1
|
Karim A. Unveiling the Potential of Probiotics in Osteoarthritis Management. Curr Rheumatol Rep 2024; 27:2. [PMID: 39579259 DOI: 10.1007/s11926-024-01166-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2024] [Indexed: 11/25/2024]
Abstract
PURPOSE OF REVIEW Osteoarthritis (OA), a highly prevalent degenerative joint disease, is of increasing concern due to its debilitating nature and negative impact on quality of life. Recent investigations have explored the therapeutic potential of probiotics to alleviate OA. This review summarizes the emerging evidence for the potential role of probiotics in managing OA symptoms and disease progression. The link between gut dysbiosis and chronic inflammation, a key player in OA progression is discussed in this review. RECENT FINDINGS Probiotics may modulate gut microbiota composition, potentially reducing systemic inflammation and alleviating OA symptoms, including joint pain and function. Possible mechanisms through which probiotics may exert these effects, including dampening inflammatory pathways and enhancing intestinal barrier integrity have been highlighted. Promising results from preclinical and clinical studies investigating the specific beneficial effects of specific probiotic strain(s) for OA management have been highlighted. Finally, limitations in current research and future directions, emphasizing the need for well-designed, large-scale clinical trials to definitively establish the therapeutic potential of probiotics in OA treatment have been discussed.
Collapse
Affiliation(s)
- Asima Karim
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, 27272, Sharjah, United Arab Emirates.
- Iron Biology Research Group, Sharjah Institute of Medical and Health Sciences, University of Sharjah, 27272, Sharjah, United Arab Emirates.
| |
Collapse
|
2
|
Adu Y, Ring D, Teunis T. Randomized Controlled Trials Studying Nonoperative Treatments of Osteoarthritis Often Use Misleading and Uninformative Control Groups: A Systematic Review. Clin Orthop Relat Res 2024:00003086-990000000-01759. [PMID: 39453403 DOI: 10.1097/corr.0000000000003273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/16/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Because there are no known treatments that alter the natural course of the pathophysiology of osteoarthritis, nonoperative treatment needs to be compared with known effective treatments that seek to mitigate symptoms or with similarly invasive inert (placebo) treatments to determine effectiveness. Comparing a treatment to an uninformative control group may inappropriately legitimize and support the use of potentially ineffective treatments. We therefore investigated the prevalence of inappropriate control groups in musculoskeletal research and asked whether these are associated with reporting a positive treatment effect. QUESTIONS/PURPOSES We systematically reviewed randomized trials of nonoperative treatments of osteoarthritis and asked: (1) What proportion of randomized trials use uninformative control groups (defined as a treatment less invasive than the tested treatment, or a treatment that might possibly not outperform placebo but is not acknowledged as such)? (2) Is the use of uninformative control groups independently associated with reporting a positive treatment effect (defined as p < 0.05 in favor of the intervention, or as making a recommendation favoring the intervention over the control treatment)? METHODS In a systematic review following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we searched PubMed, Cochrane, and Embase up to September 2023 for randomized controlled trials published between 2020 to 2022 that compared one or more nonoperative treatments for the symptoms of osteoarthritis. We excluded studies that contained a surgical treatment group. We identified 103 trials that met eligibility criteria, with a total of 15,491 patients. The risk of bias was high in 60% (n = 62) of trials using the Cochrane Risk of Bias Tool, version 2. Although the high risk of bias in the included studies is concerning, it does not invalidate our design; instead, it highlights that some studies may use flawed methods to recommend treatments with unproven effectiveness beyond nonspecific effects because the kinds of bias observed would tend to increase the apparent benefit of the treatment(s) being evaluated. We used logistic regression to test the association of uninformative control groups with a positive treatment effect, accounting for potential confounders such as conflict of interest and study bias using the Cochrane Risk of Bias score. RESULTS The use of uninformative control groups (treatments less invasive than the tested treatment, or treatments that might not outperform placebo but are not acknowledged as such) was found in 46% (47 of 103) of included studies. After accounting for potential confounding, there was no association between reporting positive treatment effects and the use of an uninformative control group. Studies with a low risk of bias had a lower likelihood of reporting a positive treatment effect (OR 0.2 [95% confidence interval 0.05 to 0.9]; p = 0.04, model pseudo R2 = 0.21). CONCLUSION The finding that recent studies that mimic high-level evidence often use uninformative control groups that do not adequately account for nonspecific effects (perceived treatment benefits unrelated to a treatment's direct physiological effects) points to a high risk of legitimizing ineffective treatments. This raises the ethical imperative for patients, clinicians, journal peer reviewers, and journal editors to hold researchers to the standard of an adequate, informative control group. Awareness and risk of bias checklists might help patients and clinicians forgo new treatments based on seemingly high-level evidence that may carry only iatrogenic, financial, and psychological harm (false hope, in particular). LEVEL OF EVIDENCE Level I, therapeutic study.
Collapse
Affiliation(s)
- Yaw Adu
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - David Ring
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Teun Teunis
- Department of Plastic Surgery, University Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Jang D, Shin J, Shim E, Ohtani N, Jeon OH. The connection between aging, cellular senescence and gut microbiome alterations: A comprehensive review. Aging Cell 2024; 23:e14315. [PMID: 39148278 PMCID: PMC11464129 DOI: 10.1111/acel.14315] [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: 03/26/2024] [Revised: 07/24/2024] [Accepted: 08/06/2024] [Indexed: 08/17/2024] Open
Abstract
The intricate interplay between cellular senescence and alterations in the gut microbiome emerges as a pivotal axis in the aging process, increasingly recognized for its contribution to systemic inflammation, physiological decline, and predisposition to age-associated diseases. Cellular senescence, characterized by a cessation of cell division in response to various stressors, induces morphological and functional changes within tissues. The complexity and heterogeneity of senescent cells, alongside the secretion of senescence-associated secretory phenotype, exacerbate the aging process through pro-inflammatory pathways and influence the microenvironment and immune system. Concurrently, aging-associated changes in gut microbiome diversity and composition contribute to dysbiosis, further exacerbating systemic inflammation and undermining the integrity of various bodily functions. This review encapsulates the burgeoning research on the reciprocal relationship between cellular senescence and gut dysbiosis, highlighting their collective impact on age-related musculoskeletal diseases, including osteoporosis, sarcopenia, and osteoarthritis. It also explores the potential of modulating the gut microbiome and targeting cellular senescence as innovative strategies for healthy aging and mitigating the progression of aging-related conditions. By exploring targeted interventions, including the development of senotherapeutic drugs and probiotic therapies, this review aims to shed light on novel therapeutic avenues. These strategies leverage the connection between cellular senescence and gut microbiome alterations to advance aging research and development of interventions aimed at extending health span and improving the quality of life in the older population.
Collapse
Affiliation(s)
- Dong‐Hyun Jang
- Department of Biomedical SciencesKorea University College of MedicineSeoulRepublic of Korea
| | - Ji‐Won Shin
- Department of Biomedical SciencesKorea University College of MedicineSeoulRepublic of Korea
| | - Eunha Shim
- Department of Biomedical SciencesKorea University College of MedicineSeoulRepublic of Korea
| | - Naoko Ohtani
- Department of PathophysiologyOsaka Metropolitan University Graduate School of MedicineOsakaJapan
| | - Ok Hee Jeon
- Department of Biomedical SciencesKorea University College of MedicineSeoulRepublic of Korea
| |
Collapse
|
4
|
Dolatkhah N, Jafari A, Eslamian F, Toopchizadeh V, Saleh P, Hashemian M. Saccharomyces boulardii improves clinical and paraclinical indices in overweight/obese knee osteoarthritis patients: a randomized triple-blind placebo-controlled trial. Eur J Nutr 2024; 63:2291-2305. [PMID: 38761281 DOI: 10.1007/s00394-024-03428-5] [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: 12/14/2023] [Accepted: 05/08/2024] [Indexed: 05/20/2024]
Abstract
PURPOSE This study aimed to determine the effect of the probiotic Saccharomyces boulardii (S. boulardii) in patients with knee osteoarthritis (KOA). METHODS In this study, 70 patients with KOA were recruited via outpatient clinics between 2020 and 2021 and randomly assigned to receive probiotics or placebo supplements for 12 weeks. The primary outcome was a change in pain intensity according to the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) pain score. RESULTS Sixty-three patients completed the trial. A linear mixed analysis of covariance (ANCOVA) model analysis showed that probiotic was better than placebo in decreasing the pain intensity measured by visual analogue scale (VAS) [-2.11 (-2.59, -1.62) in probiotic group and -0.90 (-1.32, -0.48) in placebo group, p = 0.002] and WOMAC pain score [-3.57 (-4.66, -2.49) in probiotic group and -1.43 (-2.33, -0.53) in placebo group, p < 0.001]. The daily intake of acetaminophen for pain management significantly decreased in the probiotic group [-267.18 (-400.47, -133.89) mg, p < 0.001] that was significantly better than placebo (p = 0.006). Probiotic significantly decreased the serum levels of high-sensitivity C-reactive protein (hs-CRP) inflammatory index [-2.72 (-3.24, -2.20) µg/ml] and malondialdehyde (MDA) oxidative stress index [-1.61 (-2.11, -1.11) nmol/ml] compared to the placebo (p = 0.002 and p < 0.001, respectively). Probiotic was better than placebo in increasing the scores of role disorder due to physical health (p = 0.023), pain (p = 0.048) and physical health (p = 0.031). CONCLUSION Probiotic S. boulardii supplementation in patients with KOA significantly improved pain intensity, some dimensions of QoL, and inflammatory and oxidative stress biomarkers with no severe side effects. TRIAL REGISTRY Registered on the Iranian clinical trial website ( http://www.irct.ir : IRCT20161022030424N4) on 2019-09-02.
Collapse
Affiliation(s)
- Neda Dolatkhah
- Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Alireza Jafari
- Department pf Physical Medicine and Rehabilitation, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Eslamian
- Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahideh Toopchizadeh
- Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parviz Saleh
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Hashemian
- Department of Biology, School of Arts and Sciences, Utica University, Utica, NY, USA
| |
Collapse
|
5
|
Moyseos M, Michael J, Ferreira N, Sophocleous A. The Effect of Probiotics on the Management of Pain and Inflammation in Osteoarthritis: A Systematic Review and Meta-Analysis of Clinical Studies. Nutrients 2024; 16:2243. [PMID: 39064686 PMCID: PMC11279588 DOI: 10.3390/nu16142243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Osteoarthritis (OA) is one of the most common musculoskeletal disorders. Recently, research has focused on the role of intestinal microbiome dysbiosis in OA. The aim of this study was to systematically review randomized intervention clinical studies investigating the effect of probiotics on the management of OA-related pain and inflammation. Pre-clinical studies and non-randomized trials were excluded. A literature search was conducted using MEDLINE, EMBASE, and Web of Science. Study quality was assessed with the Cochrane risk of bias (RoB2) tool and the Risk of Bias in N-of-1 Trials (RoBiNT) scale. RevMan was used for the meta-analysis. Outcome measures assessed self-reported pain, stiffness and impediment, and serum hs-CRP. Three studies, with 501 participants, were considered eligible for qualitative synthesis and meta-analysis. A significant reduction in symptoms across all outcomes measured, except stiffness, was evident with Lactobacillus casei Shirota. However, all other probiotics reviewed did not seem to have any effect on the measured outcomes. Pre-clinical evidence, along with the RCTs reviewed, suggests that probiotics of the Lactobacillus strains might be of use for managing pain and inflammation in OA. Considering the small number of studies included in the present review and the possible risk of bias, we conclude that further studies on the role of probiotics in humans with OA are warranted.
Collapse
Affiliation(s)
- Maria Moyseos
- Department of Life Sciences, School of Sciences, European University of Cyprus, 6, Diogenes Str., Nicosia 2404, Cyprus; (M.M.); (J.M.)
- Cyprus Research & Innovation Centre (CYRIC), 72, 28th October Avenue, Nicosia 2414, Cyprus
| | - Jenny Michael
- Department of Life Sciences, School of Sciences, European University of Cyprus, 6, Diogenes Str., Nicosia 2404, Cyprus; (M.M.); (J.M.)
| | - Nuno Ferreira
- Department of Social Sciences, University of Nicosia, 46, Makedonitissas Avenue, Nicosia 2417, Cyprus;
| | - Antonia Sophocleous
- Department of Life Sciences, School of Sciences, European University of Cyprus, 6, Diogenes Str., Nicosia 2404, Cyprus; (M.M.); (J.M.)
| |
Collapse
|
6
|
Cuomo A, Parascandolo I. Role of Nutrition in the Management of Patients with Chronic Musculoskeletal Pain. J Pain Res 2024; 17:2223-2238. [PMID: 38947129 PMCID: PMC11214565 DOI: 10.2147/jpr.s456202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/13/2024] [Indexed: 07/02/2024] Open
Abstract
Chronic musculoskeletal pain (CMP), defined as persistent discomfort in musculoskeletal tissues persisting for over 3 months, afflicts an estimated 1.71 billion people globally, leading to significant functional impairments and psychological distress, thereby detrimentally affecting individuals' quality of life. The objective of this narrative review is to elucidate the complex relationship among dietary habits, sarcopenia, and gut microbiota composition, with an eye toward enhancing patient management and outcomes. Given the burgeoning interest in the influence of diet on CMP, a detailed examination of the current literature is warranted. Nutritional intake is a critical determinant of the gut microbiota profile, which, in turn, is linked to musculature integrity and performance, potentially leading to sarcopenia. The development of sarcopenia can aggravate CMP owing to diminished muscular strength and functionality. Additionally, disruptions in the gut microbiota may directly modulate nociception, intensifying CMP manifestations. Thus, nutritional optimization emerges as a viable approach to CMP management. Emphasizing a diet conducive to a healthy gut microbiome could forestall or mitigate sarcopenia, thereby attenuating CMP intensity. Nevertheless, the domain calls for further empirical exploration to unravel the nuances of these interactions and to forge efficacious dietary strategies for individuals with CMP. Beyond mere analgesia, comprehensive patient care for CMP requires acknowledgment of the complex and multifactorial nature of pain and its foundational elements. Embracing an integrative treatment model allows healthcare practitioners to promise better patient prognoses, enriched life quality, and a decrease in the sustained healthcare costs associated with CMP.
Collapse
Affiliation(s)
- Arturo Cuomo
- Division of Anesthesia and Pain Medicine, Istituto Nazionale Tumori, IRCCS - Fondazione G Pascale, Naples, Italy
| | | |
Collapse
|
7
|
Lashkarbolouk N, Mazandarani M, Pakmehr A, Ejtahed HS. Evaluating the Role of Probiotics, Prebiotics, and Synbiotics Supplementation in Age-related Musculoskeletal Disorders in Older Adults: A Systematic Review. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10306-3. [PMID: 38907826 DOI: 10.1007/s12602-024-10306-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 06/24/2024]
Abstract
The aim of this systematic review is to evaluate musculoskeletal changes in response to prebiotics, probiotics, or synbiotics supplementation in older adults or in animal models of aging musculoskeletal disorders. A comprehensive search was conducted on electronic databases, including PubMed/Medline, Cochrane, and Web of Science until April 2024. The quality assessment of clinical trials was conducted using the Cochrane Collaboration tool and for animal studies, the SYRCLE's tool was used. Our literature search resulted in 652 studies. After removing duplicates and screening the articles based on their titles and abstracts, we assessed the full text of 112 articles, which yielded 20 clinical trials and 30 animal studies in our systematic review. Most of human and animal studies reported an improvement in physical performance, a decrease in frailty index, and a lower reduction in bone mineral density in the intervention groups. Body composition tends to increase in muscle ratio, muscle mass, and reduce in appendicular lean mass and muscle atrophy. Also, the intervention induced bone turnover and mineral absorption, significantly increasing Ca, P, and Mg absorption and short-chain fatty acid concentration. Additionally, levels of inflammatory markers such as IL1, IL6, IL17, T helper 17, and TNF-α exhibited a decreasing trend, while an increase in IL10 and IFN-γ was observed. Prebiotics, probiotics, or synbiotics supplementations could effectively improve the physical performance and muscle strength and reduce the risk of bone loss and frailty in the elderly.
Collapse
Affiliation(s)
- Narges Lashkarbolouk
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Golestan University of Medical Sciences, Gorgan, Iran
| | - Mahdi Mazandarani
- Elderly Health Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Azin Pakmehr
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanieh-Sadat Ejtahed
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Corriero A, Giglio M, Soloperto R, Inchingolo F, Varrassi G, Puntillo F. Microbial Symphony: Exploring the Role of the Gut in Osteoarthritis-Related Pain. A Narrative Review. Pain Ther 2024; 13:409-433. [PMID: 38678155 PMCID: PMC11111653 DOI: 10.1007/s40122-024-00602-9] [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: 02/22/2024] [Accepted: 04/03/2024] [Indexed: 04/29/2024] Open
Abstract
One of the most common musculoskeletal disorders, osteoarthritis (OA), causes worldwide disability, morbidity, and poor quality of life by degenerating articular cartilage, modifying subchondral bone, and inflaming synovial membranes. OA pathogenesis pathways must be understood to generate new preventative and disease-modifying therapies. In recent years, it has been acknowledged that gut microbiota (GM) can significantly contribute to the development of OA. Dysbiosis of GM can disrupt the "symphony" between the host and the GM, leading to a host immunological response that activates the "gut-joint" axis, ultimately worsening OA. This narrative review summarizes research supporting the "gut-joint axis" hypothesis, focusing on the interactions between GM and the immune system in its two main components, innate and adaptive immunity. Furthermore, the pathophysiological sequence of events that link GM imbalance to OA and OA-related pain is broken down and further investigated. We also suggest that diet and prebiotics, probiotics, nutraceuticals, exercise, and fecal microbiota transplantation could improve OA management and represent a new potential therapeutic tool in the light of the scarce panorama of disease-modifying osteoarthritis drugs (DMOADs). Future research is needed to elucidate these complex interactions, prioritizing how a particular change in GM, i.e., a rise or a drop of a specific bacterial strain, correlates with a certain OA subset to pinpoint the associated signaling pathway that leads to OA.
Collapse
Affiliation(s)
- Alberto Corriero
- Department of Interdisciplinary Medicine - ICU Section, University of Bari Aldo Moro, Piazza G. Cesare 11, 70124, Bari, Italy.
| | - Mariateresa Giglio
- Department of Interdisciplinary Medicine - ICU Section, University of Bari Aldo Moro, Piazza G. Cesare 11, 70124, Bari, Italy
| | - Rossana Soloperto
- Department of Intensive Care, Brussels' University Hospital (HUB), Rue de Lennik 808, 1070, Brussels, Belgium
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124, Bari, Italy
| | | | - Filomena Puntillo
- Department of Interdisciplinary Medicine - ICU Section, University of Bari Aldo Moro, Piazza G. Cesare 11, 70124, Bari, Italy.
| |
Collapse
|
9
|
Longo UG, Lalli A, Bandini B, de Sire R, Angeletti S, Lustig S, Ammendolia A, Budhiparama NC, de Sire A. Role of the Gut Microbiota in Osteoarthritis, Rheumatoid Arthritis, and Spondylarthritis: An Update on the Gut-Joint Axis. Int J Mol Sci 2024; 25:3242. [PMID: 38542216 PMCID: PMC10970477 DOI: 10.3390/ijms25063242] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/16/2024] [Accepted: 03/11/2024] [Indexed: 01/06/2025] Open
Abstract
Dysregulation of the gut microbiota and their metabolites is involved in the pathogenic process of intestinal diseases, and several pieces of evidence within the current literature have also highlighted a possible connection between the gut microbiota and the unfolding of inflammatory pathologies of the joints. This dysregulation is defined as the "gut-joint axis" and is based on the joint-gut interaction. It is widely recognized that the microbiota of the gut produce a variety of compounds, including enzymes, short-chain fatty acids, and metabolites. As a consequence, these proinflammatory compounds that bacteria produce, such as that of lipopolysaccharide, move from the "leaky gut" to the bloodstream, thereby leading to systemic inflammation which then reaches the joints, with consequences such as osteoarthritis, rheumatoid arthritis, and spondylarthritis. In this state-of-the-art research, the authors describe the connections between gut dysbiosis and osteoarthritis, rheumatoid arthritis, and spondylarthritis. Moreover, the diagnostic tools, outcome measures, and treatment options are elucidated. There is accumulating proof suggesting that the microbiota of the gut play an important part not only in immune-mediated, metabolic, and neurological illnesses but also in inflammatory joints. According to the authors, future studies should concentrate on developing innovative microbiota-targeted treatments and their effects on joint pathology as well as on organizing screening protocols to predict the onset of inflammatory joint disease based on gut dysbiosis.
Collapse
Affiliation(s)
- Umile Giuseppe Longo
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (U.G.L.); (A.L.); (B.B.)
- Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Alberto Lalli
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (U.G.L.); (A.L.); (B.B.)
- Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Benedetta Bandini
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (U.G.L.); (A.L.); (B.B.)
- Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Roberto de Sire
- Gastroenterology, Endoscopy Unit, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy;
- Gastroenterology Unit, Department of Clinical Medicine and Surgery, University Federico II of Naples, 80126 Naples, Italy
| | - Silvia Angeletti
- Unit of Clinical Laboratory Science, University Campus Bio-Medico of Rome, 00128 Rome, Italy;
| | - Sebastien Lustig
- Orthopaedic Department, Lyon North University Hospital, Hôpital de La Croix Rousse, Hospices Civils de Lyon, 103 Grande Rue de la Croix Rousse, 69004 Lyon, France;
| | - Antonio Ammendolia
- Department of Medical and Surgical Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy;
- Research Center on Musculoskeletal Health, MusculoSkeletalHealth@UMG, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy
| | | | - Alessandro de Sire
- Department of Medical and Surgical Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy;
- Research Center on Musculoskeletal Health, MusculoSkeletalHealth@UMG, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy
| |
Collapse
|
10
|
Amin U, Jiang R, Raza SM, Fan M, Liang L, Feng N, Li X, Yang Y, Guo F. Gut-joint axis: Oral Probiotic ameliorates Osteoarthritis. J Tradit Complement Med 2024; 14:26-39. [PMID: 38223812 PMCID: PMC10785157 DOI: 10.1016/j.jtcme.2023.06.002] [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: 12/30/2022] [Revised: 03/10/2023] [Accepted: 06/13/2023] [Indexed: 01/16/2024] Open
Abstract
Osteoarthritis (OA) etiology is multifactorial, and its prevalence is growing globally. The Gut microbiota shapes our immune system and impacts all aspects of health and disease. The idea of utilizing probiotics to treat different conditions prevails. Concerning musculoskeletal illness and health, current data lack the link to understand the interactions between the host and microbiome. We report that S. thermophilus, L. pentosus (as probiotics), and γ-aminobutyric acid (GABA) harbour against osteoarthritis in vivo and alleviate IL-1β induced changes in chondrocytes in vitro. We examined the increased GABA concentration in mice's serum and small intestine content followed by bacterial treatment. The treatment inhibited the catabolism of cartilage and rescued mice joints from degradation. Furthermore, the anabolic markers upregulated and decreased inflammatory markers in mice knee joints and chondrocytes. This study is the first to represent GABA's chondrogenic and chondroprotective effects on joints and human chondrocytes. This data provides a foundation for future studies to elucidate the role of GABA in regulating chondrocyte cell proliferation. These findings opened future horizons to understanding the gut-joint axis and OA treatment. Thus, probiotic/GABA therapy shields OA joints in mice and could at least serve as adjuvant therapy to treat osteoarthritis.
Collapse
Affiliation(s)
- Uzma Amin
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Microbiology, Government College University, Faisalabad, 38000, Punjab, Pakistan
| | - Rong Jiang
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Shahid Masood Raza
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Department of Microbiology, Government College University, Faisalabad, 38000, Punjab, Pakistan
| | - Mengtian Fan
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Li Liang
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Naibo Feng
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Xiaoli Li
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Yuyou Yang
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Fengjin Guo
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Rahman SO, Bariguian F, Mobasheri A. The Potential Role of Probiotics in the Management of Osteoarthritis Pain: Current Status and Future Prospects. Curr Rheumatol Rep 2023; 25:307-326. [PMID: 37656392 PMCID: PMC10754743 DOI: 10.1007/s11926-023-01108-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2023] [Indexed: 09/02/2023]
Abstract
PURPOSE OF REVIEW This narrative review article comprehensively explains the pathophysiology of osteoarthritis (OA) pain perception, how the gut microbiota is correlated with it, possible molecular pathways involved in probiotics-mediated OA pain reduction, limitations in the current research approaches, and future perspectives. RECENT FINDINGS The initiation and progression of OA, including the development of chronic pain, is intricately associated with activation of the innate immune system and subsequent inflammatory responses. Trauma, lifestyle (e.g., obesity and metabolic disease), and chronic antibiotic treatment can disrupt commensal homeostasis of the human microbiome, thereby affecting intestinal integrity and promoting leakage of bacterial endotoxins and metabolites such as lipopolysaccharides (LPS) into circulation. Increased level of LPS is associated with knee osteophyte severity and joint pain. Both preclinical and clinical studies strongly suggest that probiotics may benefit patients with OA pain through positive gut microbiota modulation and attenuating low-grade inflammation via multiple pathways. Patent data also suggests increased interest in the development of new innovations that involve probiotic use for reducing OA and joint pain. Recent data suggest that probiotics are attracting more and more attention for OA pain management. The advancement of knowledge in this area may pave the way for developing different probiotic strains that can be used to support joint health, improve treatment outcomes in OA, and reduce the huge impact of the disease on healthcare systems worldwide.
Collapse
Affiliation(s)
| | - Frédérique Bariguian
- Haleon (Formerly GSK Consumer Healthcare), Route de L'Etraz 2, Case Postale 1279, 1260, Nyon 1, Switzerland.
| | - Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, 90014, Oulu, FI, Finland.
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.
- 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
|
12
|
Wang Y, Li Y, Bo L, Zhou E, Chen Y, Naranmandakh S, Xie W, Ru Q, Chen L, Zhu Z, Ding C, Wu Y. Progress of linking gut microbiota and musculoskeletal health: casualty, mechanisms, and translational values. Gut Microbes 2023; 15:2263207. [PMID: 37800576 PMCID: PMC10561578 DOI: 10.1080/19490976.2023.2263207] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 09/21/2023] [Indexed: 10/07/2023] Open
Abstract
The musculoskeletal system is important for balancing metabolic activity and maintaining health. Recent studies have shown that distortions in homeostasis of the intestinal microbiota are correlated with or may even contribute to abnormalities in musculoskeletal system function. Research has also shown that the intestinal flora and its secondary metabolites can impact the musculoskeletal system by regulating various phenomena, such as inflammation and immune and metabolic activities. Most of the existing literature supports that reasonable nutritional intervention helps to improve and maintain the homeostasis of intestinal microbiota, and may have a positive impact on musculoskeletal health. The purpose of organizing, summarizing and discussing the existing literature is to explore whether the intervention methods, including nutritional supplement and moderate exercise, can affect the muscle and bone health by regulating the microecology of the intestinal flora. More in-depth efficacy verification experiments will be helpful for clinical applications.
Collapse
Affiliation(s)
- Yu Wang
- Department of Health and Kinesiology, School of Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Bo
- Department of Rheumatology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Enyuan Zhou
- Department of Health and Kinesiology, School of Physical Education, Jianghan University, Wuhan, China
| | - Yanyan Chen
- Department of Health and Kinesiology, School of Physical Education, Jianghan University, Wuhan, China
| | - Shinen Naranmandakh
- School of Arts and Sciences, National University of Mongolia, Ulaanbaatar, Mongolia
| | - Wenqing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qin Ru
- Department of Health and Kinesiology, School of Physical Education, Jianghan University, Wuhan, China
| | - Lin Chen
- Department of Health and Kinesiology, School of Physical Education, Jianghan University, Wuhan, China
| | - Zhaohua Zhu
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Changhai Ding
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Orthopaedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Yuxiang Wu
- Department of Health and Kinesiology, School of Physical Education, Jianghan University, Wuhan, China
| |
Collapse
|
13
|
Morimoto T, Kobayashi T, Kakiuchi T, Esaki M, Tsukamoto M, Yoshihara T, Hirata H, Yabuki S, Mawatari M. Gut-spine axis: a possible correlation between gut microbiota and spinal degenerative diseases. Front Microbiol 2023; 14:1290858. [PMID: 37965563 PMCID: PMC10641865 DOI: 10.3389/fmicb.2023.1290858] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/10/2023] [Indexed: 11/16/2023] Open
Abstract
As society ages, the number of patients with spinal degenerative diseases (SDD) is increasing, posing a major socioeconomic problem for patients and their families. SDD refers to a generic term for degenerative diseases of spinal structures, including osteoporosis (bone), facet osteoarthritis (joint), intervertebral disk degeneration (disk), lumbar spinal canal stenosis (yellow ligament), and spinal sarcopenia (muscle). We propose the term "gut-spine axis" for the first time, given the influence of gut microbiota (GM) on the metabolic, immune, and endocrine environment in hosts through various potential mechanisms. A close cross-talk is noted between the aforementioned spinal components and degenerative diseases. This review outlines the nature and role of GM, highlighting GM abnormalities associated with the degeneration of spinal components. It also summarizes the evidence linking GM to various SDD. The gut-spine axis perspective can provide novel insights into the pathogenesis and treatment of SDD.
Collapse
Affiliation(s)
- Tadatsugu Morimoto
- Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Takaomi Kobayashi
- Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Toshihiko Kakiuchi
- Department of Pediatrics, Faculty of Medicine, Saga University, Saga, Japan
| | - Motohiro Esaki
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Masatsugu Tsukamoto
- Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Tomohito Yoshihara
- Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Hirohito Hirata
- Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Shoji Yabuki
- Fukushima Medical University School of Health Sciences, Fukushima, Japan
| | - Masaaki Mawatari
- Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
14
|
Oh DK, Na HS, Jhun JY, Lee JS, Um IG, Lee SY, Park MS, Cho ML, Park SH. Bifidobacterium longum BORI inhibits pain behavior and chondrocyte death, and attenuates osteoarthritis progression. PLoS One 2023; 18:e0286456. [PMID: 37352198 PMCID: PMC10289443 DOI: 10.1371/journal.pone.0286456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/16/2023] [Indexed: 06/25/2023] Open
Abstract
Osteoarthritis (OA), the most common form of arthritis, is characterized by pain and cartilage damage; it usually exhibits gradual development. However, the pathogenesis of OA remains unclear. This study was undertaken to improve the understanding and treatment of OA. OA was induced in 7-week-old Wistar rats by intra-articular injection of monosodium iodoacetate (MIA); subsequently, the rats underwent oral administration of Bifidobacterium longum BORI (B. BORI). The effects of B. BORI were examined in chondrocytes and an MIA-induced OA rat model. In the rats, B. BORI-mediated effects on pain severity, cartilage destruction, and inflammation were recorded. Additional effects on mRNA and cytokine secretion were analyzed by quantitative polymerase chain reaction and enzyme-linked immunosorbent assay. Paw withdrawal threshold, paw withdrawal latency, and weight-bearing assessments revealed that pain severity in MIA-induced OA rats was decreased after B. BORI treatment. Histopathology analyses and three-dimensional surface renderings of rat femurs from micro-computed tomography images revealed cartilage protection and cartilage loss inhibition effects in B. BORI-treated OA rats. Immunohistochemical analyses of inflammatory cytokines and catabolic markers (e.g., matrix metalloproteinases) showed that the expression levels of both were reduced in tissue from B. BORI-treated OA rats. Furthermore, B. BORI treatment decreased the expression levels of the inflammatory cytokine monocyte chemoattractant protein-1 and inflammatory gene factors (e.g., inflammatory cell death markers) in chondrocytes. The findings indicate that oral administration of B. BORI has therapeutic potential in terms of reducing pain, progression, and inflammation in OA.
Collapse
Affiliation(s)
- Dong Keon Oh
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
| | - Hyun Sik Na
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
| | - Joo Yeon Jhun
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
| | - Jeong Su Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
| | - In Gyu Um
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
| | - Seung Yoon Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
| | | | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, Catholic University of Korea, Seoul, Korea
| |
Collapse
|
15
|
Sun C, Zhou X, Guo T, Meng J. The immune role of the intestinal microbiome in knee osteoarthritis: a review of the possible mechanisms and therapies. Front Immunol 2023; 14:1168818. [PMID: 37388748 PMCID: PMC10306395 DOI: 10.3389/fimmu.2023.1168818] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 06/02/2023] [Indexed: 07/01/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease characterized by cartilage damage and synovial inflammation and carries an enormous public health and economic burden. It is crucial to uncover the potential mechanisms of OA pathogenesis to develop new targets for OA treatment. In recent years, the pathogenic role of the gut microbiota in OA has been well recognized. Gut microbiota dysbiosis can break host-gut microbe equilibrium, trigger host immune responses and activate the "gut-joint axis", which aggravates OA. However, although the role of the gut microbiota in OA is well known, the mechanisms modulating the interactions between the gut microbiota and host immunity remain unclear. This review summarizes research on the gut microbiota and the involved immune cells in OA and interprets the potential mechanisms for the interactions between the gut microbiota and host immune responses from four aspects: gut barrier, innate immunity, adaptive immunity and gut microbiota modulation. Future research should focus on the specific pathogen or the specific changes in the gut microbiota composition to identify the related signaling pathways involved in the pathogenesis of OA. In addition, future studies should include more novel interventions on immune cell modifications and gene regulation of specific gut microbiota related to OA to validate the application of gut microbiota modulation in the onset of OA.
Collapse
Affiliation(s)
- Chang Sun
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xing Zhou
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Ting Guo
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jia Meng
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
16
|
Liu S, Li G, Xu H, Wang Q, Wei Y, Yang Q, Xiong A, Yu F, Weng J, Zeng H. "Cross-talk" between gut microbiome dysbiosis and osteoarthritis progression: a systematic review. Front Immunol 2023; 14:1150572. [PMID: 37180142 PMCID: PMC10167637 DOI: 10.3389/fimmu.2023.1150572] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/27/2023] [Indexed: 05/15/2023] Open
Abstract
Objectives The aim of this systematic review was to summarize the available literature on gut microbiome (GMB) and osteoarthritis (OA), analyze the correlation between GMB and OA, and explore potential underlying mechanisms. Methods A systematic search of the PubMed, Embase, Cochrane, and Web of Science with the keywords "Gut Microbiome" and "Osteoarthritis" was conducted to identify the human and animal studies exploring the association between GMB and OA. The retrieval time range was from the database inception to July 31, 2022. Studies reported the other arthritic diseases without OA, reviews, and studies focused on the microbiome in other parts of the body with OA, such as oral or skin, were excluded. The included studies were mainly reviewed for GMB composition, OA severity, inflammatory factors, and intestinal permeability. Results There were 31 studies published met the inclusion criteria and were analyzed, including 10 human studies and 21 animal studies. Human and animal studies have reached a consistent conclusion that GMB dysbiosis could aggravate OA. In addition, several studies have found that alterations of GMB composition can increase intestinal permeability and serum levels of inflammatory factors, while regulating GMB can alleviate the changes. Owing to the susceptibility of GMB to internal and external environments, genetics, and geography, the included studies were not consistent in GMB composition analysis. Conclusion There is a lack of high-quality studies evaluating the effects of GMB on OA. Available evidence indicated that GMB dysbiosis aggravated OA through activating the immune response and subsequent induction of inflammation. Future studies should focus on more prospective, cohort studies combined with multi-omics to further clarify the correlation.
Collapse
Affiliation(s)
- Su Liu
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Guoqing Li
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Huihui Xu
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Qichang Wang
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yihao Wei
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Qi Yang
- Department of Ultrasonography, Peking University Shenzhen Hospital, Shenzhen, China
| | - Ao Xiong
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Fei Yu
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jian Weng
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Hui Zeng
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
17
|
Illikoud N, Mantel M, Rolli-Derkinderen M, Gagnaire V, Jan G. Dairy starters and fermented dairy products modulate gut mucosal immunity. Immunol Lett 2022; 251-252:91-102. [DOI: 10.1016/j.imlet.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/24/2022] [Accepted: 11/01/2022] [Indexed: 11/07/2022]
|
18
|
Cho KH, Na HS, Jhun J, Woo JS, Lee AR, Lee SY, Lee JS, Um IG, Kim SJ, Park SH, Cho ML. Lactobacillus (LA-1) and butyrate inhibit osteoarthritis by controlling autophagy and inflammatory cell death of chondrocytes. Front Immunol 2022; 13:930511. [PMID: 36325344 PMCID: PMC9619036 DOI: 10.3389/fimmu.2022.930511] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 09/21/2022] [Indexed: 11/14/2022] Open
Abstract
Osteoarthritis (OA) reduces the quality of life as a result of the pain caused by continuous joint destruction. Inactivated Lactobacillus (LA-1) ameliorated osteoarthritis and protected cartilage by modulating inflammation. In this study, we evaluated the mechanism by which live LA-1 ameliorated OA. To investigate the effect of live LA-1 on OA progression, we administered LA-1 into monosodium iodoacetate (MIA)-induced OA animals. The pain threshold, cartilage damage, and inflammation of the joint synovial membrane were improved by live LA-1. Furthermore, the analysis of intestinal tissues and feces in the disease model has been shown to affect the systems of the intestinal system and improve the microbiome environment. Interestingly, inflammation of the intestinal tissue was reduced, and the intestinal microbiome was altered by live LA-1. Live LA-1 administration led to an increase in the level of Faecalibacterium which is a short-chain fatty acid (SCFA) butyrate-producing bacteria. The daily supply of butyrate, a bacterial SCFA, showed a tendency to decrease necroptosis, a type of abnormal cell death, by inducing autophagy and reversing impaired autophagy by the inflammatory environment. These results suggest that OA is modulated by changes in the gut microbiome, suggesting that activation of autophagy can reduce aberrant cell death. In summary, live LA-1 or butyrate ameliorates OA progression by modulating the gut environment and autophagic flux. Our findings suggest the regulation of the gut microenvironment as a therapeutic target for OA.
Collapse
Affiliation(s)
- Keun-Hyung Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine & Health Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Hyun Sik Na
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine & Health Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - JooYeon Jhun
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine & Health Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Jin Seok Woo
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
| | - A Ram Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine & Health Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Seung Yoon Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine & Health Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Jeong Su Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine & Health Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - In Gyu Um
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine & Health Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Seok Jung Kim
- Department of Orthopedic Surgery, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine & Health Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
- Department of Medical Life Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
- *Correspondence: Mi-La Cho,
| |
Collapse
|
19
|
Zemanova N, Omelka R, Mondockova V, Kovacova V, Martiniakova M. Roles of Gut Microbiome in Bone Homeostasis and Its Relationship with Bone-Related Diseases. BIOLOGY 2022; 11:1402. [PMID: 36290306 PMCID: PMC9598716 DOI: 10.3390/biology11101402] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/14/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022]
Abstract
The extended microbial genome-the gut microbiome (GM)-plays a significant role in host health and disease. It is able to influence a number of physiological functions. During dysbiosis, GM is associated with the development of various chronic diseases with impaired bone quality. In general, GM is important for bone homeostasis and can affect it via several mechanisms. This review describes the roles of GM in bone homeostasis through influencing the immune and endocrine functions, short-chain fatty acids production, calcium absorption and the gut-brain axis. The relationship between GM composition and several bone-related diseases, specifically osteoporosis, osteoarthritis, rheumatoid arthritis, diabetes mellitus, obesity and bone cancer, is also highlighted and summarized. GM manipulation may become a future adjuvant therapy in the prevention of many chronic diseases. Therefore, the beneficial effects of probiotic therapy to improve the health status of individuals with aforementioned diseases are provided, but further studies are needed to clearly confirm its effectiveness. Recent evidence suggests that GM is responsible for direct and indirect effects on drug efficacy. Accordingly, various GM alterations and interactions related to the treatment of bone-related diseases are mentioned as well.
Collapse
Affiliation(s)
- Nina Zemanova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia
| | - Radoslav Omelka
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia
| | - Vladimira Mondockova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia
| | - Veronika Kovacova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia
| | - Monika Martiniakova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia
| |
Collapse
|
20
|
Liu L, Tian F, Li GY, Xu W, Xia R. The effects and significance of gut microbiota and its metabolites on the regulation of osteoarthritis: Close coordination of gut-bone axis. Front Nutr 2022; 9:1012087. [PMID: 36204373 PMCID: PMC9530816 DOI: 10.3389/fnut.2022.1012087] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/07/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is a common chronic degenerative disease of articular cartilage in middle-aged and older individuals, which can result in the joint pain and dysfunction, and even cause the joint deformity or disability. With the enhancing process of global aging, OA has gradually become a major public health problem worldwide. Explaining pathogenesis of OA is critical for the development of new preventive and therapeutic interventions. In recent years, gut microbiota (GM) has been generally regarded as a “multifunctional organ,” which is closely relevant with a variety of immune, metabolic and inflammatory functions. Meanwhile, more and more human and animal researches have indicated the existence of gut-bone axis and suggested that GM and its metabolites are closely involved in the pathogenic process of OA, which might become a potential and promising intervention target. Based on the close coordination of gut-bone axis, this review aims to summarize and discuss the mechanisms of GM and its metabolites influencing OA from the aspects of the intestinal mucosal barrier modulation, intestinal metabolites modulation, immune modulation and strategies for the prevention or treatment of OA based on perspectives of GM and its metabolites, thus providing a profound knowledge and recognition of it.
Collapse
|
21
|
Gleason B, Chisari E, Parvizi J. Osteoarthritis Can Also Start in the Gut: The Gut-Joint Axis. Indian J Orthop 2022; 56:1150-1155. [PMID: 35813544 PMCID: PMC9232669 DOI: 10.1007/s43465-021-00473-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 07/31/2021] [Indexed: 02/04/2023]
Abstract
Background Osteoarthritis is a common cause of pain and disability with an increasing prevalence among the global population (Hunter and Bierma-Zeinstra in Lancet 393(10182):1745-1759, 2019; Zhang and Jordan in Clinics in Geriatric Medicine 26(3):355-369, 2010). Altered immune responses and low-grade systemic inflammation driven by gut dysbiosis are being increasingly recognized as contributing factors to the pathophysiology of OA (Tan et al. in International Journal of Rheumatic Diseases. https://doi.org/10.1111/1756-185X.14123, 2021; Binvignat et al. in Joint, Bone, Spine 88(5):105203, 2021; Ramasamy et al. in Nutrients 13(4):1272, 2021), which increased the interest in the so-called "gut-joint axis". The various microbiota in the gastrointestinal tract is commonly referred to as the gut microbiome. The gut microbiome is affected by age, sex, and immune system activity as well as medications, environment, and diet (Arumugam in Nature. https://doi.org/10.1038/nature09944, 2011). The microbiome is pivotal to maintain host health and contributes to nutrition, host defense, and immune development (Nishida et al. in Clinical Journal of Gastroenterology 11:1-10, 2018). Alterations in this microbiome can induce dysbiosis, which is associated with many human disease states including allergies, autoimmune disease, diabetes, and cancer (Lin and Zhang in BMC Immunology 18(1):2, 2017). A gut-joint axis is proposed as a link involving the gastrointestinal microbiome, the immune response that it induces, and joint health. Results Emerging evidence has shown that there are specific changes in the microbiome that are associated with osteoarthritis, including increased Firmicutes/Bacteroides ratio, Streptococcus spp. prevalence, and local inflammation (Collins in Osteoarthritis and Cartilage. https://doi.org/10.1016/j.joca.2015.03.014, 2015; Rios in Science and Reports. https://doi.org/10.1038/s41598-019-40601-x, 2019; Schott in JCI insight. https://doi.org/10.1172/jci.insight.95997, 2018; Boer et al. in Nature Communications 10:4881, 2019). Both the innate and adaptive immune systems are affected by the gut microbiome and can become dysregulated in dysbiosis which ultimately triggers events associated with joint OA. Conclusions The gut is an intriguing and novel target for OA therapy. Dietary modification or supplementation with fiber, probiotics, or prebiotics could provide a positive impact on the gut joint axis.
Collapse
Affiliation(s)
- Brendan Gleason
- Rothman Orthopaedic Institute at Thomas Jefferson University, 125 S 9th St. Ste 1000, Philadelphia, PA 19107 USA
| | - Emanuele Chisari
- Rothman Orthopaedic Institute at Thomas Jefferson University, 125 S 9th St. Ste 1000, Philadelphia, PA 19107 USA
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Javad Parvizi
- Rothman Orthopaedic Institute at Thomas Jefferson University, 125 S 9th St. Ste 1000, Philadelphia, PA 19107 USA
| |
Collapse
|
22
|
A systematic review of microbiome composition in osteoarthritis subjects. Osteoarthritis Cartilage 2022; 30:786-801. [PMID: 34958936 DOI: 10.1016/j.joca.2021.12.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/30/2021] [Accepted: 12/19/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) started to be associated to shifted microbiota composition recently. This systematic review aims to elucidate if there is a common microbiota composition linked with OA between different studies. METHODS We screened PubMed, Scopus, Web of Science and Cochrane databases up to July 26th 2021 to identify original studies in which microbiome was assessed from OA individuals, both in human and laboratory animals' studies. Bacteria associated with OA were summarized to find common patterns between the studies. RESULTS We identified 37 original studies where the microbiota composition was assessed in OA subjects. We identified some bacteria (Clostridium, Streptococcus, Bacteroides and Firmicutes) that were reported to be upregulated in OA subjects, whereas Lactobacillus and Bifidobacterium longum were associated with improved OA outcomes. The heterogeneity of sampling and analysis methods, different taxonomical levels reported and the lack of healthy controls in several studies made it difficult to compare the studies and reach conclusions about a potential causal link. CONCLUSIONS The current study demonstrated that some bacteria were identified as regulators of OA. Future works following standardized methodologies with more proper controls are needed to elucidate our understanding of the role of the microbiota in OA pathogenesis and progress towards new treatments.
Collapse
|
23
|
Chen Y, Wang X, Zhang C, Liu Z, Li C, Ren Z. Gut Microbiota and Bone Diseases: A Growing Partnership. Front Microbiol 2022; 13:877776. [PMID: 35602023 PMCID: PMC9121014 DOI: 10.3389/fmicb.2022.877776] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/19/2022] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota is key to human health and disease. Convincing studies have demonstrated that dysbiosis in the commensal gut microbiota is associated with intestinal and extra-intestinal diseases. Recent explorations have significantly contributed to the understanding of the relationship between gut microbiota and bone diseases (osteoporosis, osteoarthritis, rheumatoid arthritis, and bone cancer). Gut microbiota and its metabolites may become associated with the development and progression of bone disorders owing to their critical role in nutrient absorption, immunomodulation, and the gut–brain–bone axis (regulation hormones). In this work, we review the recent developments addressing the effect of gut microbiota modulation on skeletal diseases and explore a feasible preventive approach and therapy for bone diseases.
Collapse
Affiliation(s)
- Yu Chen
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xin Wang
- Department of Orthopaedic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunlei Zhang
- Bone Tumour and Bone Disease Department II, Zhengzhou Orthopaedic Hospital, Zhengzhou, China
| | - Zhiyong Liu
- Department of Orthopaedic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Chao Li
- Department of Orthopaedic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhigang Ren
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Zhigang Ren,
| |
Collapse
|
24
|
Gayathri D, Prashantkumar CS, Vasudha M. Current Insights on the Modulation of Gut Microbiome and Its Effect on Human Health. MICROBIOME-GUT-BRAIN AXIS 2022:29-51. [DOI: 10.1007/978-981-16-1626-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
25
|
Liu C, Tseng YP, Chan LP, Liang CH. The potential of Streptococcus thermophiles (TCI633) in the anti-aging. J Cosmet Dermatol 2021; 21:2635-2647. [PMID: 34587358 DOI: 10.1111/jocd.14445] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/19/2021] [Accepted: 08/27/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Streptococcus thermophilus (TCI633) is a probiotic that has been newly isolated from human breast milk, and it can produce hyaluronic acid (HA) when colonizing the gastrointestinal (GI) tract of rodents and humans. A recent study has the established that TCI633 can alleviate synovial tissue inflammation and has potential to mitigate the progression of osteoarthritis. OBJECTIVE TCI633 has not been available for use in skincare and this preliminary clinical study will assess its improvement of the skin. METHODS In this study, DNA protection, Hyaluronidase assay, cell viability, and collagen synthesis on human fibroblasts of TCI633 were assessed. Subjects were enrolled in this clinical study and randomly assigned to the TCI633 or placebo group. Each subject was informed to intake two tablets daily for 8 weeks. Each subject was required to undergo skin condition inspection at weeks 0, 4, and 8 and hematology tests to monitor HA, superoxide dismutase (SOD) and catalase levels, and kidney and liver function at weeks 0 and 8. RESULTS The effects of TCI633 supplementation, including the promotion of skin cell proliferation, the increase of their collagen content, their protection against DNA damage, and the inhibition of hyaluronidase activities, are investigated. Subjects were recruited for an 8-week long clinical trial to confirm the efficacy of TCI633 in improving the serum biochemical HA, SOD and catalase levels, and anti-skin age markers. CONCLUSIONS This work provides an alternative approach to improving health, indicating the potential of TCI633 supplementation to delay the aging of skin and improve its condition.
Collapse
Affiliation(s)
- Cheng Liu
- Department of Health and Beauty, Kaohsiung, Taiwan.,Department of Optometry, Kaohsiung, Taiwan.,Department of Dental Technology, Shu-Zen Junior College of Medicine and Management, Kaohsiung, Taiwan
| | - Ya-Ping Tseng
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Leong-Perng Chan
- Department of Otorhinolaryngology-Head and Neck Surgery, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Hua Liang
- Department of Cosmetic Science and Institute of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| |
Collapse
|
26
|
Abstract
PURPOSE OF THE REVIEW The human gut harbors a complex community of microbes that influence many processes regulating musculoskeletal development and homeostasis. This review gives an update on the current knowledge surrounding the impact of the gut microbiota on musculoskeletal health, with an emphasis on research conducted over the last three years. RECENT FINDINGS The gut microbiota and their metabolites are associated with sarcopenia, osteoporosis, osteoarthritis, and rheumatoid arthritis. The field is moving fast from describing simple correlations to pursue establishing causation through clinical trials. The gut microbiota and their microbial-synthesized metabolites hold promise for offering new potential alternatives for the prevention and treatment of musculoskeletal diseases given its malleability and response to environmental stimuli.
Collapse
Affiliation(s)
- R Li
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - C G Boer
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - L Oei
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
27
|
Binvignat M, Sokol H, Mariotti-Ferrandiz E, Berenbaum F, Sellam J. Osteoarthritis and gut microbiome. Joint Bone Spine 2021; 88:105203. [PMID: 33962035 DOI: 10.1016/j.jbspin.2021.105203] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2021] [Indexed: 12/20/2022]
Affiliation(s)
- Marie Binvignat
- Sorbonne Université, Department of Rheumatology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Inserm UMRS_938, FHU PaCeMM, 184, Rue du Faubourg Saint-Antoine, 75012 Paris, France
| | - Harry Sokol
- Sorbonne Université, Department of Gastroenterology, AP-HP, Hôpital Saint-Antoine, Inserm UMRS_938, FHU PaCeMM, 75012 Paris, France
| | - Encarnita Mariotti-Ferrandiz
- Sorbonne Université, Department of Immunology-Immunopathology- Immunotherapy- Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Inserm URMS_959, 75013 Paris, France
| | - Francis Berenbaum
- Sorbonne Université, Department of Rheumatology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Inserm UMRS_938, FHU PaCeMM, 184, Rue du Faubourg Saint-Antoine, 75012 Paris, France
| | - Jérémie Sellam
- Sorbonne Université, Department of Rheumatology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Inserm UMRS_938, FHU PaCeMM, 184, Rue du Faubourg Saint-Antoine, 75012 Paris, France.
| |
Collapse
|
28
|
Oral Administration of Lactobacillus rhamnosus Ameliorates the Progression of Osteoarthritis by Inhibiting Joint Pain and Inflammation. Cells 2021; 10:cells10051057. [PMID: 33946919 PMCID: PMC8146916 DOI: 10.3390/cells10051057] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 12/14/2022] Open
Abstract
Osteoarthritis (OA) is the most common form of arthritis and age-related degenerative joint disorder, which adversely affects quality of life and causes disability. However, the pathogenesis of OA remains unclear. This study was performed to examine the effects of Lactobacillus rhamnosus in OA progression. OA was induced in 6-week-old male Wistar rats by monosodium iodoacetate (MIA) injection, and the effects of oral administration of L. rhamnosus were examined in this OA rat model. Pain severity, cartilage destruction, and inflammation were measured in MIA-induced OA rats. The small intestines were isolated from OA rats, and the intestinal structure and inflammation were measured. Protein expression in the dorsal root ganglion was analyzed by immunohistochemistry. The effects of L. rhamnosus on mRNA and protein expression in chondrocytes stimulated with interleukin (IL)-1β and lipopolysaccharide (LPS) were analyzed by real-time polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA). Pain severity was decreased in L. rhamnosus-treated MIA-induced OA rats. The levels of expression of MCP-1, a potential inflammatory cytokine, and its receptor, CCR2, were decreased, and GABA and PPAR-γ expression were increased in L. rhamnosus-treated OA rats. The inflammation, as determined by IL-1β, and cartilage destruction, as determined by MMP3, were also significantly decreased by L. rhamnosus in OA rats. Additionally, intestinal damage and inflammation were improved by L. rhamnosus. In human OA chondrocytes, TIMP1, TIMP3, SOX9, and COL2A1 which are tissue inhibitors of MMP, and IL-10, an anti-inflammatory cytokine, were increased by L. rhamnosus. L. rhamnosus treatment led to decreased pain severity and cartilage destruction in a rat model of OA. Intestinal damage and inflammation were also decreased by L. rhamnosus treatment. Our findings suggested the therapeutic potential of L. rhamnosus in OA.
Collapse
|
29
|
Papageorgiou M, Biver E. Interactions of the microbiome with pharmacological and non-pharmacological approaches for the management of ageing-related musculoskeletal diseases. Ther Adv Musculoskelet Dis 2021; 13:1759720X211009018. [PMID: 34104230 PMCID: PMC8172340 DOI: 10.1177/1759720x211009018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/18/2021] [Indexed: 12/18/2022] Open
Abstract
Despite major progress in the understanding of the pathophysiology and therapeutic options for common ageing-related musculoskeletal conditions (i.e. osteoporosis and associated fractures, sarcopenia and osteoarthritis), there is still a considerable proportion of patients who respond sub optimally to available treatments or experience adverse effects. Emerging microbiome research suggests that perturbations in microbial composition, functional and metabolic capacity (i.e. dysbiosis) are associated with intestinal and extra-intestinal disorders including musculoskeletal diseases. Besides its contributions to disease pathogenesis, the role of the microbiome is further extended to shaping individuals' responses to disease therapeutics (i.e. pharmacomicrobiomics). In this review, we focus on the reciprocal interactions between the microbiome and therapeutics for osteoporosis, sarcopenia and osteoarthritis. Specifically, we identify the effects of therapeutics on microbiome's configurations, functions and metabolic output, intestinal integrity and immune function, but also the effects of the microbiome on the metabolism of these therapeutics, which in turn, may influence their bioavailability, efficacy and side-effect profile contributing to variable treatment responses in clinical practice. We further discuss emerging strategies for microbiota manipulation as preventive or therapeutic (alone or complementary to available treatments) approaches for improving outcomes of musculoskeletal health and disease.
Collapse
Affiliation(s)
- Maria Papageorgiou
- Division of Bone Diseases, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Emmanuel Biver
- Division of Bone Diseases, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Rue Gabrielle Perret Gentil 4, Geneva 1205, Switzerland
| |
Collapse
|
30
|
Probiotics for pain of osteoarthritis; An N-of-1 trial of individual effects. Complement Ther Med 2020; 54:102548. [PMID: 33183666 DOI: 10.1016/j.ctim.2020.102548] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE This study aimed to investigate the safety and effectiveness of probiotics in osteoarthritic pain for one individual. METHODS The study was an N-of-1 trial design, divided into 3 blocks of 10 weeks. Each block included one pair of randomized interventions (AB), separated by a washout period. The trial took place in a private naturopathic practice in Sydney, Australia. The participant was a 67 year old female with osteoarthritis in her lower back and right ankle. The active intervention was two daily capsules that contained Lactobacillus rhamnosus (LGG®), Saccharomyces cerevisiae (boulardii) and Bifidobacterium animalis ssp lactis. The placebo was an identical capsule that did not contain probiotics. The primary outcome was daily pain scores, measured by the participant on a Visual Analogue Scale (VAS). Secondary outcome measures included patient preference (of intervention), General Health Questionnaire (GHQ-12), Patient Specific Functional Scale (PSFS), Comprehensive Digestive Stool Analysis (CDSA) and rescue medication usage. A dependent t-test analysed mean pain scores for the last week of each intervention across the three blocks of the study. RESULTS The probiotic intervention was associated with lower pain scores and was the preferred intervention chosen by the participant. The mean pain score on the VAS was 4.9 ± 2.2 in the placebo condition compared to 4.0 ± 1.7 in the probiotic condition (t(20) = 2.2, p = 0.04, difference = 0.9, 95 % CI [0.04, 1.77]). CONCLUSIONS The reduction in pain scores associated with the probiotic intervention was small but clinically significant for this patient. A holistic view of the patient focusing on digestive integrity and function may be crucial for clinical applications of interventions such as probiotics. N-of-1 trial designs allow for the measurement of a holistic approach to an individual, which is aligned with naturopathic practice. Further trials are required to generate data to enable reliable estimation of population effects.
Collapse
|