1
|
Tang R, Harasymowicz NS, Wu CL, Choi YR, Lenz K, Oswald SJ, Guilak F. Gene therapy for fat-1 prevents obesity-induced metabolic dysfunction, cellular senescence, and osteoarthritis. Proc Natl Acad Sci U S A 2024; 121:e2402954121. [PMID: 39401356 PMCID: PMC11513907 DOI: 10.1073/pnas.2402954121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 08/16/2024] [Indexed: 10/18/2024] Open
Abstract
Obesity is one of the primary risk factors for osteoarthritis (OA), acting through cross talk among altered biomechanics, metabolism, adipokines, and dietary free fatty acid (FA) composition. Obesity and aging have been linked to cellular senescence in various tissues, resulting in increased local and systemic inflammation and immune dysfunction. We hypothesized that obesity and joint injury lead to cellular senescence that is typically associated with increased OA severity or with aging and that the ratio of omega-6 (ω-6) to omega-3 (ω-3) FAs regulates these pathologic effects. Mice were placed on an ω-6-rich high-fat diet or a lean control diet and underwent destabilization of the medial meniscus to induce OA. Obesity and joint injury significantly increased cellular senescence in subcutaneous and visceral fat as well as joint tissues such as synovium and cartilage. Using adeno-associated virus (AAV) gene therapy for fat-1, a fatty acid desaturase that converts ω-6 to ω-3 FAs, decreasing the serum ω-6:ω-3 FA ratio had a strong senomorphic and therapeutic effect, mitigating metabolic dysfunction, cellular senescence, and joint degeneration. In vitro coculture of bone marrow-derived macrophages and chondrocytes from control and AAV8-fat1-treated mice were used to examine the roles of various FA mediators in regulating chondrocyte senescence. Our results suggest that obesity and joint injury result in a premature "aging" of the joint as measured by senescence markers, and these changes can be ameliorated by altering FA composition using fat-1 gene therapy. These findings support the potential for fat-1 gene therapy to treat obesity- and/or injury-induced OA clinically.
Collapse
Affiliation(s)
- Ruhang Tang
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO63110
- Shriners Hospitals for Children, St. Louis, MO63110
- Center of Regenerative Medicine, Washington University, St. Louis, MO63110
| | - Natalia S. Harasymowicz
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO63110
- Shriners Hospitals for Children, St. Louis, MO63110
- Center of Regenerative Medicine, Washington University, St. Louis, MO63110
| | - Chia-Lung Wu
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO63110
- Shriners Hospitals for Children, St. Louis, MO63110
| | - Yun-Rak Choi
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO63110
- Shriners Hospitals for Children, St. Louis, MO63110
- Center of Regenerative Medicine, Washington University, St. Louis, MO63110
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul03722, South Korea
| | - Kristin Lenz
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO63110
- Shriners Hospitals for Children, St. Louis, MO63110
- Center of Regenerative Medicine, Washington University, St. Louis, MO63110
| | - Sara J. Oswald
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO63110
- Shriners Hospitals for Children, St. Louis, MO63110
- Center of Regenerative Medicine, Washington University, St. Louis, MO63110
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO63110
- Shriners Hospitals for Children, St. Louis, MO63110
- Center of Regenerative Medicine, Washington University, St. Louis, MO63110
| |
Collapse
|
2
|
Oestreich AK, Harasymowicz NS, Savadipour A, Harissa Z, Rashidi N, Luhmann MK, Kuziez M, Moley KH, Guilak F. Maternal high-fat high-sugar diet impairs bone quality and strength but not cartilage in aging mice. J Orthop Res 2024. [PMID: 39342461 DOI: 10.1002/jor.25980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 07/15/2024] [Accepted: 09/05/2024] [Indexed: 10/01/2024]
Abstract
Osteoarthritis (OA) is a prevalent aging disorder of synovial joints and recent work suggests that a parental high-fat diet increases OA severity following joint injury in offspring. We hypothesized that a maternal high-fat high-sugar (HFHS) diet would promote spontaneous osteoarthritis-related cartilage and bone changes in 1-year-old offspring. Female C57BL/6 J mice were placed on either a chow control or HFHS diet for 6 weeks before mating to a chow-fed C57BL/6 J male and maintained on their assigned diets throughout pregnancy and lactation. Male and female offspring were weaned onto a chow diet, raised to 1 year of age, and evaluated for cartilage and bone changes indicative of OA. However, offspring did not show early signs of OA as measured by histological Mankin scoring, mechanical testing of the pericellular matrix, histological synovitis scoring, or subchondral bone thickening as measured by microcomputed Tomography. On the other hand, male offspring from HFHS-fed dams had reduced trabecular bone quality in the tibial metaphysis and decreased cortical thickness. Although maternal HFHS diet did not impact trabecular or cortical bone quality in tibias of female offspring, the radii of these animals had decreased cortical thickness, increased medullary area, and impaired breaking strength compared to those of control-fed dams. Finally, we evaluated bone quality and strength in male and female F2 offspring and found that the grandmaternal diet modestly impacted radial bone geometry but not strength. Together these results suggest that maternal HFHS diet impairs F1 offspring skeletal integrity in a sex and bone site-specific manner.
Collapse
Affiliation(s)
- Arin K Oestreich
- Department of Orthopaedic Surgery, Washington University in St. Louis, St Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Natalia S Harasymowicz
- Department of Orthopaedic Surgery, Washington University in St. Louis, St Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Alireza Savadipour
- Department of Orthopaedic Surgery, Washington University in St. Louis, St Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Mechanical Engineering, Washington University, St. Louis, Missouri, USA
| | - Zainab Harissa
- Department of Orthopaedic Surgery, Washington University in St. Louis, St Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Neda Rashidi
- Department of Orthopaedic Surgery, Washington University in St. Louis, St Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Mechanical Engineering, Washington University, St. Louis, Missouri, USA
| | - Meredith K Luhmann
- Department of Orthopaedic Surgery, Washington University in St. Louis, St Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Mohammed Kuziez
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Kelle H Moley
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University, St. Louis, Missouri, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University in St. Louis, St Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Mechanical Engineering, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
3
|
Zhang Y, Zhang Z, Yu Q, Lan B, Shi Q, Liu Y, Zhang W, Li F. Dual-factor model of sleep and diet: a new approach to understanding central fatigue. Front Neurosci 2024; 18:1465568. [PMID: 39355851 PMCID: PMC11442446 DOI: 10.3389/fnins.2024.1465568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 08/23/2024] [Indexed: 10/03/2024] Open
Abstract
Background Numerous studies have recently examined the impact of dietary factors such as high-fat diets on fatigue. Our study aims to investigate whether high-fat diet (HFD) alone or combined with alternate-day fasting (ADF) can lead to the central fatigue symptoms and to investigate the potential integration of dietary and sleep variables in the development of central fatigue models. Methods Seventy-five male Wistar rats were divided into five groups: control, HFD, HFD + ADF, modified multiple platform method (MMPM), and MMPM+HFD + ADF. Each group underwent a 21-day modeling period according to their respective protocol. Their behavioral characteristics, fatigue biochemical markers, hippocampal pathological changes, mitochondrial ultrastructure, and oxidative stress damage were analyzed. Results Our findings demonstrate that using only HFD did not cause central fatigue, but combining it with ADF did. This combination led to reduced exercise endurance, decreased locomotor activity, impaired learning and memory abilities, along with alterations in serum levels of alanine aminotransferase (ALT), creatine kinase (CK), and lactate (LAC), as well as hippocampal pathological damage and other central fatigue symptoms. Moreover, the MMPM+HFD + ADF method led to the most obvious central fatigue symptoms in rats, including a variety of behavioral changes, alterations in fatigue-related biochemical metabolic markers, prominent pathological changes in hippocampal tissue, severe damage to the ultrastructure of mitochondria in hippocampal regions, changes in neurotransmitters, and evident oxidative stress damage. Additionally, it was observed that rats subjected to HFD + ADF, MMPM, and MMPM+HFD + ADF modeling method exhibited significant brain oxidative stress damage. Conclusion We have demonstrated the promotive role of dietary factors in the development of central fatigue and have successfully established a more stable and clinically relevant animal model of central fatigue by integrating dietary and sleep factors.
Collapse
Affiliation(s)
- Yifei Zhang
- School of Tradional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zehan Zhang
- School of Tradional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qingqian Yu
- School of Tradional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Bijuan Lan
- School of Tradional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qinghuan Shi
- School of Tradional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Liu
- School of Tradional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Weiyue Zhang
- School of Nursing, Beijing University of Chinese Medicine, Beijing, China
| | - Feng Li
- School of Tradional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
4
|
Kobak KA, Batushansky A, Jopkiewicz A, Peelor FF, Kinter MT, Miller BF, Griffin TM. Effect of biological sex and short-term high-fat diet on cellular proliferation, ribosomal biogenesis, and targeted protein abundance in murine articular cartilage. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100495. [PMID: 39040627 PMCID: PMC11260562 DOI: 10.1016/j.ocarto.2024.100495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/13/2024] [Indexed: 07/24/2024] Open
Abstract
Objective To identify factors contributing to sex-differences in OA risk by evaluating the short-term effect of high-fat (HF) diet on sex-specific changes in cartilage cell proliferation, ribosomal biogenesis, and targeted extra-cellular and cellular protein abundance. Materials and methods Knee cartilage was harvested to the subchondral bone from 20-week-old female and male C57BL/6J mice fed a low-fat or HF diet for 4 weeks and labeled with deuterium oxide for 1, 3, 5, 7, 15, or 21 days. Deuterium enrichment was quantified in isolated DNA and RNA to measure cell proliferation and ribosomal biogenesis, respectively. Protein concentration was measured using targeted high resolution accurate mass spectrometry. Results HF diet increased the maximal deuterium incorporation into DNA from approximately 40 to 50%, albeit at a slower rate. These findings, which were magnified in female versus male mice, indicate a greater number of proliferating cells with longer half-lives under HF diet conditions. HF diet caused distinct sex-dependent effects on deuterium incorporation into RNA, increasing the fraction of ribosomes undergoing biogenesis in male mice and doubling the rate of ribosome biogenesis in female mice. HF diet altered cartilage protein abundance similarly in both sexes, except for matrilin-3, which was more abundant in HF versus LF conditions in female mice only. Overall, HF diet treatment had a stronger effect than sex on cartilage protein abundance, with most changes involving extracellular matrix and matrix-associated proteins. Conclusions Short-term HF diet broadly altered cartilage matrix protein abundance, while sex-dependent effects primarily involved differences in cell proliferation and ribosomal biogenesis.
Collapse
Affiliation(s)
- Kamil A. Kobak
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Albert Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Anita Jopkiewicz
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Frederick F. Peelor
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Michael T. Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Benjamin F. Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Veterans Affairs Medical Center, Oklahoma City, OK, 73104, USA
| | - Timothy M. Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Veterans Affairs Medical Center, Oklahoma City, OK, 73104, USA
| |
Collapse
|
5
|
Binvignat M, Sellam J, Berenbaum F, Felson DT. The role of obesity and adipose tissue dysfunction in osteoarthritis pain. Nat Rev Rheumatol 2024; 20:565-584. [PMID: 39112603 DOI: 10.1038/s41584-024-01143-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2024] [Indexed: 08/29/2024]
Abstract
Obesity has a pivotal and multifaceted role in pain associated with osteoarthritis (OA), extending beyond the mechanistic influence of BMI. It exerts its effects both directly and indirectly through various modifiable risk factors associated with OA-related pain. Adipose tissue dysfunction is highly involved in OA-related pain through local and systemic inflammation, immune dysfunction, and the production of pro-inflammatory cytokines and adipokines. Adipose tissue dysfunction is intricately connected with metabolic syndrome, which independently exerts specific effects on OA-related pain, distinct from its association with BMI. The interplay among obesity, adipose tissue dysfunction and metabolic syndrome influences OA-related pain through diverse pain mechanisms, including nociceptive pain, peripheral sensitization and central sensitization. These complex interactions contribute to the heightened pain experience observed in individuals with OA and obesity. In addition, pain management strategies are less efficient in individuals with obesity. Importantly, therapeutic interventions targeting obesity and metabolic syndrome hold promise in managing OA-related pain. A deeper understanding of the intricate relationship between obesity, metabolic syndrome and OA-related pain is crucial and could have important implications for improving pain management and developing innovative therapeutic options in OA.
Collapse
Affiliation(s)
- Marie Binvignat
- Department of Rheumatology, Sorbonne University, AP-HP Saint-Antoine hospital, Paris, France
- Sorbonne University, INSERM UMRS_938, Centre de Recherche Saint-Antoine (CRSA), Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- Sorbonne University, INSERM UMRS_959, I3 Lab Immunology Immunopathology Immunotherapy, Paris, France
| | - Jérémie Sellam
- Department of Rheumatology, Sorbonne University, AP-HP Saint-Antoine hospital, Paris, France.
- Sorbonne University, INSERM UMRS_938, Centre de Recherche Saint-Antoine (CRSA), Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.
| | - Francis Berenbaum
- Department of Rheumatology, Sorbonne University, AP-HP Saint-Antoine hospital, Paris, France
- Sorbonne University, INSERM UMRS_938, Centre de Recherche Saint-Antoine (CRSA), Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - David T Felson
- Boston University School of Medicine, Department of Medicine, Section of Rheumatology, Boston, MA, USA
| |
Collapse
|
6
|
Iacobini C, Vitale M, Haxhi J, Menini S, Pugliese G. Impaired Remodeling of White Adipose Tissue in Obesity and Aging: From Defective Adipogenesis to Adipose Organ Dysfunction. Cells 2024; 13:763. [PMID: 38727299 PMCID: PMC11083890 DOI: 10.3390/cells13090763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
The adipose organ adapts and responds to internal and environmental stimuli by remodeling both its cellular and extracellular components. Under conditions of energy surplus, the subcutaneous white adipose tissue (WAT) is capable of expanding through the enlargement of existing adipocytes (hypertrophy), followed by de novo adipogenesis (hyperplasia), which is impaired in hypertrophic obesity. However, an impaired hyperplastic response may result from various defects in adipogenesis, leading to different WAT features and metabolic consequences, as discussed here by reviewing the results of the studies in animal models with either overexpression or knockdown of the main molecular regulators of the two steps of the adipogenesis process. Moreover, impaired WAT remodeling with aging has been associated with various age-related conditions and reduced lifespan expectancy. Here, we delve into the latest advancements in comprehending the molecular and cellular processes underlying age-related changes in WAT function, their involvement in common aging pathologies, and their potential as therapeutic targets to influence both the health of elderly people and longevity. Overall, this review aims to encourage research on the mechanisms of WAT maladaptation common to conditions of both excessive and insufficient fat tissue. The goal is to devise adipocyte-targeted therapies that are effective against both obesity- and age-related disorders.
Collapse
|
7
|
Zhang Q, Lu C, Lu F, Liao Y, Cai J, Gao J. Challenges and opportunities in obesity: the role of adipocytes during tissue fibrosis. Front Endocrinol (Lausanne) 2024; 15:1365156. [PMID: 38686209 PMCID: PMC11056552 DOI: 10.3389/fendo.2024.1365156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
Obesity is a chronic disease that affects the energy balance of the whole body. In addition to increasing fat mass, tissue fibrosis occurred in white adipose tissue in obese condition. Fibrosis is the over-activation of fibroblasts leading to excessive accumulation of extracellular matrix, which could be caused by various factors, including the status of adipocytes. The morphology of adipocytes responds rapidly and dynamically to nutrient fluctuations. Adaptive hypertrophy of normal adipocytes protects peripheral organs from damage from lipotoxicity. However, the biological behavior of hypertrophic adipocytes in chronic obesity is abnormally altered. Adipocytes lead to fibrotic remodeling of the extracellular matrix by inducing unresolved chronic inflammation, persistent hypoxia, and increasing myofibroblast numbers. Moreover, adipocyte-induced fibrosis not only restricts the flexible expansion and contraction of adipose tissue but also initiates the development of various diseases through cellular autonomic and paracrine effects. Regarding anti-fibrotic therapy, dysregulated intracellular signaling and epigenetic changes represent potential candidate targets. Thus, modulation of adipocytes may provide potential therapeutic avenues for reversing pathological fibrosis in adipose tissue and achieving the anti-obesity purpose.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chongxuan Lu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yunjun Liao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junrong Cai
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianhua Gao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Lee Y, Sarkar A, Tassey J, Levi JN, Lee S, Liu NQ, Drake AC, Magallanes J, Stevic U, Lu J, Ge D, Tang H, Mkaratigwa T, Bian F, Shkhyan R, Bonaguidi M, Evseenko D. Inactivation of a non-canonical gp130 signaling arm attenuates chronic systemic inflammation and multimorbidity induced by a high-fat diet. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588362. [PMID: 38645030 PMCID: PMC11030339 DOI: 10.1101/2024.04.08.588362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Interleukin-6 (IL-6) is a major pro-inflammatory cytokine for which the levels in plasma demonstrate a robust correlation with age and body mass index (BMI) as part of the senescence-associated secretory phenotype. IL-6 cytokines also play a crucial role in metabolic homeostasis and regenerative processes, primarily via the canonical STAT3 pathway. Thus, selective modulation of IL-6 signaling may offer a unique opportunity for therapeutic interventions. Recently, we discovered that a non-canonical signaling pathway downstream of tyrosine (Y) 814 within the intracellular domain of gp130, the IL-6 co-receptor, is responsible for the recruitment and activation of SRC family of kinases (SFK). Mice with constitutive genetic inactivation of gp130 Y814 (F814 mice) show accelerated resolution of inflammatory response and superior regenerative outcomes in skin wound healing and posttraumatic models of osteoarthritis. The current study was designed to explore if selective genetic or pharmacological inhibition of the non-canonical gp130-Y814/SFK signaling reduces systemic chronic inflammation and multimorbidity in a high-fat diet (HFD)-induced model of accelerated aging. F814 mice showed significantly reduced inflammatory response to HFD in adipose and liver tissue, with significantly reduced levels of systemic inflammation compared to wild type mice. F814 mice were also protected from HFD-induced bone loss and cartilage degeneration. Pharmacological inhibition of gp130-Y814/SFK in mice on HFD mirrored the effects observed in F814 mice on HFD; furthermore, this pharmacological treatment also demonstrated a marked increase in physical activity levels and protective effects against inflammation-associated suppression of neurogenesis in the brain tissue compared to the control group. These findings suggest that selective inhibition of SFK signaling downstream of gp130 receptor represents a promising strategy to alleviate systemic chronic inflammation. Increased degenerative changes and tissue senescence are inevitable in obese and aged organisms, but we demonstrated that the systemic response and inflammation-associated multi-morbidity can be therapeutically mitigated.
Collapse
|
9
|
Karjalainen K, Tanska P, Collins KH, Herzog W, Korhonen RK, Moo EK. Independent and combined effects of obesity and traumatic joint injury to the structure and composition of rat knee cartilage. Connect Tissue Res 2024; 65:117-132. [PMID: 38530304 DOI: 10.1080/03008207.2024.2310838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 01/22/2024] [Indexed: 03/27/2024]
Abstract
Osteoarthritis (OA) is a multifactorial joint disease characterized by articular cartilage degradation. Risk factors for OA include joint trauma, obesity, and inflammation, each of which can affect joint health independently, but their interaction and the associated consequences of such interaction were largely unexplored. Here, we studied compositional and structural alterations in knee joint cartilages of Sprague-Dawley rats exposed to two OA risk factors: joint injury and diet-induced obesity. Joint injury was imposed by surgical transection of anterior cruciate ligaments (ACLx), and obesity was induced by a high fat/high sucrose diet. Depth-dependent proteoglycan (PG) content and collagen structural network of cartilage were measured from histological sections collected previously in Collins et al.. (2015). We found that ACLx primarily affected the superficial cartilages. Compositionally, ACLx led to reduced PG content in lean animals, but increased PG content in obese rats. Structurally, ACLx caused disorganization of collagenous network in both lean and obese animals through increased collagen orientation in the superficial tissues and a change in the degree of fibrous alignment. However, the cartilage degradation attributed to joint injury and obesity was not necessarily additive when the two risk factors were present simultaneously, particularly for PG content and collagen orientation in the superficial tissues. Interestingly, sham surgeries caused a through-thickness disorganization of collagen network in lean and obese animals. We conclude that the interactions of multiple OA risk factors are complex and their combined effects cannot be understood by superposition principle. Further research is required to elucidate the interactive mechanism between OA subtypes.
Collapse
Affiliation(s)
- Kalle Karjalainen
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Petri Tanska
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Kelsey H Collins
- Laboratory of Musculoskeletal Crosstalk, Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, USA
| | - Walter Herzog
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, Canada
| | - Rami K Korhonen
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Eng Kuan Moo
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- Department of Mechanical and Aerospace Engineering, Carleton University, Ottawa, Canada
| |
Collapse
|
10
|
Minervini G, Franco R, Marrapodi MM, Almeida LE, Ronsivalle V, Cicciù M. Prevalence of temporomandibular disorders (TMD) in obesity patients: A systematic review and meta-analysis. J Oral Rehabil 2023; 50:1544-1553. [PMID: 37635375 DOI: 10.1111/joor.13573] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/28/2023] [Accepted: 08/08/2023] [Indexed: 08/29/2023]
Abstract
OBJECTIVE Obesity is a growing epidemic in the world. It increases the risk for severe health conditions, including diabetes, heart disease and stroke. Recent research has found that obesity is associated with an increased risk for temporomandibular disorders (TMDs). The purpose of this literature review with meta-analysis is to analyse the possible association between obesity and temporomandibular disorders. MATERIALS AND METHODS Pubmed, Web of Science and Lilacs were systematically searched until 01/03/2000. Articles dealing with TMD and obesity were selected, and numbers that might be useful in the meta-analysis were extracted. RESULTS Fifty articles were chosen after using all three search engines. Thirty-five articles were considered during the initial screening step. Four papers were chosen for title and abstract screening based on the PECO model. A manual search of bibliographies and the Cochrane database was also carried out, but no publications that matched the inclusion criteria were located. The articles and meta-analysis did not show a clear association between obesity and temporomandibular disorders. CONCLUSION There is no evidence regarding the reliability of either method. Both have superimposable results.
Collapse
Affiliation(s)
- Giuseppe Minervini
- Multidisciplinary Department of Medical-Surgical and Odontostomatological Specialties, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Rocco Franco
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Maria Maddalena Marrapodi
- Department of Woman, Child and General and Specialist Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Luis Eduardo Almeida
- Surgical Sciences Department, School of Dentistry, Marquette University, Milwaukee, Wisconsin, USA
| | - Vincenzo Ronsivalle
- Department of Biomedical and Surgical and Biomedical Sciences, Catania University, Catania, Italy
| | - Marco Cicciù
- Department of Biomedical and Surgical and Biomedical Sciences, Catania University, Catania, Italy
| |
Collapse
|
11
|
Amorim D, Fonseca-Rodrigues D, David-Pereira A, Costa O, Lima AP, Nogueira R, Cruz R, Martins AS, Sousa L, Oliveira F, Pereira H, Pirraco R, Pertovaara A, Almeida A, Pinto-Ribeiro F. Injection of kaolin/carrageenan in the rat knee joint induces progressive experimental knee osteoarthritis. Pain 2023; 164:2477-2490. [PMID: 37390363 DOI: 10.1097/j.pain.0000000000002954] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 04/25/2023] [Indexed: 07/02/2023]
Abstract
ABSTRACT Osteoarthritis (OA), the most common joint disorder worldwide, is characterized by progressive degeneration of articular and periarticular structures, leading to physical and emotional impairments that greatly affect the quality of life of patients. Unfortunately, no therapy has been able to halt the progression of the disease. Owing to the complexity of OA, most animal models are only able to mimic a specific stage or feature of the human disorder. In this work, we demonstrate the intraarticular injection of kaolin or carrageenan leads to the progressive degeneration of the rat's knee joint, accompanied by mechanical hyperalgesia and allodynia, gait impairments (reduced contact area of the affected limb), and radiological and histopathological findings concomitant with the development of human grade 4 OA. In addition, animals also display emotional impairments 4 weeks after induction, namely, anxious and depressive-like behaviour, important and common comorbidities of human OA patients. Overall, prolonging kaolin or carrageenan-induced monoarthritis mimics several important physical and psychological features of human OA in both male and female rodents and could be further applied in long-term studies of OA-associated chronic pain.
Collapse
Affiliation(s)
- Diana Amorim
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Institute of Biomedicine/Physiology, University of Helsinki, Helsinki, Finland
| | - Diana Fonseca-Rodrigues
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana David-Pereira
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Wolfson CARD, Institute of Psychology, Psychiatry and Neuroscience, King's College London, London, United Kingdom
| | - Octávia Costa
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Antónia Palhares Lima
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rosete Nogueira
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rute Cruz
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Sofia Martins
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Liliana Sousa
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | | - Hélder Pereira
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, Minho University, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
| | - Rogério Pirraco
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, Minho University, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
| | - Antti Pertovaara
- Institute of Biomedicine/Physiology, University of Helsinki, Helsinki, Finland
| | - Armando Almeida
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Filipa Pinto-Ribeiro
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
12
|
Ely EV, Kapinski AT, Paradi SG, Tang R, Guilak F, Collins KH. Designer Fat Cells: Adipogenic Differentiation of CRISPR-Cas9 Genome-Engineered Induced Pluripotent Stem Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.26.564206. [PMID: 37961399 PMCID: PMC10634849 DOI: 10.1101/2023.10.26.564206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Adipose tissue is an active endocrine organ that can signal bidirectionally to many tissues and organ systems in the body. With obesity, adipose tissue is a source of low-level inflammation that contributes to various co-morbidities and damage to downstream effector tissues. The ability to synthesize genetically engineered adipose tissue could have critical applications in studying adipokine signaling and the use of adipose tissue for novel therapeutic strategies. This study aimed to develop a method for non-viral adipogenic differentiation of genome-edited murine induced pluripotent stem cells (iPSCs) and to test the ability of such cells to engraft in mice in vivo . Designer adipocytes were created from iPSCs, which can be readily genetically engineered using CRISPR-Cas9 to knock out or insert individual genes of interest. As a model system for adipocyte-based drug delivery, an existing iPSC cell line that transcribes interleukin 1 receptor antagonist under the endogenous macrophage chemoattractant protein-1 promoter was tested for adipogenic capabilities under these same differentiation conditions. To understand the role of various adipocyte subtypes and their impact on health and disease, an efficient method was devised for inducing browning and whitening of IPSC-derived adipocytes in culture. Finally, to study the downstream effects of designer adipocytes in vivo , we transplanted the designer adipocytes into fat-free lipodystrophic mice as a model system for studying adipose signaling in different models of disease or repair. This novel translational tissue engineering and regenerative medicine platform provides an innovative approach to studying the role of adipose interorgan communication in various conditions.
Collapse
|
13
|
Shin H, Prasad V, Lupancu T, Malik S, Achuthan A, Biondo M, Kingwell BA, Thiem M, Gottschalk M, Weighardt H, Förster I, de Steiger R, Hamilton JA, Lee KMC. The GM-CSF/CCL17 pathway in obesity-associated osteoarthritic pain and disease in mice. Osteoarthritis Cartilage 2023; 31:1327-1341. [PMID: 37225052 DOI: 10.1016/j.joca.2023.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 05/08/2023] [Accepted: 05/18/2023] [Indexed: 05/26/2023]
Abstract
OBJECTIVES We have previously identified a granulocyte macrophage-colony stimulating factor (GM-CSF)/C-C motif ligand 17 (CCL17) pathway in monocytes/macrophages, in which GM-CSF regulates the formation of CCL17, and it is important for an experimental osteoarthritis (OA) model. We explore here additional OA models, including in the presence of obesity, such as a requirement for this pathway. DESIGN The roles of GM-CSF, CCL17, CCR4, and CCL22 in various experimental OA models, including those incorporating obesity (eight-week high-fat diet), were investigated using gene-deficient male mice. Pain-like behavior and arthritis were assessed by relative static weight distribution and histology, respectively. Cell populations (flow cytometry) and cytokine messenger RNA (mRNA) expression (qPCR) in knee infrapatellar fat pad were analyzed. Human OA sera were collected for circulating CCL17 levels (ELISA) and OA knee synovial tissue for gene expression (qPCR). RESULTS We present evidence that: i) GM-CSF, CCL17, and CCR4, but not CCL22, are required for the development of pain-like behavior and optimal disease in three experimental OA models, as well as for exacerbated OA development due to obesity, ii) obesity alone leads to spontaneous knee joint damage in a GM-CSF- and CCL17-dependent manner, and iii) in knee OA patients, early indications are that BMI correlates with a lower Oxford Knee Score (r = -0.458 and p = 0.0096), with elevated circulating CCL17 levels (r = 0.2108 and p = 0.0153) and with elevated GM-CSF and CCL17 gene expression in OA synovial tissue. CONCLUSIONS The above findings indicate that GM-CSF, CCL17, and CCR4 are involved in obesity-associated OA development, broadening their potential as targets for possible treatments for OA.
Collapse
Affiliation(s)
- Heonsu Shin
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria 3050 Australia; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Varun Prasad
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria 3050 Australia; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Tanya Lupancu
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria 3050 Australia; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Shveta Malik
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria 3050 Australia; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Adrian Achuthan
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria 3050 Australia; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Mark Biondo
- CSL Ltd, Bio21 Institute, Parkville, Victoria 3050 Australia; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Bronwyn A Kingwell
- CSL Ltd, Bio21 Institute, Parkville, Victoria 3050 Australia; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Manja Thiem
- Immunology and Environment, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Marlene Gottschalk
- Immunology and Environment, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Heike Weighardt
- Immunology and Environment, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Irmgard Förster
- Immunology and Environment, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Richard de Steiger
- Department of Surgery, Epworth Healthcare, University of Melbourne, Richmond, Victoria 3121, Australia; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - John A Hamilton
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria 3050 Australia; Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St. Albans, Victoria 3021, Australia; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Kevin M-C Lee
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria 3050 Australia; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia.
| |
Collapse
|
14
|
Hsu GCY. A Novel Burn / Synovectomy Mouse Model for Temporomandibular Joint Osteoarthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.07.552340. [PMID: 37609135 PMCID: PMC10441340 DOI: 10.1101/2023.08.07.552340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Temporomandibular Disorders (TMDs) is present in 33% of the U.S. population. Currently available animal models do not faithfully simulate the native disease progression of TMJ OA. The initiation of TMJ OA requires both local trauma and systemic inflammation. In this study, we present a novel mouse model which reproduces these two conditions. This is achieved by a procedure involving both synovectomy (local trauma) and a distant burn injury (systemic inflammation). Its efficacy at inducing TMJ OA was assessed with histomorphology and radiographic evaluation at 1,3, and 9 weeks after the procedure. We found that burn/synovectomy mice demonstrated significantly more degenerative changes in TMJ than uninjured control mice or synovotomy mice. The observed histology in burn/synoectomy mice mimicked native TMJ OA disease progression in a faithful manner. This animal model is invaluable in future research of the mechanism and risk factors of TMJ OA.
Collapse
|
15
|
Toupet K, Jorgensen C, Noël D. An injectable copolymer of fatty acids (ARA 3000 BETA) as a promising treatment for osteoarthritis. Sci Rep 2023; 13:7783. [PMID: 37179423 PMCID: PMC10183038 DOI: 10.1038/s41598-023-34206-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent rheumatic disease and a fast growing cause of disability. Current pharmacological treatments include antalgics and non-steroid anti-inflammatory drugs to control pain and inflammation as well as slow acting drugs such as intra-articular (IA) administration of hyaluronic acid. Oral supplementation or diet rich in polyunsaturated free fatty acids are proposed but evidence for benefit is still under debate. We here investigated the therapeutic potential of ARA 3000 BETA, an injectable copolymer of fatty acids, at the structural level in OA. Collagenase-induced osteoarthritis model was induced in C57BL/6 mice by collagenase injection into knee joint. Mice were treated with one or two IA or four intra-muscular injections (IM) of ARA 3000 BETA. At sacrifice, knee joints were recovered for cartilage analysis by confocal laser scanning microscopy (CLSM) and bone analysis by micro-computed tomography system. OA histological scoring was performed after safranin O/fast green staining. Histological analysis revealed a protective effect against cartilage degradation in treated knee joints after IM and IA administration. This was confirmed by CLSM with a significant improvement of all articular cartilage parameters, including thickness, volume and surface degradation whatever the administration route. A slight protective effect was also noticed on subchondral bone parameters and knee joint calcification after IM administration and to a lesser extent, two IA injections. We demonstrated the therapeutic efficacy of injectable ARA 3000 BETA in OA with a protection against cartilage and bone alterations providing the proof-of-concept that clinical translation might be envisioned to delay disease progression.
Collapse
Affiliation(s)
- Karine Toupet
- IRMB, University of Montpellier, INSERM, Montpellier, France
- ECELLFRANCE, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Christian Jorgensen
- IRMB, University of Montpellier, INSERM, Montpellier, France
- ECELLFRANCE, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Hôpital Lapeyronie, Montpellier, France
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, Montpellier, France.
- ECELLFRANCE, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France.
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Hôpital Lapeyronie, Montpellier, France.
| |
Collapse
|
16
|
Sobieh BH, El-Mesallamy HO, Kassem DH. Beyond mechanical loading: The metabolic contribution of obesity in osteoarthritis unveils novel therapeutic targets. Heliyon 2023; 9:e15700. [PMID: 37180899 PMCID: PMC10172930 DOI: 10.1016/j.heliyon.2023.e15700] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/16/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent progressive disease that frequently coexists with obesity. For several decades, OA was thought to be the result of ageing and mechanical stress on cartilage. Researchers' perspective has been greatly transformed when cumulative findings emphasized the role of adipose tissue in the diseases. Nowadays, the metabolic effect of obesity on cartilage tissue has become an integral part of obesity research; hoping to discover a disease-modifying drug for OA. Recently, several adipokines have been reported to be associated with OA. Particularly, metrnl (meteorin-like) and retinol-binding protein 4 (RBP4) have been recognized as emerging adipokines that can mediate OA pathogenesis. Accordingly, in this review, we will summarize the latest findings concerned with the metabolic contribution of obesity in OA pathogenesis, with particular emphasis on dyslipidemia, insulin resistance and adipokines. Additionally, we will discuss the most recent adipokines that have been reported to play a role in this context. Careful consideration of these molecular mechanisms interrelated with obesity and OA will undoubtedly unveil new avenues for OA treatment.
Collapse
Affiliation(s)
- Basma H. Sobieh
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Hala O. El-Mesallamy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- Faculty of Pharmacy, Sinai University, Sinai, Egypt
| | - Dina H. Kassem
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- Corresponding author. Associate Professor of Biochemistry Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, street of African Union Organization, 11566, Cairo, Egypt.
| |
Collapse
|
17
|
Savadipour A, Palmer D, Ely EV, Collins KH, Garcia-Castorena JM, Harissa Z, Kim YS, Oestrich A, Qu F, Rashidi N, Guilak F. The role of PIEZO ion channels in the musculoskeletal system. Am J Physiol Cell Physiol 2023; 324:C728-C740. [PMID: 36717101 PMCID: PMC10027092 DOI: 10.1152/ajpcell.00544.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/23/2023] [Accepted: 01/23/2023] [Indexed: 02/01/2023]
Abstract
PIEZO1 and PIEZO2 are mechanosensitive cation channels that are highly expressed in numerous tissues throughout the body and exhibit diverse, cell-specific functions in multiple organ systems. Within the musculoskeletal system, PIEZO1 functions to maintain muscle and bone mass, sense tendon stretch, and regulate senescence and apoptosis in response to mechanical stimuli within cartilage and the intervertebral disc. PIEZO2 is essential for transducing pain and touch sensations as well as proprioception in the nervous system, which can affect musculoskeletal health. PIEZO1 and PIEZO2 have been shown to act both independently as well as synergistically in different cell types. Conditions that alter PIEZO channel mechanosensitivity, such as inflammation or genetic mutations, can have drastic effects on these functions. For this reason, therapeutic approaches for PIEZO-related disease focus on altering PIEZO1 and/or PIEZO2 activity in a controlled manner, either through inhibition with small molecules, or through dietary control and supplementation to maintain a healthy cell membrane composition. Although many opportunities to better understand PIEZO1 and PIEZO2 remain, the studies summarized in this review highlight how crucial PIEZO channels are to musculoskeletal health and point to promising possible avenues for their modulation as a therapeutic target.
Collapse
Affiliation(s)
- Alireza Savadipour
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
| | - Daniel Palmer
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| | - Erica V Ely
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| | - Kelsey H Collins
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Jaquelin M Garcia-Castorena
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Zainab Harissa
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| | - Yu Seon Kim
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Arin Oestrich
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Feini Qu
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Neda Rashidi
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| |
Collapse
|
18
|
Min Y, Ahn D, Truong TMT, Kim M, Heo Y, Jee Y, Son YO, Kang I. Excessive sucrose exacerbates high fat diet-induced hepatic inflammation and fibrosis promoting osteoarthritis in mice model. J Nutr Biochem 2023; 112:109223. [PMID: 36410638 DOI: 10.1016/j.jnutbio.2022.109223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/21/2022]
Abstract
Osteoarthritis (OA) is marked by chronic low-grade systemic inflammation and cartilage destruction. High fat diet causes obesity and increases the risk of knee OA-development. However, the impact of high dietary sugar intake on OA pathogenesis has not been elucidated yet. Therefore, we investigated the effects of a high-fat and high-sucrose (HF+HS) diet in experimental OA mouse models. Eight-week-old male C57BL/6J mice were fed a standard chow (n=6), high-fat (HF) (n=5), or HF+HS (n=7) diets for 12 weeks; thereafter, the mice underwent surgical destabilization of the medial meniscus (DMM) and received the same experimental diets for an additional 8 weeks. The pathogenesis of knee OA, obesogenic parameters, and inflammation levels in the liver and adipose tissue were investigated. HF+HS diet induced severe cartilage erosion with osteophyte development and subchondral bone plate thickening, indicating that HF+HS diet exacerbated OA. Despite marginal differences in metabolic parameters, hepatic free cholesterol accumulation increased in mice with DMM-induced OA fed on HF+HS diet than in those fed HF diet. Notably, the levels of inflammatory cytokines and fibrosis markers were greater in the livers of mice with DMM-induced OA, fed on HF+HS diet than those in the control group. However, adipose tissue remodeling was not affected by the HF+HS diet. These findings indicate that excess sucrose intake along with a HF diet triggers hepatic inflammation and fibrosis, thereby, contributing to OA pathogenesis.
Collapse
Affiliation(s)
- Yunhui Min
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju Special Self-Governing Province, Republic of Korea
| | - Dohyun Ahn
- Department of Food Science and Nutrition, Jeju National University Jeju Special Self-Governing Province, Republic of Korea
| | - Thi My Tien Truong
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju Special Self-Governing Province, Republic of Korea
| | - Mangeun Kim
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju Special Self-Governing Province, Republic of Korea
| | - Yunji Heo
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju Special Self-Governing Province, Republic of Korea
| | - Youngheun Jee
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju Special Self-Governing Province, Republic of Korea; Department of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju Special Self-Governing Province, Republic of Korea
| | - Young-Ok Son
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju Special Self-Governing Province, Republic of Korea; Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju Special Self-Governing Province, Republic of Korea; Bio-Health Materials Core-Facility Center, Jeju National University, Jeju Special Self-Governing Province, Republic of Korea; Practical Translational Research Center, Jeju National University, Jeju Special Self-Governing Province,, Republic of Korea.
| | - Inhae Kang
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju Special Self-Governing Province, Republic of Korea; Department of Food Science and Nutrition, Jeju National University Jeju Special Self-Governing Province, Republic of Korea.
| |
Collapse
|
19
|
Collins KH, Guilak F. Trimming the fat - is leptin crosstalk the link between obesity and osteoarthritis? Osteoarthritis Cartilage 2023; 31:23-25. [PMID: 36273787 DOI: 10.1016/j.joca.2022.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/14/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Affiliation(s)
- K H Collins
- Department of Orthopedic Surgery, Washington University, St Louis, MO, USA; Shriners Hospitals for Children, St Louis, MO, USA; Center of Regenerative Medicine, Washington University, St Louis, MO, USA
| | - F Guilak
- Department of Orthopedic Surgery, Washington University, St Louis, MO, USA; Shriners Hospitals for Children, St Louis, MO, USA; Center of Regenerative Medicine, Washington University, St Louis, MO, USA.
| |
Collapse
|
20
|
Zhu S, Donovan EL, Makosa D, Mehta-D'souza P, Jopkiewicz A, Batushansky A, Cortassa D, Simmons AD, Lopes EBP, Kinter M, Griffin TM. Sirt3 Promotes Chondrogenesis, Chondrocyte Mitochondrial Respiration and the Development of High-Fat Diet-Induced Osteoarthritis in Mice. J Bone Miner Res 2022; 37:2531-2547. [PMID: 36214465 PMCID: PMC10091721 DOI: 10.1002/jbmr.4721] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 09/09/2022] [Accepted: 10/06/2022] [Indexed: 12/31/2022]
Abstract
Understanding how obesity-induced metabolic stress contributes to synovial joint tissue damage is difficult because of the complex role of metabolism in joint development, maintenance, and repair. Chondrocyte mitochondrial dysfunction is implicated in osteoarthritis (OA) pathology, which motivated us to study the mitochondrial deacetylase enzyme sirtuin 3 (Sirt3). We hypothesized that combining high-fat-diet (HFD)-induced obesity and cartilage Sirt3 loss at a young age would impair chondrocyte mitochondrial function, leading to cellular stress and accelerated OA. Instead, we unexpectedly found that depleting cartilage Sirt3 at 5 weeks of age using Sirt3-flox and Acan-CreERT2 mice protected against the development of cartilage degeneration and synovial hyperplasia following 20 weeks of HFD. This protection was associated with increased cartilage glycolysis proteins and reduced mitochondrial fatty acid metabolism proteins. Seahorse-based assays supported a mitochondrial-to-glycolytic shift in chondrocyte metabolism with Sirt3 deletion. Additional studies with primary murine juvenile chondrocytes under hypoxic and inflammatory conditions showed an increased expression of hypoxia-inducible factor (HIF-1) target genes with Sirt3 deletion. However, Sirt3 deletion impaired chondrogenesis using a murine bone marrow stem/stromal cell pellet model, suggesting a context-dependent role of Sirt3 in cartilage homeostasis. Overall, our data indicate that Sirt3 coordinates HFD-induced changes in mature chondrocyte metabolism that promote OA. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Shouan Zhu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Elise L Donovan
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Dawid Makosa
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Padmaja Mehta-D'souza
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Anita Jopkiewicz
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Albert Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Dominic Cortassa
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Aaron D Simmons
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | | | - Michael Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Timothy M Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.,Department of Biochemistry and Molecular Biology, Department of Physiology, Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, USA.,Biomedical Laboratory Research and Development, Veterans Affairs Medical Center, Oklahoma City, USA
| |
Collapse
|
21
|
Rios JL, Sapède D, Djouad F, Rapp AE, Lang A, Larkin J, Ladel C, Mobasheri A. Animal Models of Osteoarthritis Part 1-Preclinical Small Animal Models: Challenges and Opportunities for Drug Development. Curr Protoc 2022; 2:e596. [PMID: 36342311 DOI: 10.1002/cpz1.596] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Osteoarthritis (OA) is the most common form of arthritis and a major source of pain and disability in the adult population. There is a significant unmet medical need for the development of effective pharmacological therapies for the treatment of OA. In addition to spontaneously occurring animal models of OA, many experimental animal models have been developed to provide insights into mechanisms of pathogenesis and progression. Many of these animal models are also being used in the drug development pipeline. Here, we provide an overview of commonly used and emerging preclinical small animal models of OA and highlight the strengths and limitations of small animal models in the context of translational drug development. There is limited information in the published literature regarding the technical reliability of these small animal models and their ability to accurately predict clinical drug development outcomes. The cost and complexity of the available models however is an important consideration for pharmaceutical companies, biotechnology startups, and contract research organizations wishing to incorporate preclinical models in target validation, discovery, and development pipelines. Further considerations relevant to industry include timelines, methods of induction, the key issue of reproducibility, and appropriate outcome measures needed to objectively assess outcomes of experimental therapeutics. Preclinical small animal models are indispensable tools that will shine some light on the pathogenesis of OA and its molecular endotypes in the context of drug development. This paper will focus on small animal models used in preclinical OA research. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Jaqueline Lourdes Rios
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Percuros BV, Leiden, The Netherlands
| | - Dora Sapède
- IRMB, Université de Montpellier, INSERM, Montpellier, France
| | - Farida Djouad
- IRMB, Université de Montpellier, INSERM, Montpellier, France
| | - Anna E Rapp
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopaedics (Friedrichsheim), University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Annemarie Lang
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany
| | | | | | - Ali Mobasheri
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Departments of Orthopaedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Liege, Belgium
| |
Collapse
|
22
|
Hosokawa H, Akagi R, Watanabe S, Horii M, Shinohara M, Mikami Y, Toguchi K, Kimura S, Yamaguchi S, Ohtori S, Sasho T. Nuclear receptor subfamily 1 group D member 1 in the pathology of obesity-induced osteoarthritis progression. J Orthop Res 2022; 41:930-941. [PMID: 36102152 DOI: 10.1002/jor.25440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/28/2022] [Accepted: 09/02/2022] [Indexed: 02/04/2023]
Abstract
Mechanical overload and chemical factors are both related to obesity-induced progression of knee osteoarthritis. The circadian rhythm is related to the development of metabolic syndrome and the progression of osteoarthritis, and the core clock genes nuclear receptor subfamily 1 group D member 1 (NR1D1) and brain and muscle arnt-like protein 1 (BMAL1) are dysregulated in cartilage from patients with osteoarthritis. Here, we focused on NR1D1 and investigated osteoarthritis-related changes and gene expression in a mouse model of diet-induced obesity. A high-fat diet was provided to C57BL6/J mice, and changes in body weight, blood lipids, and gene expression were investigated. Destabilization of the medial meniscus or sham surgery was performed on mice fed a high-fat diet or normal diet, and histological osteoarthritis-related changes and NR1D1 expression were investigated. The effects of the NR1D1 agonist SR9009 were also assessed. Mice fed a high-fat diet developed significant obesity and dyslipidemia. Nr1d1 and Bmal1 gene expression levels decreased in the liver and knee joints. Moreover, increased osteoarthritis progression and decreased NR1D1 protein expression were observed in high-fat diet-fed mice after surgical osteoarthritis induction. SR9009 decreased the progression of obesity, dyslipidemia, and osteoarthritis. Overall, obesity and dyslipidemia induced by the high-fat diet led to osteoarthritis progression and decreased NR1D1 expression. Thus, NR1D1 may play an important role in obesity-induced osteoarthritis.
Collapse
Affiliation(s)
- Hiroaki Hosokawa
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan.,Center for Preventive Medicine, Musculoskeletal Disease and Pain, Chiba University, Chiba, Japan
| | - Ryuichiro Akagi
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Shotaro Watanabe
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan.,Center for Preventive Medicine, Musculoskeletal Disease and Pain, Chiba University, Chiba, Japan
| | - Manato Horii
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Masashi Shinohara
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Yukio Mikami
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Kaoru Toguchi
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Seiji Kimura
- Department of Orthopaedic Surgery, Center for Advanced Joint Function and Reconstructive Spine Surgery Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Satoshi Yamaguchi
- Graduate School of Global and Transdisciplinary Studies, Chiba University, Chiba, Japan
| | - Seiji Ohtori
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Takahisa Sasho
- Center for Preventive Medicine, Musculoskeletal Disease and Pain, Chiba University, Chiba, Japan
| |
Collapse
|
23
|
Neurobiological Mechanisms Modulating Emotionality, Cognition and Reward-Related Behaviour in High-Fat Diet-Fed Rodents. Int J Mol Sci 2022; 23:ijms23147952. [PMID: 35887310 PMCID: PMC9317076 DOI: 10.3390/ijms23147952] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 01/27/2023] Open
Abstract
Affective and substance-use disorders are associated with overweight and obesity-related complications, which are often due to the overconsumption of palatable food. Both high-fat diets (HFDs) and psychostimulant drugs modulate the neuro-circuitry regulating emotional processing and metabolic functions. However, it is not known how they interact at the behavioural level, and whether they lead to overlapping changes in neurobiological endpoints. In this literature review, we describe the impact of HFDs on emotionality, cognition, and reward-related behaviour in rodents. We also outline the effects of HFD on brain metabolism and plasticity involving mitochondria. Moreover, the possible overlap of the neurobiological mechanisms produced by HFDs and psychostimulants is discussed. Our in-depth analysis of published results revealed that HFDs have a clear impact on behaviour and underlying brain processes, which are largely dependent on the developmental period. However, apart from the studies investigating maternal exposure to HFDs, most of the published results involve only male rodents. Future research should also examine the biological impact of HFDs in female rodents. Further knowledge about the molecular mechanisms linking stress and obesity is a crucial requirement of translational research and using rodent models can significantly advance the important search for risk-related biomarkers and the development of clinical intervention strategies.
Collapse
|
24
|
Lu Z, Zhang A, Wang J, Han K, Gao H. Estrogen alleviates post-traumatic osteoarthritis progression and decreases p-EGFR levels in female mouse cartilage. BMC Musculoskelet Disord 2022; 23:685. [PMID: 35854298 PMCID: PMC9295391 DOI: 10.1186/s12891-022-05608-y] [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] [Received: 05/30/2022] [Accepted: 06/03/2022] [Indexed: 11/16/2022] Open
Abstract
Objective To investigate the effect of estrogen on the progression of post-traumatic osteoarthritis (PTOA) in mice and its possible mechanism. Methods Twelve-week-old ICR mice were divided into Group A (female control group), group B (ovariectomized(OVX) group), group C (OVX group supplemented with estrogen), and group D (male group) by destabilization of the medial meniscus (DMM)or sham operation. Safranin O staining was performed at 8 weeks and 12 weeks after operation, and the degree of articular cartilage lesion was evaluated using Mankin score. Twelve weeks after the operation, tissue sections were stained to analyze the matrix metalloproteinase 13(MMP13), phosphorylated epidermal growth factor receptor (p-EGFR) expression and apoptosis of chondrocytes. Results Decreased estrogen can significantly increase the weight of mice in female mice. The degree of cartilage damage in the knee joint on the DMM side of female mice was significantly severer than that on the Sham side. The DMM side also showed higher MMP13 expression and increased apoptotic chondrocytes. The degree of cartilage damage in the knee joint on the DMM side of female mice was significantly reduced after estrogen supplementation, and cartilage damage in the knee joint on the DMM side of female mice was less serious than that of male mice. As estrogen levels decreased, the severity of cartilage erosion in the knee joint on the DMM side was aggravated, and p-EGFR expression in the cartilage surface was also higher in female mice contrast to that in male mice. However, minimal changes in p-EGFR expression in the cartilage surface of bilateral knee joints of male mice were observe. Conclusion Estrogen has a regulatory effect on PTOA and its inhibits the expression of p-EGFR in cartilage on the knee joint surface and has a protective effect on articular cartilage in female mice.
Collapse
Affiliation(s)
- Zhihua Lu
- Yangzhou Polytechnic College, Yangzhou, Jiangsu, 225009, People's Republic of China
| | - Aihua Zhang
- Department of Rehabilitation Medicine, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, People's Republic of China
| | - Jingcheng Wang
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, People's Republic of China
| | - Kuijing Han
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, People's Republic of China.
| | - Han Gao
- Department of Doppler Ultrasonic, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, People's Republic of China.
| |
Collapse
|
25
|
A musculoskeletal finite element model of rat knee joint for evaluating cartilage biomechanics during gait. PLoS Comput Biol 2022; 18:e1009398. [PMID: 35657996 PMCID: PMC9166403 DOI: 10.1371/journal.pcbi.1009398] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 04/26/2022] [Indexed: 12/02/2022] Open
Abstract
Abnormal loading of the knee due to injuries or obesity is thought to contribute to the development of osteoarthritis (OA). Small animal models have been used for studying OA progression mechanisms. However, numerical models to study cartilage responses under dynamic loading in preclinical animal models have not been developed. Here we present a musculoskeletal finite element model of a rat knee joint to evaluate cartilage biomechanical responses during a gait cycle. The rat knee joint geometries were obtained from a 3-D MRI dataset and the boundary conditions regarding loading in the joint were extracted from a musculoskeletal model of the rat hindlimb. The fibril-reinforced poroelastic (FRPE) properties of the rat cartilage were derived from data of mechanical indentation tests. Our numerical results showed the relevance of simulating anatomical and locomotion characteristics in the rat knee joint for estimating tissue responses such as contact pressures, stresses, strains, and fluid pressures. We found that the contact pressure and maximum principal strain were virtually constant in the medial compartment whereas they showed the highest values at the beginning of the gait cycle in the lateral compartment. Furthermore, we found that the maximum principal stress increased during the stance phase of gait, with the greatest values at midstance. We anticipate that our approach serves as a first step towards investigating the effects of gait abnormalities on the adaptation and degeneration of rat knee joint tissues and could be used to evaluate biomechanically-driven mechanisms of the progression of OA as a consequence of joint injury or obesity. Osteoarthritis is a disease of the musculoskeletal system which is characterized by the degradation of articular cartilage. Changes in the knee loading after injuries or obesity contribute to the development of cartilage degeneration. Since injured cartilage cannot be reversed back to intact conditions, small animal models have been widely used for investigating osteoarthritis progression mechanisms. Moreover, experimental studies have been complemented with numerical models to overcome inherent limitations such as cost, difficulties to obtain accurate measures and replicate degenerative situations in the knee joint. However, computational models to study articular cartilage responses under dynamic loading in small animal models have not been developed. Thus, here we present a musculoskeletal finite element model (MSFE) of a rat knee joint to evaluate cartilage biomechanical responses during gait. Our computational model considers both the anatomical and locomotion characteristics of the rat knee joint for estimating mechanical responses in the articular cartilage. We suggest that our approach can be used to investigate tissue adaptations based on the mechanobiological responses of the cartilage to prevent the progression of osteoarthritis.
Collapse
|
26
|
Zhao Y, An Y, Zhou L, Wu F, Wu G, Wang J, Chen L. Animal Models of Temporomandibular Joint Osteoarthritis: Classification and Selection. Front Physiol 2022; 13:859517. [PMID: 35574432 PMCID: PMC9095932 DOI: 10.3389/fphys.2022.859517] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/04/2022] [Indexed: 01/11/2023] Open
Abstract
Temporomandibular joint osteoarthritis (TMJOA) is a common degenerative joint disease that can cause severe pain and dysfunction. It has a serious impact on the quality of lives of patients. Since mechanism underlying the pathogenesis of TMJOA is not fully understood, the development of effective tools for early diagnosis and disease-modifying therapies has been hindered. Animal models play a key role in understanding the pathological process of diseases and evaluating new therapeutic interventions. Although some similarities in disease processes between animals and humans are known, no one animal model is sufficient for studying all characteristics of TMJOA, as each model has different translatability to human clinical conditions. For the past 4 decades, TMJOA animal models have been studied by numerous researchers and can be broadly divided into induced, naturally occurring, and genetically modified models. The induced models can be divided into invasive models (intra-articular injection and surgical induction) or non-invasive models (mechanical loading, high-fat diet, and sleep deprivation). Different types of animal models simulate different pathological expressions of TMJOA and have their unique characteristics. Currently, mice, rats, and rabbits are commonly used in the study of TMJOA. This review sought to provide a general description of current experimental models of TMJOA and assist researchers in selecting the most appropriate models for different kinds of research.
Collapse
Affiliation(s)
- Yuqing Zhao
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
- School of Stomatology, Heilongjiang Key Lab of Oral Biomedicine Materials and Clinical Application & Experimental Center for Stomatology Engineering, Jiamusi University, Jiamusi, China
| | - Yanxin An
- Department of General Surgery, The First Affiliated Hospital of Xi’an Medical University, Xi’an, China
| | - Libo Zhou
- School of Basic Medicine, Heilongjiang Key Lab of Oral Biomedicine Materials and Clinical Application & Experimental Center for Stomatology Engineering, Jiamusi University, Jiamusi, China
| | - Fan Wu
- School of Stomatology, Heilongjiang Key Lab of Oral Biomedicine Materials and Clinical Application & Experimental Center for Stomatology Engineering, Jiamusi University, Jiamusi, China
| | - Gaoyi Wu
- School of Stomatology, Heilongjiang Key Lab of Oral Biomedicine Materials and Clinical Application & Experimental Center for Stomatology Engineering, Jiamusi University, Jiamusi, China
| | - Jing Wang
- Department of Oral Implants, School of Stomatology, National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, The Fourth Military Medical University, Xi’an, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, China
| | - Lei Chen
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| |
Collapse
|
27
|
Zaki S, Blaker CL, Little CB. OA foundations - experimental models of osteoarthritis. Osteoarthritis Cartilage 2022; 30:357-380. [PMID: 34536528 DOI: 10.1016/j.joca.2021.03.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/01/2021] [Accepted: 03/10/2021] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is increasingly recognised as a disease of diverse phenotypes with variable clinical presentation, progression, and response to therapeutic intervention. This same diversity is readily apparent in the many animal models of OA. However, model selection, study design, and interpretation of resultant findings, are not routinely done in the context of the target human (or veterinary) patient OA sub-population or phenotype. This review discusses the selection and use of animal models of OA in discovery and therapeutic-development research. Beyond evaluation of the different animal models on offer, this review suggests focussing the approach to OA-animal model selection on study objective(s), alignment of available models with OA-patient sub-types, and the resources available to achieve valid and translatable results. How this approach impacts model selection is discussed and an experimental design checklist for selecting the optimal model(s) is proposed. This approach should act as a guide to new researchers and a reminder to those already in the field, as to issues that need to be considered before embarking on in vivo pre-clinical research. The ultimate purpose of using an OA animal model is to provide the best possible evidence if, how, when and where a molecule, pathway, cell or process is important in clinical disease. By definition this requires both model and study outcomes to align with and be predictive of outcomes in patients. Keeping this at the forefront of research using pre-clinical OA models, will go a long way to improving the quality of evidence and its translational value.
Collapse
Affiliation(s)
- S Zaki
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Australia; Raymond Purves Bone and Joint Research Laboratory, Australia.
| | - C L Blaker
- Raymond Purves Bone and Joint Research Laboratory, Australia; Murray Maxwell Biomechanics Laboratory, The Kolling Institute, University of Sydney Faculty of Medicine and Health, At Royal North Shore Hospital, Australia.
| | - C B Little
- Raymond Purves Bone and Joint Research Laboratory, Australia.
| |
Collapse
|
28
|
Wang Y, Meng F, Wu J, Long H, Li J, Wu Z, He H, Wang H, Wang N, Xie D. Associations between adipokines gene polymorphisms and knee osteoarthritis: a meta-analysis. BMC Musculoskelet Disord 2022; 23:166. [PMID: 35193537 PMCID: PMC8864815 DOI: 10.1186/s12891-022-05111-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 02/10/2022] [Indexed: 02/06/2023] Open
Abstract
Background Adipokines gene polymorphisms are speculated to be associated with the risk of knee osteoarthritis (OA), but evidence remains conflicting. This study therefore aimed to examine whether associations exist between adipokines gene polymorphisms and knee OA by considering the evidence collected from eligible studies through a meta-analysis. Methods A systematic search was performed on PubMed, Embase, Web of Science, China National Knowledge Infrastructure (CNKI), and Wanfang up to March 31, 2020. Meta-analysis was carried out by focusing on the associations between adipokines gene polymorphisms and knee OA with the allele model, dominant model, and recessive model. Results The present meta-analysis included 5 eligible studies for ADIPOQ rs1501299 with 1,021 cases and 1,097 controls, 3 eligible studies for ADIPOQ rs2241766 with 549 cases and 544 controls, 3 eligible studies for LEPR rs1137101 with 808 cases and 856 controls, 2 eligible studies for VISFATIN rs4730153 with 339 cases and 680 controls and 2 eligible studies for VISFATIN rs16872158 with 339 cases and 680 controls. Significant association was observed between LEPR rs1137101 and knee OA in the overall population (recessive: OR = 0.40, 95% CI 0.21–0.79). Limited data revealed that associations may exist between ADIPOQ rs2241766 and knee OA in Asians (dominant: OR = 1.35, 95% CI 1.03–1.78), between VISFATIN rs4730153 and knee OA in Asians (allele: OR = 0.58, 95% CI 0.41–0.83; dominant: OR = 0.57, 95% CI 0.39–0.83), and between VISFATIN rs16872158 and knee OA in Asians (allele: OR = 1.84, 95% CI 1.26–2.68; dominant: OR = 1.94, 95% CI 1.31–2.89). Conclusions Adipokines gene polymorphisms may be associated with knee OA. The association was observed in LEPR rs1137101 in the present study. In addition, limited data revealed that associations may also exist in ADIPOQ rs2241766, VISFATIN rs4730153 and VISFATIN rs16872158. Prospero registration CRD42020187664. Supplementary Information The online version contains supplementary material available at 10.1186/s12891-022-05111-4.
Collapse
Affiliation(s)
- Yuqing Wang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fanqiang Meng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing Wu
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
| | - Huizhong Long
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiatian Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziying Wu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongyi He
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haochen Wang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ning Wang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dongxing Xie
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China. .,Hunan Engineering Research Center for Osteoarthritis, Changsha, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
29
|
Miller RE, Malfait AM. Can we prevent chronic osteoarthritis pain? A view from the bench. Osteoarthritis Cartilage 2021; 29:1635-1637. [PMID: 34628019 PMCID: PMC8791333 DOI: 10.1016/j.joca.2021.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/02/2021] [Indexed: 02/02/2023]
|
30
|
Harasymowicz NS, Azfer A, Burnett R, Simpson H, Salter DM. Chondrocytes from osteoarthritic cartilage of obese patients show altered adiponectin receptors expression and response to adiponectin. J Orthop Res 2021; 39:2333-2339. [PMID: 33484170 DOI: 10.1002/jor.24993] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/27/2020] [Accepted: 01/19/2021] [Indexed: 02/04/2023]
Abstract
Obesity and osteoarthritis (OA) are well-known comorbidities and their precise molecular interactions are still unidentified. Adiponectin, a major adipokine, known to have an anti-inflammatory effect in atherosclerosis or Type 2 Diabetes Mellitus (T2DM), has also been postulated to be pro-inflammatory in OA. This dual role of adiponectin is still not explained. The precise mechanism by which adiponectin affects cartilage and chondrocytes remains to be elucidated. In the present observational study chondrocytes from 30 patients with OA (18 females and 12 males) undergoing total knee replacement (TKR) were isolated. Expression of adiponectin receptors 1 and 2 (ADIPOR1 and ADIPOR2) was examined both at gene and protein levels in chondrocytes. The difference in adiponectin receptor expression between lean and obese patients with OA and the role of adiponectin in regulating pro-inflammatory genes (MCP-1, IL-6, and VCAM-1, MMP-1, MMP-2, and TIMP-1) has been investigated. We found that ADIPOR1 represented the most abundant adiponectin receptor in primary OA chondrocytes. ADIPOR1 and ADIPOR2 genes and ADIPOR1 protein were differently expressed in OA chondrocytes obtained from obese compared with lean patients with OA. Adiponectin induced gene expression of MCP-1, IL-6, and MMP-1 in all OA patients' chondrocytes. In contrast, VCAM-1 and MMP-2 were differently regulated by adiponectin depending on the patient's body mass index. This study suggests that adiponectin and ADIPOR1 may have important roles in the pathogenesis of cartilage degeneration in OA of obese subjects.
Collapse
Affiliation(s)
| | - Asim Azfer
- Bone Research Group, Center for Genomics and Experimental Medicine, The University of Edinburgh, Edinburgh, UK
| | - Richard Burnett
- Department of Orthopaedics and Trauma, Royal Infirmary of Edinburgh, Edinburgh, Edinburgh, UK
| | - Hamish Simpson
- Department of Orthopaedics and Trauma, The University of Edinburgh, Edinburgh, UK
| | - Donald M Salter
- Bone Research Group, Center for Genomics and Experimental Medicine, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
31
|
Hahn AK, Batushansky A, Rawle RA, Prado Lopes EB, June RK, Griffin TM. Effects of long-term exercise and a high-fat diet on synovial fluid metabolomics and joint structural phenotypes in mice: an integrated network analysis. Osteoarthritis Cartilage 2021; 29:1549-1563. [PMID: 34461226 PMCID: PMC8542629 DOI: 10.1016/j.joca.2021.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/18/2021] [Accepted: 08/04/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To explore how systemic factors that modify knee osteoarthritis risk are connected to 'whole-joint' structural changes by evaluating the effects of high-fat diet and wheel running exercise on synovial fluid (SF) metabolomics. METHODS Male mice were fed a defined control or high-fat (60% kcal fat) diet from 6 to 52 weeks of age, and half the animals were housed with running wheels from 26 to 52 weeks of age (n = 9-13 per group). Joint tissue structure and osteoarthritis pathology were evaluated by histology and micro-computed tomography. Systemic metabolic and inflammatory changes were evaluated by body composition, glucose tolerance testing, and serum biomarkers. SF metabolites were analyzed by high performance-liquid chromatography mass spectrometry. We built correlation-based network models to evaluate the connectivity between systemic and local metabolic biomarkers and osteoarthritis structural pathology within each experimental group. RESULTS High-fat diet caused moderate osteoarthritis, including cartilage pathology, synovitis and increased subchondral bone density. In contrast, voluntary exercise had a negligible effect on these joint structure components. 1,412 SF metabolite features were detected, with high-fat sedentary mice being the most distinct. Diet and activity uniquely altered SF metabolites attributed to amino acids, lipids, and steroids. Notably, high-fat diet increased network connections to systemic biomarkers such as interleukin-1β and glucose intolerance. In contrast, exercise increased local joint-level network connections, especially among subchondral bone features and SF metabolites. CONCLUSION Network mapping showed that obesity strengthened SF metabolite links to blood glucose and inflammation, whereas exercise strengthened SF metabolite links to subchondral bone structure.
Collapse
Affiliation(s)
- A K Hahn
- Molecular Biosciences Program, Montana State University, Bozeman, MT, 59717, USA; Department of Cell Biology & Neuroscience, Montana State University, Bozeman, MT, 59717, USA; Department of Biological and Environmental Sciences, Carroll College, Helena, MT, 59625, USA
| | - A Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104, USA
| | - R A Rawle
- Molecular Biosciences Program, Montana State University, Bozeman, MT, 59717, USA; Department of Microbiology & Immunology, Montana State University, Bozeman, MT, 59717, USA
| | - E B Prado Lopes
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104, USA
| | - R K June
- Molecular Biosciences Program, Montana State University, Bozeman, MT, 59717, USA; Department of Cell Biology & Neuroscience, Montana State University, Bozeman, MT, 59717, USA; Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, USA.
| | - T M Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104, USA; Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Veterans Affairs Medical Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
32
|
Li X, Li Y, Yang X, Liao R, Chen L, Guo Q, Yang J. PR11-364P22.2/ATF3 protein interaction mediates IL-1β-induced catabolic effects in cartilage tissue and chondrocytes. J Cell Mol Med 2021; 25:6188-6202. [PMID: 34037306 PMCID: PMC8256372 DOI: 10.1111/jcmm.16561] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/10/2021] [Accepted: 03/24/2021] [Indexed: 12/31/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease which lacks effective medical treatment due to ill-defined molecular mechanisms underlying the pathology. Inflammation is a key factor that induces and aggravates OA. Therefore, the current study aims to explore roles of the dysregulated long non-coding RNAs in the pro-inflammatory cytokine IL-1β-mediated catabolic effects in cartilage tissue and chondrocytes. We identified RP11-364P22.2 as dysregulated in OA patient-derived cartilage tissues and highly responsive to IL-1β stimulus. RNA pull-down coupled with mass spectrometry demonstrated that RP11-364P22.2 physically binds to activating transcription factor 3 (ATF3) and thus increases the protein stability and facilitates its nuclear translocation. Loss- and gain-of-function assays indicated that the interaction between RP11-364P22.2 and ATF3 is indispensable for the detrimental effects of IL-1β including growth inhibition, apoptosis induction as well as degradation of the key chondrocyte structural proteins of type II collage and Aggrecan and synthesis of the extracellular matrix-degrading enzyme MMP13 in chondrocytes. In vivo, depletion of the RP11-364P22.2 effector ATF3 drastically prevented OA development in the rats with surgical destabilization of the medial meniscus (DMM). These results highlight the important roles of lncRNAs in the pathogenesis of OA and indicate the RP11-364P22.2/ATF3 regulatory axis as a potential therapeutic target of inflammation-induced OA.
Collapse
Affiliation(s)
- Xilei Li
- Department of AnesthesiologyXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaChina
| | - Yusheng Li
- Department of OrthopaedicsXiangya HospitalCentral South UniversityChangshaChina
| | - Xucheng Yang
- Department of OrthopaedicsXiangya HospitalCentral South UniversityChangshaChina
| | - Runzhi Liao
- Department of OrthopaedicsXiangya HospitalCentral South UniversityChangshaChina
| | - Liang Chen
- Department of OrthopaedicsXiangya HospitalCentral South UniversityChangshaChina
| | - Qulian Guo
- Department of AnesthesiologyXiangya HospitalCentral South UniversityChangshaChina
| | - Junxiao Yang
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaChina
- Department of OrthopaedicsXiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
33
|
Boyd JT, LoCoco PM, Furr AR, Bendele MR, Tram M, Li Q, Chang FM, Colley ME, Samenuk GM, Arris DA, Locke EE, Bach SBH, Tobon A, Ruparel SB, Hargreaves KM. Elevated dietary ω-6 polyunsaturated fatty acids induce reversible peripheral nerve dysfunction that exacerbates comorbid pain conditions. Nat Metab 2021; 3:762-773. [PMID: 34140694 PMCID: PMC8287645 DOI: 10.1038/s42255-021-00410-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 05/12/2021] [Indexed: 02/05/2023]
Abstract
Chronic pain is the leading cause of disability worldwide1 and is commonly associated with comorbid disorders2. However, the role of diet in chronic pain is poorly understood. Of particular interest is the Western-style diet, enriched with ω-6 polyunsaturated fatty acids (PUFAs) that accumulate in membrane phospholipids and oxidise into pronociceptive oxylipins3,4. Here we report that mice administered an ω-6 PUFA-enriched diet develop persistent nociceptive hypersensitivities, spontaneously active and hyper-responsive glabrous afferent fibres and histologic markers of peripheral nerve damage reminiscent of a peripheral neuropathy. Linoleic and arachidonic acids accumulate in lumbar dorsal root ganglia, with increased liberation via elevated phospholipase (PLA)2 activity. Pharmacological and molecular inhibition of PLA2G7 or diet reversal with high levels of ω-3 PUFAs attenuate nociceptive behaviours, neurophysiologic abnormalities and afferent histopathology induced by high ω-6 intake. Additionally, ω-6 PUFA accumulation exacerbates allodynia observed in preclinical inflammatory and neuropathic pain models and is strongly correlated with multiple pain indices of clinical diabetic neuropathy. Collectively, these data reveal dietary enrichment with ω-6 PUFAs as a new aetiology of peripheral neuropathy and risk factor for chronic pain and implicate multiple therapeutic considerations for clinical pain management.
Collapse
Affiliation(s)
- Jacob T Boyd
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Peter M LoCoco
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ashley R Furr
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Michelle R Bendele
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Meilinn Tram
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Qun Li
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Fang-Mei Chang
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Madeline E Colley
- Department of Chemistry, University of Texas San Antonio, San Antonio, TX, USA
| | - Grace M Samenuk
- Department of Chemistry, University of Texas San Antonio, San Antonio, TX, USA
| | - Dominic A Arris
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Erin E Locke
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Stephan B H Bach
- Department of Chemistry, University of Texas San Antonio, San Antonio, TX, USA
| | - Alejandro Tobon
- Department of Neurology, South Texas Veterans Health Care System, San Antonio, TX, USA
| | - Shivani B Ruparel
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Kenneth M Hargreaves
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
34
|
Zheng S, Wu F, Winzenberg T, Cicuttini F, Wluka AE, Antony B, Aitken D, Blizzard L, Ding C. The cross-sectional and longitudinal associations of dietary patterns with knee symptoms and MRI detected structure in patients with knee osteoarthritis. Osteoarthritis Cartilage 2021; 29:527-535. [PMID: 33588084 DOI: 10.1016/j.joca.2020.12.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 12/07/2020] [Accepted: 12/14/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVES To examine the cross-sectional and longitudinal associations of dietary patterns with knee symptoms and structures in patients with knee osteoarthritis (OA). METHODS Participants with symptomatic knee OA were recruited from a randomised, placebo-controlled trial conducted in Tasmania (N = 259) and Victoria (N = 133). Diet was assessed by the Anti-Cancer Council of Victoria food frequency questionnaire. Factor analysis was used to identify dietary patterns. Knee symptoms were assessed using Western Ontario and McMaster Universities Arthritis Index (WOMAC) and structures using MRI. Multivariable linear regressions were used to examine associations. RESULTS Three dietary patterns ("high-fat", "healthy" and "mixed") were identified in whole sample. Participants with higher "healthy pattern" score had lower total WOMAC, pain, function and stiffness scores at baseline but the associations were not significant over 24 months. Three ("western", "vegetable and meat" and "mediterranean") and two ("processed" and "vegetable") patterns were identified in Tasmania and Victoria, respectively. Cross-sectionally, only "mediterranean pattern" and "vegetable pattern" scores were significantly and negatively associated with total WOMAC or function scores. Longitudinally, participants with higher "western pattern" had worsening function (β: 0.35, 95%CI: 0.03, 0.67) and total WOMAC (β: 0.40, 95%CI: 0.07, 0.72) scores; furthermore, "vegetable pattern" was associated with decreased WOMAC stiffness score (β: -0.47, 95%CI: -0.93, -0.02). In contrast, dietary patterns were largely not associated with structural changes. CONCLUSION Some healthy dietary patterns were associated with reduced joint symptoms but dietary patterns were not associated with joint structure in this sample with knee OA. Further studies are required to confirm these findings.
Collapse
Affiliation(s)
- S Zheng
- Department of Rheumatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, PR China; Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - F Wu
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - T Winzenberg
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - F Cicuttini
- Department of Epidemiology and Preventive Medicine, Monash University, Victoria, Australia
| | - A E Wluka
- Department of Epidemiology and Preventive Medicine, Monash University, Victoria, Australia
| | - B Antony
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - D Aitken
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - L Blizzard
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - C Ding
- Department of Rheumatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, PR China; Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia; Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
35
|
Kerr GJ, To B, White I, Millecamps M, Beier F, Grol MW, Stone LS, Séguin CA. Diet-induced obesity leads to behavioral indicators of pain preceding structural joint damage in wild-type mice. Arthritis Res Ther 2021; 23:93. [PMID: 33752736 PMCID: PMC7983381 DOI: 10.1186/s13075-021-02463-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
Introduction Obesity is one of the largest modifiable risk factors for the development of musculoskeletal diseases, including intervertebral disc (IVD) degeneration and back pain. Despite the clinical association, no studies have directly assessed whether diet-induced obesity accelerates IVD degeneration, back pain, or investigated the biological mediators underlying this association. In this study, we examine the effects of chronic consumption of a high-fat or high-fat/high-sugar (western) diet on the IVD, knee joint, and pain-associated outcomes. Methods Male C57BL/6N mice were randomized into one of three diet groups (chow control; high-fat; high-fat, high-sugar western diet) at 10 weeks of age and remained on the diet for 12, 24, or 40 weeks. At endpoint, animals were assessed for behavioral indicators of pain, joint tissues were collected for histological and molecular analysis, serum was collected to assess for markers of systemic inflammation, and IBA-1, GFAP, and CGRP were measured in spinal cords by immunohistochemistry. Results Animals fed obesogenic (high-fat or western) diets showed behavioral indicators of pain beginning at 12 weeks and persisting up to 40 weeks of diet consumption. Histological indicators of moderate joint degeneration were detected in the IVD and knee following 40 weeks on the experimental diets. Mice fed the obesogenic diets showed synovitis, increased intradiscal expression of inflammatory cytokines and circulating levels of MCP-1 compared to control. Linear regression modeling demonstrated that age and diet were both significant predictors of most pain-related behavioral outcomes, but not histopathological joint degeneration. Synovitis was associated with alterations in spontaneous activity. Conclusion Diet-induced obesity accelerates IVD degeneration and knee OA in mice; however, pain-related behaviors precede and are independent of histopathological structural damage. These findings contribute to understanding the source of obesity-related back pain and the contribution of structural IVD degeneration.
Collapse
Affiliation(s)
- Geoffrey J Kerr
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, Bone and Joint Institute, The University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Bethia To
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, Bone and Joint Institute, The University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Ian White
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, Bone and Joint Institute, The University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Magali Millecamps
- Alan Edwards Centre for Research on Pain, Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
| | - Frank Beier
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, Bone and Joint Institute, The University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Matthew W Grol
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, Bone and Joint Institute, The University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Laura S Stone
- Department of Anesthesiology, Faculty of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Cheryle A Séguin
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, Bone and Joint Institute, The University of Western Ontario, London, Ontario, N6A 5C1, Canada.
| |
Collapse
|
36
|
Tan L, Harper LR, Armstrong A, Carlson CS, Yammani RR. Dietary saturated fatty acid palmitate promotes cartilage lesions and activates the unfolded protein response pathway in mouse knee joints. PLoS One 2021; 16:e0247237. [PMID: 33617553 PMCID: PMC7899342 DOI: 10.1371/journal.pone.0247237] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 02/03/2021] [Indexed: 01/10/2023] Open
Abstract
Increased intake of dietary saturated fatty acids has been linked to obesity and the development of Osteoarthritis (OA). However, the mechanism by which these fats promote cartilage degradation and the development of OA is not clearly understood. Here, we report the effects of consumption of common dietary saturated and unsaturated fatty acids, palmitate and oleate, respectively, on body weight, metabolic factors, and knee articular cartilage in a mouse model of diet-induced obesity. Mice fed on a diet rich in saturated or unsaturated fatty acid gained an equal amount of weight; however, mice fed a palmitate diet, but not a control or oleate diet, exhibited more cartilage lesions and increased expression of 1) unfolded protein response (UPR)/endoplasmic reticulum (ER) stress markers including BIP, P-IRE1α, XBP1, ATF4, and CHOP; 2) apoptosis markers CC3 and C-PARP; and 3) negative cell survival regulators Nupr1 and TRB3, in knee articular cartilage. Palmitate-induced apoptosis was confirmed by TUNEL staining. Likewise, dietary palmitate was also increased the circulatory levels of classic proinflammatory cytokines, including IL-6 and TNF-α. Taken together, our results demonstrate that increased weight gain is not sufficient for the development of obesity-linked OA and suggest that dietary palmitate promotes UPR/ER stress and cartilage lesions in mouse knee joints. This study validates our previous in vitro findings and suggests that ER stress could be the critical metabolic factor contributing to the development of diet/obesity induced OA.
Collapse
Affiliation(s)
- Li Tan
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Lindsey R. Harper
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States of America
| | - Alexandra Armstrong
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States of America
| | - Cathy S. Carlson
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States of America
| | - Raghunatha R. Yammani
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
- * E-mail:
| |
Collapse
|
37
|
Zhang Y, Sun L, Liu X, Zhu D, Dang J, Xue Y, Fan H. Investigating the protective effect of tanshinone IIA against chondrocyte dedifferentiation: a combined molecular biology and network pharmacology approach. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:249. [PMID: 33708876 PMCID: PMC7940936 DOI: 10.21037/atm-20-4023] [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] [Indexed: 11/10/2022]
Abstract
Background Osteoarthritis (OA) is a common degenerative disease with multifactorial etiology. The dedifferentiation of chondrocytes can accelerate the progress of OA. Tanshinone IIA (TIIA) has been widely used to treat OA for many years and has proved to be effective in inhibiting chondrocyte dedifferentiation. Until now, the precise mechanism of TIIA’s effect against dedifferentiation has not been well understood. Methods The targets of TIIA were explored from public databases using various methods. The related targets of OA were obtained from the GeneCards database and the Online Mendelian Inheritance in Man (OMIM) database. The potential targets and signaling pathways were determined using protein-protein interaction (PPI), Gene Ontology (GO), and the Kyoto Encyclopedia of Genes and Genomes (KEGG). Cell viability, proliferation, and metabolic activity were analyzed in vitro. The effects of TIIA on chondrocyte dedifferentiation were evaluated by assessing morphological changes, glycosaminoglycan (GAG) production, and messenger RNA (mRNA) levels of cartilage-related genes. After 48 hours of culture in medium with 100 μg/mL TIIA, chondrocytes/hydrogel spheres were implanted to repair cartilage defects in a rat model. The harvested specimens were examined with hematoxylin and eosin (H&E) staining and immunohistochemistry to evaluate cartilage regeneration. Results The results showed that there were 28 genes potentially interacting in the TIIA-chondrocyte dedifferentiation network, and nine hub genes were identified. In vitro experiments showed an inhibitory effect of TIIA on chondrocyte dedifferentiation. The proliferation and viability of chondrocytes were promoted by TIIA at a concentration of 100–200 μg/mL, but inhibited by TIIA at 400 μg/mL. Furthermore, the histology results showed that chondrocyte/hydrogel spheres pre-treated with TIIA had better cartilage repair. Conclusions This study revealed a systematic network pharmacology approach and provided a basis for the future study of TIIA as an effective treatment for cartilage regeneration. Moreover, in vitro and in vivo results confirmed the protective effects of TIIA against chondrocyte dedifferentiation.
Collapse
Affiliation(s)
- Yushen Zhang
- Department of Orthopedic Surgery, Xi-jing Hospital, Fourth Military Medical University, Xi'an, China
| | - Liguo Sun
- Department of Orthopedic Surgery, Xi-jing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xincheng Liu
- Department of Orthopedic Surgery, Xi-jing Hospital, Fourth Military Medical University, Xi'an, China
| | - Dongze Zhu
- Department of Orthopedic Surgery, Xi-jing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jingyi Dang
- Department of Orthopedic Surgery, Xi-jing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yingsen Xue
- Department of Orthopedic Surgery, Northwest Women and Children's Hospital, Xi'an, China
| | - Hongbin Fan
- Department of Orthopedic Surgery, Xi-jing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
38
|
Collins AT, Hu G, Newman H, Reinsvold MH, Goldsmith MR, Twomey-Kozak JN, Leddy HA, Sharma D, Shen L, DeFrate LE, Karner CM. Obesity alters the collagen organization and mechanical properties of murine cartilage. Sci Rep 2021; 11:1626. [PMID: 33452305 PMCID: PMC7810701 DOI: 10.1038/s41598-020-80599-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis is a debilitating disease characterized by cartilage degradation and altered cartilage mechanical properties. Furthermore, it is well established that obesity is a primary risk factor for osteoarthritis. The purpose of this study was to investigate the influence of obesity on the mechanical properties of murine knee cartilage. Two-month old wild type mice were fed either a normal diet or a high fat diet for 16 weeks. Atomic force microscopy-based nanoindentation was used to quantify the effective indentation modulus of medial femoral condyle cartilage. Osteoarthritis progression was graded using the OARSI system. Additionally, collagen organization was evaluated with picrosirius red staining imaged using polarized light microscopy. Significant differences between diet groups were assessed using t tests with p < 0.05. Following 16 weeks of a high fat diet, no significant differences in OARSI scoring were detected. However, we detected a significant difference in the effective indentation modulus between diet groups. The reduction in cartilage stiffness is likely the result of disrupted collagen organization in the superficial zone, as indicated by altered birefringence on polarized light microscopy. Collectively, these results suggest obesity is associated with changes in knee cartilage mechanical properties, which may be an early indicator of disease progression.
Collapse
Affiliation(s)
- Amber T Collins
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
| | - Guoli Hu
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
| | - Hunter Newman
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
| | - Michael H Reinsvold
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
| | - Monique R Goldsmith
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
| | - John N Twomey-Kozak
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
| | - Holly A Leddy
- Shared Materials Instrumentation Facility, Pratt School of Engineering, Duke University, Durham, NC, 27710, USA
| | - Deepika Sharma
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
| | - Leyao Shen
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
| | - Louis E DeFrate
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA.
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27710, USA.
- Department of Mechanical Engineering and Materials Science, Pratt School of Engineering, Duke University, Durham, NC, 27710, USA.
| | - Courtney M Karner
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
39
|
Collins KH, Lenz KL, Pollitt EN, Ferguson D, Hutson I, Springer LE, Oestreich AK, Tang R, Choi YR, Meyer GA, Teitelbaum SL, Pham CTN, Harris CA, Guilak F. Adipose tissue is a critical regulator of osteoarthritis. Proc Natl Acad Sci U S A 2021; 118:e2021096118. [PMID: 33443201 PMCID: PMC7817130 DOI: 10.1073/pnas.2021096118] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Osteoarthritis (OA), the leading cause of pain and disability worldwide, disproportionally affects individuals with obesity. The mechanisms by which obesity leads to the onset and progression of OA are unclear due to the complex interactions among the metabolic, biomechanical, and inflammatory factors that accompany increased adiposity. We used a murine preclinical model of lipodystrophy (LD) to examine the direct contribution of adipose tissue to OA. Knee joints of LD mice were protected from spontaneous or posttraumatic OA, on either a chow or high-fat diet, despite similar body weight and the presence of systemic inflammation. These findings indicate that adipose tissue itself plays a critical role in the pathophysiology of OA. Susceptibility to posttraumatic OA was reintroduced into LD mice using implantation of a small adipose tissue depot derived from wild-type animals or mouse embryonic fibroblasts that undergo spontaneous adipogenesis, implicating paracrine signaling from fat, rather than body weight, as a mediator of joint degeneration.
Collapse
Affiliation(s)
- Kelsey H Collins
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110
- Shriners Hospitals for Children, St. Louis, MO 63110
- Center of Regenerative Medicine, Washington University, St. Louis, MO 63110
| | - Kristin L Lenz
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110
- Shriners Hospitals for Children, St. Louis, MO 63110
- Center of Regenerative Medicine, Washington University, St. Louis, MO 63110
| | - Eleanor N Pollitt
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110
- Shriners Hospitals for Children, St. Louis, MO 63110
- Center of Regenerative Medicine, Washington University, St. Louis, MO 63110
| | - Daniel Ferguson
- Division of Endocrinology, Washington University, St. Louis, MO 63110
| | - Irina Hutson
- Division of Endocrinology, Washington University, St. Louis, MO 63110
| | - Luke E Springer
- Division of Rheumatology, Washington University, St. Louis, MO 63110
| | - Arin K Oestreich
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110
- Shriners Hospitals for Children, St. Louis, MO 63110
- Center of Regenerative Medicine, Washington University, St. Louis, MO 63110
| | - Ruhang Tang
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110
- Shriners Hospitals for Children, St. Louis, MO 63110
- Center of Regenerative Medicine, Washington University, St. Louis, MO 63110
| | - Yun-Rak Choi
- Yonsei University College of Medicine, Seoul 120-752, South Korea
| | - Gretchen A Meyer
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110
- Program in Physical Therapy, Washington University, St. Louis, MO 63110
| | - Steven L Teitelbaum
- Department of Pathology and Immunology, Washington University, St. Louis, MO 63110
| | | | - Charles A Harris
- Division of Endocrinology, Washington University, St. Louis, MO 63110
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110;
- Shriners Hospitals for Children, St. Louis, MO 63110
- Center of Regenerative Medicine, Washington University, St. Louis, MO 63110
| |
Collapse
|
40
|
Mok SW, Fu SC, Cheuk YC, Chu IM, Chan KM, Qin L, Yung SH, Kevin Ho KW. Intra-Articular Delivery of Quercetin Using Thermosensitive Hydrogel Attenuate Cartilage Degradation in an Osteoarthritis Rat Model. Cartilage 2020; 11:490-499. [PMID: 30160166 PMCID: PMC7488941 DOI: 10.1177/1947603518796550] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE Quercetin (Que), a bioflavonoid, is both anti-inflammatory and antioxidative. Que has been used as an oral supplement for osteoarthritis (OA) with inconsistent findings because of its low bioavailability. We encapsulated Que in a mPEG-polypeptide thermogel to prolong its bioactivity. The efficacy of this formulation was evaluated in a posttraumatic OA rat model. DESIGN Methoxy-poly(ethylene glycol)-l-poly(alanine) (mPEG-PA) polymer was synthesized and characterized in terms of cytotoxicity and release kinetics in vitro. At 12 weeks old, Sprague-Dawley rats underwent anterior cruciate ligament transection (ACLT). At 24 weeks post-operation, rats received either an intra-articular (IA) injection of saline, hydrogel, or hydrogel with Que (50 or 500 μg). Gait analysis was performed at pre-ACLT, pre-treatment, and at 4, 8, and 12 weeks post-treatment. At 12 weeks post-treatment, knee joints were collected for histopathological evaluation. RESULTS In vitro studies showed that chondrocytes were viable after 72 hours of incubation with mPEG-PA, and the release of Que could be sustained for >28 days. Among all OA rats, the limb idleness index (LII) were significantly increased at 24 weeks post-ACLT. Rats that received hydrogel with Que (50 μg) showed the most reduction in LII at both 4 and 8 weeks post-treatment. The Osteoarthritis Research Society International score of rats received hydrogel with Que (50 μg) was significantly lower than the control group. All rats suffered from low-grade synovitis (Krenn score: 2-4). CONCLUSION This study suggests that a sustained delivery of Que (50 μg) could provide symptom relief and also delay the progression of OA in the knee.
Collapse
Affiliation(s)
- Sze-Wing Mok
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR,Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR
| | - Sai-Chuen Fu
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR,Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR
| | - Yau-Chuk Cheuk
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR,Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR
| | - I-Ming Chu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Kai-Ming Chan
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR,Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR
| | - Ling Qin
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR,Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR
| | - Shu-Hang Yung
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR,Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR
| | - Ki-Wai Kevin Ho
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR,Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR,Ki-Wai Kevin Ho, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Rm 74034, 5/F, Lui Che Woo Clinical Science Building, Prince of Wales Hospital, Shatin, Hong Kong SAR.
| |
Collapse
|
41
|
Jacobs BY, Allen KD. Factors affecting the reliability of behavioral assessments for rodent osteoarthritis models. Lab Anim 2020; 54:317-329. [PMID: 31431137 PMCID: PMC7830740 DOI: 10.1177/0023677219867715] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The translational value of osteoarthritis (OA) models is often debated because numerous studies have shown that animal models frequently fail to predict the efficacy of therapies in humans. In part, this failing may be due to the paucity of preclinical studies that include behavioral assessments in their metrics. Behavioral assessments of animal OA models can provide valuable data on the pain and disability associated with disease-sequelae of significant clinical relevance. Clinical definitions of efficacy for OA therapeutics often center on their palliative effects. Thus, the widespread inclusion of behaviors indicative of pain and disability in preclinical animal studies may contribute to greater success identifying clinically relevant interventions. Unfortunately, studies that include behavioral assays still frequently encounter pitfalls in assay selection, protocol consistency, and data/methods transparency. Targeted selection of behavioral assays, with consideration of the array of clinical OA phenotypes and the limitations of individual behavioral assays, is necessary to identify clinically relevant outcomes in OA animal models appropriately. Furthermore, to facilitate accurate comparisons across research groups and studies, it is necessary to improve the transparency of methods. Finally, establishing agreed-upon and clear definitions of behavioral data will reduce the convolution of data both within and between studies. Improvement in these areas is critical to the continued benefit of preclinical animal studies as translationally relevant data in OA research. As such, this review highlights the current state of behavioral analyses in preclinical OA models.
Collapse
Affiliation(s)
- Brittany Y Jacobs
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Kyle D Allen
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| |
Collapse
|
42
|
Pathogenesis of Osteoarthritis: Risk Factors, Regulatory Pathways in Chondrocytes, and Experimental Models. BIOLOGY 2020; 9:biology9080194. [PMID: 32751156 PMCID: PMC7464998 DOI: 10.3390/biology9080194] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 12/28/2022]
Abstract
As the most common chronic degenerative joint disease, osteoarthritis (OA) is the leading cause of pain and physical disability, affecting millions of people worldwide. Mainly characterized by articular cartilage degradation, osteophyte formation, subchondral bone remodeling, and synovial inflammation, OA is a heterogeneous disease that impacts all component tissues of the articular joint organ. Pathological changes, and thus symptoms, vary from person to person, underscoring the critical need of personalized therapies. However, there has only been limited progress towards the prevention and treatment of OA, and there are no approved effective disease-modifying osteoarthritis drugs (DMOADs). Conventional treatments, including non-steroidal anti-inflammatory drugs (NSAIDs) and physical therapy, are still the major remedies to manage the symptoms until the need for total joint replacement. In this review, we provide an update of the known OA risk factors and relevant mechanisms of action. In addition, given that the lack of biologically relevant models to recapitulate human OA pathogenesis represents one of the major roadblocks in developing DMOADs, we discuss current in vivo and in vitro experimental OA models, with special emphasis on recent development and application potential of human cell-derived microphysiological tissue chip platforms.
Collapse
|
43
|
Masson AO, Krawetz RJ. Understanding cartilage protection in OA and injury: a spectrum of possibilities. BMC Musculoskelet Disord 2020; 21:432. [PMID: 32620156 PMCID: PMC7334861 DOI: 10.1186/s12891-020-03363-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 05/25/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a prevalent musculoskeletal disease resulting in progressive degeneration of the hyaline articular cartilage within synovial joints. Current repair treatments for OA often result in poor quality tissue that is functionally ineffective compared to the hyaline cartilage and demonstrates increased failure rates post-treatment. Complicating efforts to improve clinical outcomes, animal models used in pre-clinical research show significant heterogeneity in their regenerative and degenerative responses associated with their species, age, genetic/epigenetic traits, and context of cartilage injury or disease. These can lead to variable outcomes when testing and validating novel therapeutic approaches for OA. Furthermore, it remains unclear whether protection against OA among different model systems is driven by inhibition of cartilage degeneration, enhancement of cartilage regeneration, or any combination thereof. MAIN TEXT Understanding the mechanistic basis underlying this context-dependent duality is essential for the rational design of targeted cartilage repair and OA therapies. Here, we discuss some of the critical variables related to the cross-species paradigm of degenerative and regenerative abilities found in pre-clinical animal models, to highlight that a gradient of regenerative competence within cartilage may exist across species and even in the greater human population, and likely influences clinical outcomes. CONCLUSIONS A more complete understanding of the endogenous regenerative potential of cartilage in a species specific context may facilitate the development of effective therapeutic approaches for cartilage injury and/or OA.
Collapse
Affiliation(s)
- Anand O Masson
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB, Canada.,Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Roman J Krawetz
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB, Canada. .,Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada. .,Department Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada. .,Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
44
|
Kimmerling KA, Oswald SJ, Huebner JL, Little D, Kraus VB, Kang JX, Wu CL, Guilak F. Transgenic conversion of ω-6 to ω-3 polyunsaturated fatty acids via fat-1 reduces the severity of post-traumatic osteoarthritis. Arthritis Res Ther 2020; 22:83. [PMID: 32295649 PMCID: PMC7160898 DOI: 10.1186/s13075-020-02170-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 03/30/2020] [Indexed: 12/22/2022] Open
Abstract
Background Dietary fatty acid (FA) content has been shown to influence the development of post-traumatic osteoarthritis (PTOA) in obesity. We used the fat-1 transgenic mouse to examine the hypothesis that endogenous reduction of ω-6 to ω-3 FA ratio, under the same dietary conditions, would mitigate metabolic inflammation and the pathogenesis of PTOA in obese male and female mice. Methods Male and female fat-1 and wild-type littermates were fed either a control diet or an ω-6 FA-rich high-fat diet and underwent destabilization of the medial meniscus (DMM) surgery to induce PTOA. OA severity, synovitis, and osteophyte formation were determined histologically, while biomarker and lipidomic analyses were performed to evaluate levels of adipokines, insulin, pro-/anti-inflammatory cytokines, and FAs in serum and joint synovial fluid. Multivariable models were performed to elucidate the associations of dietary, metabolic, and mechanical factors with PTOA. Results We found that elevated serum levels of ω-3 FAs in fat-1 mice as compared to wild-type controls fed the same diet resulted in reduced OA and synovitis in a sex- and diet-dependent manner, despite comparable body weights. The fat-1 mice showed trends toward decreased serum pro-inflammatory cytokines and increased anti-inflammatory cytokines. Multivariable analysis for variables predicting OA severity in mice resulted in correlations with serum FA levels, but not with body weight. Conclusions This study provides further evidence that circulating FA composition and systemic metabolic inflammation, rather than body weight, may be the major risk factor for obesity-associated OA. We also demonstrate the potential genetic use of ω-3 FA desaturase in mitigating PTOA in obese patients following injury.
Collapse
Affiliation(s)
- Kelly A Kimmerling
- Department of Orthopaedic Surgery, Washington University in St. Louis, Campus Box 8233, Couch Biomedical Research Bldg, Room 3121, St. Louis, MO, 63110, USA.,Shriners Hospitals for Children - St. Louis, St. Louis, MO, USA
| | - Sara J Oswald
- Department of Orthopaedic Surgery, Washington University in St. Louis, Campus Box 8233, Couch Biomedical Research Bldg, Room 3121, St. Louis, MO, 63110, USA.,Shriners Hospitals for Children - St. Louis, St. Louis, MO, USA
| | - Janet L Huebner
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Dianne Little
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, USA.,Department of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Virginia B Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA.,Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Jing X Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Chia-Lung Wu
- Department of Orthopaedic Surgery, Washington University in St. Louis, Campus Box 8233, Couch Biomedical Research Bldg, Room 3121, St. Louis, MO, 63110, USA. .,Shriners Hospitals for Children - St. Louis, St. Louis, MO, USA.
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University in St. Louis, Campus Box 8233, Couch Biomedical Research Bldg, Room 3121, St. Louis, MO, 63110, USA. .,Shriners Hospitals for Children - St. Louis, St. Louis, MO, USA.
| |
Collapse
|
45
|
Tan L, Harper L, McNulty MA, Carlson CS, Yammani RR. High-fat diet induces endoplasmic reticulum stress to promote chondrocyte apoptosis in mouse knee joints. FASEB J 2020; 34:5818-5826. [PMID: 32124494 DOI: 10.1096/fj.201902746r] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/31/2022]
Abstract
Mice fed a high-fat diet (HFD) become obese and develop osteoarthritis (OA)-like lesions, including chondrocyte apoptosis, in the knee joints. However, the mechanism by which HFD/obesity induces chondrocyte apoptosis is not clearly understood. In the present study, male mice were fed a low-fat diet (LFD, 10% kcal), HFD (45% kcal), or a HFD administered with 0.5 g/kg bodyweight of 4-phenyl butyric acid (PBA, a small chaperone known to ease endoplasmic reticulum [ER] stress), via the drinking water. At the end of the 18-week study, stifle (knee) joints from all animals were collected, fixed, paraffin embedded, and sectioned. Immunostaining of joints from the HFD group showed increased expression of ER stress and apoptotic markers and increased expression of nuclear protein 1 and tribbles related protein-3 compared to the LFD group. Mice on HFD also showed higher percentage of chondrocyte death, lower chondrocyte numbers per cartilage area, and thickening of subchondral bone. Administration of PBA alleviated all of the HFD-induced symptoms. Our study demonstrated that HFD induces ER stress to promote chondrocyte death and subchondral bone thickening, which could be relieved by alleviating ER stress via PBA administration, suggesting that ER stress could play an important role in obesity-linked OA and could be targeted for OA therapeutics.
Collapse
Affiliation(s)
- Li Tan
- Section of Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Lindsey Harper
- Veterinary Clinical Sciences Department, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, USA
| | - Margaret A McNulty
- Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cathy S Carlson
- Veterinary Clinical Sciences Department, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, USA
| | - Raghunatha R Yammani
- Section of Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
46
|
Collins KH, MacDonald GZ, Hart DA, Seerattan RA, Rios JL, Reimer RA, Herzog W. Impact of age on host responses to diet-induced obesity: Development of joint damage and metabolic set points. JOURNAL OF SPORT AND HEALTH SCIENCE 2020; 9:132-139. [PMID: 32099721 PMCID: PMC7031772 DOI: 10.1016/j.jshs.2019.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 03/01/2019] [Accepted: 03/12/2019] [Indexed: 05/08/2023]
Abstract
BACKGROUND Osteoarthritis is one of the leading causes of pain and disability worldwide, and a large percentage of patients with osteoarthritis are individuals who are also obese. In recent years, a series of animal models have demonstrated that obesity-inducing diets can result in synovial joint damage (both with and without the superimposition of trauma), which may be related to changes in percentage of body fat and a series of low-level systemic inflammatory mediators. Of note, there is a disparity between whether the dietary challenges commence at weaning, representing a weanling onset, or at skeletal maturity, representing an adult onset of obesity. We wished to evaluate the effect of the dietary exposure time and the age at which animals are exposed to a high-fat and high-sucrose (HFS) diet to determine whether these factors may result in disparate outcomes, as there is evidence suggesting that these factors result in differential metabolic disturbances. Based on dietary exposure time, we hypothesized that rats fed an HFS diet for 14 weeks from weaning (HFS Weanling) would demonstrate an increase in knee joint damage scores, whereas rats exposed to the HFS diet for 4 weeks, starting at 12 weeks of age (HFS Adult) and rats exposed to a standard chow diet (Chow) would not display an increase in knee joint damage scores. METHODS Male Sprague-Dawley rats were fed either an HFS diet for 14 weeks from weaning (HFS Weanling) or an HFS diet for 4 weeks, starting at 12 weeks of age (HFS Adult). At sacrifice, joints were scored using the modified Mankin Criteria, and serum was analyzed for a defined subset of inflammatory markers (Interleukin-6, leptin, monocyte chemoattractant protein-1, and tumor necrosis factor α). RESULTS When the HFS Weanling and HFS Adult groups were compared, both groups had a similar percent of body fat, although the HFS Weanling group had a significantly greater body mass than the HFS Adult group. The HFS Weanling and HFS Adult animals had a significant increase in body mass and percentage of body fat when compared to the Chow group. Although knee joint damage scores were low in all 3 groups, we found, contrary to our hypothesis, that the HFS Adult group had statistically significant greater knee joint damage scores than the Chow and HFS Weanling groups. Furthermore, we observed that the HFS Weanling group did not have significant differences in knee joint damage scores relative to the Chow group. CONCLUSION These findings indicate that the HFS Weanling animals were better able to cope with the dietary challenge of an HFS diet than the HFS Adult group. Interestingly, when assessing various serum proinflammatory markers, no significant differences were detected between the HFS Adult and HFS Weanling groups. Although details regarding the mechanisms underlying an increase in knee joint damage scores in the HFS Adult group remain to be elucidated, these findings indicate that dietary exposure time maybe less important than the age at which an HFS diet is introduced. Moreover, increases in serum proinflammatory mediators do not appear to be directly linked to knee joint damage scores in the HFS Weanling group animals but may be partially responsible for the observed knee joint damage in the adults over the very short time of exposure to the HFS diet.
Collapse
Affiliation(s)
- Kelsey H Collins
- Human Performance Laboratory, University of Calgary, Calgary, AB T2N 1N4, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, AB T2N 1N4, Canada; Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63108, USA
| | - Graham Z MacDonald
- Human Performance Laboratory, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - David A Hart
- Human Performance Laboratory, University of Calgary, Calgary, AB T2N 1N4, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, AB T2N 1N4, Canada; Bone & Joint Health Strategic Clinical Network, Alberta Health Services, Edmonton, AB T5E 4E3, Canada
| | - Ruth A Seerattan
- Human Performance Laboratory, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Jaqueline L Rios
- Human Performance Laboratory, University of Calgary, Calgary, AB T2N 1N4, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, AB T2N 1N4, Canada
| | - Raylene A Reimer
- Human Performance Laboratory, University of Calgary, Calgary, AB T2N 1N4, Canada; Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, AB T2N 1N4, Canada
| | - Walter Herzog
- Human Performance Laboratory, University of Calgary, Calgary, AB T2N 1N4, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, AB T2N 1N4, Canada.
| |
Collapse
|
47
|
Griffin TM, Batushansky A, Hudson J, Lopes EBP. Correlation network analysis shows divergent effects of a long-term, high-fat diet and exercise on early stage osteoarthritis phenotypes in mice. JOURNAL OF SPORT AND HEALTH SCIENCE 2020; 9:119-131. [PMID: 32099720 PMCID: PMC7031811 DOI: 10.1016/j.jshs.2019.05.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/02/2019] [Accepted: 04/23/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Obesity increases knee osteoarthritis (OA) risk through metabolic, inflammatory, and biomechanical factors, but how these systemic and local mediators interact to drive OA pathology is not well understood. We tested the effect of voluntary running exercise after chronic diet-induced obesity on knee OA-related cartilage and bone pathology in mice. We then used a correlation-based network analysis to identify systemic and local factors associated with early-stage knee OA phenotypes among the different diet and exercise groups. METHODS Male C57BL/6J mice were fed a defined control (10% kcal fat) or high fat (HF) (60% kcal fat) diet from 6 to 37 weeks of age. At 25 weeks, one-half of the mice from each diet group were housed in cages with running wheels for the remainder of the study. Histology, micro computed tomography, and magnetic resonance imaging were used to evaluate changes in joint tissue structure and OA pathology. These local variables were then compared to systemic metabolic (body mass, body fat, and glucose tolerance), inflammatory (serum adipokines and inflammatory mediators), and functional (mechanical tactile sensitivity and grip strength) outcomes using a correlation-based network analysis. Diet and exercise effects were evaluated by two-way analysis of variance. RESULTS An HF diet increased the infrapatellar fat pad size and posterior joint osteophytes, and wheel running primarily altered the subchondral cortical and trabecular bone. Neither HF diet nor exercise altered average knee cartilage OA scores compared to control groups. However, the coefficient of variation was ≥25% for many outcomes, and some mice in both diet groups developed moderate OA (≥33% maximum score). This supported using correlation-based network analyses to identify systemic and local factors associated with early-stage knee OA phenotypes. In wheel-running cohorts, an HF diet reduced the network size compared to the control diet group despite similar running distances, suggesting that diet-induced obesity dampens the effects of exercise on systemic and local OA-related factors. Each of the 4 diet and activity groups showed mostly unique networks of local and systemic factors correlated with early-stage knee OA. CONCLUSION Despite minimal group-level effects of chronic diet-induced obesity and voluntary wheel running on knee OA pathology under the current test durations, diet and exercise substantially altered the relationships among systemic and local variables associated with early-stage knee OA. These results suggest that distinct pre-OA phenotypes may exist prior to the development of disease.
Collapse
MESH Headings
- Adipokines/blood
- Animals
- Cartilage, Articular/pathology
- Cartilage, Articular/physiopathology
- Diet, High-Fat/adverse effects
- Disease Models, Animal
- Hand Strength
- Hyperalgesia/physiopathology
- Inflammation Mediators/blood
- Male
- Mice, Inbred C57BL
- Obesity/complications
- Obesity/physiopathology
- Osteoarthritis, Knee/etiology
- Osteoarthritis, Knee/metabolism
- Osteoarthritis, Knee/pathology
- Osteoarthritis, Knee/physiopathology
- Physical Conditioning, Animal
Collapse
Affiliation(s)
- Timothy M Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK 73104, USA; Reynolds Oklahoma Center on Aging and Departments of Biochemistry and Molecular Biology, Physiology, and Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | - Albert Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK 73104, USA
| | - Joanna Hudson
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK 73104, USA
| | - Erika Barboza Prado Lopes
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK 73104, USA
| |
Collapse
|
48
|
Gao YH, Zhao CW, Liu B, Dong N, Ding L, Li YR, Liu JG, Feng W, Qi X, Jin XH. An update on the association between metabolic syndrome and osteoarthritis and on the potential role of leptin in osteoarthritis. Cytokine 2020; 129:155043. [PMID: 32078923 DOI: 10.1016/j.cyto.2020.155043] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/22/2020] [Accepted: 02/11/2020] [Indexed: 12/23/2022]
Abstract
Metabolic syndrome (MetS) has been associated with osteoarthritis (OA). Leptin, which is one of the markers of MetS, has been associated with OA pathophysiology. This study aimed to provide an update on the association between MetS and OA and on the potential role of leptin in OA. In this review, we summarized the current knowledge of the association between MetS and OA and updated the evidence on the potential role of leptin in OA. Clinical studies have investigated the epidemiologic association between MetS or its components and OA. Results suggested strong epidemiologic associations between MetS and OA, especially in the Asian population. Animal studies also indicated that metabolic dysregulation may lead to OA pathogenesis. The systemic role of MetS in OA pathophysiology is associated with obesity-related inflammation, the beneficial role of n-3 polyunsaturated fatty acids and deleterious role of cholesterol, physical inactivity, hypertension-induced subchondral ischemia, dyslipidemia-induced ectopic lipid deposition in chondrocytes, hyperglycemia-induced local effects of oxidative stress and advanced glycation end-products, low-grade systemic inflammation, and obesity-related adipokines by inducing the expression of proinflammtory factors. Leptin levels in serum/plasma and synovial fluid were associated with joint pain, radiographic progression, bone formation biomarkers, cartilage volume, knee OA incidence, and total joint arthroplasty in OA patients. Elevated leptin expression and increased effect of leptin on infrapatellar fat pad, synovium, articular cartilage, and bone were also involved in the pathogenesis of OA. Current knowledge indicates a convincing epidemiologic association between MetS and OA, especially in the Asian population. Animal studies have also shown that metabolic dysregulation may lead to OA pathogenesis. Accumulating evidence suggests that leptin may play a potential role in OA pathogenesis. Therefore, leptin and its receptor may be an emerging target for intervention in metabolic-associated OA.
Collapse
Affiliation(s)
- Yu-Hang Gao
- Department of Orthopaedic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Cheng-Wu Zhao
- Department of Sports Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Bo Liu
- Department of Ultrasound, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Ning Dong
- Department of Pediatric Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Lu Ding
- Department of Orthopaedic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Ye-Ran Li
- Department of Orthopaedic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Jian-Guo Liu
- Department of Orthopaedic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Wei Feng
- Department of Orthopaedic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xin Qi
- Department of Orthopaedic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Xian-Hua Jin
- Department of Dermatology, The Second Hospital of Jilin University, Changchun, Jilin 130022, China.
| |
Collapse
|
49
|
Kremer M, Becker LJ, Barrot M, Yalcin I. How to study anxiety and depression in rodent models of chronic pain? Eur J Neurosci 2020; 53:236-270. [DOI: 10.1111/ejn.14686] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/06/2020] [Accepted: 01/14/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Mélanie Kremer
- Centre National de la Recherche Scientifique Institut des Neurosciences Cellulaires et Intégratives Université de Strasbourg Strasbourg France
| | - Léa J. Becker
- Centre National de la Recherche Scientifique Institut des Neurosciences Cellulaires et Intégratives Université de Strasbourg Strasbourg France
| | - Michel Barrot
- Centre National de la Recherche Scientifique Institut des Neurosciences Cellulaires et Intégratives Université de Strasbourg Strasbourg France
| | - Ipek Yalcin
- Centre National de la Recherche Scientifique Institut des Neurosciences Cellulaires et Intégratives Université de Strasbourg Strasbourg France
| |
Collapse
|
50
|
Sansone V, Applefield RC, De Luca P, Pecoraro V, Gianola S, Pascale W, Pascale V. Does a high-fat diet affect the development and progression of osteoarthritis in mice?: A systematic review. Bone Joint Res 2020; 8:582-592. [PMID: 31934329 PMCID: PMC6946912 DOI: 10.1302/2046-3758.812.bjr-2019-0038.r1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
Aims The aim of this study was to systematically review the literature for evidence of the effect of a high-fat diet (HFD) on the onset or progression of osteoarthritis (OA) in mice. Methods A literature search was performed in PubMed, Embase, Web of Science, and Scopus to find all studies on mice investigating the effects of HFD or Western-type diet on OA when compared with a control diet (CD). The primary outcome was the determination of cartilage loss and alteration. Secondary outcomes regarding local and systemic levels of proteins involved in inflammatory processes or cartilage metabolism were also examined when reported. Results In total, 14 publications met our inclusion criteria and were included in our review. Our meta-analysis showed that, when measured by the modified Mankin Histological-Histochemical Grading System, there was a significantly higher rate of OA in mice fed a HFD than in mice on a CD (standardized mean difference (SMD) 1.27, 95% confidence interval (CI) 0.63 to 1.91). Using the Osteoarthritis Research Society International (OARSI) score, there was a trend towards HFD causing OA (SMD 0.78, 95% CI -0.04 to 1.61). In terms of OA progression, a HFD consistently worsened the progression of surgically induced OA when compared with a CD. Finally, numerous inflammatory cytokines such as tumour necrosis factor alpha (TNF-α), interleukin (IL)-1β, and leptin, among others, were found to be altered by a HFD. Conclusion A HFD seems to induce or exacerbate the progression of OA in mice. The metabolic changes and systemic inflammation brought about by a HFD appear to be key players in the onset and progression of OA. Cite this article:Bone Joint Res 2019;8:582–592.
Collapse
Affiliation(s)
- Valerio Sansone
- Department of Orthopaedics, Universitá degli Studi di Milano and IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | | | | | - Valentina Pecoraro
- Department of Laboratory Medicine, Ospedale Civile Sant'Agostino Estense di Baggiovra, Baggiovara, Italy
| | | | | | - Valerio Pascale
- Department of Orthopaedics, Universitá degli Studi di Milano and IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| |
Collapse
|