1
|
Atasoy-Zeybek A, Showel KK, Nagelli CV, Westendorf JJ, Evans CH. The intersection of aging and estrogen in osteoarthritis. NPJ WOMEN'S HEALTH 2025; 3:15. [PMID: 40017990 PMCID: PMC11860234 DOI: 10.1038/s44294-025-00063-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/03/2025] [Indexed: 03/01/2025]
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by cartilage degradation, inflammation, and pain. While multiple factors contribute to OA development, age and sex are primary risk factors, particularly affecting postmenopausal women. The dramatic increase in OA risk after menopause suggests estrogen deficiency accelerates disease progression. This review explores the molecular mechanisms connecting aging and estrogen deficiency in OA development, focusing on key genes and pathways identified through RNA sequencing.
Collapse
Affiliation(s)
- Aysegul Atasoy-Zeybek
- Musculoskeletal Gene Therapy Research Laboratory, Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN USA
| | - Kelly K. Showel
- Musculoskeletal Gene Therapy Research Laboratory, Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN USA
- Department of Pharmacology, Mayo Clinic, Rochester, MN USA
| | - Christopher V. Nagelli
- Musculoskeletal Gene Therapy Research Laboratory, Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN USA
| | | | - Christopher H. Evans
- Musculoskeletal Gene Therapy Research Laboratory, Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN USA
| |
Collapse
|
2
|
Antoinette AY, Ziemian SN, Brown AR, Hudson EB, Chlebek C, Wright TM, Goldring SR, Goldring MB, Otero M, van der Meulen MC. PTH treatment before cyclic joint loading improves cartilage health and attenuates load-induced osteoarthritis development in mice. SCIENCE ADVANCES 2024; 10:eadk8402. [PMID: 38640238 PMCID: PMC11029811 DOI: 10.1126/sciadv.adk8402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 03/18/2024] [Indexed: 04/21/2024]
Abstract
Osteoarthritis (OA) treatment is limited by the lack of effective nonsurgical interventions to slow disease progression. Here, we examined the contributions of the subchondral bone properties to OA development. We used parathyroid hormone (PTH) to modulate bone mass before OA initiation and alendronate (ALN) to inhibit bone remodeling during OA progression. We examined the spatiotemporal progression of joint damage by combining histopathological and transcriptomic analyses across joint tissues. The additive effect of PTH pretreatment before OA initiation and ALN treatment during OA progression most effectively attenuated load-induced OA pathology. Individually, PTH directly improved cartilage health and slowed the development of cartilage damage, whereas ALN primarily attenuated subchondral bone changes associated with OA progression. Joint damage reflected early transcriptomic changes. With both treatments, the structural changes were associated with early modulation of immunoregulation and immunoresponse pathways that may contribute to disease mechanisms. Overall, our results demonstrate the potential of subchondral bone-modifying therapies to slow the progression of OA.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Miguel Otero
- Hospital for Special Surgery, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | | |
Collapse
|
3
|
Liao S, Yang M, Li D, Wu Y, Sun H, Lu J, Liu X, Deng T, Wang Y, Xie N, Tang D, Nie G, Fan X. Comprehensive bulk and single-cell transcriptome profiling give useful insights into the characteristics of osteoarthritis associated synovial macrophages. Front Immunol 2023; 13:1078414. [PMID: 36685529 PMCID: PMC9849898 DOI: 10.3389/fimmu.2022.1078414] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/06/2022] [Indexed: 01/07/2023] Open
Abstract
Background Osteoarthritis (OA) is a common chronic joint disease, but the association between molecular and cellular events and the pathogenic process of OA remains unclear. Objective The study aimed to identify key molecular and cellular events in the processes of immune infiltration of the synovium in OA and to provide potential diagnostic and therapeutic targets. Methods To identify the common differential expression genes and function analysis in OA, we compared the expression between normal and OA samples and analyzed the protein-protein interaction (PPI). Additionally, immune infiltration analysis was used to explore the differences in common immune cell types, and Gene Set Variation Analysis (GSVA) analysis was applied to analyze the status of pathways between OA and normal groups. Furthermore, the optimal diagnostic biomarkers for OA were identified by least absolute shrinkage and selection operator (LASSO) models. Finally, the key role of biomarkers in OA synovitis microenvironment was discussed through single cell and Scissor analysis. Results A total of 172 DEGs (differentially expressed genes) associated with osteoarticular synovitis were identified, and these genes mainly enriched eight functional categories. In addition, immune infiltration analysis found that four immune cell types, including Macrophage, B cell memory, B cell, and Mast cell were significantly correlated with OA, and LASSO analysis showed that Macrophage were the best diagnostic biomarkers of immune infiltration in OA. Furthermore, using scRNA-seq dataset, we also analyzed the cell communication patterns of Macrophage in the OA synovial inflammatory microenvironment and found that CCL, MIF, and TNF signaling pathways were the mainly cellular communication pathways. Finally, Scissor analysis identified a population of M2-like Macrophages with high expression of CD163 and LYVE1, which has strong anti-inflammatory ability and showed that the TNF gene may play an important role in the synovial microenvironment of OA. Conclusion Overall, Macrophage is the best diagnostic marker of immune infiltration in osteoarticular synovitis, and it can communicate with other cells mainly through CCL, TNF, and MIF signaling pathways in microenvironment. In addition, TNF gene may play an important role in the development of synovitis.
Collapse
Affiliation(s)
- Shengyou Liao
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Ming Yang
- Department of Otolaryngology, Shenzhen First People’s Hospital, The Affiliated Hospital of Jinan University, Shenzhen, Guangdong, China
| | - Dandan Li
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, China
| | - Ye Wu
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China,Department of Otolaryngology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hong Sun
- The Bio-bank of Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Jingxiao Lu
- The Bio-bank of Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Xinying Liu
- The Bio-bank of Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Tingting Deng
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Yujie Wang
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Ni Xie
- The Bio-bank of Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Donge Tang
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, China
| | - Guohui Nie
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China,State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China,*Correspondence: Guohui Nie, ; Xiaoqin Fan,
| | - Xiaoqin Fan
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China,The Bio-bank of Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China,*Correspondence: Guohui Nie, ; Xiaoqin Fan,
| |
Collapse
|
4
|
Direct comparison of non-osteoarthritic and osteoarthritic synovial fluid-induced intracellular chondrocyte signaling and phenotype changes. Osteoarthritis Cartilage 2023; 31:60-71. [PMID: 36150677 DOI: 10.1016/j.joca.2022.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Since the joint microenvironment and tissue homeostasis are highly dependent on synovial fluid, we aimed to compare the essential chondrocyte signaling signatures of non-osteoarthritic vs end-stage osteoarthritic knee synovial fluid. Moreover, we determined the phenotypic consequence of the distinct signaling patterns on articular chondrocytes. METHODS Protein profiling of synovial fluid was performed using antibody arrays. Chondrocyte signaling and phenotypic changes induced by non-osteoarthritic and osteoarthritic synovial fluid were analyzed using a phospho-kinase array, luciferase-based transcription factor activity assays, and RT-qPCR. The origin of osteoarthritic synovial fluid signaling was evaluated by comparing the signaling responses of conditioned media from cartilage, synovium, infrapatellar fat pad and meniscus. Osteoarthritic synovial fluid induced pathway-phenotype relationships were evaluated using pharmacological inhibitors. RESULTS Compared to non-osteoarthritic synovial fluid, osteoarthritic synovial fluid was enriched in cytokines, chemokines and growth factors that provoked differential MAPK, AKT, NFκB and cell cycle signaling in chondrocytes. Functional pathway analysis confirmed increased activity of these signaling events upon osteoarthritic synovial fluid stimulation. Tissue secretomes of osteoarthritic cartilage, synovium, infrapatellar fat pad and meniscus activated several inflammatory signaling routes. Furthermore, the distinct pathway signatures of osteoarthritic synovial fluid led to accelerated chondrocyte dedifferentiation via MAPK/ERK signaling, increased chondrocyte fibrosis through MAPK/JNK and PI3K/AKT activation, an elevated inflammatory response mediated by cPKC/NFκB, production of extracellular matrix-degrading enzymes by MAPK/p38 and PI3K/AKT routes, and enabling of chondrocyte proliferation. CONCLUSION This study provides the first mechanistic comparison between non-osteoarthritic and osteoarthritic synovial fluid, highlighting MAPKs, cPKC/NFκB and PI3K/AKT as crucial OA-associated intracellular signaling routes.
Collapse
|
5
|
Jiang P, Sun S, Zhang J, Li C, Ma G, Wang J, Chen F, Liao DJ. RNA expression profiling from the liquid fraction of synovial fluid in knee joint osteoarthritis patients. Am J Transl Res 2022; 14:6782-6791. [PMID: 36247259 PMCID: PMC9556501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/20/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVE To investigate the RNA profile of synovial fluid (SF) from osteoarthritis (OA) patients and carry out cluster analysis of OA-related genes. METHODS RNA of SF from OA patients was isolated using RNA-specific Trizol. A cDNA library was built and subjected to the second-generation sequencing using HisSeq4000 with a data size of 8G. The sequencing reads were aligned to the UCSC human reference genome (hg38) using Tophat with default parameters. Gene function enrichment was generated using DAVID. RESULTS The minimum weight 0.096 µg RNA of SF sample was used for sequencing analysis, which produced 66,154,562 clean reads, 91.28% of which were matched to the reference with 2,682 genes identified. Some of the unmatchable reads matched RNAs of bacteria, mainly Pseudomonas. The detected human RNAs in samples fell into different categories of genes, including protein-coding ones, processed and unprocessed pseudogenes, and long noncoding, antisense and miscellaneous RNAs that mediate various biological functions. Interestingly, 80% of the expressed genes belonged to the mitochondrial genome. CONCLUSION These results suggest that less than 0.1 µg RNA is sufficient for establishing a cDNA library and deep sequencing, and that the liquid fraction of SF contains a whole RNA repertoire that may reflect a history of previous microorganism infections.
Collapse
Affiliation(s)
- Peng Jiang
- School of Clinical Medicine, Shandong UniversityJinan 250100, Shandong Province, China
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan 250021, Shandong Province, China
| | - Shui Sun
- School of Clinical Medicine, Shandong UniversityJinan 250100, Shandong Province, China
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan 250021, Shandong Province, China
| | - Ju Zhang
- CAS Key Laboratory of Genomics Sciences and Information, Beijing Institute of Genomics, Chinese Academy of SciencesBeijing 100101, China
| | - Cuidan Li
- CAS Key Laboratory of Genomics Sciences and Information, Beijing Institute of Genomics, Chinese Academy of SciencesBeijing 100101, China
| | - Guannan Ma
- CAS Key Laboratory of Genomics Sciences and Information, Beijing Institute of Genomics, Chinese Academy of SciencesBeijing 100101, China
| | - Jian Wang
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan 250021, Shandong Province, China
| | - Fei Chen
- CAS Key Laboratory of Genomics Sciences and Information, Beijing Institute of Genomics, Chinese Academy of SciencesBeijing 100101, China
| | - Dezhong Joshua Liao
- Department of Pathology, and Key Lab of Endemic and Ethnic Diseases of the Ministry of Education of China in Guizhou Medical UniversityGuiyang 550004, Guizhou Province, China
| |
Collapse
|
6
|
Wang K, Esbensen Q, Karlsen T, Eftang C, Owesen C, Aroen A, Jakobsen R. Low-Input RNA-Sequencing in Patients with Cartilage Lesions, Osteoarthritis, and Healthy Cartilage. Cartilage 2021; 13:550S-562S. [PMID: 34775802 PMCID: PMC8808811 DOI: 10.1177/19476035211057245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
OBJECTIVE To analyze and compare cartilage samples from 3 groups of patients utilizing low-input RNA-sequencing. DESIGN Cartilage biopsies were collected from patients in 3 groups (n = 48): Cartilage lesion (CL) patients had at least ICRS grade 2, osteoarthritis (OA) samples were taken from patients undergoing knee replacement, and healthy cartilage (HC) was taken from ACL-reconstruction patients without CLs. RNA was isolated using an optimized protocol. RNA samples were assessed for quality and sequenced with a low-input SmartSeq2 protocol. RESULTS RNA isolation yielded 48 samples with sufficient quality for sequencing. After quality control, 13 samples in the OA group, 9 in the HC group, and 9 in the CL group were included in the analysis. There was a high degree of co-clustering between the HC and CL groups with only 6 genes significantly up- or downregulated. OA and the combined HC/CL group clustered significantly separate from each other, yielding 659 significantly upregulated and 1,369 downregulated genes. GO-term analysis revealed that genes matched to cartilage and connective tissue development terms. CONCLUSION The gene expression profiles from the 3 groups suggest that there are no major differences in gene expression between cartilage from knees with a cartilage injury and knees without an apparent cartilage injury. OA cartilage, as expected, showed markedly different gene expression from the other 2 groups. The gene expression profiles resulting from this low-input RNA-sequencing study offer opportunities to discover new pathways not previously recognized that may be explored in future studies.
Collapse
Affiliation(s)
- Katherine Wang
- Faculty of Medicine, University of
Oslo, Oslo, Norway,Oslo Sports Trauma Research Center,
Norwegian School of Sports Sciences, Oslo, Norway,Department of Orthopaedic Surgery,
Akershus University Hospital, Lørenskog, Norway,Katherine Wang, Faculty of Medicine,
University of Oslo, P.O. Box 1072 Blindern, 0316 Oslo, Norway.
| | - Q.Y. Esbensen
- Department of Clinical Molecular
Biology (EpiGen), Akershus University Hospital, Lørenskog, Norway,Department of Clinical Molecular
Biology, University of Oslo, Oslo, Norway
| | - T.A. Karlsen
- Norwegian Center for Stem Cell
Research, Department of Immunology and Transfusion Medicine, Oslo University
Hospital, Rikshospitalet, Oslo, Norway
| | - C.N. Eftang
- Department of Pathology, Akershus
University Hospital, Lørenskog, Norway
| | - C. Owesen
- Department of Orthopaedic Surgery,
Akershus University Hospital, Lørenskog, Norway
| | - A. Aroen
- Oslo Sports Trauma Research Center,
Norwegian School of Sports Sciences, Oslo, Norway,Department of Orthopaedic Surgery,
Akershus University Hospital, Lørenskog, Norway,Institute of Clinical Medicine, Faculty
of Medicine, University of Oslo, Oslo, Norway
| | - R.B. Jakobsen
- Department of Orthopaedic Surgery,
Akershus University Hospital, Lørenskog, Norway,Department of Health Management and
Health Economics, Institute of Health and Society, Faculty of Medicine, University
of Oslo, Oslo, Norway
| |
Collapse
|
7
|
Pemmari A, Leppänen T, Hämäläinen M, Moilanen T, Moilanen E. Chondrocytes from Osteoarthritis Patients Adopt Distinct Phenotypes in Response to Central T H1/T H2/T H17 Cytokines. Int J Mol Sci 2021; 22:ijms22179463. [PMID: 34502384 PMCID: PMC8431052 DOI: 10.3390/ijms22179463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/30/2021] [Accepted: 08/06/2021] [Indexed: 12/27/2022] Open
Abstract
Chronic low-grade inflammation plays a central role in the pathogenesis of osteoarthritis (OA), and several pro- and anti-inflammatory cytokines have been implicated to mediate and regulate this process. Out of these cytokines, particularly IFNγ, IL-1β, IL-4 and IL-17 are associated with different phenotypes of T helper (TH) cells and macrophages, both examples of cells known for great phenotypic and functional heterogeneity. Chondrocytes also display various phenotypic changes during the course of arthritis. We set out to study the hypothesis of whether chondrocytes might adopt polarized phenotypes analogous to TH cells and macrophages. We studied the effects of IFNγ, IL-1β, IL-4 and IL-17 on gene expression in OA chondrocytes with RNA-Seq. Chondrocytes were harvested from the cartilage of OA patients undergoing knee replacement surgery and then cultured with or without the cytokines for 24 h. Total RNA was isolated and sequenced, and GO (Gene Ontology) functional analysis was performed. We also separately investigated genes linked to OA in recent genome wide expression analysis (GWEA) studies. The expression of more than 2800 genes was significantly altered in chondrocytes treated with IL-1β [in the C(IL-1β) phenotype] with a fold change (FC) > 2.5 in either direction. These included a large number of genes associated with inflammation, cartilage degradation and attenuation of metabolic signaling. The profile of genes differentially affected by IFNγ (the C(IFNγ) phenotype) was relatively distinct from that of the C(IL-1β) phenotype and included several genes associated with antigen processing and presentation. The IL-17-induced C(IL-17) phenotype was characterized by the induction of a more limited set of proinflammatory factors compared to C(IL-1β) cells. The C(IL-4) phenotype induced by IL-4 displayed a differential expression of a rather small set of genes compared with control, primarily those associated with TGFβ signaling and the regulation of inflammation. In conclusion, our results show that OA chondrocytes can adopt diverse phenotypes partly analogously to TH cells and macrophages. This phenotypic plasticity may play a role in the pathogenesis of arthritis and open new therapeutic avenues for the development of disease-modifying treatments for (osteo)arthritis.
Collapse
Affiliation(s)
- Antti Pemmari
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, University of Tampere and Tampere University Hospital, 33100 Tampere, Finland; (A.P.); (T.L.); (M.H.)
| | - Tiina Leppänen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, University of Tampere and Tampere University Hospital, 33100 Tampere, Finland; (A.P.); (T.L.); (M.H.)
| | - Mari Hämäläinen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, University of Tampere and Tampere University Hospital, 33100 Tampere, Finland; (A.P.); (T.L.); (M.H.)
| | - Teemu Moilanen
- Coxa Hospital for Joint Replacement, 33520 Tampere, Finland;
| | - Eeva Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, University of Tampere and Tampere University Hospital, 33100 Tampere, Finland; (A.P.); (T.L.); (M.H.)
- Correspondence:
| |
Collapse
|
8
|
Kubrova E, Su M, Galeano-Garces C, Galvan ML, Jerez S, Dietz AB, Smith J, Qu W, van Wijnen AJ. Differences in Cytotoxicity of Lidocaine, Ropivacaine, and Bupivacaine on the Viability and Metabolic Activity of Human Adipose-Derived Mesenchymal Stem Cells. Am J Phys Med Rehabil 2021; 100:82-91. [PMID: 32657816 PMCID: PMC11784493 DOI: 10.1097/phm.0000000000001529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE We evaluated biological effects of distinct local anesthetics on human adipose-derived mesenchymal stem cells when applied to reduce periprocedural pain during mesenchymal stem cell injections. METHODS AND MATERIALS Metabolic activity (MTS assay), viability (Live/Dead stain), and gene expression (quantitative real-time reverse-transcriptase polymerase chain reaction) were measured in mesenchymal stem cells incubated with various concentrations of lidocaine, ropivacaine, or bupivacaine during a 12-hr time course. RESULTS Cell viability and metabolic activity decreased in a dose, time, and substance-specific manner after exposure to lidocaine, ropivacaine, and bupivacaine, with ropivacaine being the least cytotoxic. Cell viability decreases after brief exposure (<1.5 hrs) at clinically relevant concentrations (eg, 8 mg/ml of lidocaine, 2.5 mg/ml of ropivacaine or bupivacaine). Mesenchymal stem cells exposed to local anesthetics change their expression of mRNA biomarkers for stress response (EGR1, EGR2), proliferation (MKI67, HIST2H4A), ECM (COL1A1, COL3A1), and cell surface marker (CD105). CONCLUSIONS Local anesthetics are cytotoxic to clinical-grade human mesenchymal stem cells in a dose-, time-, and agent-dependent manner and change expression of ECM, proliferation, and cell surface markers. Lidocaine and bupivacaine are more cytotoxic than ropivacaine. Single-dose injections of local anesthetics may affect the biological properties of mesenchymal stem cells in vitro but may not affect the effective dose of MSCs in a clinical setting.
Collapse
Affiliation(s)
- Eva Kubrova
- From the Department of Physical Medicine & Rehabilitation, Mayo Clinic, Rochester, Minnesota (EK, MS, JS, WQ); Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota (EK, MS, CG-G, MLG, SJ, AJvW); Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, Minnesota (MS, CG-G, SJ, AJvW); Department of Physical Medicine & Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, China (MS); and Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (ABD)
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Pemmari A, Leppänen T, Hämäläinen M, Moilanen T, Vuolteenaho K, Moilanen E. Widespread regulation of gene expression by glucocorticoids in chondrocytes from patients with osteoarthritis as determined by RNA-Seq. Arthritis Res Ther 2020; 22:271. [PMID: 33203447 PMCID: PMC7670667 DOI: 10.1186/s13075-020-02289-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023] Open
Abstract
Background Intra-articular glucocorticoid (GC) injections are widely used as a symptomatic treatment for osteoarthritis (OA). However, there are also concerns about their potentially harmful effects, and their detailed effects on chondrocyte phenotype remain poorly understood. Methods We studied the effects of dexamethasone on gene expression in OA chondrocytes with RNA-Seq. Chondrocytes were isolated from the cartilage from OA patients undergoing knee replacement surgery and cultured with or without dexamethasone for 24 h. Total RNA was isolated and sequenced, and functional analysis was performed against the Gene Ontology (GO) database. Results for selected genes were confirmed with RT-PCR. We also investigated genes linked to OA in recent genome-wide expression analysis (GWEA) studies. Results Dexamethasone increased the expression of 480 and reduced that of 755 genes with a fold change (FC) 2.0 or greater. Several genes associated with inflammation and cartilage anabolism/catabolism as well as lipid and carbohydrate metabolism were among the most strongly affected genes. In the GO analysis, genes involved in the extracellular matrix organization, cell proliferation and adhesion, inflammation, and collagen synthesis were enriched among the significantly affected genes. In network analysis, NGF, PI3KR1, and VCAM1 were identified as central genes among those most strongly affected by dexamethasone. Conclusions This is the first study investigating the genome-wide effects of GCs on the gene expression in OA chondrocytes. In addition to clear anti-inflammatory and anticatabolic effects, GCs affect lipid and glucose metabolism in chondrocytes, an observation that might be particularly important in the metabolic phenotype of OA.
Collapse
Affiliation(s)
- Antti Pemmari
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Tiina Leppänen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Mari Hämäläinen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Teemu Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland.,Coxa Hospital for Joint Replacement, Tampere, Finland
| | - Katriina Vuolteenaho
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Eeva Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland.
| |
Collapse
|
10
|
Schivo S, Khurana S, Govindaraj K, Scholma J, Kerkhofs J, Zhong L, Huang X, van de Pol J, Langerak R, van Wijnen AJ, Geris L, Karperien M, Post JN. ECHO, the executable CHOndrocyte: A computational model to study articular chondrocytes in health and disease. Cell Signal 2020; 68:109471. [DOI: 10.1016/j.cellsig.2019.109471] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/01/2019] [Accepted: 11/12/2019] [Indexed: 12/18/2022]
|
11
|
Kubrova E, Qu W, Galvan ML, Paradise CR, Yang J, Dietz AB, Dudakovic A, Smith J, van Wijnen AJ. Hypothermia and nutrient deprivation alter viability of human adipose-derived mesenchymal stem cells. Gene 2019; 722:144058. [PMID: 31494240 DOI: 10.1016/j.gene.2019.144058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 08/17/2019] [Indexed: 12/11/2022]
Abstract
PURPOSE Adipose-derived mesenchymal stem cells (MSCs) are attractive biological agents in regenerative medicine. To optimize cell therapies, it is necessary to determine the most effective delivery method for MSCs. Therefore, we evaluated the biological properties of MSCs after exposure to various temperatures to define optimal storage conditions prior to therapeutic delivery of MSCs. DESIGN Prospective observational study. METHODS AND MATERIALS Adherent and non-adherent MSCs were incubated at multiple temperatures (i.e., 4, 23 and 37 °C) in Lactated Ringers (LR) solution lacking essential cell growth ingredients, or in culture media which is optimized for cell growth. Cells were assessed either after the temperature changes (4 h) or after recovery (24 h). Metabolic activity of MSCs, cell number and expression of representative mRNA biomarkers were evaluated to assess the biological effects of temperature. We monitored changes in mRNAs expression related to cytoprotective- or stress-related responses (e.g., FOS, JUN, ATF1, ATF4, EGR1, EGR2, MYC), proliferation (e.g., HIST2H4, CCNB2), and extracellular matrix production (ECM; e.g., COL3A1, COL1A1) by quantitative real time reverse-transcriptase polymerase chain reaction (RT-qPCR) analysis. RESULTS Our study demonstrates that storing MSCs in Lactated Ringers (LR) solution for 4 h decreases cell number and metabolic activity. The number of viable MSCs decreased significantly when cultured at physiological temperature (37 °C) and severe hypothermia (4 °C), while cells grown at ambient temperature (23 °C) exhibited the least detrimental effects. There were no appreciable biological differences in mRNA markers for proliferation or ECM deposition at any of the temperatures. However, biomarkers related to cytoprotective- or stress-responses were selectively elevated depending on temperature or media type (i.e., LR versus standard media). CONCLUSION The biological impact of nutrient-free media and temperature changes after 4 h exposure persists after a 24 h recovery period. Hence, storage temperature and media conditions should be optimized to improve effective dosing of MSCs.
Collapse
Affiliation(s)
- Eva Kubrova
- Department of Physical Medicine &Rehabilitation, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, United States of America; Department of Orthopedic Surgery, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, United States of America; Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Wenchun Qu
- Department of Physical Medicine &Rehabilitation, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, United States of America.
| | - M Lizeth Galvan
- Department of Orthopedic Surgery, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, United States of America; Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Christopher R Paradise
- Department of Orthopedic Surgery, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, United States of America; Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, United States of America; Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States of America
| | - Juan Yang
- Department of Physical Medicine &Rehabilitation, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Allan B Dietz
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States of America
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, United States of America; Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Jay Smith
- Department of Physical Medicine &Rehabilitation, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, United States of America; Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, United States of America.
| |
Collapse
|
12
|
Nie H, Kubrova E, Wu T, Denbeigh JM, Hunt C, Dietz AB, Smith J, Qu W, van Wijnen AJ. Effect of Lidocaine on Viability and Gene Expression of Human Adipose-derived Mesenchymal Stem Cells: An in vitro Study. PM R 2019; 11:1218-1227. [PMID: 30784215 DOI: 10.1002/pmrj.12141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/28/2019] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To assess the biologic effects of lidocaine on the viability, proliferation, and function of human adipose tissue-derived mesenchymal stromal/stem cells (MSCs) in vitro. METHODS Adipose-derived MSCs from three donors were exposed to lidocaine at various dilutions (2 mg/mL to 8 mg/mL) and exposure times (0.5 to 4 hours). Cell number and viability, mitochondrial activity, and real-time reverse-transcriptase quantitative polymerase chain reaction (RT-qPCR) were analyzed at 0 (immediate effects) or 24 and 48 hours (recovery effects) after treatment with lidocaine. RESULTS Trypan blue staining showed that increasing concentrations of lidocaine decreased the number of observable viable cells. 3-[4,5,dimethylthiazol-2-yl]-5-[3-carboxymethoxy-phenyl]-2-[4-sulfophenyl]-2H-tetrazolium (MTS) assays revealed a concentration- and time- dependent decline of mitochondrial activity and proliferative ability. Gene expression analysis by RT-qPCR revealed that adipose-derived MSCs exposed to lidocaine express robust levels of stress response/cytoprotective genes. However, higher concentrations of lidocaine caused a significant downregulation of these genes. No significant differences were observed in expression of extracellular matrix (ECM) markers COL1A1 and DCN except for COL3A1 (P < .05). Levels of messenger RNA (mRNA) for proliferation markers (CCNB2, HIST2H4A, P < .001) and MKI67 (P < .001) increased at 24 and 48 hours. Expression levels of several transcription factors- including SP1, PRRX1, and ATF1-were modulated in the same manner. MSC surface markers CD44 and CD105 demonstrated decreased expression immediately after treatment, but at 24 and 48 hours postexposure, the MSC markers showed no significant difference among groups. CONCLUSION Lidocaine is toxic to MSCs in a dose- and time- dependent manner. MSC exposure to high (4-8 mg/mL) concentrations of lidocaine for prolonged periods can affect their biologic functions. Although the exposure time in vivo is short, it is essential to choose safe concentrations when applying lidocaine along with MSCs to avoid compromising the viability and potency of the stem cell therapy.
Collapse
Affiliation(s)
- Hai Nie
- Department of Orthopedic Surgery Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN.,Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN
| | - Eva Kubrova
- Department of Orthopedic Surgery Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN.,Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN
| | - Tao Wu
- Department of Physical Medicine & Rehabilitation, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN
| | - Janet M Denbeigh
- Department of Orthopedic Surgery Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN
| | - Christine Hunt
- Department of Physical Medicine & Rehabilitation, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN
| | - Allan B Dietz
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN
| | - Jay Smith
- Department of Physical Medicine & Rehabilitation, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN
| | - Wenchun Qu
- Department of Physical Medicine & Rehabilitation, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN
| | - Andre J van Wijnen
- Department of Orthopedic Surgery Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN.,Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
13
|
Chen YJ, Chang WA, Wu LY, Hsu YL, Chen CH, Kuo PL. Systematic Analysis of Transcriptomic Profile of Chondrocytes in Osteoarthritic Knee Using Next-Generation Sequencing and Bioinformatics. J Clin Med 2018; 7:E535. [PMID: 30544699 PMCID: PMC6306862 DOI: 10.3390/jcm7120535] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/05/2018] [Accepted: 12/07/2018] [Indexed: 12/28/2022] Open
Abstract
The phenotypic change of chondrocytes and the interplay between cartilage and subchondral bone in osteoarthritis (OA) has received much attention. Structural changes with nerve ingrowth and vascular penetration within OA cartilage may contribute to arthritic joint pain. The aim of this study was to identify differentially expressed genes and potential miRNA regulations in OA knee chondrocytes through next-generation sequencing and bioinformatics analysis. Results suggested the involvement of SMAD family member 3 (SMAD3) and Wnt family member 5A (WNT5A) in the growth of blood vessels and cell aggregation, representing features of cartilage damage in OA. Additionally, 26 dysregulated genes with potential miRNA⁻mRNA interactions were identified in OA knee chondrocytes. Myristoylated alanine rich protein kinase C substrate (MARCKS), epiregulin (EREG), leucine rich repeat containing 15 (LRRC15), and phosphodiesterase 3A (PDE3A) expression patterns were similar among related OA cartilage, subchondral bone and synovial tissue arrays in Gene Expression Omnibus database. The Ingenuity Pathway Analysis identified MARCKS to be associated with the outgrowth of neurite, and novel miRNA regulations were proposed to play critical roles in the pathogenesis of the altered OA knee joint microenvironment. The current findings suggest new perspectives in studying novel genes potentially contributing to arthritic joint pain in knee OA, which may assist in finding new targets for OA treatment.
Collapse
Affiliation(s)
- Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Wei-An Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Ling-Yu Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chia-Hsin Chen
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 804, Taiwan.
| |
Collapse
|
14
|
Paradise CR, Galeano-Garces C, Galeano-Garces D, Dudakovic A, Milbrandt TA, Saris DBF, Krych AJ, Karperien M, Ferguson GB, Evseenko D, Riester SM, van Wijnen AJ, Larson AN. Molecular characterization of physis tissue by RNA sequencing. Gene 2018; 668:87-96. [PMID: 29775757 PMCID: PMC5994380 DOI: 10.1016/j.gene.2018.05.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 05/11/2018] [Indexed: 12/15/2022]
Abstract
The physis is a well-established and anatomically distinct cartilaginous structure that is crucial for normal long-bone development and growth. Abnormalities in physis function are linked to growth plate disorders and other pediatric musculoskeletal diseases. Understanding the molecular pathways operative in the physis may permit development of regenerative therapies to complement surgically-based procedures that are the current standard of care for growth plate disorders. Here, we performed next generation RNA sequencing on mRNA isolated from human physis and other skeletal tissues (e.g., articular cartilage and bone; n = 7 for each tissue). We observed statistically significant enrichment of gene sets in the physis when compared to the other musculoskeletal tissues. Further analysis of these upregulated genes identified physis-specific networks of extracellular matrix proteins including collagens (COL2A1, COL6A1, COL9A1, COL14A1, COL16A1) and matrilins (MATN1, MATN2, MATN3), and signaling proteins in the WNT pathway (WNT10B, FZD1, FZD10, DKK2) or the FGF pathway (FGF10, FGFR4). Our results provide further insight into the gene expression networks that contribute to the physis' unique structural composition and regulatory signaling networks. Physis-specific expression profiles may guide ongoing initiatives in tissue engineering and cell-based therapies for treatment of growth plate disorders and growth modulation therapies. Furthermore, our findings provide new leads for therapeutic drug discovery that would permit future intervention through pharmacological rather than surgical strategies.
Collapse
Affiliation(s)
- Christopher R Paradise
- Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA; Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
| | - Catalina Galeano-Garces
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA; Department of Developmental BioEngineering, University of Twente, Enschede, Netherlands
| | | | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Todd A Milbrandt
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Daniel B F Saris
- Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA; Department of Developmental BioEngineering, University of Twente, Enschede, Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Aaron J Krych
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA; Department of Sports Medicine, Mayo Clinic, Rochester, MN, USA
| | - Marcel Karperien
- Department of Developmental BioEngineering, University of Twente, Enschede, Netherlands
| | - Gabriel B Ferguson
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, CA, USA
| | - Denis Evseenko
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, CA, USA
| | - Scott M Riester
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Andre J van Wijnen
- Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| | - A Noelle Larson
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
15
|
Galuzzi M, Perteghella S, Antonioli B, Tosca MC, Bari E, Tripodo G, Sorrenti M, Catenacci L, Mastracci L, Grillo F, Marazzi M, Torre ML. Human Engineered Cartilage and Decellularized Matrix as an Alternative to Animal Osteoarthritis Model. Polymers (Basel) 2018; 10:738. [PMID: 30960663 PMCID: PMC6403588 DOI: 10.3390/polym10070738] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 06/28/2018] [Accepted: 07/03/2018] [Indexed: 01/05/2023] Open
Abstract
(1) Objective: to obtain a reproducible, robust, well-defined, and cost-affordable in vitro model of human cartilage degeneration, suitable for drug screening; (2) Methods: we proposed 3D models of engineered cartilage, considering two human chondrocyte sources (articular/nasal) and five culture methods (pellet, alginate beads, silk/alginate microcarriers, and decellularized cartilage). Engineered cartilages were treated with pro-inflammatory cytokine IL-1β to promote cartilage degradation; (3) Results: articular chondrocytes have been rejected since they exhibit low cellular doubling with respect to nasal cells, with longer culture time for cell expansion; furthermore, pellet and alginate bead cultures lead to insufficient cartilage matrix production. Decellularized cartilage resulted as good support for degeneration model, but long culture time and high cell amount are required to obtain the adequate scaffold colonization. Here, we proposed, for the first time, the combined use of decellularized cartilage, as aggrecanase substrate, with pellet, alginate beads, or silk/alginate microcarriers, as polymeric scaffolds for chondrocyte cultures. This approach enables the development of suitable models of cartilaginous pathology. The results obtained after cryopreservation also demonstrated that beads and microcarriers are able to preserve chondrocyte functionality and metabolic activity; (4) Conclusions: alginate and silk/alginate-based scaffolds can be easily produced and cryopreserved to obtain a cost-affordable and ready-to-use polymer-based product for the subsequent screening of anti-inflammatory drugs for cartilage diseases.
Collapse
Affiliation(s)
- Marta Galuzzi
- Tissue Therapy Unit, ASST Niguarda Hospital, Piazza Ospedale Maggiore 3, 20162 Milan, Italy.
| | - Sara Perteghella
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
- PharmaExceed S.r.l., 27100 Pavia, Italy.
| | - Barbara Antonioli
- Tissue Therapy Unit, ASST Niguarda Hospital, Piazza Ospedale Maggiore 3, 20162 Milan, Italy.
| | - Marta Cecilia Tosca
- Tissue Therapy Unit, ASST Niguarda Hospital, Piazza Ospedale Maggiore 3, 20162 Milan, Italy.
| | - Elia Bari
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| | - Giuseppe Tripodo
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| | - Milena Sorrenti
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| | - Laura Catenacci
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| | - Luca Mastracci
- Section of Histopathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), IRCCS San Martino IST Hospital, University of Genoa, Largo R. Benzi 8, 16121 Genoa, Italy.
| | - Federica Grillo
- Section of Histopathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), IRCCS San Martino IST Hospital, University of Genoa, Largo R. Benzi 8, 16121 Genoa, Italy.
| | - Mario Marazzi
- Tissue Therapy Unit, ASST Niguarda Hospital, Piazza Ospedale Maggiore 3, 20162 Milan, Italy.
| | - Maria Luisa Torre
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
- PharmaExceed S.r.l., 27100 Pavia, Italy.
| |
Collapse
|
16
|
Sieker JT, Proffen BL, Waller KA, Chin K, Karamchedu NP, Akelman MR, Perrone GS, Kiapour AM, Konrad J, Murray MM, Fleming BC. Transcriptional profiling of articular cartilage in a porcine model of early post-traumatic osteoarthritis. J Orthop Res 2018; 36:318-329. [PMID: 28671352 PMCID: PMC5752630 DOI: 10.1002/jor.23644] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 06/25/2017] [Indexed: 02/04/2023]
Abstract
To identify the molecular pathophysiology present in early post-traumatic osteoarthritis (PTOA), the transcriptional profile of articular cartilage and its response to surgical PTOA induction were determined. Thirty six Yucatan minipigs underwent anterior cruciate ligament (ACL) transection and were randomly assigned in equal numbers to no further treatment, reconstruction or ligament repair. Cartilage was harvested at 1 and 4 weeks post-operatively and histology and RNA-sequencing were performed and compared to controls. Microscopic cartilage scores significantly worsened at 1 (p = 0.028) and 4 weeks (p = 0.001) post-surgery relative to controls, but did not differ between untreated, reconstruction or repair groups. Gene expression after ACL reconstruction and ACL transection were similar, with only 0.03% (including SERPINB7 and CR2) and 0.2% of transcripts (including INHBA) differentially expressed at 1 and 4 weeks respectively. COL2A1, COMP, SPARC, CHAD, and EF1ALPHA were the most highly expressed non ribosomal, non mitochondrial genes in the controls and remained abundant after surgery. A total of 1,275 genes were differentially expressed between 1 and 4 weeks post-surgery. With the treatment groups pooled, 682 genes were differentially expressed at both time-points, with the most significant changes observed in MMP1, COCH, POSTN, CYTL1, and PTGFR. This study confirmed the development of a microscopic PTOA stage after ACL surgery in the porcine model. Upregulation of multiple proteases (including MMP1 and ADAMTS4) were found; however, the level of expression remained orders of magnitude below that of extracellular matrix protein-coding genes (including COL2A1 and ACAN). In summary, genes with established roles in PTOA as well as novel targets for specific intervention were identified. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:318-329, 2018.
Collapse
Affiliation(s)
- Jakob T. Sieker
- Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | | | - Kimberly A. Waller
- Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI
| | - Kaitlyn Chin
- Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI
| | - Naga P. Karamchedu
- Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI
| | - Matthew R. Akelman
- Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI
| | | | - Ata M. Kiapour
- Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | - Johannes Konrad
- Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | | | - Braden C. Fleming
- Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI
| |
Collapse
|
17
|
Lewallen EA, Salib CG, Trousdale WH, Berry CE, Hanssen GM, Robin JX, Tibbo ME, Viste A, Reina N, Morrey ME, Sanchez-Sotelo J, Hanssen AD, Berry DJ, van Wijnen AJ, Abdel MP. Molecular pathology of total knee arthroplasty instability defined by RNA-seq. Genomics 2017; 110:247-256. [PMID: 29174847 DOI: 10.1016/j.ygeno.2017.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 11/03/2017] [Accepted: 11/04/2017] [Indexed: 11/15/2022]
Abstract
Total knee arthroplasty (TKA) is a durable and reliable procedure to alleviate pain and improve joint function. However, failures related to flexion instability sometimes occur. The goal of this study was to define biological differences between tissues from patients with and without flexion instability of the knee after TKA. Human knee joint capsule tissues were collected at the time of primary or revision TKAs and analyzed by RT-qPCR and RNA-seq, revealing novel patterns of differential gene expression between the two groups. Interestingly, genes related to collagen production and extracellular matrix (ECM) degradation were higher in samples from patients with flexion instability. Partitioned clustering analyses further emphasized differential gene expression patterns between sample types that may help guide clinical interpretations of this complication. Future efforts to disentangle the effects of physical and biological (e.g., transcriptomic modifications) risk factors will aid in further characterizing and avoiding flexion instability after TKA.
Collapse
Affiliation(s)
- Eric A Lewallen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States; Department of Biological Sciences, Hampton University, Hampton, VA, United States.
| | - Christopher G Salib
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States.
| | - William H Trousdale
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States.
| | - Charlotte E Berry
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States
| | | | - Joseph X Robin
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Meagan E Tibbo
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States.
| | - Anthony Viste
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Nicolas Reina
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Mark E Morrey
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States.
| | | | - Arlen D Hanssen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States.
| | - Daniel J Berry
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States.
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States; Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN, United States.
| | - Matthew P Abdel
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
18
|
Galeano-Garces C, Camilleri ET, Riester SM, Dudakovic A, Larson DR, Qu W, Smith J, Dietz AB, Im HJ, Krych AJ, Larson AN, Karperien M, van Wijnen AJ. Molecular Validation of Chondrogenic Differentiation and Hypoxia Responsiveness of Platelet-Lysate Expanded Adipose Tissue-Derived Human Mesenchymal Stromal Cells. Cartilage 2017; 8:283-299. [PMID: 28618870 PMCID: PMC5625857 DOI: 10.1177/1947603516659344] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE To determine the optimal environmental conditions for chondrogenic differentiation of human adipose tissue-derived mesenchymal stromal/stem cells (AMSCs). In this investigation we specifically investigate the role of oxygen tension and 3-dimensional (3D) culture systems. DESIGN Both AMSCs and primary human chondrocytes were cultured for 21 days in chondrogenic media under normoxic (21% oxygen) or hypoxic (2% oxygen) conditions using 2 distinct 3D culture methods (high-density pellets and poly-ε-caprolactone [PCL] scaffolds). Histologic analysis of chondro-pellets and the expression of chondrocyte-related genes as measured by reverse transcriptase quantitative polymerase chain reaction were used to evaluate the efficiency of differentiation. RESULTS AMSCs are capable of expressing established cartilage markers including COL2A1, ACAN, and DCN when grown in chondrogenic differentiation media as determined by gene expression and histologic analysis of cartilage markers. Expression of several cartilage-related genes was enhanced by low oxygen tension, including ACAN and HAPLN1. The pellet culture environment also promoted the expression of hypoxia-inducible cartilage markers compared with cells grown on 3D scaffolds. CONCLUSIONS Cell type-specific effects of low oxygen and 3D environments indicate that mesenchymal cell fate and differentiation potential is remarkably sensitive to oxygen. Genetic programming of AMSCs to a chondrocytic phenotype is effective under hypoxic conditions as evidenced by increased expression of cartilage-related biomarkers and biosynthesis of a glycosaminoglycan-positive matrix. Lower local oxygen levels within cartilage pellets may be a significant driver of chondrogenic differentiation.
Collapse
Affiliation(s)
- Catalina Galeano-Garces
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA,Department of Developmental Bioengineering, University of Twente, Enschede, Netherlands
| | | | - Scott M. Riester
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Dirk R. Larson
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Wenchun Qu
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Jay Smith
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Allan B. Dietz
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Hee-Jeong Im
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Aaron J. Krych
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - A. Noelle Larson
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Marcel Karperien
- Department of Developmental Bioengineering, University of Twente, Enschede, Netherlands
| | - Andre J. van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA,Andre J. van Wijnen, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| |
Collapse
|