1
|
Zhao F, Qiu Y, Liu W, Zhang Y, Liu J, Bian L, Shao L. Biomimetic Hydrogels as the Inductive Endochondral Ossification Template for Promoting Bone Regeneration. Adv Healthc Mater 2024; 13:e2303532. [PMID: 38108565 DOI: 10.1002/adhm.202303532] [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: 10/14/2023] [Revised: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Repairing critical size bone defects (CSBD) is a major clinical challenge and requires effective intervention by biomaterial scaffolds. Inspired by the fact that the cartilaginous template-based endochondral ossification (ECO) process is crucial to bone healing and development, developing biomimetic biomaterials to promote ECO is recognized as a promising approach for repairing CSBD. With the unique highly hydrated 3D polymeric network, hydrogels can be designed to closely emulate the physiochemical properties of cartilage matrix to facilitate ECO. In this review, the various preparation methods of hydrogels possessing the specific physiochemical properties required for promoting ECO are introduced. The materiobiological impacts of the physicochemical properties of hydrogels, such as mechanical properties, topographical structures and chemical compositions on ECO, and the associated molecular mechanisms related to the BMP, Wnt, TGF-β, HIF-1α, FGF, and RhoA signaling pathways are further summarized. This review provides a detailed coverage on the materiobiological insights required for the design and preparation of hydrogel-based biomaterials to facilitate bone regeneration.
Collapse
Affiliation(s)
- Fujian Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Yonghao Qiu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Wenjing Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Yanli Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Jia Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Longquan Shao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, P. R. China
| |
Collapse
|
2
|
Liu N, Zheng Q, Zhang Y, Wang H, Zhang Z, He L, Wei C, Xia H, Liu Y, Wang X. Hypoxia differently regulates the proportion of ALDH hi cells in lung squamous carcinoma H520 and adenocarcinoma A549 cells via the Wnt/β-catenin pathway. Thorac Cancer 2024; 15:1419-1428. [PMID: 38736300 PMCID: PMC11194122 DOI: 10.1111/1759-7714.15328] [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: 02/06/2024] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are a specific subpopulation of cancer cells with the ability of self-renewal, infinite proliferation, multidifferentiation and tumorigenicity, and play critical roles in cancer progression and treatment resistance. CSCs are tightly regulated by the tumor microenvironment, such as hypoxia; however, how hypoxia regulates CSCs in non-small cell lung cancer (NSCLC) remains unclear. METHODS The proportion of ALDHhi cells was examined using the Aldefluor assay. Tankyrase inhibitor XAV939 and siRNA were used to inhibit β-catenin while pcDNA3-β-catenin (S33Y) plasmid enhanced the expression of β-catenin. Western blot was administered for protein detection. The mRNA expression was measured by quantitative real-time PCR. RESULTS We found that hypoxia led to an increase in the proportion of ALDHhi cells in lung squamous carcinoma (LUSC) H520 cells, while causing a decrease in the ALDHhi cell proportion in lung adenocarcinoma (LUAD) A549 cells. Similarly, β-catenin expression was upregulated in H520 cells but downregulated in A549 cells upon exposure to hypoxia. Mechanically, the proportion of ALDHhi cells in both cell lines was decreased by β-catenin inhibitor or siRNA knockdown, whereas increased after β-catenin overexpression. Furthermore, hypoxia treatment suppressed E-cadherin expression in H520 cells and enhanced N-cadherin and β-catenin expression, while this effect was completely opposite in A549 cells. CONCLUSION The hypoxia-EMT-β-catenin axis functions as an important regulator for the proportion of CSCs in NSCLC and could potentially be explored as therapeutic targets in the future.
Collapse
Affiliation(s)
- Ni Liu
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
| | - Qi Zheng
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
| | - Yuqing Zhang
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
| | - Huimin Wang
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
- Department of OncologyYantai Affiliated Hospital of Binzhou Medical UniversityYantaiChina
| | - Zhihui Zhang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Long He
- Department of OncologyThe Third Hospital of JinanJinanChina
| | - Chenxi Wei
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
| | - Handai Xia
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
| | - Yanguo Liu
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
| | - Xiuwen Wang
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
| |
Collapse
|
3
|
Zhang Y, Li G, Wang J, Zhou F, Ren X, Su J. Small Joint Organoids 3D Bioprinting: Construction Strategy and Application. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2302506. [PMID: 37814373 DOI: 10.1002/smll.202302506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 09/28/2023] [Indexed: 10/11/2023]
Abstract
Osteoarthritis (OA) is a chronic disease that causes pain and disability in adults, affecting ≈300 million people worldwide. It is caused by damage to cartilage, including cellular inflammation and destruction of the extracellular matrix (ECM), leading to limited self-repairing ability due to the lack of blood vessels and nerves in the cartilage tissue. Organoid technology has emerged as a promising approach for cartilage repair, but constructing joint organoids with their complex structures and special mechanisms is still challenging. To overcome these boundaries, 3D bioprinting technology allows for the precise design of physiologically relevant joint organoids, including shape, structure, mechanical properties, cellular arrangement, and biological cues to mimic natural joint tissue. In this review, the authors will introduce the biological structure of joint tissues, summarize key procedures in 3D bioprinting for cartilage repair, and propose strategies for constructing joint organoids using 3D bioprinting. The authors also discuss the challenges of using joint organoids' approaches and perspectives on their future applications, opening opportunities to model joint tissues and response to joint disease treatment.
Collapse
Affiliation(s)
- Yuan Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Guangfeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- Department of Trauma Orthopedics, Zhongye Hospital, Shanghai, 200941, China
| | - Jian Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Fengjin Zhou
- Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China
| | - Xiaoxiang Ren
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
4
|
Yoo S, Choi S, Kim I, Kim IS. Hypoxic regulation of extracellular vesicles: Implications for cancer therapy. J Control Release 2023; 363:201-220. [PMID: 37739015 DOI: 10.1016/j.jconrel.2023.09.034] [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: 03/01/2023] [Revised: 08/18/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
Extracellular vesicles (EVs) play a pivotal role in intercellular communication and have been implicated in cancer progression. Hypoxia, a pervasive hallmark of cancer, is known to regulate EV biogenesis and function. Hypoxic EVs contain a specific set of proteins, nucleic acids, lipids, and metabolites, capable of reprogramming the biology and fate of recipient cells. Enhancing the intrinsic therapeutic efficacy of EVs can be achieved by strategically modifying their structure and contents. Moreover, the use of EVs as drug delivery vehicles holds great promise for cancer treatment. However, various hurdles must be overcome to enable their clinical application as cancer therapeutics. In this review, we aim to discuss the current knowledge on the hypoxic regulation of EVs. Additionally, we will describe the underlying mechanisms by which EVs contribute to cancer progression in hypoxia and outline the progress and limitations of hypoxia-related EV therapeutics for cancer.
Collapse
Affiliation(s)
- Seongkyeong Yoo
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, South Korea; Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, South Korea
| | - Sanga Choi
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, South Korea; Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, South Korea
| | - Iljin Kim
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, South Korea; Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, South Korea.
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, South Korea; Chemical and Biological Integrative Research Center, Biomedical Research Institute, Korea Institute Science and Technology, Seoul 02792, South Korea.
| |
Collapse
|
5
|
Stampoultzis T, Guo Y, Nasrollahzadeh N, Rana VK, Karami P, Pioletti DP. Low-oxygen tension augments chondrocyte sensitivity to biomimetic thermomechanical cues in cartilage-engineered constructs. iScience 2023; 26:107491. [PMID: 37599834 PMCID: PMC10432199 DOI: 10.1016/j.isci.2023.107491] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/14/2023] [Accepted: 07/23/2023] [Indexed: 08/22/2023] Open
Abstract
Chondrocytes respond to various biophysical cues, including oxygen tension, transient thermal signals, and mechanical stimuli. However, understanding how these factors interact to establish a unique regulatory microenvironment for chondrocyte function remains unclear. Herein, we explore these interactions using a joint-simulating bioreactor that independently controls the culture's oxygen concentration, evolution of temperature, and mechanical loading. Our analysis revealed significant coupling between these signals, resulting in a remarkable ∼14-fold increase in collagen type II (COL2a) and aggrecan (ACAN) mRNA expression. Furthermore, dynamic thermomechanical stimulation enhanced glycosaminoglycan and COL2a protein synthesis, with the magnitude of the biosynthetic changes being oxygen dependent. Additionally, our mechanistic study highlighted the crucial role of SRY-box transcription factor 9 (SOX9) as a major regulator of chondrogenic response, specifically expressed in response to combined biophysical signals. These findings illuminate the integration of various mechanobiological cues by chondrocytes and provide valuable insights for improving the extracellular matrix content in cartilage-engineered constructs.
Collapse
Affiliation(s)
- Theofanis Stampoultzis
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Yanheng Guo
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Naser Nasrollahzadeh
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Vijay Kumar Rana
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Peyman Karami
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Dominique P. Pioletti
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| |
Collapse
|
6
|
Kuca-Warnawin E, Kurowska W, Plebańczyk M, Wajda A, Kornatka A, Burakowski T, Janicka I, Syrówka P, Skalska U. Basic Properties of Adipose-Derived Mesenchymal Stem Cells of Rheumatoid Arthritis and Osteoarthritis Patients. Pharmaceutics 2023; 15:pharmaceutics15031003. [PMID: 36986863 PMCID: PMC10051260 DOI: 10.3390/pharmaceutics15031003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Rheumatoid arthritis (RA) and osteoarthritis (OA) are destructive joint diseases, the development of which are associated with the expansion of pathogenic T lymphocytes. Mesenchymal stem cells may be an attractive therapeutic option for patients with RA or OA due to the regenerative and immunomodulatory abilities of these cells. The infrapatellar fat pad (IFP) is a rich and easily available source of mesenchymal stem cells (adipose-derived stem cells, ASCs). However, the phenotypic, potential and immunomodulatory properties of ASCs have not been fully characterised. We aimed to evaluate the phenotype, regenerative potential and effects of IFP-derived ASCs from RA and OA patients on CD4+ T cell proliferation. The MSC phenotype was assessed using flow cytometry. The multipotency of MSCs was evaluated on the basis of their ability to differentiate into adipocytes, chondrocytes and osteoblasts. The immunomodulatory activities of MSCs were examined in co-cultures with sorted CD4+ T cells or peripheral blood mononuclear cells. The concentrations of soluble factors involved in ASC-dependent immunomodulatory activities were assessed in co-culture supernatants using ELISA. We found that ASCs with PPIs from RA and OA patients maintain the ability to differentiate into adipocytes, chondrocytes and osteoblasts. ASCs from RA and OA patients also showed a similar phenotype and comparable abilities to inhibit CD4+ T cell proliferation, which was dependent on the induction of soluble factors The results of our study constitute the basis for further research on the therapeutic potential of ASCs in the treatment of patients with RA and OA.
Collapse
Affiliation(s)
- Ewa Kuca-Warnawin
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Weronika Kurowska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Magdalena Plebańczyk
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Anna Wajda
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Anna Kornatka
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Tomasz Burakowski
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Iwona Janicka
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Piotr Syrówka
- Rheumaorthopedics Clinic and Polyclinic, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Urszula Skalska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| |
Collapse
|
7
|
RhoA Signaling in Neurodegenerative Diseases. Cells 2022; 11:cells11091520. [PMID: 35563826 PMCID: PMC9103838 DOI: 10.3390/cells11091520] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/25/2022] [Accepted: 04/29/2022] [Indexed: 02/04/2023] Open
Abstract
Ras homolog gene family member A (RhoA) is a small GTPase of the Rho family involved in regulating multiple signal transduction pathways that influence a diverse range of cellular functions. RhoA and many of its downstream effector proteins are highly expressed in the nervous system, implying an important role for RhoA signaling in neurons and glial cells. Indeed, emerging evidence points toward a role of aberrant RhoA signaling in neurodegenerative diseases such as Parkinson’s disease, Alzheimer’s disease, Huntington’s disease, and amyotrophic lateral sclerosis. In this review, we summarize the current knowledge of RhoA regulation and downstream cellular functions with an emphasis on the role of RhoA signaling in neurodegenerative diseases and the therapeutic potential of RhoA inhibition in neurodegeneration.
Collapse
|
8
|
Kahraman E, Ribeiro R, Lamghari M, Neto E. Cutting-Edge Technologies for Inflamed Joints on Chip: How Close Are We? Front Immunol 2022; 13:802440. [PMID: 35359987 PMCID: PMC8960235 DOI: 10.3389/fimmu.2022.802440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/18/2022] [Indexed: 11/17/2022] Open
Abstract
Osteoarthritis (OA) is a painful and disabling musculoskeletal disorder, with a large impact on the global population, resulting in several limitations on daily activities. In OA, inflammation is frequent and mainly controlled through inflammatory cytokines released by immune cells. These outbalanced inflammatory cytokines cause cartilage extracellular matrix (ECM) degradation and possible growth of neuronal fibers into subchondral bone triggering pain. Even though pain is the major symptom of musculoskeletal diseases, there are still no effective treatments to counteract it and the mechanisms behind these pathologies are not fully understood. Thus, there is an urgent need to establish reliable models for assessing the molecular mechanisms and consequently new therapeutic targets. Models have been established to support this research field by providing reliable tools to replicate the joint tissue in vitro. Studies firstly started with simple 2D culture setups, followed by 3D culture focusing mainly on cell-cell interactions to mimic healthy and inflamed cartilage. Cellular approaches were improved by scaffold-based strategies to enhance cell-matrix interactions as well as contribute to developing mechanically more stable in vitro models. The progression of the cartilage tissue engineering would then profit from the integration of 3D bioprinting technologies as these provide 3D constructs with versatile structural arrangements of the 3D constructs. The upgrade of the available tools with dynamic conditions was then achieved using bioreactors and fluid systems. Finally, the organ-on-a-chip encloses all the state of the art on cartilage tissue engineering by incorporation of different microenvironments, cells and stimuli and pave the way to potentially simulate crucial biological, chemical, and mechanical features of arthritic joint. In this review, we describe the several available tools ranging from simple cartilage pellets to complex organ-on-a-chip platforms, including 3D tissue-engineered constructs and bioprinting tools. Moreover, we provide a fruitful discussion on the possible upgrades to enhance the in vitro systems making them more robust regarding the physiological and pathological modeling of the joint tissue/OA.
Collapse
Affiliation(s)
- Emine Kahraman
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Rua Dr. Roberto Frias, Porto, Portugal
| | - Ricardo Ribeiro
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Meriem Lamghari
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Estrela Neto
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| |
Collapse
|
9
|
Hu X, Zhang W, Li X, Zhong D, Li Y, Li J, Jin R. Strategies to Modulate the Redifferentiation of Chondrocytes. Front Bioeng Biotechnol 2021; 9:764193. [PMID: 34881234 PMCID: PMC8645990 DOI: 10.3389/fbioe.2021.764193] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/01/2021] [Indexed: 01/17/2023] Open
Abstract
Because of the low self-healing capacity of articular cartilage, cartilage injuries and degenerations triggered by various diseases are almost irreversible. Previous studies have suggested that human chondrocytes cultured in vitro tend to dedifferentiate during the cell-amplification phase and lose the physiological properties and functions of the cartilage itself, which is currently a critical limitation in the cultivation of cartilage for tissue engineering. Recently, numerous studies have focused on the modulation of chondrocyte redifferentiation. Researchers discovered the effect of various conditions (extracellular environment, cell sources, growth factors and redifferentiation inducers, and gene silencing and overexpression) on the redifferentiation of chondrocytes during the in vitro expansion of cells, and obtained cartilage tissue cultured in vitro that exhibited physiological characteristics and functions that were similar to those of human cartilage tissue. Encouragingly, several studies reported positive results regarding the modulation of the redifferentiation of chondrocytes in specific conditions. Here, the various factors and conditions that modulate the redifferentiation of chondrocytes, as well as their limitations and potential applications and challenges are reviewed. We expect to inspire research in the field of cartilage repair toward the future treatment of arthropathy.
Collapse
Affiliation(s)
- Xiaoshen Hu
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Weiyang Zhang
- Shool of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Xiang Li
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dongling Zhong
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuxi Li
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Juan Li
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rongjiang Jin
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
Li X, Li S, Qian J, Chen Y, Zhou Y, Fu P. Early Efficacy of Type I Collagen-Based Matrix-Assisted Autologous Chondrocyte Transplantation for the Treatment of Articular Cartilage Lesions. Front Bioeng Biotechnol 2021; 9:760179. [PMID: 34778233 PMCID: PMC8584836 DOI: 10.3389/fbioe.2021.760179] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Articular cartilage is a complex structure that allows for low frictional gliding and effective shock absorption. Various sports injuries and inflammatory conditions can lead to lesions in the articular cartilage, which has limited regenerative potential. Type I collagen combined with autologous chondrocytes in a three-dimensional culture were used to induce the regeneration of single-layer autologous expanded chondrocytes without chondrogenic differentiation. Purpose: To assess the clinical, radiological, and histological changes following collagen-based autologous chondrocyte transplantation (MACT) for chondral knee lesions. Methods: The study prospectively enrolled 20 patients with symptomatic knee chondral lesions (mean size lesion was 2.41 ± 0.43 cm2, range: 2.0-3.4 cm2) in the lateral femoral condyle and femoral groove who underwent type I collagen-based MACT between July 2017 and July 2019. knee injury and osteoarthritis outcome score (KOOS) was assessed before the procedure, and periodic clinical follow-up was conducted every 3 months for a maximum of 12 months following the procedure and at 1-year intervals thereafter. Magnetic resonance imaging (MRI) T2 mapping of repaired cartilage was also used for the quantitative analysis of regeneration. In one patient, second-look arthroscopy was performed to assess cartilage regeneration characteristics, and a portion of regenerated cartilage was harvested for histological evaluation 12 months after implantation. Results: At pre-operation and at three, six, 12, and 24 months after the operation, KOOS pain, symptoms, daily life activities, sports and recreation, as well as the quality of life were significantly improved between every two time points. Hematoxylin and eosin (HE) staining indicated that the newly formed cartilage was comprised of naive chondrocytes. Safranin O-fast (S-O) green staining of the regenerated tissue revealed fibroblast-like cells surrounded by glycosaminoglycans. Immunohistochemistry (IHC) analysis indicated that collagen type II was uniformly distributed at the deep zone of articular cartilage and type I collagen mainly depositing in the superficial cartilage layer. The T2 values for repaired tissue gradually decreased, eventually approaching near-average values. Conclusion: The present study demonstrated that type I collagen-based MACT is a clinically effective treatment for improving functionality and pain levels. Histological evidence confirmed hyaline cartilage induction and showed that repaired cartilage tended to emerge from the deep to the superficial layer. The quantitative MRI T2 mapping test indicated that there still was a difference between the transplanted cartilage and the surrounding hyaline cartilage. Taken together, the current method represents an efficient approach for the restoration of knee cartilage lesions.
Collapse
Affiliation(s)
- Xiang Li
- Department of Arthroplasty Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Shiao Li
- Department of Arthroplasty Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jiatian Qian
- Department of Arthroplasty Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yancheng Chen
- Department of Arthroplasty Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yiqin Zhou
- Department of Arthroplasty Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Peiliang Fu
- Department of Arthroplasty Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
11
|
Govoni M, Muscari C, Bonafè F, Morselli PG, Cortesi M, Dallari D, Giordano E. A brief very-low oxygen tension regimen is sufficient for the early chondrogenic commitment of human adipose-derived mesenchymal stem cells. Adv Med Sci 2021; 66:98-104. [PMID: 33461101 DOI: 10.1016/j.advms.2020.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 10/19/2020] [Accepted: 12/23/2020] [Indexed: 12/30/2022]
Abstract
PURPOSE The aim of this study was to evaluate the effects exerted over chondrogenic commitment of human adipose-derived mesenchymal stem cells (ADSCs) by a very low oxygen tension (<1% pO2). MATERIALS/METHODS Cell morphology, mRNA levels of chondrocyte-specific marker genes and the involvement of p38 MAPK signalling were monitored in human ADSCs under a very low oxygen tension. RESULTS Cell morphology was significantly changed after two days of hypoxic preconditioning when they featured as elongated spindle-shaped cells. SRY-box containing gene 9, aggrecan and collagen type II mRNA levels were enhanced under severe hypoxic culture conditions. Moreover, the inhibition of p38 MAPK resulted in a substantial reduction in transcription of the above-mentioned specific genes, proving the pivotal role of this pathway in the transcriptional regulation of chondrogenesis. CONCLUSIONS Here, we propose a protocol showing the early commitment of stem cells towards the chondrogenic phenotype in only 2 days of culture via a very low hypoxic environment, in the absence of growth factors added in the culture medium.
Collapse
|
12
|
Otsu K, Ida-Yonemochi H, Ikezaki S, Ema M, Hitomi J, Ohshima H, Harada H. Oxygen regulates epithelial stem cell proliferation via RhoA-actomyosin-YAP/TAZ signal in mouse incisor. Development 2021; 148:dev.194787. [PMID: 33472844 DOI: 10.1242/dev.194787] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 01/11/2021] [Indexed: 12/22/2022]
Abstract
Stem cells are maintained in specific niches that strictly regulate their proliferation and differentiation for proper tissue regeneration and renewal. Molecular oxygen (O2) is an important component of the niche microenvironment, but little is known about how O2 governs epithelial stem cell (ESC) behavior. Here, we demonstrate that O2 plays a crucial role in regulating the proliferation of ESCs using the continuously growing mouse incisors. We have revealed that slow-cycling cells in the niche are maintained under relatively hypoxic conditions compared with actively proliferating cells, based on the blood vessel distribution and metabolic status. Mechanistically, we have demonstrated that, during hypoxia, HIF1α upregulation activates the RhoA signal, thereby promoting cortical actomyosin and stabilizing the adherens junction complex, including merlin. This leads to the cytoplasmic retention of YAP/TAZ to attenuate cell proliferation. These results shed light on the biological significance of blood-vessel geometry and the signaling mechanism through microenvironmental O2 to orchestrate ESC behavior, providing a novel molecular basis for the microenvironmental O2-mediated stem cell regulation during tissue development and renewal.
Collapse
Affiliation(s)
- Keishi Otsu
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University, 1-1-1, Idaidori, Yahaba, Iwate 028-3694, Japan
| | - Hiroko Ida-Yonemochi
- Division of Anatomy and Cell Biology of the Hard Tissue, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, 2-5274 Gakkocho-dori, Chuo-ku, Niigata 951-8514, Japan
| | - Shojiro Ikezaki
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University, 1-1-1, Idaidori, Yahaba, Iwate 028-3694, Japan
| | - Masatsugu Ema
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Jiro Hitomi
- Division of Human Embryology, Department of Anatomy, Iwate Medical University, 1-1-1, Idaidori, Yahaba, Iwate 028-3694, Japan
| | - Hayato Ohshima
- Division of Anatomy and Cell Biology of the Hard Tissue, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, 2-5274 Gakkocho-dori, Chuo-ku, Niigata 951-8514, Japan
| | - Hidemitsu Harada
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University, 1-1-1, Idaidori, Yahaba, Iwate 028-3694, Japan
| |
Collapse
|
13
|
Cheng J, Song Q, Yang Y, Sun Z, Tian X, Tian X, Feng L. Lipolysis by downregulating miR-92a activates the Wnt/β-catenin signaling pathway in hypoxic rats. Biomed Rep 2020; 13:33. [PMID: 32793347 PMCID: PMC7418506 DOI: 10.3892/br.2020.1340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 07/03/2020] [Indexed: 11/30/2022] Open
Abstract
The aim of the present study was to investigate the role of miR-92a in lipid metabolism in hypoxic rats. Microarray analysis and reverse transcription-quantitative (RT-q)PCR were used to detect changes in the mRNA expression levels of miR-92a in the epididymal fat of hypoxic and normoxic rats. The downstream target mRNA of miR-92a was predicted using bioinformatics analysis and verified using a dual luciferase reporter assay. Changes in the expression of frizzled (Fzd)10 and c-Myc in the epididymal fat were detected using RT-qPCR and western blotting. Microarray analysis and RT-qPCR results showed that the expression of miR-92a was significantly lower in the fat tissues of the hypoxic rats compared with the normoxic rats. The results of the dual luciferase reporter assay showed that the target gene of miR-92a was Fzd10, which is an acceptor in the Wnt pathway. Fzd10 expression was upregulated in the hypoxic rats. The mRNA expression levels of c-Myc, which is located downstream of the Wnt pathway, was increased significantly. The increase in the mRNA and protein expression levels of Fzd10 and c-Myc may be associated with miR-92a downregulation. Downregulation of miR-92a in-turn may result in lipolysis through the regulation of the Wnt/β-catenin signaling pathway, and thus weight loss in the rats.
Collapse
Affiliation(s)
- Jingjing Cheng
- Department of Sports and Health, Shandong Sport University, Jinan, Shandong 250102, P.R. China
| | - Qipeng Song
- Department of Sports and Health, Shandong Sport University, Jinan, Shandong 250102, P.R. China
| | - Yingjie Yang
- Department of Sports and Health, Shandong Sport University, Jinan, Shandong 250102, P.R. China
| | - Zhiyuan Sun
- Department of Sports and Health, Shandong Sport University, Jinan, Shandong 250102, P.R. China
| | - Xiaoyi Tian
- Department of Sports and Health, Shandong Sport University, Jinan, Shandong 250102, P.R. China
| | - Xuewen Tian
- Department of Sports and Health, Shandong Sport University, Jinan, Shandong 250102, P.R. China.,Biology Center, China Institute of Sport Science, Beijing 100061, P.R. China
| | - Lianshi Feng
- Biology Center, China Institute of Sport Science, Beijing 100061, P.R. China
| |
Collapse
|
14
|
He A, Ye A, Song N, Liu N, Zhou G, Liu Y, Ye X. Phenotypic redifferentiation of dedifferentiated microtia chondrocytes through a three-dimensional chondrogenic culture system. Am J Transl Res 2020; 12:2903-2915. [PMID: 32655818 PMCID: PMC7344067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 06/02/2020] [Indexed: 06/11/2023]
Abstract
Chondrocytes from microtia patients are a valuable cell source for the tissue-engineering of auricles. However, dedifferentiation of microtia chondrocytes remains an obstacle for clinical translation. Strategies, such as three-dimensional (3D) culture systems, and the use of chondrogenic growth factors, have successfully induced redifferentiation of dedifferentiated chondrocytes from healthy individuals. However, it remains unknown whether these strategies are similarly effective for microtia patient-derived chondrocytes, which may carry genomic defects. To address this issue, dedifferentiated microtia chondrocytes (DMCs) were cultured in a 3D chondrogenic culture system for 4-8 weeks to investigate their redifferentiated properties and to generate redifferentiated microtia chondrocytes (RMCs). To predict the degree and course of redifferentiation, RMCs at different time points were harvested and examined for cell morphology, cell proliferation, type II collagen expression at passaging, and chondrogenic capacity. We show that a 3D chondrogenic culture system can effectively induce DMCs to become redifferentiated, functional chondrocytes, enabling them to regenerate mature cartilage. Furthermore, RMCs achieved their full original function after culture in the chondrogenic culture system for 6-8 weeks. Interestingly, redifferentiation of microtia chondrocytes exhibited a time-dependent trend. Although the primary mechanism by which the 3D chondrogenic culture system regulated the transition of DMCs into RMCs remains unknown, the current study provides deeper insight into microtia chondrocytes and promotes clinical translation of tissue-engineered auricles.
Collapse
Affiliation(s)
- Aijuan He
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan UniversityShanghai, P. R. China
- Department of Plastic and Reconstructive Surgery, Shanghai 9 People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai, P. R. China
| | - Anqi Ye
- Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of MedicineShanghai, P. R. China
| | - Nan Song
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan UniversityShanghai, P. R. China
| | - Ninghua Liu
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan UniversityShanghai, P. R. China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai 9 People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai, P. R. China
- Research Institute of Plastic Surgery, Wei Fang Medical CollegeWeifang, Shandong, China
| | - Yanqun Liu
- Department of Plastic and Reconstructive Surgery, Shanghai 9 People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai, P. R. China
- Research Institute of Plastic Surgery, Wei Fang Medical CollegeWeifang, Shandong, China
| | - Xinhai Ye
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan UniversityShanghai, P. R. China
| |
Collapse
|
15
|
Huang X, Zhong L, Hendriks J, Post JN, Karperien M. Different response of human chondrocytes from healthy looking areas and damaged regions to IL1β stimulation under different oxygen tension. J Orthop Res 2019; 37:84-93. [PMID: 30255592 DOI: 10.1002/jor.24142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 08/25/2018] [Indexed: 02/04/2023]
Abstract
Due to its avascular nature, articular cartilage is relatively hypoxic. The aim of this study was to elucidate the functional changes of macroscopically healthy looking areas chondrocytes (MHC) and macroscopically damaged regions chondrocytes (MDC) at a cellular level in response to the inflammatory cytokine IL1β under different oxygen tension levels. In this study, two-dimensional (2-D) expanded MHC and MDC were redifferentiated in 3-D pellet cultures in chondrogenic differentiation medium, supplemented with or without IL1β at conventional culture (normoxia) or 2.5% O2 (hypoxia) for 3 weeks. qPCR, immunohistochemistry and ELISA were used to detect the expression of anabolic and catabolic gene expression. Alcian blue/Safranin O staining and GAG assay were used to measure cartilage matrix production. Cell proliferation and apoptosis were assessed by EdU staining and TUNEL assay, respectively. The results showed that hypoxia enhanced matrix production in both MHC and MDC and this effect was stronger on MDC. Under normoxia, MHC showed higher expression of cartilage markers and lower catabolic genes expression than MDC. Interestingly, hypoxia diminished the difference between MHC and MDC. IL1β potently induced MMPs expression regardless of cell population and oxygen tension. The fold induction of these MMPs in hypoxia was however much higher than in normoxia. In addition, hypoxia promoted the expression of HIF1α and HIF2α in MHC, while it only enhanced HIF1α expression but decreased the HIF2α expression in MDC. We concluded that hypoxia stimulated the redifferentiation of cultured chondrocytes, particularly in MDC derived from macroscopically diseased cartilage. Oxygen tension may profoundly and differentially influence inflammation-associated cartilage injury and diseases by regulating the expression of HIF1α and HIF2α. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 9999:XX-XX, 2018.
Collapse
Affiliation(s)
- Xiaobin Huang
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, 7500 AE, The Netherlands
| | - Leilei Zhong
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, 7500 AE, The Netherlands
| | - Jan Hendriks
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, 7500 AE, The Netherlands
| | - Janine N Post
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, 7500 AE, The Netherlands
| | - Marcel Karperien
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, 7500 AE, The Netherlands
| |
Collapse
|
16
|
Delve E, Parreno J, Co V, Wu PH, Chong J, Di Scipio M, Kandel RA. CDC42 regulates the expression of superficial zone molecules in part through the actin cytoskeleton and myocardin-related transcription factor-A. J Orthop Res 2018. [PMID: 29537109 DOI: 10.1002/jor.23892] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Osteoarthritis (OA) is a degenerative disease that initially manifests as loss of the superficial zone (SZ) of articular cartilage. SZ chondrocytes (SZC) differ in morphology from other chondrocytes as they are elongated and oriented parallel to the tissue surface. Proteoglycan 4 (PRG4) and tenascin C (TNC) are molecules expressed by SZC, which have been shown to be chondroprotective. Identification of the signalling pathway(s) regulating expression of SZ molecules may lead to a therapeutic target that can be used to delay or prevent the onset of OA. The hypothesis of this study is that expression of SZ molecules are regulated in part, by the CDC42-actin-myocardin-related transcription factor-A (MRTF-A) signaling pathway. SZC from bovine metacarpal-phalangeal joints were isolated and grown in monolayer culture. Each target in the CDC42-actin-MRTF-A pathway was inhibited and the effect on cell shape, actin cytoskeleton status, and expression of PRG4 and TNC were determined. Treatment with the CDC42 inhibitor ML141 decreased PRG4 and TNC expression, and correlated with increased cell circularity and G-/F-actin ratio. PRG4 and TNC expression were differentially regulated by actin depolymerizing agents, latrunculin B and cytochalasin D. Chemical inhibition of MRTF-A resulted in decreased expression of both PRG4 and TNC; however, specific knockdown by small interfering RNA only decreased expression of TNC indicating that TNC, but not PRG4, is regulated by MRTF-A. Although PRG4 and TNC expression are both regulated by CDC42 and actin, it appears to occur through different downstream signaling pathways. Further study is required to elucidate the pathway regulating PRG4. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:2421-2430, 2018.
Collapse
Affiliation(s)
- Elizabeth Delve
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario
| | - Justin Parreno
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Vivian Co
- University of Ontario Institute of Technology, Oshawa, Ontario
| | - Po-Han Wu
- The Department of Human Biology, University of Toronto, Toronto, Ontario
| | - Jasmine Chong
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario
| | - Matteo Di Scipio
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario
| | - Rita A Kandel
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario.,Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario.,Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario
| |
Collapse
|
17
|
Kim JG, Islam R, Cho JY, Jeong H, Cap KC, Park Y, Hossain AJ, Park JB. Regulation of RhoA GTPase and various transcription factors in the RhoA pathway. J Cell Physiol 2018; 233:6381-6392. [PMID: 29377108 DOI: 10.1002/jcp.26487] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 01/11/2018] [Indexed: 12/11/2022]
Abstract
RhoA GTPase plays a variety of functions in regulation of cytoskeletal proteins, cellular morphology, and migration along with various proliferation and transcriptional activity in cells. RhoA activity is regulated by guanine nucleotide exchange factors (GEFs), GTPase activating proteins (GAPs), and the guanine nucleotide dissociation factor (GDI). The RhoA-RhoGDI complex exists in the cytosol and the active GTP-bound form of RhoA is located to the membrane. GDI displacement factors (GDFs) including IκB kinase γ (IKKγ) dissociate the RhoA-GDI complex, allowing activation of RhoA through GEFs. In addition, modifications of Tyr42 phosphorylation and Cys16/20 oxidation in RhoA and Tyr156 phosphorylation and oxidation of RhoGDI promote the dissociation of the RhoA-RhoGDI complex. The expression of RhoA is regulated through transcriptional factors such as c-Myc, HIF-1α/2α, Stat 6, and NF-κB along with several reported microRNAs. As the role of RhoA in regulating actin-filament formation and myosin-actin interaction has been well described, in this review we focus on the transcriptional activity of RhoA and also the regulation of RhoA message itself. Of interest, in the cytosol, activated RhoA induces transcriptional changes through filamentous actin (F-actin)-dependent ("actin switch") or-independent means. RhoA regulates the activity of several transcription regulators such as serum response factor (SRF)/MAL, AP-1, NF-κB, YAP/TAZ, β-catenin, and hypoxia inducible factor (HIF)-1α. Interestingly, RhoA also itself is localized to the nucleus by an as-yet-undiscovered mechanism.
Collapse
Affiliation(s)
- Jae-Gyu Kim
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon, Kangwon-do, Republic of Korea
| | - Rokibul Islam
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon, Kangwon-do, Republic of Korea
| | - Jung Y Cho
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon, Kangwon-do, Republic of Korea.,Department of Biochemistry, Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon, Kangwon-do, Republic of Korea
| | - Hwalrim Jeong
- Department of Paediatrics, Chuncheon Sacred Hospital Hallym University, Chuncheon, Kangwon-do, Republic of Korea
| | - Kim-Cuong Cap
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon, Kangwon-do, Republic of Korea
| | - Yohan Park
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon, Kangwon-do, Republic of Korea
| | - Abu J Hossain
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon, Kangwon-do, Republic of Korea.,Department of Biochemistry, Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon, Kangwon-do, Republic of Korea
| | - Jae-Bong Park
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon, Kangwon-do, Republic of Korea.,Department of Biochemistry, Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon, Kangwon-do, Republic of Korea
| |
Collapse
|
18
|
Krajewska-Włodarczyk M, Owczarczyk-Saczonek A, Placek W, Osowski A, Wojtkiewicz J. Articular Cartilage Aging-Potential Regenerative Capacities of Cell Manipulation and Stem Cell Therapy. Int J Mol Sci 2018; 19:E623. [PMID: 29470431 PMCID: PMC5855845 DOI: 10.3390/ijms19020623] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 02/11/2018] [Accepted: 02/16/2018] [Indexed: 12/13/2022] Open
Abstract
Changes in articular cartilage during the aging process are a stage of natural changes in the human body. Old age is the major risk factor for osteoarthritis but the disease does not have to be an inevitable consequence of aging. Chondrocytes are particularly prone to developing age-related changes. Changes in articular cartilage that take place in the course of aging include the acquisition of the senescence-associated secretory phenotype by chondrocytes, a decrease in the sensitivity of chondrocytes to growth factors, a destructive effect of chronic production of reactive oxygen species and the accumulation of the glycation end products. All of these factors affect the mechanical properties of articular cartilage. A better understanding of the underlying mechanisms in the process of articular cartilage aging may help to create new therapies aimed at slowing or inhibiting age-related modifications of articular cartilage. This paper presents the causes and consequences of cellular aging of chondrocytes and the biological therapeutic outlook for the regeneration of age-related changes of articular cartilage.
Collapse
Affiliation(s)
- Magdalena Krajewska-Włodarczyk
- Department of Rheumatology, Municipal Hospital in Olsztyn, 10-900 Olsztyn, Poland.
- Department of Internal Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Agnieszka Owczarczyk-Saczonek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Waldemar Placek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Adam Osowski
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Joanna Wojtkiewicz
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
- Laboratory for Regenerative Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| |
Collapse
|