1
|
Kumar A, Suryakumar G, Singh SN, Rathor R. A comprehensive review on physiological and biological activities of carnosine: turning from preclinical facts to potential clinical applications. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03427-7. [PMID: 39302423 DOI: 10.1007/s00210-024-03427-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 08/30/2024] [Indexed: 09/22/2024]
Abstract
Carnosine, a compound with plethora of benefits, was originally discovered in 1900 and is formed by the amide linkage of β-alanine and L-histidine. Carnosine production is limited by β-alanine whereas the imidazole ring of histidine moiety makes it a suitable buffer in physiological pH range. It is reported to be found in the skeletal muscle, brain, heart, and gastrointestinal tissues of humans. This review focuses on the biological properties of carnosine including pH buffering ability, antioxidant activity, anti-inflammatory activity, anti-aging effect, enhancement of cognitive function, and immunomodulation. The relevance of carnosine in muscle function attributing to enhancement of physical performance has also been highlighted. Studies spanning several years have proved the preclinical effectiveness of carnosine in treating diverse pathological diseases. A complete summary of all key activities of carnosine from in vivo investigations and clinical trials has been compiled. Considering its numerous advantages, carnosine may be a promising option for the development of a nutraceutical.
Collapse
Affiliation(s)
- Akshita Kumar
- Soldier Performance Division, Defence Institute of Physiology & Allied Sciences (DIPAS), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Geetha Suryakumar
- Soldier Performance Division, Defence Institute of Physiology & Allied Sciences (DIPAS), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Som Nath Singh
- Soldier Performance Division, Defence Institute of Physiology & Allied Sciences (DIPAS), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Richa Rathor
- Soldier Performance Division, Defence Institute of Physiology & Allied Sciences (DIPAS), Lucknow Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
2
|
Bačenková D, Trebuňová M, Demeterová J, Živčák J. Human Chondrocytes, Metabolism of Articular Cartilage, and Strategies for Application to Tissue Engineering. Int J Mol Sci 2023; 24:17096. [PMID: 38069417 PMCID: PMC10707713 DOI: 10.3390/ijms242317096] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Hyaline cartilage, which is characterized by the absence of vascularization and innervation, has minimal self-repair potential in case of damage and defect formation in the chondral layer. Chondrocytes are specialized cells that ensure the synthesis of extracellular matrix components, namely type II collagen and aggregen. On their surface, they express integrins CD44, α1β1, α3β1, α5β1, α10β1, αVβ1, αVβ3, and αVβ5, which are also collagen-binding components of the extracellular matrix. This article aims to contribute to solving the problem of the possible repair of chondral defects through unique methods of tissue engineering, as well as the process of pathological events in articular cartilage. In vitro cell culture models used for hyaline cartilage repair could bring about advanced possibilities. Currently, there are several variants of the combination of natural and synthetic polymers and chondrocytes. In a three-dimensional environment, chondrocytes retain their production capacity. In the case of mesenchymal stromal cells, their favorable ability is to differentiate into a chondrogenic lineage in a three-dimensional culture.
Collapse
Affiliation(s)
- Darina Bačenková
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Košice, Letná 9, 042 00 Košice, Slovakia; (M.T.); (J.D.); (J.Ž.)
| | | | | | | |
Collapse
|
3
|
Arias C, Vásquez B, Salazar LA. Propolis as a Potential Therapeutic Agent to Counteract Age-Related Changes in Cartilage: An In Vivo Study. Int J Mol Sci 2023; 24:14272. [PMID: 37762574 PMCID: PMC10532056 DOI: 10.3390/ijms241814272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Aging is intricately linked to chronic low-grade systemic inflammation, which plays a significant role in various age-related conditions, including osteoarthritis (OA). The aging process significantly influences the development of OA due to alterations in cartilage composition, reduced proteoglycan content, dysregulation of growth factor signaling, and heightened oxidative stress. Propolis, a natural product renowned for its potent antioxidant and anti-inflammatory properties, has the potential to mitigate age-induced changes in cartilage. The primary objective of this study was to rigorously assess the impact of in vivo propolis treatment on the histopathological characteristics of knee articular cartilage in senescent rats. This study involved a cohort of twenty male Sprague-Dawley rats, randomly allocated into four distinct groups for comparative analysis: YR (control group consisting of young rats), SR (senescent rats), SR-EEP (senescent rats treated with an ethanolic extract of propolis, EEP), and SR-V (senescent rats administered with a control vehicle). This study employed comprehensive histological and stereological analyses of knee articular cartilage. Propolis treatment exhibited a significant capacity to alleviate the severity of osteoarthritis, enhance the structural integrity of cartilage, and augment chondrocyte density. These promising findings underscore the potential of propolis as a compelling therapeutic agent to counteract age-related alterations in cartilage and, importantly, to potentially forestall the onset of osteoarthritis.
Collapse
Affiliation(s)
- Consuelo Arias
- Escuela de Kinesiología, Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Santiago 8380000, Chile;
| | - Bélgica Vásquez
- Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Avenida Francisco Salazar 01145, Temuco 4811230, Chile
- Centre of Excellence in Morphological and Surgical Studies, Universidad de La Frontera, Avenida Francisco Salazar 01145, Temuco 4811230, Chile
| | - Luis A. Salazar
- Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Avenida Francisco Salazar 01145, Temuco 4811230, Chile
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Avenida Francisco Salazar 01145, Temuco 4811230, Chile
| |
Collapse
|
4
|
Potential Methods of Targeting Cellular Aging Hallmarks to Reverse Osteoarthritic Phenotype of Chondrocytes. BIOLOGY 2022; 11:biology11070996. [PMID: 36101377 PMCID: PMC9312132 DOI: 10.3390/biology11070996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/12/2022] [Accepted: 06/20/2022] [Indexed: 01/15/2023]
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease that causes pain, physical disability, and life quality impairment. The pathophysiology of OA remains largely unclear, and currently no FDA-approved disease-modifying OA drugs (DMOADs) are available. As has been acknowledged, aging is the primary independent risk factor for OA, but the mechanisms underlying such a connection are not fully understood. In this review, we first revisit the changes in OA chondrocytes from the perspective of cellular hallmarks of aging. It is concluded that OA chondrocytes share many alterations similar to cellular aging. Next, based on the findings from studies on other cell types and diseases, we propose methods that can potentially reverse osteoarthritic phenotype of chondrocytes back to a healthier state. Lastly, current challenges and future perspectives are summarized.
Collapse
|
5
|
Diniz FC, Hipkiss AR, Ferreira GC. The Potential Use of Carnosine in Diabetes and Other Afflictions Reported in Long COVID Patients. Front Neurosci 2022; 16:898735. [PMID: 35812220 PMCID: PMC9257001 DOI: 10.3389/fnins.2022.898735] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/18/2022] [Indexed: 12/15/2022] Open
Abstract
Carnosine is a dipeptide expressed in both the central nervous system and periphery. Several biological functions have been attributed to carnosine, including as an anti-inflammatory and antioxidant agent, and as a modulator of mitochondrial metabolism. Some of these mechanisms have been implicated in the pathophysiology of coronavirus disease-2019 (COVID-19). COVID-19 is caused by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2). The clinical manifestation and recovery time for COVID-19 are variable. Some patients are severely affected by SARS-CoV-2 infection and may experience respiratory failure, thromboembolic disease, neurological symptoms, kidney damage, acute pancreatitis, and even death. COVID-19 patients with comorbidities, including diabetes, are at higher risk of death. Mechanisms underlying the dysfunction of the afflicted organs in COVID-19 patients have been discussed, the most common being the so-called cytokine storm. Given the biological effects attributed to carnosine, adjuvant therapy with this dipeptide could be considered as supportive treatment in patients with either COVID-19 or long COVID.
Collapse
Affiliation(s)
- Fabiola Cardoso Diniz
- Laboratório de Erros Inatos do Metabolismo, Programa de Bioquímica e Biofísica Celular, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Ciências Biológicas - Biofísica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, United States
| | - Alan Roger Hipkiss
- Aston Research Centre for Healthy Ageing, Aston University, Birmingham, United Kingdom
| | - Gustavo Costa Ferreira
- Laboratório de Erros Inatos do Metabolismo, Programa de Bioquímica e Biofísica Celular, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Ciências Biológicas - Biofísica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Química Biológica, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
6
|
Application of Alginate Hydrogels for Next-Generation Articular Cartilage Regeneration. Int J Mol Sci 2022; 23:ijms23031147. [PMID: 35163071 PMCID: PMC8835677 DOI: 10.3390/ijms23031147] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 12/28/2022] Open
Abstract
The articular cartilage has insufficient intrinsic healing abilities, and articular cartilage injuries often progress to osteoarthritis. Alginate-based scaffolds are attractive biomaterials for cartilage repair and regeneration, allowing for the delivery of cells and therapeutic drugs and gene sequences. In light of the heterogeneity of findings reporting the benefits of using alginate for cartilage regeneration, a better understanding of alginate-based systems is needed in order to improve the approaches aiming to enhance cartilage regeneration with this compound. This review provides an in-depth evaluation of the literature, focusing on the manipulation of alginate as a tool to support the processes involved in cartilage healing in order to demonstrate how such a material, used as a direct compound or combined with cell and gene therapy and with scaffold-guided gene transfer procedures, may assist cartilage regeneration in an optimal manner for future applications in patients.
Collapse
|
7
|
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
|
8
|
Njoto I. Hyperglycemia Duration Impact On Anatomical Damage Level Of Osteoarthritic Articular Cartilage In Rat Models With Diabetes Mellitus Type 1. RUSSIAN OPEN MEDICAL JOURNAL 2021. [DOI: 10.15275/rusomj.2021.0304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background — Diabetes mellitus caused alteration of chondrocytes morphology of superficial layer on osteoarthritic articular (OA) cartilage in an articular cartilage rat model. These results need to be analyzed in relation to hyperglycemia duration. Objective — This study evaluates the influence of hyperglycemia on microscopic anatomical damage progression in OA cartilage. Material and Methods — Thirty-five adult male rats were divided into seven groups: control group, three OA groups, and three OA groups with type 1 diabetes mellitus (DMT-1). For OA groups, the first, second, and third group was sacrificed on the third, fourth, and sixth week respectively after two months maintenance. OA with DMT-1 groups were performed anterior cruciate ligament transaction (ACLT) and were injected streptozotocin intraperitoneally to promote DMT-1 for one-month maintenance. DMT-1.1, DMT-1.2, and DMT-1.3 group was sacrificed on the third, fourth, and sixth week respectively after two months maintenance. The right knee cartilage was taken and processed for histopathology with hematoxylin and eosin staining, then analyzed using a Pritzker scale. Results — In OA group with DMT-1, hyperglycemia duration (6th>4th>3th weeks exposure) increased the level of damage in the OA cartilage compared with the OA group. Pritzker scale observe on deeper abrasiveness of the superficial articular layer, cartilage fissure reaching the middle layer, a more severe decrease in the chondrocytes columnar pattern, changing of matrix integrity, and many sclerotic conditions were provoked by increasing the hyperglycemia duration. Conclusion — Hyperglycemia duration influenced the damage level in the articular cartilage, increasing the progression of OA disease in animal models.
Collapse
Affiliation(s)
- Ibrahim Njoto
- University of Wijaya Kusuma Surabaya, Surabaya, East Java, Indonesia
| |
Collapse
|
9
|
CircSERPINE2 weakens IL-1β-caused apoptosis and extracellular matrix degradation of chondrocytes by regulating miR-495/TGFBR2 axis. Biosci Rep 2021; 40:226746. [PMID: 33094798 PMCID: PMC7610148 DOI: 10.1042/bsr20201601] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 12/14/2022] Open
Abstract
The dysregulated circular RNAs (circRNAs) are relevant to the development of osteoarthritis (OA). The circRNA serpin family E member 2 (circSERPINE2) is dysregulated in OA, while the role and mechanism of circSERPINE2 in OA are largely unknown. The aim of our research is to explore how and whether circSERPINE2 regulates interleukin-1β (IL-1β)-caused chondrocyte damage in OA. In the present study, the chondrocytes (CHON-001 cells) were exposed to IL-1β to mimic the injury in OA. CircSERPINE2, microRNA-495 (miR-495) and transforming growth factor-β receptor 2 (TGFBR2) abundances were detected via quantitative reverse-transcription polymerase chain reaction (qRT-PCR) or Western blot. Cell apoptosis was assessed via viability, apoptotic rate and caspase-3 activity. Extracellular matrix was investigated by levels of Sry-type high-mobility-group box 9 (SOX9), collagen type II α 1 (COL2A1) and Aggrecan using Western blot. The interaction among circSERPINE2, miR-495 and TGFBR2 was assessed via dual-luciferase reporter analysis and RNA immunoprecipitation (RIP). The results showed that circSERPINE2 expression was reduced in OA patients and IL-1β-treated chondrocytes. CircSERPINE2 overexpression mitigated IL-1β-caused apoptosis and extracellular matrix degradation. miR-495 was targeted by circSERPINE2 and up-regulated in OA patients and IL-1β-treated chondrocytes. miR-495 up-regulation reversed overexpression of circSERPINE2-mediated inhibition of apoptosis and extracellular matrix degradation. TGFBR2 was targeted by miR-495 and lowly expressed in OA patients and IL-1β-treated chondrocytes. CircSERPINE2 could mediate TGFBR2 expression by binding with miR-495. As a conclusion, circSERPINE2 attenuated IL-1β-caused apoptosis and extracellular matrix degradation of chondrocytes by regulating miR-495/TGFBR2 axis, indicating a new target for OA treatment.
Collapse
|
10
|
Arias C, Saavedra N, Leal K, Vásquez B, Abdalla DSP, Salazar LA. Histological Evaluation and Gene Expression Profiling of Autophagy-Related Genes for Cartilage of Young and Senescent Rats. Int J Mol Sci 2020; 21:ijms21228607. [PMID: 33203108 PMCID: PMC7697851 DOI: 10.3390/ijms21228607] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/13/2020] [Accepted: 11/13/2020] [Indexed: 01/06/2023] Open
Abstract
Autophagy is a cellular mechanism that protects cells from stress by digesting non-functional cellular components. In the cartilage, chondrocytes depend on autophagy as a principal mechanism to maintain cellular homeostasis. This protective role diminishes prior to the structural damage that normally occurs during aging. Considering that aging is the main risk factor for osteoarthritis, evaluating the expression of genes associated with autophagy in senescent cartilage might allow for the identification of potential therapeutic targets for treatment. Thus, we studied two groups of young and senescent rats. A histological analysis of cartilage and gene expression quantification for autophagy-related genes were performed. In aged cartilage, morphological changes were observed, such as an increase in cartilage degeneration as measured by the modified Mankin score, a decrease in the number of chondrocytes and collagen II (Col2a1), and an increase in matrix metalloproteinase 13 (Mmp13). Moreover, 84 genes associated with autophagy were evaluated by a PCR array analysis, and 15 of them were found to be significantly decreased with aging. Furthermore, an in silico analysis based on by two different bioinformatics software tools revealed that several processes including cellular homeostasis, autophagosome assembly, and aging—as well as several biological pathways such as autophagy, insulin-like growth factor 1 (IGF-1) signaling, PI3K (phosphoinositide 3-kinase)/AKT (serine/threonine kinase) signaling, and mammalian target of rapamycin (mTOR) signaling—were enriched. In conclusion, the analysis identified some potential targets for osteoarthritis treatment that would allow for the development of new therapeutic strategies for this chronic disease.
Collapse
Affiliation(s)
- Consuelo Arias
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Av. Francisco Salazar 01145, Temuco 4811230, Chile; (C.A.); (N.S.); (K.L.)
- Carrera de Kinesiología, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Av. Alemania 1090, Temuco 4810101, Chile
| | - Nicolás Saavedra
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Av. Francisco Salazar 01145, Temuco 4811230, Chile; (C.A.); (N.S.); (K.L.)
| | - Karla Leal
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Av. Francisco Salazar 01145, Temuco 4811230, Chile; (C.A.); (N.S.); (K.L.)
| | - Bélgica Vásquez
- Facultad de Ciencias de la Salud, Universidad de Tarapacá, Av. General Velásquez 1775, Arica 1000007, Chile;
| | - Dulcineia S. P. Abdalla
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, Universidade de São Paulo, Avenida Professor Lineu Prestes 580, São Paulo CEP 05508-000, SP, Brazil;
| | - Luis A. Salazar
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Av. Francisco Salazar 01145, Temuco 4811230, Chile; (C.A.); (N.S.); (K.L.)
- Correspondence: ; Tel.: +56-45-259-6724
| |
Collapse
|
11
|
Allas L, Brochard S, Rochoux Q, Ribet J, Dujarrier C, Veyssiere A, Aury-Landas J, Grard O, Leclercq S, Vivien D, Ea HK, Maubert E, Cohen-Solal M, Boumediene K, Agin V, Baugé C. EZH2 inhibition reduces cartilage loss and functional impairment related to osteoarthritis. Sci Rep 2020; 10:19577. [PMID: 33177650 PMCID: PMC7658239 DOI: 10.1038/s41598-020-76724-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/29/2020] [Indexed: 12/19/2022] Open
Abstract
Histone methyltransferase EZH2 is upregulated during osteoarthritis (OA), which is the most widespread rheumatic disease worldwide, and a leading cause of disability. This study aimed to assess the impact of EZH2 inhibition on cartilage degradation, inflammation and functional disability. In vitro, gain and loss of EZH2 function were performed in human articular OA chondrocytes stimulated with IL-1β. In vivo, the effects of EZH2 inhibition were investigated on medial meniscectomy (MMX) OA mouse model. The tissue alterations were assayed by histology and the functional disabilities of the mice by actimetry and running wheel. In vitro, EZH2 overexpression exacerbated the action of IL-1β in chondrocytes increasing the expression of genes involved in inflammation, pain (NO, PGE2, IL6, NGF) and catabolism (MMPs), whereas EZH2 inhibition by a pharmacological inhibitor, EPZ-6438, reduced IL-1β effects. Ex vivo, EZH2 inhibition decreased IL-1β-induced degradation of cartilage. In vivo, intra-articular injections of the EZH2 inhibitor reduced cartilage degradation and improved motor functions of OA mice. This study demonstrates that the pharmacological inhibition of the histone methyl-transferase EZH2 slows the progression of osteoarthritis and improves motor functions in an experimental OA model, suggesting that EZH2 could be an effective target for the treatment of OA by reducing catabolism, inflammation and pain.
Collapse
Affiliation(s)
- Lyess Allas
- EA7451 BioConnect, Normandie Univ, Université de Caen, 14032, Caen, France
| | - Sybille Brochard
- EA7451 BioConnect, Normandie Univ, Université de Caen, 14032, Caen, France
| | - Quitterie Rochoux
- EA7451 BioConnect, Normandie Univ, Université de Caen, 14032, Caen, France
- Service de Rhumatologie, CHU, Caen, France
| | - Jules Ribet
- EA7451 BioConnect, Normandie Univ, Université de Caen, 14032, Caen, France
- Service de Rhumatologie, CHU, Caen, France
| | - Cleo Dujarrier
- UMRS1237 PhIND, INSERM, Normandie Univ, Institut Blood and Brain @ Caen-Normandie, Université de Caen, Caen, France
| | - Alexis Veyssiere
- EA7451 BioConnect, Normandie Univ, Université de Caen, 14032, Caen, France
- Service de Chirurgie Maxillo-Faciale, CHU, Caen, France
| | | | - Ophélie Grard
- EA7451 BioConnect, Normandie Univ, Université de Caen, 14032, Caen, France
| | - Sylvain Leclercq
- EA7451 BioConnect, Normandie Univ, Université de Caen, 14032, Caen, France
- Service de Chirurgie Orthopédique, Clinique Saint-Martin, Caen, France
| | - Denis Vivien
- UMRS1237 PhIND, INSERM, Normandie Univ, Institut Blood and Brain @ Caen-Normandie, Université de Caen, Caen, France
- Department of Clinical Research, CHU Caen-Normandie, Caen, France
| | - Hang-Korng Ea
- UMR-1132 BIOSCAR, INSERM, Université de Paris, Paris, France
| | - Eric Maubert
- UMRS1237 PhIND, INSERM, Normandie Univ, Institut Blood and Brain @ Caen-Normandie, Université de Caen, Caen, France
| | | | - Karim Boumediene
- EA7451 BioConnect, Normandie Univ, Université de Caen, 14032, Caen, France
| | - Véronique Agin
- UMRS1237 PhIND, INSERM, Normandie Univ, Institut Blood and Brain @ Caen-Normandie, Université de Caen, Caen, France
| | - Catherine Baugé
- EA7451 BioConnect, Normandie Univ, Université de Caen, 14032, Caen, France.
| |
Collapse
|
12
|
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
|
13
|
Tan GK, Pryce BA, Stabio A, Brigande JV, Wang C, Xia Z, Tufa SF, Keene DR, Schweitzer R. Tgfβ signaling is critical for maintenance of the tendon cell fate. eLife 2020; 9:52695. [PMID: 31961320 PMCID: PMC7025861 DOI: 10.7554/elife.52695] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 01/17/2020] [Indexed: 12/12/2022] Open
Abstract
Studies of cell fate focus on specification, but little is known about maintenance of the differentiated state. In this study, we find that the mouse tendon cell fate requires continuous maintenance in vivo and identify an essential role for TGFβ signaling in maintenance of the tendon cell fate. To examine the role of TGFβ signaling in tenocyte function the TGFβ type II receptor (Tgfbr2) was targeted in the Scleraxis-expressing cell lineage using the ScxCre deletor. Tendon development was not disrupted in mutant embryos, but shortly after birth tenocytes lost differentiation markers and reverted to a more stem/progenitor state. Viral reintroduction of Tgfbr2 to mutants prevented and even rescued tenocyte dedifferentiation suggesting a continuous and cell autonomous role for TGFβ signaling in cell fate maintenance. These results uncover the critical importance of molecular pathways that maintain the differentiated cell fate and a key role for TGFβ signaling in these processes.
Collapse
Affiliation(s)
- Guak-Kim Tan
- Research Division, Shriners Hospital for Children, Portland, United States
| | - Brian A Pryce
- Research Division, Shriners Hospital for Children, Portland, United States
| | - Anna Stabio
- Research Division, Shriners Hospital for Children, Portland, United States
| | - John V Brigande
- Oregon Hearing Research Center, Oregon Health & Science University, Portland, United States
| | - ChaoJie Wang
- Computational Biology Program, Oregon Health & Science University, Portland, United States
| | - Zheng Xia
- Computational Biology Program, Oregon Health & Science University, Portland, United States
| | - Sara F Tufa
- Research Division, Shriners Hospital for Children, Portland, United States
| | - Douglas R Keene
- Research Division, Shriners Hospital for Children, Portland, United States
| | - Ronen Schweitzer
- Research Division, Shriners Hospital for Children, Portland, United States.,Department of Orthopedics, Oregon Health & Science University, Portland, United States
| |
Collapse
|
14
|
Pinto-Cardoso R, Pereira-Costa F, Pedro Faria J, Bandarrinha P, Bessa-Andrês C, Correia-de-Sá P, Bernardo Noronha-Matos J. Adenosinergic signalling in chondrogenesis and cartilage homeostasis: Friend or foe? Biochem Pharmacol 2019; 174:113784. [PMID: 31884043 DOI: 10.1016/j.bcp.2019.113784] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Abstract
Chondrocytes and their mesenchymal cell progenitors secrete a variety of bioactive molecules, including adenine nucleotides and nucleosides, but these molecules are not usually highlighted in review papers about the secretome of these cells. Ageing and inflammatory insults compromise chondrocytes ability to keep ATP/adenosine synthesis, release and turnover. Cartilage homeostasis depends on extracellular adenosine levels, which acting via four P1 purinoceptor subtypes modulates the release of pro-inflammatory mediators, including NO, PGE2 and several cytokines. Native articular cartilage is challenged by synovial fluid flow during normal joint motion transiently increasing ATP release and adenosine formation in the joint microenvironment. Excessive joint motion and shockwave trauma are deleterious to cartilage homeostasis due to HIF-1α overexpression, resulting in disproportionate ecto-5'-nucleotidase/CD73 production, adenosine accumulation and superfluous A2B receptors activation. Scarcity of data however exists on the putative interplay between coexistent high affinity (A2A and A3) and low affinity (A2B) adenosine receptors activation affecting stem cells fate towards preferential chondrogenic or osteogenic lineages in the human cartilage. Hints gathered in this commentary result mainly from studies using human immortalized cell lines and animal (e.g. rodent, equine, bovine) tissue samples. The available data point towards adenosine A2A and A3 receptors having cartilage protective roles, while excessive adenosine accumulation may be detrimental via low affinity A2B receptors activation, with little reference to the putative role of the adenosine forming enzyme ecto-5'-nucleotidase/CD73. Thus, emphasizing the multiple pathways responsible for controlling adenosine signalling in cartilage will certainly impact on the search for novel therapeutic targets for highly disabling articular disorders.
Collapse
Affiliation(s)
- Rui Pinto-Cardoso
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal
| | - Flávio Pereira-Costa
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal
| | - João Pedro Faria
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal
| | - Patrícia Bandarrinha
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal
| | - Catarina Bessa-Andrês
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal.
| | - José Bernardo Noronha-Matos
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal.
| |
Collapse
|
15
|
Charlier E, Deroyer C, Ciregia F, Malaise O, Neuville S, Plener Z, Malaise M, de Seny D. Chondrocyte dedifferentiation and osteoarthritis (OA). Biochem Pharmacol 2019; 165:49-65. [DOI: 10.1016/j.bcp.2019.02.036] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 02/28/2019] [Indexed: 02/08/2023]
|
16
|
Njoto I, Soekanto A, Ernawati E, Abdurrachman A, Kalim H, Handono K, Soeatmadji DW, Fatchiyah F. Chondrocyte Intracellular Matrix Strain Fields of Articular Cartilage Surface in Hyperglycemia Model of Rat: Cellular Morphological Study. Med Arch 2019; 72:348-351. [PMID: 30524167 PMCID: PMC6282912 DOI: 10.5455/medarh.2018.72.348-351] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Introduction: Chondrocyte is one cell in articular cartilage was products many proteins, molecules, and other factors. The external influence of cartilage, such as: hyperglycemia was entering joint capsule and impact to the chondrocytes and the cartilage. Hyperglycemia caused modification of heparan sulfate proteoglycan 2 (perlecan) proteins through glycation process. Aim: The aim of this study was to analyze morphological changing of chondrocytes pericellular matrix by the influence of hyperglycemia. Material and Methods: Eighteen adult male rats were divided into six groups: control, rat treated with sugar intake was 0.5 mg/kg, 0.75 mg/ kg, 1mg/kg, 1.5 g/kg and 2 mg/kg of body weight. The animal model was dislocated and left knee was taken to observe changing of chondrocytes pericellular matrix strain fields by Scanning Electron Microscope (SEM) from perpendicular to femoral condylus cartilage. Results: Changing of chondrocytes intracellular matrix strain fields as changing of cell diameters and cell distances at group control and group I to group V, which cell diameters was lower level and cell distances was the highest level at over diet 2. This changing of intracellular matrix strain fields was corresponding to changing chondrocytes morphology in hyperglycemia condition, due to hypertrophic stage as adaptive responses. This research as based on next research for accomplish of hyperglycemia influence to morphology articular cartilage changing to prevent degeneration of cartilage towards osteoarthritis. Conclusions: Present study concludes that hyperglycemia influence to chondrocyte intracellular matrix strain fields changing.
Collapse
Affiliation(s)
- Ibrahim Njoto
- Anatomy Department, Faculty of Medicine, Wijaya Kusuma Surabaya University
| | - Ayly Soekanto
- Anatomy Department, Faculty of Medicine, Wijaya Kusuma Surabaya University
| | - Ernawati Ernawati
- Farmacology Department, Faculty of Medicine, Wijaya Kusuma Surabaya University
| | | | - Handono Kalim
- Proffessor of Internal Medicine at Doctoral Programme, Faculty of Medicine, Brawijaya University, Malang
| | - Kusworini Handono
- Proffessor of Clinical Pathology Doctoral Programme, Faculty of Medicine, Brawijaya University, Malang
| | - Djoko W Soeatmadji
- Proffessor of Internal Medicine at Doctoral Programme, Faculty of Medicine, Brawijaya University, Malang
| | - Fatchiyah Fatchiyah
- Research Group of Smart Molecule of Natural Genetics Resources UB, and Department of Biology, Faculty of Sciences, Brawijaya University
| |
Collapse
|
17
|
Gurusinghe S, Bandara N, Hilbert B, Trope G, Wang L, Strappe P. Lentiviral vector expression of Klf4 enhances chondrogenesis and reduces hypertrophy in equine chondrocytes. Gene 2018; 680:9-19. [PMID: 30205175 DOI: 10.1016/j.gene.2018.09.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 08/14/2018] [Accepted: 09/06/2018] [Indexed: 12/26/2022]
Abstract
Monolayer expansion of chondrocytes in culture results in the dedifferentiation of chondrocytes with inferior cartilage specific extracellular matrix synthesis and proliferation when compared with its native counterpart. We aimed to enhance chondrocyte proliferation and articular cartilage specific gene expression through ectopic expression of the major pluripotency transcription factors (Oct4, Sox2, Klf4 and c-Myc). We also aimed to provide insights to the modulation of TGFβ receptor mRNA with Klf4 overexpression. Equine chondrocytes pooled from three donors were transduced with lentiviral vectors expressing the induced pluripotency factors, Oct4, Sox2. Klf4 and c-Myc (OSKM), singly, or in combination or together with green fluorescent protein (GFP) as a control. Klf4 and c-Myc overexpressing chondrocytes showed a significant increase in mitosis when compared to the control (P < 0.01 and P < 0.0001 respectively). Furthermore, overexpression of Klf4 or OSKM in three dimensional (3D) culture of equine chondrocytes resulted in a significant increase in Col2a1 mRNA levels relative to the controls (P < 0.05 and P < 0.01 respectively) while all transcription factors significantly lowered the mRNA of the fibrocartilage marker Col1a1. We also employed a Col2a1 promoter driven GFP reporter for real time monitoring of Col2a1 gene activation in 3D micromass culture, which showed significantly higher promoter activity when cultures were treated with the growth factor TGFβ3 (P < 0.05). The chondrogenic properties of Klf4 transduced chondrocytes at a lower passage (P4) showed significant increases in Sox9 (P < 0.001), Col2a1 (P < 0.05) and TGFβ receptor I (P < 0.05) and II (P < 0.001) expression relative to the DS-Red expressing control. The chondrocyte dedifferentiation marker Col1a1 and hypertrophic marker Col10a1 were significantly downregulated with the inclusion of Klf4 (P < 0.01 and P < 0.05 respectively). In Conclusion, chondrogenic re-differentiation and proliferation of equine chondrocytes is promoted through ectopic expression of Klf4 while suppressing chondrocyte dedifferentiation.
Collapse
Affiliation(s)
- Saliya Gurusinghe
- Graham Centre for Agricultural Innovation, Charles Sturt University, Wagga Wagga, NSW 2678, Australia
| | - Nadeeka Bandara
- St. Vincent's Institute for Medical Research, Melbourne, VIC 3000, Australia
| | - Bryan Hilbert
- School of Animal and Veterinary Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia
| | - Gareth Trope
- School of Animal and Veterinary Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia
| | - Lexin Wang
- School of Biomedical Science, Charles Sturt University, Wagga Wagga, NSW 2678, Australia
| | - Padraig Strappe
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, Qld 4701, Australia.
| |
Collapse
|
18
|
Carnosine and the processes of ageing. Maturitas 2016; 93:28-33. [DOI: 10.1016/j.maturitas.2016.06.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 05/31/2016] [Accepted: 06/01/2016] [Indexed: 12/19/2022]
|
19
|
Lewis R, Barrett-Jolley R. Changes in Membrane Receptors and Ion Channels as Potential Biomarkers for Osteoarthritis. Front Physiol 2015; 6:357. [PMID: 26648874 PMCID: PMC4664663 DOI: 10.3389/fphys.2015.00357] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/11/2015] [Indexed: 01/01/2023] Open
Abstract
Osteoarthritis (OA), a degenerative joint condition, is currently difficult to detect early enough for any of the current treatment options to be completely successful. Early diagnosis of this disease could increase the numbers of patients who are able to slow its progression. There are now several diseases where membrane protein biomarkers are used for early diagnosis. The numbers of proteins in the membrane is vast and so it is a rich source of potential biomarkers for OA but we need more knowledge of these before they can be considered practical biomarkers. How are they best measured and are they selective to OA or even certain types of OA? The first step in this process is to identify membrane proteins that change in OA. Here, we summarize several ion channels and receptors that change in OA models and/or OA patients, and may thus be considered candidates as novel membrane biomarkers of OA.
Collapse
Affiliation(s)
- Rebecca Lewis
- Faculty of Health and Medical Sciences, School of Veterinary Medicine and Science, University of Surrey Guildford, UK
| | - Richard Barrett-Jolley
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease, University of Liverpool Liverpool, UK
| |
Collapse
|
20
|
Use of Adult Stem Cells for Cartilage Tissue Engineering: Current Status and Future Developments. Stem Cells Int 2015; 2015:438026. [PMID: 26246809 PMCID: PMC4515346 DOI: 10.1155/2015/438026] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 03/16/2015] [Accepted: 03/17/2015] [Indexed: 02/06/2023] Open
Abstract
Due to their low self-repair ability, cartilage defects that result from joint injury, aging, or osteoarthritis, are the most often irreversible and are a major cause of joint pain and chronic disability. So, in recent years, researchers and surgeons have been working hard to elaborate cartilage repair interventions for patients who suffer from cartilage damage. However, current methods do not perfectly restore hyaline cartilage and may lead to the apparition of fibro- or hypertrophic cartilage. In the next years, the development of new strategies using adult stem cells, in scaffolds, with supplementation of culture medium and/or culture in low oxygen tension should improve the quality of neoformed cartilage. Through these solutions, some of the latest technologies start to bring very promising results in repairing cartilage from traumatic injury or chondropathies. This review discusses the current knowledge about the use of adult stem cells in the context of cartilage tissue engineering and presents clinical trials in progress, as well as in the future, especially in the field of bioprinting stem cells.
Collapse
|
21
|
Tekari A, Luginbuehl R, Hofstetter W, Egli RJ. Transforming growth factor beta signaling is essential for the autonomous formation of cartilage-like tissue by expanded chondrocytes. PLoS One 2015; 10:e0120857. [PMID: 25775021 PMCID: PMC4361600 DOI: 10.1371/journal.pone.0120857] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 01/27/2015] [Indexed: 02/07/2023] Open
Abstract
Cartilage is a tissue with limited self-healing potential. Hence, cartilage defects require surgical attention to prevent or postpone the development of osteoarthritis. For cell-based cartilage repair strategies, in particular autologous chondrocyte implantation, articular chondrocytes are isolated from cartilage and expanded in vitro to increase the number of cells required for therapy. During expansion, the cells lose the competence to autonomously form a cartilage-like tissue, that is in the absence of exogenously added chondrogenic growth factors, such as TGF-βs. We hypothesized that signaling elicited by autocrine and/or paracrine TGF-β is essential for the formation of cartilage-like tissue and that alterations within the TGF-β signaling pathway during expansion interfere with this process. Primary bovine articular chondrocytes were harvested and expanded in monolayer culture up to passage six and the formation of cartilage tissue was investigated in high density pellet cultures grown for three weeks. Chondrocytes expanded for up to three passages maintained the potential for autonomous cartilage-like tissue formation. After three passages, however, exogenous TGF-β1 was required to induce the formation of cartilage-like tissue. When TGF-β signaling was blocked by inhibiting the TGF-β receptor 1 kinase, the autonomous formation of cartilage-like tissue was abrogated. At the initiation of pellet culture, chondrocytes from passage three and later showed levels of transcripts coding for TGF-β receptors 1 and 2 and TGF-β2 to be three-, five- and five-fold decreased, respectively, as compared to primary chondrocytes. In conclusion, the autonomous formation of cartilage-like tissue by expanded chondrocytes is dependent on signaling induced by autocrine and/or paracrine TGF-β. We propose that a decrease in the expression of the chondrogenic growth factor TGF-β2 and of the TGF-β receptors in expanded chondrocytes accounts for a decrease in the activity of the TGF-β signaling pathway and hence for the loss of the potential for autonomous cartilage-like tissue formation.
Collapse
Affiliation(s)
- Adel Tekari
- Group for Bone Biology & Orthopaedic Research, Department Clinical Research, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | | | - Willy Hofstetter
- Group for Bone Biology & Orthopaedic Research, Department Clinical Research, University of Bern, Bern, Switzerland
| | - Rainer J. Egli
- Group for Bone Biology & Orthopaedic Research, Department Clinical Research, University of Bern, Bern, Switzerland
- RMS Foundation, Bettlach, Switzerland
| |
Collapse
|
22
|
Duan L, Ma B, Liang Y, Chen J, Zhu W, Li M, Wang D. Cytokine networking of chondrocyte dedifferentiation in vitro and its implications for cell-based cartilage therapy. Am J Transl Res 2015; 7:194-208. [PMID: 25901191 PMCID: PMC4399086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/25/2014] [Indexed: 06/04/2023]
Abstract
Autologous chondrocyte implantation (ACI) is a golden treatment for large defects of the knee joint without osteoarthritis or other complications. Despite notable progresses, generation of a stable chondrocyte phenotype using progenitor cells remains a main obstacle for chondrocyte-based cartilage treatment. Monolayer chondrocyte expansion in vitro is accompanied by chondrocyte dedifferentiation, which produces a non-specific mechanically inferior extracellular matrix (ECM) unsuitable for ACI. In-depth understanding of the molecular events during chondrocyte dedifferentiation is required to maintain the capacity of in vitro expanded chondrocytes to produce hyaline cartilage-specific ECM. This review discusses key cytokines and signaling pathways involved in chondrocyte dedifferentiation from the standpoint of catabolism and anabolism. Some potential therapeutic strategies are also presented to counteract chondrocyte dedifferentiation for cell-based cartilage therapy.
Collapse
Affiliation(s)
- Li Duan
- School of Medicine, Sun Yat-sen UniversityGuangzhou 510182, Guangdong Province, China
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
- Department of Orthopedics, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
| | - Bin Ma
- Division of Immunology, University Children’s Hospital ZurichZurich 8032, Switzerland
| | - Yujie Liang
- School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate SchoolShenzhen 518055, Guangdong Province, China
| | - Jielin Chen
- School of Medicine, Sun Yat-sen UniversityGuangzhou 510182, Guangdong Province, China
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
- Department of Orthopedics, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
| | - Weimin Zhu
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
- Department of Orthopedics, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
| | - Mingtao Li
- School of Medicine, Sun Yat-sen UniversityGuangzhou 510182, Guangdong Province, China
| | - Daping Wang
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
- Department of Orthopedics, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
| |
Collapse
|
23
|
Catherine B, Girard N, Lhuissier E, Bazille C, Boumediene K. Regulation and Role of TGFβ Signaling Pathway in Aging and Osteoarthritis Joints. Aging Dis 2014; 5:394-405. [PMID: 25489490 DOI: 10.14336/ad.2014.0500394] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/04/2013] [Accepted: 12/04/2013] [Indexed: 12/20/2022] Open
Abstract
Transforming growth factor beta (TGFβ) is a major signalling pathway in joints. This superfamilly is involved in numerous cellular processes in cartilage. Usually, they are considered to favor chondrocyte differentiation and cartilage repair. However, other studies show also deleterious effects of TGFβ which may induce hypertrophy. This may be explained at least in part by alteration of TGFβ signaling pathways in aging chondrocytes. This review focuses on the functions of TGFβ in joints and the regulation of its signaling mediators (receptors, Smads) during aging and osteoarthritis.
Collapse
Affiliation(s)
| | - Nicolas Girard
- Normandie Univ, France ; UNICAEN, EA4652 MILPAT, Caen, France
| | - Eva Lhuissier
- Normandie Univ, France ; UNICAEN, EA4652 MILPAT, Caen, France
| | - Celine Bazille
- Normandie Univ, France ; UNICAEN, EA4652 MILPAT, Caen, France ; Service d'Anatomie Pathologique, CHU, Caen, France
| | | |
Collapse
|
24
|
Johno H, Kitamura M. Pathological in situ reprogramming of somatic cells by the unfolded protein response. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:644-54. [PMID: 23831328 DOI: 10.1016/j.ajpath.2013.05.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 05/07/2013] [Accepted: 05/10/2013] [Indexed: 12/19/2022]
Abstract
In response to tissue injuries, terminally differentiated cells are reprogrammed to undergo dedifferentiation to gain mitogenic and metabolic properties. The dedifferentiated cells acquire an immature phenotype, proliferate actively, produce abundant extracellular matrix, and recruit circulating leukocytes via secretion of chemokines, contributing to tissue repair and/or fibrosis. However, this remodeling process is self-limiting, and in the later phase, the activated, dedifferentiated cells are reprogrammed to redifferentiate into a mature, quiescent phenotype. Currently, molecular mechanisms underlying this bidirectional pathological reprogramming remain elusive. It is known that the unfolded protein response (UPR) is induced at local tissues under pathological situations and affects cellular fate-survival or death. It is also known that the UPR is involved in cell differentiation and organogenesis during embryonic development. In this review, we describe a hypothesis for regulatory roles of the UPR in the pathological reprogramming of somatic cells (ie, cellular dedifferentiation and redifferentiation at the sites of injury).
Collapse
Affiliation(s)
- Hisashi Johno
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | | |
Collapse
|