] [Indexed: 12/15/2022] Open
Abstract
Growing evidence indicates that infrapatellar fat pad (IPFP)-derived stem cells (IPFSCs) exert robust proliferation capacities and multilineage differentiation potentials. However, few papers summarize the advantages that the IPFP and IPFSCs have in regenerative medicine. In this review we delineate the development and anatomy of the IPFP by comparing it with an adjacent fibrous tissue, synovium, and a more frequently harvested fat depot, subcutaneous adipose tissue. Furthermore, we explore the similarities and differences of stem cells from these three tissues in terms of IPFSCs, synovium-derived stem cells and subcutaneous adipose tissue-derived stem cells in proliferation capacity and tri-lineage differentiation potentials, including chondrogenesis, osteogenesis and adipogenesis. Finally, we highlight the advantages of IPFSCs in regenerative medicine, such as the abundant accessibility and the ability to resist inflammation and senescence, two hurdles for cell-based tissue regeneration. Considering the comparative advantages of IPFSCs, the IPFP can serve as an excellent stem cell source for regenerative medicine, particularly for cartilage regeneration.
Collapse
Affiliation(s)
- Yu Sun
- Department of Orthopaedics, Orthopaedics Institute, Subei People’s Hospital of Jiangsu Province, Yangzhou, Jiangsu, China
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
| | - Song Chen
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Exercise Physiology, West Virginia University, Morgantown, WV, USA
- Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
39
|
Liu G, Wang B, Li S, Jin Q, Dai Y. Human breast cancer decellularized scaffolds promote epithelial-to-mesenchymal transitions and stemness of breast cancer cells in vitro. J Cell Physiol 2018; 234:9447-9456. [PMID: 30478896 DOI: 10.1002/jcp.27630] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 09/27/2018] [Indexed: 12/11/2022]
Abstract
Breast cancer, with unsatisfactory survival rates, is the leading cause of cancer-related death in women worldwide. Recent advances in the genetic basis of breast cancer have benefitted the development of gene-based medicines and therapies. Tissue engineering technologies, including tissue decellularizations and reconstructions, are potential therapeutic alternatives for cancer research and tissue regeneration. In our study, human breast cancer biopsies were decellularized by a detergent technique, with sodium lauryl ether sulfate (SLES) solution, for the first time. And the decellularization process was optimized to maximally maintain tissue microarchitectures and extracellular matrix (ECM) components with minimal DNA compounds preserved. Histology analysis and DNA quantification results confirmed the decellularization effect with maximal genetic compounds removal. Quantification, immunofluorescence, and histology analyses demonstrated better preservation of ECM components in 0.5% SLES-treated scaffolds. Scaffolds seeded with MCF-7 cells demonstrated the process of cell recellularization in vitro, with increased cell migration, proliferation, and epithelial-to-mesenchymal transition (EMT) process. When treated with 5-fluorouracil, the expressions of stem cell markers, including Oct4, Sox2, and CD49F, were maximally maintained in the recellularized scaffold with decreased apoptosis rates compared with monolayer cells. These results showed that the decellularized breast scaffold model with SLES treatments would help to simulate the pathogenesis of breast cancer in vitro. And we hope that this model could further accelerate the development of effective therapies for breast cancer and benefit drug screenings.
Collapse
Affiliation(s)
- Gang Liu
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, College of life science, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Biao Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, College of life science, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Shubin Li
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, College of life science, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Qin Jin
- Department of Pathlogy, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, China
| | - Yanfeng Dai
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, College of life science, Inner Mongolia University, Hohhot, Inner Mongolia, China
| |
Collapse
|
40
|
Li J, Huang Y, Song J, Li X, Zhang X, Zhou Z, Chen D, Ma PX, Peng W, Wang W, Zhou G. Cartilage regeneration using arthroscopic flushing fluid-derived mesenchymal stem cells encapsulated in a one-step rapid cross-linked hydrogel. Acta Biomater 2018; 79:202-215. [PMID: 30165202 PMCID: PMC6510982 DOI: 10.1016/j.actbio.2018.08.029] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 08/14/2018] [Accepted: 08/23/2018] [Indexed: 01/22/2023]
Abstract
UNLABELLED Many attempts have been made to repair articular cartilage defects, including mesenchymal stem cell (MSC)-based tissue engineering strategies. Although this approach shows promise, optimizing MSC sources and their delivery is challenging. This study was designed to test the feasibility of using MSCs found in the human arthroscopic flushing fluid (AFF) for cartilage regeneration, by incorporating them into a newly developed one-step rapid cross-linking hyper-branched polyPEGDA/HA hydrogel. AFF-MSCs were isolated from the original intra-articular flushing fluid of 10 patients prior to arthroscopic procedures. The hydrogel was fabricated with hyper-branched polyPEGDA and thiolated hyaluronic acid (HA). In vitro assays demonstrated that AFF-MSCs possessed the typical MSC morphology and phenotype, and maintained chondrogenic differentiation properties when encapsulated within the hydrogel. The AFF-MSC/hydrogel composite could significantly repair full-thickness cartilage defects generated in a rat model after 8 weeks of implantation; smooth cartilage was formed with evidence of hyaline cartilage formation. These data suggest that human AFF-MSCs are a novel and abundant MSC source that have high therapeutic value for cartilage regeneration. STATEMENT OF SIGNIFICANCE Many attempts have been made to repair the defects of articular cartilage, including mesenchymal stem cell (MSC)-based tissue engineering strategies. Optimizing MSC sources and their delivery approaches still remain clinically challenging. Recent studies determined that MSCs derived from synovium and synovial fluid exhibited superior chondrogenic potential. However, no feasible methods to harvest these human tissues and cells have been impeding them for clinical application. Hereby, we explored a simple and easy accessible approach to obtain a new stem cell source from arthroscopic flushing fluid (AFF-MSCs), which probably contains plenty of MSCs from synovium and synovial fluid. Further experiments demonstrated that encapsulation of these stem cells with one-step rapid cross-linked polyPEGDA/HA hydrogel held very encouraging potential for cartilage regeneration.
Collapse
Affiliation(s)
- Jun Li
- Shenzhen Key Laboratory of Anti-Ageing and Regenerative Medicine, Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Medical School, Shenzhen, Guangdong 518060, China; Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Guangdong, 518060, China
| | - Yongcan Huang
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Departments of Orthopaedics, Peking University Shenzhen Hospital, Shenzhen, Guangdong 510086, China
| | - Jun Song
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Guangdong, 518060, China
| | - Xiaolin Li
- Shenzhen Key Laboratory of Anti-Ageing and Regenerative Medicine, Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Medical School, Shenzhen, Guangdong 518060, China; The Charles Institute of Dermatology, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Xintao Zhang
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Departments of Orthopaedics, Peking University Shenzhen Hospital, Shenzhen, Guangdong 510086, China
| | - Zhiyu Zhou
- Shenzhen Key Laboratory of Anti-Ageing and Regenerative Medicine, Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Medical School, Shenzhen, Guangdong 518060, China
| | - Di Chen
- Shenzhen Key Laboratory of Anti-Ageing and Regenerative Medicine, Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Medical School, Shenzhen, Guangdong 518060, China; Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Peter X Ma
- Schools of Dentistry, Engineering and Medicine, University of Michigan, Ann Arbor, MI 48109-1078, USA
| | - Wenjing Peng
- Shenzhen Anhe Health Sciences Co. Ltd., Longhua District, Shenzhen, China
| | - Wenxin Wang
- The Charles Institute of Dermatology, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland.
| | - Guangqian Zhou
- Shenzhen Key Laboratory of Anti-Ageing and Regenerative Medicine, Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Medical School, Shenzhen, Guangdong 518060, China.
| |
Collapse
|
41
|
Chen F, Li G, Zhao ER, Li J, Hableel G, Lemaster JE, Bai Y, Sen GL, Jokerst JV. Cellular toxicity of silicon carbide nanomaterials as a function of morphology. Biomaterials 2018; 179:60-70. [PMID: 29980075 PMCID: PMC6069971 DOI: 10.1016/j.biomaterials.2018.06.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/05/2018] [Accepted: 06/19/2018] [Indexed: 02/06/2023]
Abstract
Silicon carbide has been shown to be biocompatible and is used as a coating material for implanted medical devices to prevent biofilms. Silicon carbide nanomaterials are also promising in cell tracking due to their stable and strong luminescence, but more comprehensive studies of this material on the nanoscale are needed. Here, we studied the toxicity of silicon carbide nanomaterials on human mesenchymal stem cells in terms of metabolism, viability, adhesion, proliferation, migration, oxidative stress, and differentiation ability. We compared two different shapes and found that silicon carbide nanowires are toxic to human mesenchymal stem cells but not to cancer cell lines at the concentration of 0.1 mg/mL. Control silicon carbide nanoparticles were biocompatible to human mesenchymal stem cells at 0.1 mg/mL. We studied the potential mechanistic effect of silicon carbide nanowires on human mesenchymal stem cells' phenotype, cytokine secretion, and gene expression. These findings suggest that the toxic effect of silicon carbide nanomaterials to human mesenchymal stem cells are dependent on morphology.
Collapse
Affiliation(s)
- Fang Chen
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Gongyi Li
- College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, Hunan, 410073 PR China
| | - Eric Ruike Zhao
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Jingting Li
- Departments of Dermatology and Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Ghanim Hableel
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Jeanne E Lemaster
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Yuting Bai
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - George L Sen
- Departments of Dermatology and Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Jesse V Jokerst
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Radiology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
42
|
Chen X, Li M, Yan J, Liu T, Pan G, Yang H, Pei M, He F. Alcohol Induces Cellular Senescence and Impairs Osteogenic Potential in Bone Marrow-Derived Mesenchymal Stem Cells. Alcohol Alcohol 2018; 52:289-297. [PMID: 28339869 PMCID: PMC5397879 DOI: 10.1093/alcalc/agx006] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/09/2017] [Indexed: 12/19/2022] Open
Abstract
Aims Chronic and excessive alcohol consumption is a high-risk factor for osteoporosis. Bone marrow-derived mesenchymal stem cells (BM-MSCs) play an important role in bone formation; however, they are vulnerable to ethanol (EtOH). The purpose of this research was to investigate whether EtOH could induce premature senescence in BM-MSCs and subsequently impair their osteogenic potential. Methods Human BM-MSCs were exposed to EtOH ranging from 10 to 250 mM. Senescence-associated β-galactosidase (SA-β-gal) activity, cell cycle distribution, cell proliferation and reactive oxygen species (ROS) were evaluated. Mineralization and osteoblast-specific gene expression were evaluated during osteogenesis in EtOH-treated BM-MSCs. To investigate the role of silent information regulator Type 1 (SIRT1) in EtOH-induced senescence, resveratrol (ResV) was used to activate SIRT1 in EtOH-treated BM-MSCs. Results EtOH treatments resulted in senescence-associated phenotypes in BM-MSCs, such as decreased cell proliferation, increased SA-β-gal activity and G0/G1 cell cycle arrest. EtOH also increased the intracellular ROS and the expression of senescence-related genes, such as p16INK4α and p21. The down-regulated levels of SIRT1 accompanied with suppressed osteogenic differentiation were confirmed in EtOH-treated BM-MSCs. Activation of SIRT1 by ResV partially counteracted the effects of EtOH by decreasing senescence markers and rescuing the inhibited osteogenesis. Conclusion EtOH treatments induced premature senescence in BM-MSCs in a dose-dependent manner that was responsible for EtOH-impaired osteogenic differentiation. Activation of SIRT1 was effective in ameliorating EtOH-induced senescence phenotypes in BMSCs and could potentially lead to a new strategy for clinically preventing or treating alcohol-induced osteoporosis. Short summary Ethanol (EtOH) treatments induce premature senescence in marrow-derived mesenchymal stem cells in a dose-dependent manner that is responsible for EtOH-impaired osteogenic differentiation. Activation of SIRT1 is effective in ameliorating EtOH-induced senescence phenotypes, which potentially leads to a new strategy for clinically treating alcohol-induced osteoporosis.
Collapse
Affiliation(s)
- Xi Chen
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou 215153, Jiangsu, China.,Orthopaedic Institute, Medical College, Soochow University, No. 708 Renmin Road, Suzhou 215007, China.,School of Biology and Basic Medical Sciences, Medical College, Soochow University, No. 199 Renai Road, Suzhou 215123, China
| | - Mao Li
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou 215153, Jiangsu, China.,Orthopaedic Institute, Medical College, Soochow University, No. 708 Renmin Road, Suzhou 215007, China
| | - Jinku Yan
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou 215153, Jiangsu, China.,Orthopaedic Institute, Medical College, Soochow University, No. 708 Renmin Road, Suzhou 215007, China
| | - Tao Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou 215153, Jiangsu, China
| | - Guoqing Pan
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou 215153, Jiangsu, China.,Orthopaedic Institute, Medical College, Soochow University, No. 708 Renmin Road, Suzhou 215007, China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou 215153, Jiangsu, China.,Orthopaedic Institute, Medical College, Soochow University, No. 708 Renmin Road, Suzhou 215007, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics and Division of Exercise Physiology, West Virginia University, PO Box 9196, One Medical Center Drive, Morgantown, WV 26505-9196, USA
| | - Fan He
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou 215153, Jiangsu, China.,Orthopaedic Institute, Medical College, Soochow University, No. 708 Renmin Road, Suzhou 215007, China
| |
Collapse
|
43
|
Wu Z, Qiu X, Gao B, Lian C, Peng Y, Liang A, Xu C, Gao W, Zhang L, Su P, Rong L, Huang D. Melatonin-mediated miR-526b-3p and miR-590-5p upregulation promotes chondrogenic differentiation of human mesenchymal stem cells. J Pineal Res 2018; 65:e12483. [PMID: 29498095 DOI: 10.1111/jpi.12483] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 02/14/2018] [Indexed: 12/12/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs), with inherent chondrogenic differentiation potential appear to be ideally suited for therapeutic use in cartilage regeneration. Accumulating evidence has demonstrated that melatonin can promote chondrogenic differentiation in human BMSCs. However, little is known about the mechanism. MicroRNAs (miRNAs) have been shown to regulate the differentiation of BMSCs, but their roles in melatonin-promoted chondrogenic differentiation have not been characterized. Here, we demonstrate that melatonin promoted chondrogenic differentiation of human BMSCs via upregulation of miR-526b-3p and miR-590-5p. Mechanistically, the elevated miR-526b-3p and miR-590-5p enhanced SMAD1 phosphorylation by targeting SMAD7. Additionally, administration of miR-526b-3p mimics or miR-590-5p mimics successfully promoted the chondrogenic differentiation of human BMSCs. Collectively, our study suggests that modification of BMSCs using melatonin or miRNA transduction could be an effective therapy for cartilage damage and degeneration.
Collapse
Affiliation(s)
- Zizhao Wu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xianjian Qiu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Bo Gao
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chengjie Lian
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan Peng
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Anjing Liang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Caixia Xu
- Research Centre for Translational Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenjie Gao
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Spine Surgery, Xi'an Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Liangming Zhang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Peiqiang Su
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Limin Rong
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Dongsheng Huang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
44
|
Drevet S, Gavazzi G, Grange L, Dupuy C, Lardy B. Reactive oxygen species and NADPH oxidase 4 involvement in osteoarthritis. Exp Gerontol 2018; 111:107-117. [PMID: 30012342 DOI: 10.1016/j.exger.2018.07.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/06/2018] [Accepted: 07/09/2018] [Indexed: 01/04/2023]
Abstract
Osteoarthritis (OA) is a degenerative chronic disease affecting >300,000 million people around the world as of 2016. Symptomatic measures exist, but there are hardly any curative treatments available. Disruption of the cartilage homeostasis in favor of catabolism leads to cartilage destruction. ROS-macromolecular-induced damage is significantly greater in OA cartilage and OA is described as low-grade chronic systemic inflammation. This review aimed to assess the critical role of cartilage ageing and oxidative stress in the OA process, focusing in particular on NADPH oxidase and especially Nox4 involvement. With age, hypertrophic senescent cells with an altered redox cell profile accumulated. Chondrocytes are more sensitive to oxidant-mediators and the serum level of pro-inflammatory mediators increases. Age-related advanced glycation end products impact on extra cellular matrix (ECM) properties leading to the apoptosis of chondrocytes. A focus on NADPH oxidase-mediated-ROS signaling highlighted the very specific Nox4 isoform, which plays a role on the final common pathway targeting chondrocyte cells. IL-1β-mediated Nox4 stimulation induced an increase in the levels released by the chondrocyte of MMP-1 and MMP-13 proteins, which are involved in ECM degradation. In comparison with the other Nox isoforms, Nox4 remains unusual, since it is constitutively active, does not depend on cytosolic activator proteins and seems to generate H2O2 thanks to the specific conformation of the Nox4 E-loop. Nox4-induced ROS production appears an essential actor in the OA process and it could be relevant to focus on this target in the aim of discovering and developing new therapeutic strategies.
Collapse
Affiliation(s)
- S Drevet
- Grenoble Alpes University Hospital, Orthogeriatric Unit, Geriatric Department, Grenoble Alpes University, GREPI UGA-EFS EA7408, Boulevard de la Chantourne, 38043 Grenoble Cedex 1, France.
| | - G Gavazzi
- Grenoble Alpes University Hospital, Acute Geriatric Medicine Unit, Geriatric Department, Grenoble Alpes University, GREPI UGA-EFS EA7408, Boulevard de la Chantourne, 38043 Grenoble Cedex 1, France.
| | - L Grange
- Grenoble Alpes University Hospital, Rheumatology Department, Hopital Sud, GREPI UGA-EFS EA7408, 19 avenue de Kimberley, 38130 Echirolles, France.
| | - C Dupuy
- Institut Gustave Roussy, UMR 8200 CNRS "Stabilité génétique et Oncogenèse", 114 rue Edouard Vaillant, 94805 Villejuif Cedex, France.
| | - B Lardy
- Grenoble Alpes University Hospital, Biology Department, Grenoble Alpes University, GREPI UGA-EFS EA7408, Boulevard de la Chantourne, 38043 Grenoble Cedex 1, France.
| |
Collapse
|
45
|
Yang R, Liu Y, Yu T, Liu D, Shi S, Zhou Y, Zhou Y. Hydrogen sulfide maintains dental pulp stem cell function via TRPV1-mediated calcium influx. Cell Death Discov 2018; 4:1. [PMID: 30062050 PMCID: PMC6060166 DOI: 10.1038/s41420-018-0071-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 02/17/2018] [Accepted: 02/20/2018] [Indexed: 12/21/2022] Open
Abstract
Hydrogen sulfide (H2S), an endogenous gasotransmitter, mediated a variety of biological processes through multiple signaling pathways, and aberrant H2S metabolism has been associated with mesenchymal stem cell (MSC) dysfunction. Here we employed the small interfering RNA treatment for cystathionine β-synthase (CBS), cystathionine γ-lyase, the main enzymes to synthesize H2S, and CBS-knockout mice to analyze the effect of H2S on dental pulp homeostasis. We showed that H2S deficiency attenuated dental pulp stem cell (DPSC) osteogenic/dentinogenic differentiation in vitro and in vivo with enhanced cell proliferation. Mechanically, H2S facilitated the transient receptor potential action channel subfamily V member 1-mediated calcium (Ca2+) influx, which subsequently activated the β-catenin pathway. While H2S deficiency decreased Ca2+, resulting in glycogen synthase kinase-3β-mediated β-catenin degradation, which controls proliferation and differentiation of DPSCs. Consistently, H2S-deficient mice displayed disturbed pattern of dental pulp and less dentin formation. In this study, we identified a previously unknown mechanism by which H2S regulates DPSC lineage determination and dental pulp homeostasis.
Collapse
Affiliation(s)
- Ruili Yang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, 100081 Beijing, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, 100081 Beijing, China
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, 100050 Beijing, China
| | - Tingting Yu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, 100081 Beijing, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, 100081 Beijing, China
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Dawei Liu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, 100081 Beijing, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, 100081 Beijing, China
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Songtao Shi
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Yongsheng Zhou
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, 100081 Beijing, China
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 100081 Beijing, China
| | - Yanheng Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, 100081 Beijing, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, 100081 Beijing, China
| |
Collapse
|
46
|
Immunomodulation of Human Mesenchymal Stem/Stromal Cells in Intervertebral Disc Degeneration: Insights From a Proinflammatory/Degenerative Ex Vivo Model. Spine (Phila Pa 1976) 2018; 43:E673-E682. [PMID: 29189572 DOI: 10.1097/brs.0000000000002494] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
STUDY DESIGN Ex vivo experimental study. OBJECTIVE To investigate the effect of proinflammatory/degenerative intervertebral disc (IVD) microenvironment on the regenerative and immunomodulatory behavior of mesenchymal stem/stromal cells (MSCs), using an ex vivo model from bovine origin. SUMMARY OF BACKGROUND DATA Low back pain is a cause of disability worldwide, most frequently associated with IVD degeneration and inflammation, and characterized by increased levels of inflammatory mediators, often disregarded. MSC-based therapies to low back pain have been advocated, but the involvement of inflammation in IVD remodeling mechanism, promoted by MSCs has not yet been explored. METHODS Bovine IVD organ cultures of nucleus pulposus punches were stimulated with needle puncture and culture medium supplementation with 10 ng/mL of interleukin (IL)-1β, to induce a proinflammatory/degenerative environment, as previously established. Human bone marrow-derived MSCs were cultured on top of transwells, placed above nucleus pulposus punches, for up to 16 days. MSCs were analyzed by screening cell viability/apoptosis, metabolic activity, migration, and inflammatory cytokines production in response to the proinflammatory environment. IVD extracellular matrix (ECM) remodeling, gene expression profile of IVD cells, and inflammatory cytokine profile in the presence of MSCs in basal versus proinflammatory conditions were also evaluated. RESULTS Proinflammatory/degenerative IVD conditions did not affect MSCs viability, but promoted cell migration, while increasing IL-6, IL-8, monocyte chemoattractant protein-1, and prostaglandin E2 and reducing transforming growth factor-β1 production by MSCs. MSCs did not stimulate ECM production (namely type II collagen or aggrecan) in neither basal nor inflammatory conditions, instead MSCs downregulated bovine proinflammatory IL-6, IL-8, and TNF-α gene expression levels in IL-1β-stimulated IVDs. CONCLUSION The present study provides evidence for an immunomodulatory paracrine effect of MSCs in degenerated IVD without an apparent effect in ECM remodeling, and suggest an MSCs mechanism-of-action dependent on a cytokine feedback loop. LEVEL OF EVIDENCE 5.
Collapse
|
47
|
Stem Cells for Cartilage Repair: Preclinical Studies and Insights in Translational Animal Models and Outcome Measures. Stem Cells Int 2018. [PMID: 29535784 PMCID: PMC5832141 DOI: 10.1155/2018/9079538] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Due to the restricted intrinsic capacity of resident chondrocytes to regenerate the lost cartilage postinjury, stem cell-based therapies have been proposed as a novel therapeutic approach for cartilage repair. Moreover, stem cell-based therapies using mesenchymal stem cells (MSCs) or induced pluripotent stem cells (iPSCs) have been used successfully in preclinical and clinical settings. Despite these promising reports, the exact mechanisms underlying stem cell-mediated cartilage repair remain uncertain. Stem cells can contribute to cartilage repair via chondrogenic differentiation, via immunomodulation, or by the production of paracrine factors and extracellular vesicles. But before novel cell-based therapies for cartilage repair can be introduced into the clinic, rigorous testing in preclinical animal models is required. Preclinical models used in regenerative cartilage studies include murine, lapine, caprine, ovine, porcine, canine, and equine models, each associated with its specific advantages and limitations. This review presents a summary of recent in vitro data and from in vivo preclinical studies justifying the use of MSCs and iPSCs in cartilage tissue engineering. Moreover, the advantages and disadvantages of utilizing small and large animals will be discussed, while also describing suitable outcome measures for evaluating cartilage repair.
Collapse
|
48
|
Zhou L, Chen X, Liu T, Zhu C, Si M, Jargstorf J, Li M, Pan G, Gong Y, Luo ZP, Yang H, Pei M, He F. SIRT1-dependent anti-senescence effects of cell-deposited matrix on human umbilical cord mesenchymal stem cells. J Tissue Eng Regen Med 2018; 12:e1008-e1021. [PMID: 28107614 PMCID: PMC9805355 DOI: 10.1002/term.2422] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 10/07/2016] [Accepted: 01/17/2017] [Indexed: 01/03/2023]
Abstract
Human umbilical cord-derived mesenchymal stem cells (UC-MSCs) are considered an attractive cell source for tissue regeneration. However, environmental oxidative stress can trigger premature senescence in MSCs and thus compromises their regenerative potential. Extracellular matrix (ECM) derived from MSCs has been shown to facilitate cell proliferation and multi-lineage differentiation. This investigation evaluated the effect of cell-deposited decellularized ECM (DECM) on oxidative stress-induced premature senescence in UC-MSCs. Sublethal dosages of H2 O2 , ranging from 50 μm to 200 μm, were used to induce senescence in MSCs. We found that DECM protected UC-MSCs from oxidative stress-induced premature senescence. When treated with H2 O2 at the same concentration, cell proliferation of DECM-cultured UC-MSCs was twofold higher than those on standard tissue culture polystyrene (TCPS). After exposure to 100 μm H2 O2 , fewer senescence-associated β-galactosidase-positive cells were observed on DECM than those on TCPS (17.6 ± 4.0% vs. 60.4 ± 6.2%). UC-MSCs cultured on DECM also showed significantly lower levels of senescence-related regulators, such as p16INK4α and p21. Most importantly, DECM preserved the osteogenic differentiation potential of UC-MSCs with premature senescence. The underlying molecular mechanisms involved the silent information regulator type 1 (SIRT1)-dependent signalling pathway, confirmed by the fact that the SIRT1 inhibitor nicotinamide counteracted the DECM-mediated anti-senescent effect. Collagen type I, rather than fibronectin, partially contributed to the protective effect of decellularized matrix. These findings provide a new strategy of using stem cell-deposited matrix to overcome the challenge of cellular senescence and to facilitate the clinical application of MSCs in regenerative medicine. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Long Zhou
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xi Chen
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, China
| | - Tao Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Caihong Zhu
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| | - Michelle Si
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Departments of Biology and Chemistry, Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Joseph Jargstorf
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Biology, Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Mao Li
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guoqing Pan
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| | - Yihong Gong
- School of Engineering, Sun Yat-sen University, Guangzhou, China
| | - Zong-Ping Luo
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huilin Yang
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics and Division of Exercise Physiology, West Virginia University, Morgantown, WV, USA
| | - Fan He
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
49
|
Pizzute T, Li J, Zhang Y, Davis ME, Pei M. Fibroblast Growth Factor Ligand Dependent Proliferation and Chondrogenic Differentiation of Synovium-Derived Stem Cells and Concomitant Adaptation of Wnt/Mitogen-Activated Protein Kinase Signals. Tissue Eng Part A 2017; 22:1036-46. [PMID: 27411850 DOI: 10.1089/ten.tea.2016.0102] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cell expansion techniques commonly utilize exogenous factors to increase cell proliferation and create a larger cell population for use in cell-based therapies. One strategy for cartilage regenerative therapies is autologous stem cell expansion and fibroblast growth factor (FGF) supplementation during cell expansion, particularly FGF-2. However, it is unknown whether FGF-10, another FGF implicated in limb and skeletal development, can elicit the same rejuvenation responses in terms of proliferation and differentiation of human synovium-derived stem cells (SDSCs). In this study, we expanded SDSCs in either FGF-2 or FGF-10 for 7 days; a control group had no treatment. FGF-2 and FGF-10 supplementation was also exclusively tested during the differentiation phase. Expanded SDSCs were evaluated for their ability to successfully engage in chondrogenic and osteogenic differentiation. We found that FGF-2 supplementation during proliferation, but not differentiation, was able to increase glycosaminoglycan deposition, pellet size, and chondrogenic gene expression following chondrogenic induction, as well as increased calcium deposition, alkaline phosphatase activity, and expression of vital osteogenic differentiation genes following osteogenic induction. FGF-10 did not elicit a similar preconditioning effect. We also observed changes of both Wnt signals and mitogen-activated protein kinase expression during SDSC chondrogenesis, which occurred in a manner dependent upon the supplementation phase of FGF-2 administration. These results indicated that FGF-2, but not FGF-10, may be supplemented during stem cell expansion to prime cells for successful chondrogenesis and osteogenesis.
Collapse
Affiliation(s)
- Tyler Pizzute
- 1 Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University , Morgantown, West Virginia.,2 Exercise Physiology, West Virginia University , Morgantown, West Virginia
| | - Jingting Li
- 1 Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University , Morgantown, West Virginia.,2 Exercise Physiology, West Virginia University , Morgantown, West Virginia
| | - Ying Zhang
- 1 Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University , Morgantown, West Virginia.,3 Mechanical and Aerospace Engineering, West Virginia University , Morgantown, West Virginia
| | - Mary E Davis
- 4 Department of Physiology and Pharmacology, West Virginia University , Morgantown, West Virginia
| | - Ming Pei
- 1 Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University , Morgantown, West Virginia.,2 Exercise Physiology, West Virginia University , Morgantown, West Virginia.,3 Mechanical and Aerospace Engineering, West Virginia University , Morgantown, West Virginia
| |
Collapse
|
50
|
Rahmati M, Nalesso G, Mobasheri A, Mozafari M. Aging and osteoarthritis: Central role of the extracellular matrix. Ageing Res Rev 2017; 40:20-30. [PMID: 28774716 DOI: 10.1016/j.arr.2017.07.004] [Citation(s) in RCA: 364] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 05/10/2017] [Accepted: 07/24/2017] [Indexed: 12/11/2022]
Abstract
Osteoarthritis (OA), is a major cause of severe joint pain, physical disability and quality of life impairment in the aging population across the developed and developing world. Increased catabolism in the extracellular matrix (ECM) of the articular cartilage is a key factor in the development and progression of OA. The molecular mechanisms leading to an impaired matrix turnover have not been fully clarified, however cellular senescence, increased expression of inflammatory mediators as well as oxidative stress in association with an inherently limited regenerative potential of the tissue, are all important contributors to OA development. All these factors are linked to and tend to be maximized by aging. Nonetheless the role of aging in compromising joint stability and function in OA has not been completely clarified yet. This review will systematically analyze cellular and structural changes taking place in the articular cartilage and bone in the pathogenesis of OA which are linked to aging. A particular emphasis will be placed on age-related changes in the phenotype of the articular chondrocytes.
Collapse
Affiliation(s)
- Maryam Rahmati
- Cellular and Molecular Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran; Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Giovanna Nalesso
- Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Ali Mobasheri
- The D-BOARD European Consortium for Biomarker Discovery, The APPROACH Innovative Medicines Initiative (IMI) Consortium, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK; Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, Arthritis Research UK Pain Centre, Medical Research Council and Arthritis Research UK Centre for Musculoskeletal Ageing Research, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK; Center of Excellence in Genomic Medicine Research (CEGMR), King Fahd Medical Research Center (KFMRC) and Sheik Salem Bin Mahfouz Scientific Chair for Treatment of Osteoarthritis with Stem Cells, King AbdulAziz University, Jeddah, 21589, Saudi Arabia
| | - Masoud Mozafari
- Cellular and Molecular Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran; Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran; Bioengineering Research Group, Nanotechnology and Advanced Materials Department, Materials and Energy Research Center (MERC), P.O. Box 14155-4777, Tehran, Iran.
| |
Collapse
|