1
|
Harissa Z, Kim Y, Dicks AR, Steward N, Guilak F. Skeletal dysplasia-causing mutations in TRPV4 alter the chondrocyte transcriptomic response to mechanical loading. Am J Physiol Cell Physiol 2025; 328:C1135-C1149. [PMID: 40019039 DOI: 10.1152/ajpcell.01066.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/28/2025] [Accepted: 02/21/2025] [Indexed: 03/01/2025]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a mechanosensitive ion channel highly expressed in chondrocytes that supports cartilage development and homeostasis. Mutations in the channel can cause skeletal dysplasias, including the gain-of-function mutations V620I and T89I, which lead to brachyolmia and metatropic dysplasia, respectively. These mutations suppress hypertrophic differentiation, but the mechanisms by which they alter chondrocyte response to mechanical load remain to be elucidated. To determine the effect of these mutations on chondrocyte mechanotransduction, tissue-engineered cartilage was derived from differentiated clustered regularly interspaced short palindromic repeats (CRISPR)-edited human-induced pluripotent stem cells (hiPSCs) harboring the moderate V620I or severe T89I TRPV4 mutations. Wild-type and mutant tissue-engineered hiPSC-derived cartilage contructs were subjected to compressive mechanical loading at physiological levels, and transcriptomic signatures were assessed by RNA-sequencing. Our results demonstrate that the V620I and T89I mutations diminish the mechanoresponsiveness of chondrocytes, as evidenced by reduced gene expression downstream of TRPV4 activation, including those involved in endochondral ossification. Changes in the expression of genes involved in extracellular matrix production and organization were found to contribute toward the phenotype in V620I mutant chondrocytes, whereas dysregulated retinoic acid signaling was linked to T89I, and disrupted proliferation was common to both. Our findings suggest that dysfunctional mechanotransduction due to V620I and T89I mutations in TRPV4 contribute to the developmental phenotypes, supporting TRPV4 modulation as a potential pharmacologic target.NEW & NOTEWORTHY Gain-of-function mutations in TRPV4, a mechano- and osmosensitive ion channel, are linked to skeletal dysplasias, but their effects on chondrocyte mechanotransduction remain unknown. Using human iPSCs harboring skeletal dysplasia-causing mutations, we developed and mechanically loaded tissue-engineered cartilage. Our findings show that V620I and T89I mutations reduce chondrocyte mechanoresponsiveness, evidenced by decreased gene expression downstream of TRPV4 activation, providing insight into TRPV4-related skeletal disorders and potential pharmacological targets.
Collapse
Affiliation(s)
- Zainab Harissa
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University, St. Louis, Missouri, United States
| | - Yuseon Kim
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University, St. Louis, Missouri, United States
| | - Amanda R Dicks
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University, St. Louis, Missouri, United States
| | - Nancy Steward
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University, St. Louis, Missouri, United States
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University, St. Louis, Missouri, United States
| |
Collapse
|
2
|
Yuan M, Li Q, Wang Z, Liu L, Wen C, Liu G, Yu F, Feng L, Yang L. TRPV4 Promotes Vascular Calcification by Directly Associating With and Activating β-Catenin. Arterioscler Thromb Vasc Biol 2025; 45:e101-e117. [PMID: 39973749 DOI: 10.1161/atvbaha.124.321793] [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: 09/02/2024] [Accepted: 02/07/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Vascular calcification contributes to increased cardiovascular morbidity and mortality in patients with chronic kidney disease, diabetes, and atherosclerosis. Currently, there are no effective therapeutic strategies to prevent or reverse vascular calcification. TRPV4 (transient receptor potential channel V4), a key Ca2+-permeable channel, plays an important role in various diseases. However, the role and mechanism of TRPV4 in vascular calcification have not yet been elucidated. METHODS The effects of TRPV4 on vascular calcification were explored in vitro and in vivo. TRPV4 interactome assessment and molecular docking were performed to investigate the mechanism and specific therapeutic strategy for vascular calcification. RESULTS TRPV4 was substantially upregulated in high inorganic phosphate-induced calcified vascular smooth muscle cells (SMCs) and calcified aortas from cholecalciferol (vitamin D3)-overloaded mice. TRPV4 overexpression increased the expression of the osteochondrogenic markers Runx2 (runt-related transcription factor 2), Msx2 (Msh homeobox 2), and Sox9 (SRY-box transcription factor 9) and exacerbated high inorganic phosphate-induced vascular SMC calcification in a Ca2+ influx-dependent manner. In contrast, TRPV4 deficiency or inactivation significantly inhibited vascular SMC calcification under high inorganic phosphate conditions. Moreover, compared with that in control littermates, SMC-specific TRPV4 deficiency in mice alleviated vitamin D3-induced and 5/6 nephrectomy-induced vascular calcification. Mechanistically, TRPV4 interacted with β-catenin and activated β-catenin/TCF (T-cell factor) transcriptional activity via Ca2+/ASK1 (apoptosis signal regulating kinase 1)/p38 signaling. β-Catenin knockdown abolished the effects of TRPV4 overexpression on vascular SMC calcification. TRPV4/β-catenin interaction is pivotal for maintaining TRPV4/Ca2+-induced ASK1/p38/β-catenin activation. Hesperidin, a natural product found in citrus fruits, effectively disrupted TRPV4/β-catenin interaction, thereby inhibiting ASK1/p38/β-catenin activity and preventing vascular calcification. CONCLUSIONS Our study identified TRPV4 as a new pathogenic factor for vascular calcification that directly associates with and activates β-catenin. Blocking the TRPV4/β-catenin interaction through hesperidin suppressed the progression of vascular calcification and may be an effective precision strategy to address vascular calcification.
Collapse
MESH Headings
- Animals
- TRPV Cation Channels/metabolism
- TRPV Cation Channels/genetics
- Vascular Calcification/pathology
- Vascular Calcification/metabolism
- Vascular Calcification/genetics
- Vascular Calcification/prevention & control
- beta Catenin/metabolism
- beta Catenin/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Mice, Inbred C57BL
- Disease Models, Animal
- Mice
- Male
- Humans
- Mice, Knockout
- Aortic Diseases/pathology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/prevention & control
- Cells, Cultured
- Molecular Docking Simulation
- p38 Mitogen-Activated Protein Kinases/metabolism
- Core Binding Factor Alpha 1 Subunit/metabolism
- Core Binding Factor Alpha 1 Subunit/genetics
- Cholecalciferol/pharmacology
- Phosphates/metabolism
- Calcium Signaling/drug effects
- Aorta/metabolism
- Aorta/pathology
- Aorta/drug effects
Collapse
Affiliation(s)
- Menglu Yuan
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China. MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, China
| | - Qi Li
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China. MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, China
| | - Zhiwei Wang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China. MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, China
| | - Liangju Liu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China. MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, China
| | - Chengyuan Wen
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China. MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, China
| | - Guizhu Liu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China. MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, China
| | - Fan Yu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China. MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, China
| | - Lei Feng
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China. MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, China
| | - Liu Yang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China. MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, China
| |
Collapse
|
3
|
Hernández-Vega AM, García-Villegas R, Rosenbaum T. Roles for TRPV4 in disease: A discussion of possible mechanisms. Cell Calcium 2024; 124:102972. [PMID: 39609180 DOI: 10.1016/j.ceca.2024.102972] [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: 08/01/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024]
Abstract
The transient receptor potential vanilloid 4 (TRPV4) ion channel is a ubiquitously expressed Ca2+-permeable ion channel that controls intracellular calcium ([Ca2+]i) homeostasis in various types of cells. The physiological roles for TRPV4 are tissue specific and the mechanisms behind this specificity remain mostly unclarified. It is noteworthy that mutations in the TRPV4 channel have been associated to a broad spectrum of congenital diseases, with most of these mutations mainly resulting in gain-of-function. Mutations have been identified in human patients showing a variety of phenotypes and symptoms, mostly related to skeletal and neuromuscular disorders. Since TRPV4 is so widely expressed throughout the body, it comes as no surprise that the literature is growing in evidence linking this protein to malfunction in systems other than the skeletal and neuromuscular. In this review, we summarize the expression patterns of TRPV4 in several tissues and highlight findings of recent studies that address critical structural and functional features of this channel, particularly focusing on its interactions and signaling pathways related to Ca2+ entry. Moreover, we discuss the roles of TRPV4 mutations in some diseases and pinpoint some of the mechanisms underlying pathological states where TRPV4's malfunction is prominent.
Collapse
Affiliation(s)
- Ana M Hernández-Vega
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - Refugio García-Villegas
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Ciudad de México, 07360, México
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico.
| |
Collapse
|
4
|
Ali EAM, Smaida R, Meyer M, Ou W, Li Z, Han Z, Benkirane-Jessel N, Gottenberg JE, Hua G. iPSCs chondrogenic differentiation for personalized regenerative medicine: a literature review. Stem Cell Res Ther 2024; 15:185. [PMID: 38926793 PMCID: PMC11210138 DOI: 10.1186/s13287-024-03794-1] [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: 03/28/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Cartilage, an important connective tissue, provides structural support to other body tissues, and serves as a cushion against impacts throughout the body. Found at the end of the bones, cartilage decreases friction and averts bone-on-bone contact during joint movement. Therefore, defects of cartilage can result from natural wear and tear, or from traumatic events, such as injuries or sudden changes in direction during sports activities. Overtime, these cartilage defects which do not always produce immediate symptoms, could lead to severe clinical pathologies. The emergence of induced pluripotent stem cells (iPSCs) has revolutionized the field of regenerative medicine, providing a promising platform for generating various cell types for therapeutic applications. Thus, chondrocytes differentiated from iPSCs become a promising avenue for non-invasive clinical interventions for cartilage injuries and diseases. In this review, we aim to highlight the current strategies used for in vitro chondrogenic differentiation of iPSCs and to explore their multifaceted applications in disease modeling, drug screening, and personalized regenerative medicine. Achieving abundant functional iPSC-derived chondrocytes requires optimization of culture conditions, incorporating specific growth factors, and precise temporal control. Continual improvements in differentiation methods and integration of emerging genome editing, organoids, and 3D bioprinting technologies will enhance the translational applications of iPSC-derived chondrocytes. Finally, to unlock the benefits for patients suffering from cartilage diseases through iPSCs-derived technologies in chondrogenesis, automatic cell therapy manufacturing systems will not only reduce human intervention and ensure sterile processes within isolator-like platforms to minimize contamination risks, but also provide customized production processes with enhanced scalability and efficiency.
Collapse
Affiliation(s)
- Eltahir Abdelrazig Mohamed Ali
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1260, Regenerative NanoMedicine (RNM), 1 Rue Eugène Boeckel, 67000, Strasbourg, France
- Université de Strasbourg, 67000, Strasbourg, France
| | - Rana Smaida
- Lamina Therapeutics, 1 Rue Eugène Boeckel, 67000, Strasbourg, France
| | - Morgane Meyer
- Université de Strasbourg, 67000, Strasbourg, France
- Lamina Therapeutics, 1 Rue Eugène Boeckel, 67000, Strasbourg, France
| | - Wenxin Ou
- Université de Strasbourg, 67000, Strasbourg, France
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), Service de Rhumatologie, Centre Hospitalier Universitaire de Strasbourg, 67000, Strasbourg, France
- Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Zongjin Li
- Nankai University School of Medicine, Tianjin, 300071, China
| | - Zhongchao Han
- Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health & Biotech Co, Beijing, 100176, China
| | - Nadia Benkirane-Jessel
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1260, Regenerative NanoMedicine (RNM), 1 Rue Eugène Boeckel, 67000, Strasbourg, France.
- Université de Strasbourg, 67000, Strasbourg, France.
- Lamina Therapeutics, 1 Rue Eugène Boeckel, 67000, Strasbourg, France.
| | - Jacques Eric Gottenberg
- Université de Strasbourg, 67000, Strasbourg, France.
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), Service de Rhumatologie, Centre Hospitalier Universitaire de Strasbourg, 67000, Strasbourg, France.
| | - Guoqiang Hua
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1260, Regenerative NanoMedicine (RNM), 1 Rue Eugène Boeckel, 67000, Strasbourg, France.
- Université de Strasbourg, 67000, Strasbourg, France.
| |
Collapse
|
5
|
Adegunsoye A, Gonzales NM, Gilad Y. Induced Pluripotent Stem Cells in Disease Biology and the Evidence for Their In Vitro Utility. Annu Rev Genet 2023; 57:341-360. [PMID: 37708421 DOI: 10.1146/annurev-genet-022123-090319] [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] [Indexed: 09/16/2023]
Abstract
Many human phenotypes are impossible to recapitulate in model organisms or immortalized human cell lines. Induced pluripotent stem cells (iPSCs) offer a way to study disease mechanisms in a variety of differentiated cell types while circumventing ethical and practical issues associated with finite tissue sources and postmortem states. Here, we discuss the broad utility of iPSCs in genetic medicine and describe how they are being used to study musculoskeletal, pulmonary, neurologic, and cardiac phenotypes. We summarize the particular challenges presented by each organ system and describe how iPSC models are being used to address them. Finally, we discuss emerging iPSC-derived organoid models and the potential value that they can bring to studies of human disease.
Collapse
Affiliation(s)
- Ayodeji Adegunsoye
- Genetics, Genomics, and Systems Biology, Section of Pulmonary and Critical Care, and the Department of Medicine, University of Chicago, Chicago, Illinois, USA;
| | - Natalia M Gonzales
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA; ,
| | - Yoav Gilad
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA; ,
- Department of Human Genetics, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
6
|
Eremeev A, Pikina A, Ruchko Y, Bogomazova A. Clinical Potential of Cellular Material Sources in the Generation of iPSC-Based Products for the Regeneration of Articular Cartilage. Int J Mol Sci 2023; 24:14408. [PMID: 37833856 PMCID: PMC10572671 DOI: 10.3390/ijms241914408] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/06/2023] [Accepted: 09/06/2023] [Indexed: 10/15/2023] Open
Abstract
Inflammatory joint diseases, among which osteoarthritis and rheumatoid arthritis are the most common, are characterized by progressive degeneration of the cartilage tissue, resulting in the threat of limited or lost joint functionality in the absence of treatment. Currently, treating these diseases is difficult, and a number of existing treatment and prevention measures are not entirely effective and are complicated by the patients' conditions, the multifactorial nature of the pathology, and an incomplete understanding of the etiology. Cellular technologies based on induced pluripotent stem cells (iPSCs) can provide a vast cellular resource for the production of artificial cartilage tissue for replacement therapy and allow the possibility of a personalized approach. However, the question remains whether a number of etiological abnormalities associated with joint disease are transmitted from the source cell to iPSCs and their chondrocyte derivatives. Some data state that there is no difference between the iPSCs and their derivatives from healthy and sick donors; however, there are other data indicating a dissimilarity. Therefore, this topic requires a thorough study of the differentiation potential of iPSCs and the factors influencing it, the risk factors associated with joint diseases, and a comparative analysis of the characteristics of cells obtained from patients. Together with cultivation optimization methods, these measures can increase the efficiency of obtaining cell technology products and make their wide practical application possible.
Collapse
Affiliation(s)
- Artem Eremeev
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow 119435, Russia; (A.P.); (A.B.)
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, Moscow 119334, Russia;
| | - Arina Pikina
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow 119435, Russia; (A.P.); (A.B.)
- Department of Embryology, Faculty of Biology, Lomonosov Moscow State University, GSP-1 Leninskie Gory, Moscow 119991, Russia
| | - Yevgeny Ruchko
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, Moscow 119334, Russia;
| | - Alexandra Bogomazova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow 119435, Russia; (A.P.); (A.B.)
| |
Collapse
|
7
|
Palomino Lago E, Jelbert ER, Baird A, Lam PY, Guest DJ. Equine induced pluripotent stem cells are responsive to inflammatory cytokines before and after differentiation into musculoskeletal cell types. In Vitro Cell Dev Biol Anim 2023; 59:514-527. [PMID: 37582999 PMCID: PMC10520172 DOI: 10.1007/s11626-023-00800-3] [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: 05/26/2023] [Accepted: 07/19/2023] [Indexed: 08/17/2023]
Abstract
Persistent inflammation is associated with the poor regeneration of musculoskeletal tissues. Embryonic stem cells (ESCs) have an attenuated response to inflammatory cytokines, but there are mixed reports on the response of induced pluripotent stem cells (iPSCs) to inflammation. Horses provide a relevant large animal model for studying musculoskeletal tissue diseases and the testing of novel therapies. The aim of this study was to determine if equine iPSCs are responsive to the inflammatory cytokines IL-1β, TNFα and IFN-γ in their undifferentiated state, or following differentiation into tendon and cartilage-like cells. We demonstrated that in undifferentiated iPSCs, the cytokines induce NF-κB P65 and STAT1 nuclear translocation which leads to cell death, decreased OCT4 expression and increased expression of inflammatory genes. Following differentiation towards cartilage-like cells exposure to the cytokines resulted in STAT1 nuclear translocation, changes in cartilage gene expression and increased expression of matrix metalloproteinases (MMPs) and inflammatory genes. Exposure of iPSC-derived tendon-like cells to the cytokines resulted nuclear translocation of NF-κB P65 and STAT1, altered tendon gene expression, increased MMP expression and increased expression of inflammatory genes. Equine iPSCs are therefore capable of responding to inflammatory stimulation and this may have relevance for their future clinical application.
Collapse
Affiliation(s)
- Esther Palomino Lago
- Department of Clinical Sciences and Services, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, AL9 7TA, Herts, UK
| | - Elizabeth R Jelbert
- Department of Clinical Sciences and Services, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, AL9 7TA, Herts, UK
| | - Arabella Baird
- Animal Health Trust, Lanwades Park, Kentford, Newmarket, CB8 7UU, UK
| | - Pak Y Lam
- Department of Clinical Sciences and Services, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, AL9 7TA, Herts, UK
| | - Deborah J Guest
- Department of Clinical Sciences and Services, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, AL9 7TA, Herts, UK.
| |
Collapse
|
8
|
Dicks AR, Maksaev GI, Harissa Z, Savadipour A, Tang R, Steward N, Liedtke W, Nichols CG, Wu CL, Guilak F. Skeletal dysplasia-causing TRPV4 mutations suppress the hypertrophic differentiation of human iPSC-derived chondrocytes. eLife 2023; 12:e71154. [PMID: 36810131 PMCID: PMC9949800 DOI: 10.7554/elife.71154] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 02/03/2023] [Indexed: 02/24/2023] Open
Abstract
Mutations in the TRPV4 ion channel can lead to a range of skeletal dysplasias. However, the mechanisms by which TRPV4 mutations lead to distinct disease severity remain unknown. Here, we use CRISPR-Cas9-edited human-induced pluripotent stem cells (hiPSCs) harboring either the mild V620I or lethal T89I mutations to elucidate the differential effects on channel function and chondrogenic differentiation. We found that hiPSC-derived chondrocytes with the V620I mutation exhibited increased basal currents through TRPV4. However, both mutations showed more rapid calcium signaling with a reduced overall magnitude in response to TRPV4 agonist GSK1016790A compared to wildtype (WT). There were no differences in overall cartilaginous matrix production, but the V620I mutation resulted in reduced mechanical properties of cartilage matrix later in chondrogenesis. mRNA sequencing revealed that both mutations up-regulated several anterior HOX genes and down-regulated antioxidant genes CAT and GSTA1 throughout chondrogenesis. BMP4 treatment up-regulated several essential hypertrophic genes in WT chondrocytes; however, this hypertrophic maturation response was inhibited in mutant chondrocytes. These results indicate that the TRPV4 mutations alter BMP signaling in chondrocytes and prevent proper chondrocyte hypertrophy, as a potential mechanism for dysfunctional skeletal development. Our findings provide potential therapeutic targets for developing treatments for TRPV4-mediated skeletal dysplasias.
Collapse
Affiliation(s)
- Amanda R Dicks
- Department of Biomedical Engineering, Washington University in St. LouisSt LouisUnited States
- Department of Orthopedic Surgery, Washington University School of Medicine, St. LouisSt LouisUnited States
- Shriners Hospitals for Children - St. LouisSt. LouisUnited States
| | - Grigory I Maksaev
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. LouisSt LouisUnited States
| | - Zainab Harissa
- Department of Biomedical Engineering, Washington University in St. LouisSt LouisUnited States
- Department of Orthopedic Surgery, Washington University School of Medicine, St. LouisSt LouisUnited States
- Shriners Hospitals for Children - St. LouisSt. LouisUnited States
| | - Alireza Savadipour
- Department of Orthopedic Surgery, Washington University School of Medicine, St. LouisSt LouisUnited States
- Shriners Hospitals for Children - St. LouisSt. LouisUnited States
- Department of Mechanical Engineering and Material Science, Washington University in St. LouisSt. LouisUnited States
| | - Ruhang Tang
- Department of Orthopedic Surgery, Washington University School of Medicine, St. LouisSt LouisUnited States
- Shriners Hospitals for Children - St. LouisSt. LouisUnited States
| | - Nancy Steward
- Department of Orthopedic Surgery, Washington University School of Medicine, St. LouisSt LouisUnited States
- Shriners Hospitals for Children - St. LouisSt. LouisUnited States
| | - Wolfgang Liedtke
- Department of Neurology, Duke University School of MedicineDurhamUnited States
- Department of Molecular Pathobiology - NYU College of DentistryNew YorkUnited States
| | - Colin G Nichols
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. LouisSt LouisUnited States
| | - Chia-Lung Wu
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of RochesterRochesterUnited States
| | - Farshid Guilak
- Department of Orthopedic Surgery, Washington University School of Medicine, St. LouisSt LouisUnited States
- Shriners Hospitals for Children - St. LouisSt. LouisUnited States
| |
Collapse
|
9
|
Dicks AR, Steward N, Guilak F, Wu CL. Chondrogenic Differentiation of Human-Induced Pluripotent Stem Cells. Methods Mol Biol 2023; 2598:87-114. [PMID: 36355287 PMCID: PMC9830630 DOI: 10.1007/978-1-0716-2839-3_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The generation of large quantities of genetically defined human chondrocytes remains a critical step for the development of tissue engineering strategies for cartilage regeneration and high-throughput drug screening. This protocol describes chondrogenic differentiation of human-induced pluripotent stem cells (hiPSCs), which can undergo genetic modification and the capacity for extensive cell expansion. The hiPSCs are differentiated in a stepwise manner in monolayer through the mesodermal lineage for 12 days using defined growth factors and small molecules. This is followed by 28 days of chondrogenic differentiation in a 3D pellet culture system using transforming growth factor beta 3 and specific compounds to inhibit off-target differentiation. The 6-week protocol results in hiPSC-derived cartilaginous tissue that can be characterized by histology, immunohistochemistry, and gene expression or enzymatically digested to isolate chondrocyte-like cells. Investigators can use this protocol for experiments including genetic engineering, in vitro disease modeling, or tissue engineering.
Collapse
Affiliation(s)
- Amanda R Dicks
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Shriners Hospitals for Children - St. Louis, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA
| | - Nancy Steward
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Shriners Hospitals for Children - St. Louis, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA.
- Shriners Hospitals for Children - St. Louis, St. Louis, MO, USA.
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA.
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA.
| | - Chia-Lung Wu
- Department of Orthopaedic Surgery and Rehabilitation, Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
10
|
Humphreys PA, Mancini FE, Ferreira MJS, Woods S, Ogene L, Kimber SJ. Developmental principles informing human pluripotent stem cell differentiation to cartilage and bone. Semin Cell Dev Biol 2022; 127:17-36. [PMID: 34949507 DOI: 10.1016/j.semcdb.2021.11.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 12/14/2022]
Abstract
Human pluripotent stem cells can differentiate into any cell type given appropriate signals and hence have been used to research early human development of many tissues and diseases. Here, we review the major biological factors that regulate cartilage and bone development through the three main routes of neural crest, lateral plate mesoderm and paraxial mesoderm. We examine how these routes have been used in differentiation protocols that replicate skeletal development using human pluripotent stem cells and how these methods have been refined and improved over time. Finally, we discuss how pluripotent stem cells can be employed to understand human skeletal genetic diseases with a developmental origin and phenotype, and how developmental protocols have been applied to gain a better understanding of these conditions.
Collapse
Affiliation(s)
- Paul A Humphreys
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK; Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering & Henry Royce Institute, University of Manchester, UK
| | - Fabrizio E Mancini
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Miguel J S Ferreira
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK; Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering & Henry Royce Institute, University of Manchester, UK
| | - Steven Woods
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Leona Ogene
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Susan J Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| |
Collapse
|
11
|
De Kinderen P, Meester J, Loeys B, Peeters S, Gouze E, Woods S, Mortier G, Verstraeten A. Differentiation of Induced Pluripotent Stem Cells Into Chondrocytes: Methods and Applications for Disease Modeling and Drug Discovery. J Bone Miner Res 2022; 37:397-410. [PMID: 35124831 DOI: 10.1002/jbmr.4524] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/25/2022] [Accepted: 02/01/2022] [Indexed: 11/11/2022]
Abstract
Induced pluripotent stem cell (iPSC) technology allows pathomechanistic and therapeutic investigation of human heritable disorders affecting tissue types whose collection from patients is difficult or even impossible. Among them are cartilage diseases. Over the past decade, iPSC-chondrocyte disease models have been shown to exhibit several key aspects of known disease mechanisms. Concurrently, an increasing number of protocols to differentiate iPSCs into chondrocytes have been published, each with its respective (dis)advantages. In this review we provide a comprehensive overview of the different differentiation approaches, the hitherto described iPSC-chondrocyte disease models and mechanistic and/or therapeutic insights that have been derived from their investigation, and the current model limitations. Key lessons are that the most appropriate differentiation approach is dependent upon the cartilage disease under investigation and that further optimization is still required to recapitulate the in vivo cartilage. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Pauline De Kinderen
- Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Josephina Meester
- Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Bart Loeys
- Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium.,Department of Human Genetics, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Silke Peeters
- Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Elvire Gouze
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France
| | - Steven Woods
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Geert Mortier
- Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Aline Verstraeten
- Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
12
|
The Induced Pluripotent Stem Cells in Articular Cartilage Regeneration and Disease Modelling: Are We Ready for Their Clinical Use? Cells 2022; 11:cells11030529. [PMID: 35159338 PMCID: PMC8834349 DOI: 10.3390/cells11030529] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 02/01/2023] Open
Abstract
The development of induced pluripotent stem cells has brought unlimited possibilities to the field of regenerative medicine. This could be ideal for treating osteoarthritis and other skeletal diseases, because the current procedures tend to be short-term solutions. The usage of induced pluripotent stem cells in the cell-based regeneration of cartilage damages could replace or improve on the current techniques. The patient’s specific non-invasive collection of tissue for reprogramming purposes could also create a platform for drug screening and disease modelling for an overview of distinct skeletal abnormalities. In this review, we seek to summarise the latest achievements in the chondrogenic differentiation of pluripotent stem cells for regenerative purposes and disease modelling.
Collapse
|
13
|
Hwang JJ, Choi J, Rim YA, Nam Y, Ju JH. Application of Induced Pluripotent Stem Cells for Disease Modeling and 3D Model Construction: Focus on Osteoarthritis. Cells 2021; 10:cells10113032. [PMID: 34831254 PMCID: PMC8622662 DOI: 10.3390/cells10113032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 11/16/2022] Open
Abstract
Since their discovery in 2006, induced pluripotent stem cells (iPSCs) have shown promising potential, specifically because of their accessibility and plasticity. Hence, the clinical applicability of iPSCs was investigated in various fields of research. However, only a few iPSC studies pertaining to osteoarthritis (OA) have been performed so far, despite the high prevalence rate of degenerative joint disease. In this review, we discuss some of the most recent applications of iPSCs in disease modeling and the construction of 3D models in various fields, specifically focusing on osteoarthritis and OA-related conditions. Notably, we comprehensively reviewed the successful results of iPSC-derived disease models in recapitulating OA phenotypes for both OA and early-onset OA to encompass their broad etiology. Moreover, the latest publications with protocols that have used iPSCs to construct 3D models in recapitulating various conditions, particularly the OA environment, were further discussed. With the overall optimistic results seen in both fields, iPSCs are expected to be more widely used for OA disease modeling and 3D model construction, which could further expand OA drug screening, risk assessment, and therapeutic capabilities.
Collapse
Affiliation(s)
- Joel Jihwan Hwang
- College of Public Health and Social Justice, Saint Louis University, St. Louis, MO 63103, USA;
| | - Jinhyeok Choi
- YiPSCELL, Inc., 39 Banpo-daero, Seocho-gu, Seoul 06579, Korea; (J.C.); (Y.N.)
| | - Yeri Alice Rim
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Yoojun Nam
- YiPSCELL, Inc., 39 Banpo-daero, Seocho-gu, Seoul 06579, Korea; (J.C.); (Y.N.)
| | - Ji Hyeon Ju
- YiPSCELL, Inc., 39 Banpo-daero, Seocho-gu, Seoul 06579, Korea; (J.C.); (Y.N.)
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
- Division of Rheumatology, Department of Internal Medicine, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul St. Mary’s Hospital, Seoul 06591, Korea
- Correspondence:
| |
Collapse
|
14
|
Willard VP, Leddy HA, Palmer D, Wu CL, Liedtke W, Guilak F. Transient receptor potential vanilloid 4 as a regulator of induced pluripotent stem cell chondrogenesis. Stem Cells 2021; 39:1447-1456. [PMID: 34427363 DOI: 10.1002/stem.3440] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 07/19/2021] [Indexed: 12/14/2022]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a polymodal calcium-permeable cation channel that is highly expressed in cartilage and is sensitive to a variety of extracellular stimuli. The expression of this channel has been associated with the process of chondrogenesis in adult stem cells as well as several cell lines. Here, we used a chondrogenic reporter (Col2a1-GFP) in murine induced pluripotent stem cells (iPSCs) to examine the hypothesis that TRPV4 serves as both a marker and a regulator of chondrogenesis. Over 21 days of chondrogenesis, iPSCs showed significant increases in Trpv4 expression along with the standard chondrogenic gene markers Sox9, Acan, and Col2a1, particularly in the green fluorescent protein positive (GFP+) chondroprogenitor subpopulation. Increased gene expression for Trpv4 was also reflected by the presence of TRPV4 protein and functional Ca2+ signaling. Daily activation of TRPV4 using the specific agonist GSK1016790A resulted in significant increases in cartilaginous matrix production. An improved understanding of the role of TRPV4 in chondrogenesis may provide new insights into the development of new therapeutic approaches for diseases of cartilage, such as osteoarthritis, or channelopathies and hereditary disorders that affect cartilage during development. Harnessing the role of TRPV4 in chondrogenesis may also provide a novel approach for accelerating stem cell differentiation in functional tissue engineering of cartilage replacements for joint repair.
Collapse
Affiliation(s)
| | - Holly A Leddy
- Shared Materials Instrumentation Facility, Duke University, Durham, North Carolina, USA
| | - Daniel Palmer
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA.,Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, USA.,Center of Regenerative Medicine, Washington University, St. Louis, Missouri, USA
| | - Chia-Lung Wu
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA.,Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, USA.,Center of Regenerative Medicine, Washington University, St. Louis, Missouri, USA.,Department of Orthopaedic Surgery and Rehabilitation, Center for Musculoskeletal Research, University of Rochester, Rochester, New York, USA
| | | | - Farshid Guilak
- Cytex Therapeutics, Inc, Durham, North Carolina, USA.,Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA.,Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, USA.,Center of Regenerative Medicine, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
15
|
Woods S, Humphreys PA, Bates N, Richardson SA, Kuba SY, Brooks IR, Cain SA, Kimber SJ. Regulation of TGFβ Signalling by TRPV4 in Chondrocytes. Cells 2021; 10:726. [PMID: 33805168 PMCID: PMC8064313 DOI: 10.3390/cells10040726] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/05/2021] [Accepted: 03/13/2021] [Indexed: 12/17/2022] Open
Abstract
The growth factor TGFβ and the mechanosensitive calcium-permeable cation channel TRPV4 are both important for the development and maintenance of many tissues. Although TRPV4 and TGFβ both affect core cellular functions, how their signals are integrated is unknown. Here we show that pharmacological activation of TRPV4 significantly increased the canonical response to TGFβ stimulation in chondrocytes. Critically, this increase was only observed when TRPV4 was activated after, but not before TGFβ stimulation. The increase was prevented by pharmacological TRPV4 inhibition or knockdown and is calcium/CamKII dependent. RNA-seq analysis after TRPV4 activation showed enrichment for the TGFβ signalling pathway and identified JUN and SP1 as key transcription factors involved in this response. TRPV4 modulation of TGFβ signalling represents an important pathway linking mechanical signalling to tissue development and homeostasis.
Collapse
Affiliation(s)
- Steven Woods
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; (P.A.H.); (N.B.); (S.A.R.); (S.Y.K.); (I.R.B.); (S.A.C.)
| | | | | | | | | | | | | | - Susan J. Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; (P.A.H.); (N.B.); (S.A.R.); (S.Y.K.); (I.R.B.); (S.A.C.)
| |
Collapse
|
16
|
Sun X, Kato H, Sato H, Torio M, Han X, Zhang Y, Hirofuji Y, Kato TA, Sakai Y, Ohga S, Fukumoto S, Masuda K. Impaired neurite development and mitochondrial dysfunction associated with calcium accumulation in dopaminergic neurons differentiated from the dental pulp stem cells of a patient with metatropic dysplasia. Biochem Biophys Rep 2021; 26:100968. [PMID: 33748438 PMCID: PMC7960789 DOI: 10.1016/j.bbrep.2021.100968] [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: 03/19/2020] [Revised: 12/22/2020] [Accepted: 02/22/2021] [Indexed: 12/15/2022] Open
Abstract
Transient receptor potential vanilloid member 4 (TRPV4) is a Ca2+ permeable nonselective cation channel, and mutations in the TRPV4 gene cause congenital skeletal dysplasias and peripheral neuropathies. Although TRPV4 is widely expressed in the brain, few studies have assessed the pathogenesis of TRPV4 mutations in the brain. We aimed to elucidate the pathological associations between a specific TRPV4 mutation and neurodevelopmental defects using dopaminergic neurons (DNs) differentiated from dental pulp stem cells (DPSCs). DPSCs were isolated from a patient with metatropic dysplasia and multiple neuropsychiatric symptoms caused by a gain-of-function TRPV4 mutation, c.1855C>T (p.L619F). The mutation was corrected by CRISPR/Cas9 to obtain isogenic control DPSCs. Mutant DPSCs differentiated into DNs without undergoing apoptosis; however, neurite development was significantly impaired in mutant vs. control DNs. Mutant DNs also showed accumulation of mitochondrial Ca2+ and reactive oxygen species, low adenosine triphosphate levels despite a high mitochondrial membrane potential, and lower peroxisome proliferator-activated receptor gamma coactivator 1-alpha expression and mitochondrial content. These results suggested that the persistent Ca2+ entry through the constitutively activated TRPV4 might perturb the adaptive coordination of multiple mitochondrial functions, including oxidative phosphorylation, redox control, and biogenesis, required for dopaminergic circuit development in the brain. Thus, certain mutations in TRPV4 that are associated with skeletal dysplasia might have pathogenic effects on brain development, and mitochondria might be a potential therapeutic target to alleviate the neuropsychiatric symptoms of TRPV4-related diseases.
Collapse
Key Words
- ATP, adenosine triphosphate
- DN, dopaminergic neuron
- DPSC, dental pulp stem cell
- Dental pulp stem cells
- Dopaminergic neuron
- MD, metatropic dysplasia
- MPP, mitochondrial membrane potential
- Metatropic dysplasia
- Mitochondria
- NURR1, nuclear receptor related 1
- PGC-1α, peroxisome proliferator-activated receptor gamma coactivator 1-alpha
- ROS, reactive oxygen species
- RPL13A, 60S ribosomal protein L13a
- Reactive oxygen species
- SOD, superoxide dismutase
- TRPV4, transient receptor potential vanilloid member 4
- Transient receptor potential vanilloid 4
Collapse
Affiliation(s)
- Xiao Sun
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Hiroki Kato
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Hiroshi Sato
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Michiko Torio
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Xu Han
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Yu Zhang
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Yuta Hirofuji
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Takahiro A Kato
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Yasunari Sakai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Satoshi Fukumoto
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Keiji Masuda
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
17
|
Masuda K, Han X, Kato H, Sato H, Zhang Y, Sun X, Hirofuji Y, Yamaza H, Yamada A, Fukumoto S. Dental Pulp-Derived Mesenchymal Stem Cells for Modeling Genetic Disorders. Int J Mol Sci 2021; 22:ijms22052269. [PMID: 33668763 PMCID: PMC7956585 DOI: 10.3390/ijms22052269] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/18/2021] [Accepted: 02/23/2021] [Indexed: 12/20/2022] Open
Abstract
A subpopulation of mesenchymal stem cells, developmentally derived from multipotent neural crest cells that form multiple facial tissues, resides within the dental pulp of human teeth. These stem cells show high proliferative capacity in vitro and are multipotent, including adipogenic, myogenic, osteogenic, chondrogenic, and neurogenic potential. Teeth containing viable cells are harvested via minimally invasive procedures, based on various clinical diagnoses, but then usually discarded as medical waste, indicating the relatively low ethical considerations to reuse these cells for medical applications. Previous studies have demonstrated that stem cells derived from healthy subjects are an excellent source for cell-based medicine, tissue regeneration, and bioengineering. Furthermore, stem cells donated by patients affected by genetic disorders can serve as in vitro models of disease-specific genetic variants, indicating additional applications of these stem cells with high plasticity. This review discusses the benefits, limitations, and perspectives of patient-derived dental pulp stem cells as alternatives that may complement other excellent, yet incomplete stem cell models, such as induced pluripotent stem cells, together with our recent data.
Collapse
Affiliation(s)
- Keiji Masuda
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan; (X.H.); (H.S.); (Y.Z.); (X.S.); (Y.H.); (H.Y.)
- Correspondence: (K.M.); (S.F.); Tel.: +81-92-642-6402 (K.M. & S.F.)
| | - Xu Han
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan; (X.H.); (H.S.); (Y.Z.); (X.S.); (Y.H.); (H.Y.)
| | - Hiroki Kato
- Department of Molecular Cell Biology and Oral Anatomy, Graduate School of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan;
| | - Hiroshi Sato
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan; (X.H.); (H.S.); (Y.Z.); (X.S.); (Y.H.); (H.Y.)
| | - Yu Zhang
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan; (X.H.); (H.S.); (Y.Z.); (X.S.); (Y.H.); (H.Y.)
| | - Xiao Sun
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan; (X.H.); (H.S.); (Y.Z.); (X.S.); (Y.H.); (H.Y.)
| | - Yuta Hirofuji
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan; (X.H.); (H.S.); (Y.Z.); (X.S.); (Y.H.); (H.Y.)
| | - Haruyoshi Yamaza
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan; (X.H.); (H.S.); (Y.Z.); (X.S.); (Y.H.); (H.Y.)
| | - Aya Yamada
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8577, Japan;
| | - Satoshi Fukumoto
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan; (X.H.); (H.S.); (Y.Z.); (X.S.); (Y.H.); (H.Y.)
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8577, Japan;
- Correspondence: (K.M.); (S.F.); Tel.: +81-92-642-6402 (K.M. & S.F.)
| |
Collapse
|
18
|
Induced Pluripotent Stem Cell-Differentiated Chondrocytes Repair Cartilage Defect in a Rabbit Osteoarthritis Model. Stem Cells Int 2020; 2020:8867349. [PMID: 33224204 PMCID: PMC7671807 DOI: 10.1155/2020/8867349] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/08/2020] [Accepted: 10/19/2020] [Indexed: 12/14/2022] Open
Abstract
The aim of this study was to explore the therapeutic effect of iPSC-mesenchymal stem cell (MSC)-derived chondrocytes in a rabbit osteoarthritis (OA) model. The iPSCs were characterized by gene expressions, immunostaining of pluripotent markers, and in vivo teratoma formation. iPSC-differentiated MSCs were characterized by flow cytometry and trilineage differentiation. A rabbit OA model was established by the transection of the anterior cruciate ligament. The therapeutic effect of transplanted iPSC-MSC-chondrocytes on the OA was evaluated by the histology, immunostaining, and qPCR of defective cartilage. The results showed iPSC could express pluripotency markers such as OCT4, SOX2, and NANOG and form an embryoid body and a teratoma. After differentiation of iPSCs for 30 days, MSCs were established. The iPSC-MSC could express typical MSC markers such as CD29, CD44, CD90, CD105, and HLA-ABC. They could differentiate into adipocytes, osteocytes, and chondrocytes. In this model, iPSC-MSC-chondrocytes significantly improved the histology and ICRS (International Cartilage Repair Society) scores. The transplanted cartilage expressed less IL-1β, TNF-α, and MMP13 than control cartilage. In conclusion, the iPSCs we derived might represent an emerging source for differentiated MSC-chondrocyte and might rescue cartilage defects through its anti-inflammatory and anti-catabolic effects.
Collapse
|
19
|
Hayes AJ, Melrose J. Aggrecan, the Primary Weight-Bearing Cartilage Proteoglycan, Has Context-Dependent, Cell-Directive Properties in Embryonic Development and Neurogenesis: Aggrecan Glycan Side Chain Modifications Convey Interactive Biodiversity. Biomolecules 2020; 10:E1244. [PMID: 32867198 PMCID: PMC7564073 DOI: 10.3390/biom10091244] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/19/2020] [Accepted: 08/23/2020] [Indexed: 02/06/2023] Open
Abstract
This review examines aggrecan's roles in developmental embryonic tissues, in tissues undergoing morphogenetic transition and in mature weight-bearing tissues. Aggrecan is a remarkably versatile and capable proteoglycan (PG) with diverse tissue context-dependent functional attributes beyond its established role as a weight-bearing PG. The aggrecan core protein provides a template which can be variably decorated with a number of glycosaminoglycan (GAG) side chains including keratan sulphate (KS), human natural killer trisaccharide (HNK-1) and chondroitin sulphate (CS). These convey unique tissue-specific functional properties in water imbibition, space-filling, matrix stabilisation or embryonic cellular regulation. Aggrecan also interacts with morphogens and growth factors directing tissue morphogenesis, remodelling and metaplasia. HNK-1 aggrecan glycoforms direct neural crest cell migration in embryonic development and is neuroprotective in perineuronal nets in the brain. The ability of the aggrecan core protein to assemble CS and KS chains at high density equips cartilage aggrecan with its well-known water-imbibing and weight-bearing properties. The importance of specific arrangements of GAG chains on aggrecan in all its forms is also a primary morphogenetic functional determinant providing aggrecan with unique tissue context dependent regulatory properties. The versatility displayed by aggrecan in biodiverse contexts is a function of its GAG side chains.
Collapse
Affiliation(s)
- Anthony J Hayes
- Bioimaging Research Hub, Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, Wales, UK
| | - James Melrose
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, Northern Sydney Local Health District, Royal North Shore Hospital, St. Leonards 2065, NSW, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney 2052, NSW, Australia
- Sydney Medical School, Northern, The University of Sydney, Faculty of Medicine and Health at Royal North Shore Hospital, St. Leonards 2065, NSW, Australia
| |
Collapse
|
20
|
Generation and characterization of human induced pluripotent stem cells (iPSCs) from hand osteoarthritis patient-derived fibroblasts. Sci Rep 2020; 10:4272. [PMID: 32144293 PMCID: PMC7060311 DOI: 10.1038/s41598-020-61071-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 02/17/2020] [Indexed: 12/21/2022] Open
Abstract
Knowledge and research results about hand osteoarthritis (hOA) are limited due to the lack of samples and animal models of the disease. Here, we report the generation of two induced pluripotent stem cell (iPSC)-lines from patients with radiographic hOA. Furthermore, we wondered whether these iPSC-lines carried single nucleotide polymorphisms (SNPs) within genes that have been associated with hOA. Finally, we performed chondrogenic differentiation of the iPSCs in order to prove their usefulness as cellular models of the disease. We performed a non-integrative reprogramming of dermal fibroblasts obtained from two patients with radiographic rhizarthrosis and non-erosive hOA by introducing the transcriptional factors Oct4, Sox2, Klf4 and c-Myc using Sendai virus. After reprogramming, embryonic stem cell-like colonies emerged in culture, which fulfilled all the criteria to be considered iPSCs. Both iPSC-lines carried variants associated with hOA in the four studied genes and showed differences in their chondrogenic capacity when compared with a healthy control iPSC-line. To our knowledge this is the first time that the generation of iPSC-lines from patients with rhizarthrosis and non-erosive hOA is reported. The obtained iPSC-lines might enable us to model the disease in vitro, and to deeper study both the molecular and cellular mechanisms underlying hOA.
Collapse
|
21
|
Accelerated osteoblastic differentiation in patient-derived dental pulp stem cells carrying a gain-of-function mutation of TRPV4 associated with metatropic dysplasia. Biochem Biophys Res Commun 2020; 523:841-846. [PMID: 31954514 DOI: 10.1016/j.bbrc.2019.12.123] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 12/30/2019] [Indexed: 11/20/2022]
Abstract
Metatropic dysplasia (MD) is a congenital skeletal dysplasia characterized by severe platyspondyly and dumbbell-like long-bone deformities. These skeletal phenotypes are predominantly caused by autosomal dominant gain-of-function (GOF) mutations in transient receptor potential vanilloid 4 (TRPV4), which encodes a nonselective Ca2+-permeable cation channel. Previous studies have shown that constitutive TRPV4 channel activation leads to irregular chondrogenic proliferation and differentiation, and thus to the disorganized endochondral ossification seen in MD. Therefore, the present study investigated the role of TRPV4 in osteoblast differentiation and MD pathogenesis. Specifically, the behavior of osteoblasts differentiated from patient-derived dental pulp stem cells carrying a heterozygous single base TRPV4 mutation, c.1855C > T (p.L619F) was compared to that of osteoblasts differentiated from isogenic control cells (in which the mutation was corrected using the CRISPR/Cas9 system). The mutant osteoblasts exhibited enhanced calcification (indicated by intense Alizarin Red S staining), increased intracellular Ca2+ levels, strongly upregulated runt-related transcription factor 2 and osteocalcin expression, and increased expression and nuclear translocation of nuclear factor-activated T cell c1 (NFATc1) compared to control cells. These results suggest that the analyzed TRPV4 GOF mutation disrupts osteoblastic differentiation and induces MD-associated disorganized endochondral ossification by increasing Ca2+/NFATc1 pathway activity. Thus, inhibiting the NFATc1 pathway may be a promising potential therapeutic strategy to attenuate skeletal deformities in MD.
Collapse
|
22
|
Chen MQ, Luan JJ. HMGB1 promotes bone fracture healing through activation of ERK signaling pathway in a rat tibial fracture model. Kaohsiung J Med Sci 2019; 35:550-558. [PMID: 31162822 DOI: 10.1002/kjm2.12095] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 05/12/2019] [Indexed: 01/04/2023] Open
Abstract
This work was to investigate potential roles of HMGB1-mediated ERK pathway in the healing process of bone fracture. Rat tibial fracture models were established and divided into control (rats with normal saline), HMGB1 (rats with HMGB1), and HMGB1+ PD98059 groups (rats with HMGB1 and 1 mg/kg of ERK1/2 inhibitor PD98059) with 30 rats per each. The healing of rats' fracture was observed by X-ray films, the morphological changes of bone fractures by HE staining, the callus formation by micro-CT and biomechanical test, and the expression of osteogenesis-related genes, HMGB1 and ERK-related proteins by qRT-PCR and Western blot. Rats in the HMGB1 group was increased in X-ray scores, peak torque, torsional stiffness, and the bone volume fraction (bone volume/total volume, BV/TV); meanwhile, those rats presented elevations in osteogenesis-related genes and HMGB1 expressions, as well as p-ERK/ERK ratio. However, rats in the HMGB1+ PD98059 group was significantly reduced in X-ray score, peak torque, torsional stiffness, and BV/TV, as well as the expression of osteogenesis-related genes and the ratio of p-ERK/ERK, as compared to those from HMGB1 group. HMGB1 could promote the expressions of osteogenesis-related genes and accelerate the healing process of fracture via activation of the ERK signaling pathway.
Collapse
Affiliation(s)
- Ming-Qi Chen
- Department of Traumatic Orthopedics, YanTaiShan Hospital, YanTai City, Shandong, China
| | - Jing-Jie Luan
- Department of Traumatic Orthopedics, YanTaiShan Hospital, YanTai City, Shandong, China
| |
Collapse
|
23
|
Nonaka K, Han X, Kato H, Sato H, Yamaza H, Hirofuji Y, Masuda K. Novel gain-of-function mutation of TRPV4 associated with accelerated chondrogenic differentiation of dental pulp stem cells derived from a patient with metatropic dysplasia. Biochem Biophys Rep 2019; 19:100648. [PMID: 31463371 PMCID: PMC6709385 DOI: 10.1016/j.bbrep.2019.100648] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 04/22/2019] [Accepted: 05/03/2019] [Indexed: 01/16/2023] Open
Abstract
Metatropic dysplasia is a congenital skeletal dysplasia characterized by severe platyspondyly, dumbbell-like deformity of long tubular bones, and progressive kyphoscoliosis with growth. It is caused by mutations in the gene TRPV4, encoding the transient receptor potential vanilloid 4, which acts as a calcium channel. Many heterozygous single base mutations of this gene have been associated with the disorder, showing autosomal dominant inheritance. Although abnormal endochondral ossification has been observed by histological examination of bone in a patient with lethal metatropic dysplasia, the etiology of the disorder remains largely unresolved. As dental pulp stem cells (DPSCs) are mesenchymal stem cells that differentiate into bone lineage cells, DPSCs derived from patients with congenital skeletal dysplasia might be useful as a disease-specific cellular model for etiological investigation. The purpose of this study was to clarify the pathological association between TRPV4 mutation and chondrocyte differentiation by analyzing DPSCs from a patient with non-lethal metatropic dysplasia. We identified a novel heterozygous single base mutation, c.1855C>T in TRPV4. This was predicted to be a missense mutation, p.L619F, in putative transmembrane segment 5. The mutation was repaired by CRISPR/Cas9 system to obtain isogenic control DPSCs for further analysis. The expression of stem cell markers and fibroblast-like morphology were comparable between patient-derived mutant and control DPSCs, although expression of TRPV4 was lower in mutant DPSCs than control DPSCs. Despite the lower TRPV4 expression in mutant DPSCs, the intracellular Ca2+ level was comparable at the basal level between mutant and control DPSCs, while its level was markedly higher following stimulation with 4α-phorbol 12,13-didecanoate (4αPDD), a specific agonist for TRPV4, in mutant DPSCs than in control DPSCs. In the presence of 4αPDD, we observed accelerated early chondrocyte differentiation and upregulated mRNA expression of SRY-box 9 (SOX9) in mutant DPSCs. Our findings suggested that the novel missense mutation c.1855C>T of TRPV4 was a gain-of-function mutation leading to enhanced intracellular Ca2+ level, which was associated with accelerated chondrocyte differentiation and SOX9 upregulation. Our results also suggest that patient-derived DPSCs can be a useful disease-specific cellular model for elucidating the pathological mechanism of metatropic dysplasia. Dental pulp stem cells from a patient with metatropic dysplasia were analyzed. A novel heterozygous single base mutation, c.1855C > T, was identified in TRPV4. The mutation was repaired by gene editing to obtain isogenic control cells. The intracellular Ca2+ level was enhanced in mutant cells by 4αPDD. Chondrocyte differentiation was enhanced in mutant cells by 4αPDD.
Collapse
Affiliation(s)
- Kentaro Nonaka
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Xu Han
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Hiroki Kato
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Hiroshi Sato
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Haruyoshi Yamaza
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Yuta Hirofuji
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Keiji Masuda
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
24
|
Hines SL, Richter JE, Mohammad AN, Mahim J, Atwal PS, Caulfield TR. Protein informatics combined with multiple data sources enriches the clinical characterization of novel TRPV4 variant causing an intermediate skeletal dysplasia. Mol Genet Genomic Med 2019; 7:e566. [PMID: 30693671 PMCID: PMC6418443 DOI: 10.1002/mgg3.566] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 11/02/2018] [Accepted: 12/02/2018] [Indexed: 01/19/2023] Open
Abstract
Background Transient receptor potential cation channel subfamily V member 4 (TRPV4) is an ion channel permeable to Ca2+ that is sensitive to physical, hormonal, and chemical stimuli. This protein is expressed in many cell types, including osteoclasts, chondrocytes, and sensory neurons. As such, pathogenic variants of this gene are associated with skeletal dysplasias and neuromuscular disorders. Pathogenesis of these phenotypes is not yet completely understood, but it is known that genotype–phenotype correlations for TRPV4 pathogenic variants often are not present. Methods Newly characterized, suspected pathogenic variant in TRPV4 was analyzed using protein informatics and personalized protein‐level molecular studies, genomic exome analysis, and clinical study. Results This statement is demonstrated in the family of our proband, a 47‐year‐old female having the novel c.2401A>G (p.K801E) variant of TRPV4. We discuss the common symptoms between the proband, her father, and her daughter, and compare her phenotype to known TRPV4‐associated skeletal dysplasias. Conclusions Protein informatics and molecular modeling are used to confirm the pathogenicity of the unique TRPV4 variant found in this family. Multiple data were combined in a comprehensive manner to give complete overall perspective on the patient disease and prognosis.
Collapse
Affiliation(s)
- Stephanie L Hines
- Department of Clinical Genomics, Mayo Clinic, Jacksonville, Florida.,Department of General Internal Medicine, Mayo Clinic, Jacksonville, Florida.,Center for Individualized Medicine, Mayo Clinic, Jacksonville, Florida
| | - John E Richter
- Department of Clinical Genomics, Mayo Clinic, Jacksonville, Florida
| | - Ahmed N Mohammad
- Department of Clinical Genomics, Mayo Clinic, Jacksonville, Florida
| | - Jain Mahim
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | - Thomas R Caulfield
- Center for Individualized Medicine, Mayo Clinic, Jacksonville, Florida.,Department of Neuroscience, Mayo Clinic, Jacksonville, Florida.,Mayo Graduate School, Neurobiology of Disease, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
25
|
Saitta B, Elphingstone J, Limfat S, Shkhyan R, Evseenko D. CaMKII inhibition in human primary and pluripotent stem cell-derived chondrocytes modulates effects of TGFβ and BMP through SMAD signaling. Osteoarthritis Cartilage 2019; 27:158-171. [PMID: 30205161 PMCID: PMC6309757 DOI: 10.1016/j.joca.2018.08.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 08/15/2018] [Accepted: 08/16/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Upregulation of calcium/calmodulin-dependent kinase II (CaMKII) is implicated in the pathogenesis of osteoarthritis (OA) and reactivation of articular cartilage hypertrophy. However, direct inhibition of CaMKII unexpectedly augmented symptoms of OA in animal models. The role of CaMKII in OA remains unclear and requires further investigation. METHODS Analysis of CaMKII expression was performed in normal human and OA articular chondrocytes, and signaling mechanisms were assessed in articular, fetal and Pluripotent Stem Cell (PSC)-derived human chondrocytes using pharmacological (KN93), peptide (AC3-I) and small interfering RNA (siRNA) inhibitors of CaMKII. RESULTS Expression levels of phospho-CaMKII (pCaMKII) were significantly and consistently increased in human OA specimens. BMP2/4 activated expression of pCaMKII as well as COLII and COLX in human adult articular chondrocytes, and also increased the levels and nuclear localization of SMADs1/5/8, while TGFβ1 showed minimal or no activation of the chondrogenic program in adult chondrocytes. Targeted blockade of CaMKII with specific siRNAs decreased levels of pSMADs, COLII, COLX and proteoglycans in normal and OA adult articular chondrocytes in the presence of both BMP4 and TGFβ1. Both human fetal and PSC-derived chondrocytes also demonstrated a decrease of chondrogenic differentiation in the presence of small molecule and peptide inhibitors of CaMKII. Furthermore, immunoprecipitation for SMADs1/5/8 or 2/3 followed by western blotting for pCaMKII showed direct interaction between SMADs and pCaMKII in primary chondrocytes. CONCLUSION Current study demonstrates a direct role for CaMKII in TGF-β and BMP-mediated responses in primary and PSC-derived chondrocytes. These findings have direct implications for tissue engineering of cartilage tissue from stem cells and therapeutic management of OA.
Collapse
Affiliation(s)
- Biagio Saitta
- Departments of Orthopaedic Surgery, University of Southern California, Los Angeles, CA, 90033, USA,Medicine Div. of Nephrology and Hypertension, University of Southern California, Los Angeles, CA, 90033, USA
| | - Joseph Elphingstone
- Departments of Orthopaedic Surgery, University of Southern California, Los Angeles, CA, 90033, USA
| | - Sean Limfat
- Departments of Orthopaedic Surgery, University of Southern California, Los Angeles, CA, 90033, USA
| | - Ruzanna Shkhyan
- Departments of Orthopaedic Surgery, University of Southern California, Los Angeles, CA, 90033, USA
| | - Denis Evseenko
- Departments of Orthopaedic Surgery, University of Southern California, Los Angeles, CA, 90033, USA,Stem Cell Research and Regenerative Medicine Keck School of Medicine of University of Southern California, Los Angeles, CA, 90033, USA,Corresponding Author:Denis Evseenko MD, PhD., Associate Professor of Orthopaedic Surgery, Stem Cell Research and Regenerative Medicine, Keck School of Medicine of USC, 1450 Biggy St, NRT 4509, Los Angeles, CA 90033,
| |
Collapse
|
26
|
Jin YZ, Lee JH. Mesenchymal Stem Cell Therapy for Bone Regeneration. Clin Orthop Surg 2018; 10:271-278. [PMID: 30174801 PMCID: PMC6107811 DOI: 10.4055/cios.2018.10.3.271] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/27/2018] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been used in clinic for approximately 20 years. During this period, various new populations of MSCs have been found or manipulated. However, their characters and relative strength for bone regeneration have not been well known. For a comprehensive understanding of MSCs, we reviewed the literature on the multipotent cells ranging from the definition to the current research progress for bone regeneration. Based on our literature review, bone marrow MSCs have been most widely studied and utilized in clinical settings. Among other populations of MSCs, adipose-derived MSCs and perivascular MSCs might be potential candidates for bone regeneration, whose efficacy and safety still require further investigation.
Collapse
Affiliation(s)
- Yuan-Zhe Jin
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Jae Hyup Lee
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Korea.,Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul, Korea.,Institute of Medical and Biological Engineering, Seoul National University Medical Research Center, Seoul, Korea
| |
Collapse
|
27
|
Current Therapeutic Strategies for Stem Cell-Based Cartilage Regeneration. Stem Cells Int 2018; 2018:8490489. [PMID: 29765426 PMCID: PMC5889878 DOI: 10.1155/2018/8490489] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 12/14/2017] [Accepted: 01/23/2018] [Indexed: 12/13/2022] Open
Abstract
The process of cartilage destruction in the diarthrodial joint is progressive and irreversible. This destruction is extremely difficult to manage and frustrates researchers, clinicians, and patients. Patients often take medication to control their pain. Surgery is usually performed when pain becomes uncontrollable or joint function completely fails. There is an unmet clinical need for a regenerative strategy to treat cartilage defect without surgery due to the lack of a suitable regenerative strategy. Clinicians and scientists have tried to address this using stem cells, which have a regenerative potential in various tissues. Cartilage may be an ideal target for stem cell treatment because it has a notoriously poor regenerative potential. In this review, we describe past, present, and future strategies to regenerate cartilage in patients. Specifically, this review compares a surgical regenerative technique (microfracture) and cell therapy, cell therapy with and without a scaffold, and therapy with nonaggregated and aggregated cells. We also review the chondrogenic potential of cells according to their origin, including autologous chondrocytes, mesenchymal stem cells, and induced pluripotent stem cells.
Collapse
|
28
|
Csobonyeiova M, Polak S, Zamborsky R, Danisovic L. iPS cell technologies and their prospect for bone regeneration and disease modeling: A mini review. J Adv Res 2017; 8:321-327. [PMID: 28386481 PMCID: PMC5374850 DOI: 10.1016/j.jare.2017.02.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/24/2017] [Accepted: 02/25/2017] [Indexed: 12/27/2022] Open
Abstract
Bone disorders are a group of varied acute and chronic traumatic, degenerative, malignant or congenital conditions affecting the musculoskeletal system. They are prevalent in society and, with an ageing population, the incidence and impact on the population's health is growing. Severe persisting pain and limited mobility are the major symptoms of the disorder that impair the quality of life in affected patients. Current therapies only partially treat the disorders, offering management of symptoms, or temporary replacement with inert materials. However, during the last few years, the options for the treatment of bone disorders have greatly expanded, thanks to the advent of regenerative medicine. Skeletal cell-based regeneration medicine offers promising reparative therapies for patients. Mesenchymal stem (stromal) cells from different tissues have been gradually translated into clinical practice; however, there are a number of limitations. The introduction of reprogramming methods and the subsequent production of induced pluripotent stem cells provides a possibility to create human-specific models of bone disorders. Furthermore, human-induced pluripotent stem cell-based autologous transplantation is considered to be future breakthrough in the field of regenerative medicine. The main goal of the present paper is to review recent applications of induced pluripotent stem cells in bone disease modeling and to discuss possible future therapy options. The present article contributes to the dissemination of scientific and pre-clinical results between physicians, mainly orthopedist and thus supports the translation to clinical practice.
Collapse
Affiliation(s)
- Maria Csobonyeiova
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Stefan Polak
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Radoslav Zamborsky
- Department of Orthopaedics, Faculty of Medicine, Comenius University and Children’s University Hospital, Limbova 1, 831 01 Bratislava, Slovakia
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
- Regenmed Ltd., Medena 29, 811 02 Bratislava, Slovakia
| |
Collapse
|
29
|
The potential of induced pluripotent stem cells as a tool to study skeletal dysplasias and cartilage-related pathologic conditions. Osteoarthritis Cartilage 2017; 25:616-624. [PMID: 27919783 DOI: 10.1016/j.joca.2016.11.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 11/10/2016] [Accepted: 11/28/2016] [Indexed: 02/07/2023]
Abstract
The development of induced pluripotent stem cells (iPSCs) technology has opened up new horizons for development of new research tools especially for skeletal dysplasias, which often lack human disease models. Regenerative medicine and tissue engineering could be the next areas to benefit from refinement of iPSC methods to repair focal cartilage defects, while applications for osteoarthritis (OA) and drug screening have evolved rather slowly. Although the advances in iPSC research of skeletal dysplasias and repair of focal cartilage lesions are not directly relevant to OA, they can be considered to pave the way to future prospects and solutions to OA research, too. The same problems which face the present cell-based treatments of cartilage injuries concern also the iPSC-based ones. However, established iPSC lines, which have no genomic aberrations and which efficiently differentiate into extracellular matrix secreting chondrocytes, could be an invaluable cell source for cell transplantations in the future. The safety issues concerning the recipient risks of teratoma formation and immune response still have to be solved before the potential use of iPSCs in cartilage repair of focal cartilage defects and OA.
Collapse
|
30
|
Grace MS, Bonvini SJ, Belvisi MG, McIntyre P. Modulation of the TRPV4 ion channel as a therapeutic target for disease. Pharmacol Ther 2017; 177:9-22. [PMID: 28202366 DOI: 10.1016/j.pharmthera.2017.02.019] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Transient Receptor Potential Vanilloid 4 (TRPV4) is a broadly expressed, polymodally gated ion channel that plays an important role in many physiological and pathophysiological processes. TRPV4 knockout mice and several synthetic pharmacological compounds that selectively target TRPV4 are now available, which has allowed detailed investigation in to the therapeutic potential of this ion channel. Results from animal studies suggest that TRPV4 antagonism has therapeutic potential in oedema, pain, gastrointestinal disorders, and lung diseases such as cough, bronchoconstriction, pulmonary hypertension, and acute lung injury. A lack of observed side-effects in vivo has prompted a first-in-human trial for a TRPV4 antagonist in healthy participants and stable heart failure patients. If successful, this would open up an exciting new area of research for a multitude of TRPV4-related pathologies. This review will discuss the known roles of TRPV4 in disease, and highlight the possible implications of targeting this important cation channel for therapy.
Collapse
Affiliation(s)
- Megan S Grace
- Baker Heart and Diabetes Institute, Melbourne, Australia; School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Australia; Department of Physiology, School of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia.
| | - Sara J Bonvini
- Respiratory Pharmacology, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Maria G Belvisi
- Respiratory Pharmacology, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Peter McIntyre
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Australia
| |
Collapse
|
31
|
Diederichs S, Gabler J, Autenrieth J, Kynast KL, Merle C, Walles H, Utikal J, Richter W. Differential Regulation of SOX9 Protein During Chondrogenesis of Induced Pluripotent Stem Cells Versus Mesenchymal Stromal Cells: A Shortcoming for Cartilage Formation. Stem Cells Dev 2016; 25:598-609. [DOI: 10.1089/scd.2015.0312] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Solvig Diederichs
- Research Center for Experimental Orthopedics, Orthopedic University Hospital Heidelberg, Heidelberg, Germany
| | - Jessica Gabler
- Research Center for Experimental Orthopedics, Orthopedic University Hospital Heidelberg, Heidelberg, Germany
| | - Jennifer Autenrieth
- Research Center for Experimental Orthopedics, Orthopedic University Hospital Heidelberg, Heidelberg, Germany
| | - Katharina L. Kynast
- Research Center for Experimental Orthopedics, Orthopedic University Hospital Heidelberg, Heidelberg, Germany
| | - Christian Merle
- Clinic for Orthopedics and Trauma Surgery, Orthopedic University Hospital Heidelberg, Heidelberg, Germany
| | - Heike Walles
- Department Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Würzburg, Germany
- Fraunhofer Institute for Interfacial Engineering and Biotechnology, Translational Center Würzburg ‘Regenerative Therapies in Oncology and Musculoskeletal Diseases’–Würzburg Branch, Würzburg, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology, and Allergology, University Medical Center Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Wiltrud Richter
- Research Center for Experimental Orthopedics, Orthopedic University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
32
|
An Overview of Direct Somatic Reprogramming: The Ins and Outs of iPSCs. Int J Mol Sci 2016; 17:ijms17010141. [PMID: 26805822 PMCID: PMC4730380 DOI: 10.3390/ijms17010141] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/12/2016] [Accepted: 01/13/2016] [Indexed: 02/07/2023] Open
Abstract
Stem cells are classified into embryonic stem cells and adult stem cells. An evolving alternative to conventional stem cell therapies is induced pluripotent stem cells (iPSCs), which have a multi-lineage potential comparable to conventionally acquired embryonic stem cells with the additional benefits of being less immunoreactive and avoiding many of the ethical concerns raised with the use of embryonic material. The ability to generate iPSCs from somatic cells provides tremendous promise for regenerative medicine. The breakthrough of iPSCs has raised the possibility that patient-specific iPSCs can provide autologous cells for cell therapy without the concern for immune rejection. iPSCs are also relevant tools for modeling human diseases and drugs screening. However, there are still several hurdles to overcome before iPSCs can be used for translational purposes. Here, we review the recent advances in somatic reprogramming and the challenges that must be overcome to move this strategy closer to clinical application.
Collapse
|
33
|
Nejadnik H, Diecke S, Lenkov OD, Chapelin F, Donig J, Tong X, Derugin N, Chan RCF, Gaur A, Yang F, Wu JC, Daldrup-Link HE. Improved approach for chondrogenic differentiation of human induced pluripotent stem cells. Stem Cell Rev Rep 2016; 11:242-53. [PMID: 25578634 PMCID: PMC4412587 DOI: 10.1007/s12015-014-9581-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human induced pluripotent stem cells (hiPSCs) have demonstrated great potential for hyaline cartilage regeneration. However, current approaches for chondrogenic differentiation of hiPSCs are complicated and inefficient primarily due to intermediate embryoid body formation, which is required to generate endodermal, ectodermal, and mesodermal cell lineages. We report a new, straightforward and highly efficient approach for chondrogenic differentiation of hiPSCs, which avoids embryoid body formation. We differentiated hiPSCs directly into mesenchymal stem /stromal cells (MSC) and chondrocytes. hiPSC-MSC-derived chondrocytes showed significantly increased Col2A1, GAG, and SOX9 gene expression compared to hiPSC-MSCs. Following transplantation of hiPSC-MSC and hiPSC-MSC-derived chondrocytes into osteochondral defects of arthritic joints of athymic rats, magnetic resonance imaging studies showed gradual engraftment, and histological correlations demonstrated hyaline cartilage matrix production. Results present an efficient and clinically translatable approach for cartilage tissue regeneration via patient-derived hiPSCs, which could improve cartilage regeneration outcomes in arthritic joints.
Collapse
Affiliation(s)
- Hossein Nejadnik
- Department of Radiology, and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA, 94304, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Salazar-Noratto GE, Barry FP, Guldberg RE. Application of biomaterials to in vitro pluripotent stem cell disease modeling of the skeletal system. J Mater Chem B 2016; 4:3482-3489. [DOI: 10.1039/c5tb02645h] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Disease-specific pluripotent stem cells can be derived through genetic manipulation of embryonic stem cells or by reprogramming somatic cells (induced pluripotent stem cells).
Collapse
Affiliation(s)
- Giuliana E. Salazar-Noratto
- Wallace H. Coulter Department of Biomedical Engineering
- Georgia Institute of Technology and Emory University
- Atlanta
- USA
- Parker H. Petit Institute for Bioengineering and Bioscience
| | - Frank P. Barry
- Regenerative Medicine Institute
- National University of Ireland Galway
- Biosciences
- Dangan
- Ireland
| | - Robert E. Guldberg
- Parker H. Petit Institute for Bioengineering and Bioscience
- Georgia Institute of Technology
- Atlanta
- USA
- Woodruff School of Mechanical Engineering
| |
Collapse
|
35
|
Induced Pluripotency and Gene Editing in Disease Modelling: Perspectives and Challenges. Int J Mol Sci 2015; 16:28614-34. [PMID: 26633382 PMCID: PMC4691066 DOI: 10.3390/ijms161226119] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 11/13/2015] [Accepted: 11/24/2015] [Indexed: 02/07/2023] Open
Abstract
Embryonic stem cells (ESCs) are chiefly characterized by their ability to self-renew and to differentiate into any cell type derived from the three main germ layers. It was demonstrated that somatic cells could be reprogrammed to form induced pluripotent stem cells (iPSCs) via various strategies. Gene editing is a technique that can be used to make targeted changes in the genome, and the efficiency of this process has been significantly enhanced by recent advancements. The use of engineered endonucleases, such as homing endonucleases, zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs) and Cas9 of the CRISPR system, has significantly enhanced the efficiency of gene editing. The combination of somatic cell reprogramming with gene editing enables us to model human diseases in vitro, in a manner considered superior to animal disease models. In this review, we discuss the various strategies of reprogramming and gene targeting with an emphasis on the current advancements and challenges of using these techniques to model human diseases.
Collapse
|
36
|
Tong S, Xue L, Xu DP, Liu ZM, Du Y, Wang XK. In vitro culture of hFOB1.19 osteoblast cells on TGF-β1-SF-CS three-dimensional scaffolds. Mol Med Rep 2015; 13:181-7. [PMID: 26530112 PMCID: PMC4686111 DOI: 10.3892/mmr.2015.4498] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 10/06/2015] [Indexed: 01/08/2023] Open
Abstract
The aim of the present study was to examine the biocompatibility of transforming growth factor-β1-silk fibroin-chitosan (TGF-β1-SF-CS) scaffolds. In order to provide an ideal scaffold for use in bone tissue engineering, TGF-β1 was introduced into the SF-CS scaffold in order to reconstruct a three dimensional scaffold, following which hFOB1.19 osteoblast cells were seeded onto TGF-β1-SF-CS and SF-CS scaffolds. On the TGF-β1-SF-CS and SF-CS scaffolds, the cell adhesion rate increased in a time-dependent manner. Scanning electron microscopy revealed that the cells grew actively and exhibited normal morphological features with multiple fissions, and granular and filamentous substrates were observed surrounding the cells. In addition, the cell microfilaments were closely connected with the scaffolds. The cells exhibited attached growth on the surfaces of the scaffolds, however, the growth also extended into the scaffolds. Cell Counting Kit-8 and ALP analyses revealed that TGF-β1 significantly promoted the growth and proliferation of the hFOB1.19 osteoblast cells in the SF-CS scaffolds, and the enhancement of osteoblast cell proliferation and activity by TGF-β1 occurred in a time-dependent manner. The TGF-β1-SF-CS composite material may offer potential as an ideal scaffold material for bone tissue engineering.
Collapse
Affiliation(s)
- Shuang Tong
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Liaoning Institute of Dental Research, Shenyang, Liaoning 110002, P.R. China
| | - Lei Xue
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Liaoning Institute of Dental Research, Shenyang, Liaoning 110002, P.R. China
| | - Da-Peng Xu
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Liaoning Institute of Dental Research, Shenyang, Liaoning 110002, P.R. China
| | - Zi-Mei Liu
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Liaoning Institute of Dental Research, Shenyang, Liaoning 110002, P.R. China
| | - Yang Du
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Liaoning Institute of Dental Research, Shenyang, Liaoning 110002, P.R. China
| | - Xu-Kai Wang
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Liaoning Institute of Dental Research, Shenyang, Liaoning 110002, P.R. China
| |
Collapse
|
37
|
Abstract
Introduction: Genetic skeletal diseases (GSDs) are a diverse and complex group of rare genetic conditions that affect the development and homeostasis of the skeleton. Although individually rare, as a group of related diseases, GSDs have an overall prevalence of at least 1 per 4,000 children. There are currently very few specific therapeutic interventions to prevent, halt or modify skeletal disease progression and therefore the generation of new and effective treatments requires novel and innovative research that can identify tractable therapeutic targets and biomarkers of these diseases. Areas covered: Remarkable progress has been made in identifying the genetic basis of the majority of GSDs and in developing relevant model systems that have delivered new knowledge on disease mechanisms and are now starting to identify novel therapeutic targets. This review will provide an overview of disease mechanisms that are shared amongst groups of different GSDs and describe potential therapeutic approaches that are under investigation. Expert opinion: The extensive clinical variability and genetic heterogeneity of GSDs renders this broad group of rare diseases a bench to bedside challenge. However, the evolving hypothesis that clinically different diseases might share common disease mechanisms is a powerful concept that will generate critical mass for the identification and validation of novel therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Michael D Briggs
- Newcastle University, Institute of Genetic Medicine, International Centre for Life , Central Parkway, Newcastle-upon-Tyne, NE1 3BZ, UK
| | - Peter A Bell
- Newcastle University, Institute of Genetic Medicine, International Centre for Life , Newcastle-upon-Tyne, NE1 3BZ, UK
| | - Michael J Wright
- Newcastle University, Institute of Genetic Medicine, International Centre for Life , Newcastle-upon-Tyne, NE1 3BZ, UK
| | - Katarzyna A Pirog
- Newcastle University, Institute of Genetic Medicine, International Centre for Life , Newcastle-upon-Tyne, NE1 3BZ, UK
| |
Collapse
|
38
|
McNulty AL, Leddy HA, Liedtke W, Guilak F. TRPV4 as a therapeutic target for joint diseases. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2015; 388:437-50. [PMID: 25519495 PMCID: PMC4361386 DOI: 10.1007/s00210-014-1078-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 12/04/2014] [Indexed: 02/07/2023]
Abstract
Biomechanical factors play a critical role in regulating the physiology as well as the pathology of multiple joint tissues and have been implicated in the pathogenesis of osteoarthritis. Therefore, the mechanisms by which cells sense and respond to mechanical signals may provide novel targets for the development of disease-modifying osteoarthritis drugs (DMOADs). Transient receptor potential vanilloid 4 (TRPV4) is a Ca(2+)-permeable cation channel that serves as a sensor of mechanical or osmotic signals in several musculoskeletal tissues, including cartilage, bone, and synovium. The importance of TRPV4 in joint homeostasis is apparent in patients harboring TRPV4 mutations, which result in the development of a spectrum of skeletal dysplasias and arthropathies. In addition, the genetic knockout of Trpv4 results in the development of osteoarthritis and decreased osteoclast function. In engineered cartilage replacements, chemical activation of TRPV4 can reproduce many of the anabolic effects of mechanical loading to accelerate tissue growth and regeneration. Overall, TRPV4 plays a key role in transducing mechanical, pain, and inflammatory signals within joint tissues and thus is an attractive therapeutic target to modulate the effects of joint diseases. In pathological conditions in the joint, when the delicate balance of TRPV4 activity is altered, a variety of different tools could be utilized to directly or indirectly target TRPV4 activity.
Collapse
Affiliation(s)
- Amy L. McNulty
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710
| | - Holly A. Leddy
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710
| | - Wolfgang Liedtke
- Department of Neurology and Duke University Clinics for Pain and Palliative Care, Duke University Medical Center, Durham, NC 27710
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
39
|
Riminucci M, Remoli C, Robey PG, Bianco P. Stem cells and bone diseases: new tools, new perspective. Bone 2015; 70:55-61. [PMID: 25240458 PMCID: PMC5524373 DOI: 10.1016/j.bone.2014.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 09/03/2014] [Accepted: 09/09/2014] [Indexed: 12/13/2022]
Abstract
Postnatal skeletal stem cells are a unique class of progenitors with biological properties that extend well beyond the limits of stemness as commonly defined. Skeletal stem cells sustain skeletal tissue homeostasis, organize and maintain the complex architectural structure of the bone marrow microenvironment and provide a niche for hematopoietic progenitor cells. The identification of stem cells in the human post-natal skeleton has profoundly changed our approach to the physiology and pathology of this system. Skeletal diseases have been long interpreted essentially in terms of defective function of differentiated cells and/or abnormal turnover of the matrix that they produce. The notion of a skeletal stem cell has brought forth multiple, novel concepts in skeletal biology that provide potential alternative concepts. At the same time, the recognition of the complex functions played by skeletal progenitors, such as the structural and functional organization of the bone marrow, has provided an innovative, unifying perspective for understanding bone and bone marrow changes simultaneously occurring in many disorders. Finally, the possibility to isolate and highly enrich for skeletal progenitors, enables us to reproduce perfectly normal or pathological organ miniatures. These, in turn, provide suitable models to investigate and manipulate the pathogenetic mechanisms of many genetic and non-genetic skeletal diseases. This article is part of a Special Issue entitled Stem cells and Bone.
Collapse
Affiliation(s)
- Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, Italy.
| | - Cristina Remoli
- Department of Molecular Medicine, Sapienza University of Rome, Italy
| | - Pamela G Robey
- Craniofacial and Skeletal Diseases Branch, National Institute of Craniofacial and Dental Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Paolo Bianco
- Department of Molecular Medicine, Sapienza University of Rome, Italy
| |
Collapse
|
40
|
Leddy HA, McNulty AL, Guilak F, Liedtke W. Unraveling the mechanism by which TRPV4 mutations cause skeletal dysplasias. Rare Dis 2014; 2:e962971. [PMID: 26942100 PMCID: PMC4755236 DOI: 10.4161/2167549x.2014.962971] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 09/03/2014] [Indexed: 01/02/2023] Open
Abstract
Transient Receptor Potential Vanilloid 4 (TRPV4) is a mechano- and osmosensitive cation channel that is highly expressed in chondrocytes, the cells in cartilage. A large number of mutations in TRPV4 have been linked to skeletal dysplasias, and the goal of this addendum is to shed light on the mechanisms by which mutations in TRPV4 can cause skeletal dysplasias by focusing on 3 recent publications. These papers suggest that skeletal dysplasia-causing TRPV4 mutations reprogram chondrocytes to increase follistatin production, which inhibits BMP signaling, thus slowing the process of endochondral ossification and leading to skeletal dysplasia. In spite of these important advances in our understanding of the disease mechanism, much remains to be elucidated. Nonetheless, these new data suggest that inhibiting aberrant TRPV4 activity in the cartilage may be a promising direction for therapeutic intervention.
Collapse
Affiliation(s)
- Holly A Leddy
- Department of orthopedic Surgery; Duke University Medical Center ; Durham, NC USA
| | - Amy L McNulty
- Department of orthopedic Surgery; Duke University Medical Center ; Durham, NC USA
| | - Farshid Guilak
- Department of orthopedic Surgery; Duke University Medical Center ; Durham, NC USA
| | - Wolfgang Liedtke
- Department of Neurology and Duke University Clinics for Pain and Palliative Care; Duke University Medical Center ; Durham, NC USA
| |
Collapse
|
41
|
Patterson SE, Dealy CN. Mechanisms and models of endoplasmic reticulum stress in chondrodysplasia. Dev Dyn 2014; 243:875-93. [DOI: 10.1002/dvdy.24131] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 03/10/2014] [Accepted: 03/17/2014] [Indexed: 12/14/2022] Open
Affiliation(s)
- Sara E. Patterson
- Center for Regenerative Medicine and Skeletal Development; Department of Reconstructive Sciences; University of Connecticut Health Center; Farmington Connecticut
| | - Caroline N. Dealy
- Center for Regenerative Medicine and Skeletal Development; Department of Reconstructive Sciences; University of Connecticut Health Center; Farmington Connecticut
- Center for Regenerative Medicine and Skeletal Development; Department of Orthopedic Surgery; University of Connecticut Health Center; Farmington Connecticut
| |
Collapse
|