101
|
Potential roles of 1,5-anhydro-D-fructose in modulating gut microbiome in mice. Sci Rep 2021; 11:19648. [PMID: 34608200 PMCID: PMC8490373 DOI: 10.1038/s41598-021-99052-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 09/17/2021] [Indexed: 12/23/2022] Open
Abstract
The gut microbiota has tremendous potential to affect the host’s health, in part by synthesizing vitamins and generating nutrients from food that is otherwise indigestible by the host. 1,5-Anhydro-d-fructose (1,5-AF) is a monosaccharide with a wide range of bioactive potentials, including anti-oxidant, anti-inflammatory, and anti-microbial effects. Based on its potential benefits and minimal toxicity, it is anticipated that 1,5-AF will be used as a dietary supplement to support general health. However, the effects of 1,5-AF on the gut microbiota are yet to be clarified. Here, using an unbiased metagenomic approach, we profiled the bacterial taxa and functional genes in the caecal microbiota of mice fed a diet containing either 2% 1,5-AF or a reference sweetener. Supplementation with 1,5-AF altered the composition of the gut microbiota, enriching the proportion of Faecalibacterium prausnitzii. 1,5-AF also altered the metabolomic profile of the gut microbiota, enriching genes associated with nicotinamide adenine dinucleotide biosynthesis. These findings support the potential benefits of 1,5-AF, but further studies are required to clarify the impact of 1,5-AF on health and disease.
Collapse
|
102
|
Martinez RE, Leatherwood JL, Arnold CE, Glass KG, Walter KW, Valigura HC, Norton SA, White-Springer SH. Responses to an intra-articular lipopolysaccharide challenge following dietary supplementation of Saccharomyces cerevisiae fermentation product in young horses. J Anim Sci 2021; 99:6383485. [PMID: 34619765 PMCID: PMC8557629 DOI: 10.1093/jas/skab272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/21/2021] [Indexed: 12/22/2022] Open
Abstract
Dietary intervention may be a valuable strategy to optimize the intra-articular environment in young horses to prolong their performance career. To test the hypothesis that dietary supplementation of a Saccharomyces cerevisiae fermentation product would reduce markers of joint inflammation and increase markers of cartilage metabolism following a single inflammatory insult, Quarter Horse yearlings (mean ± SD; 9 ± 1.0 mo) were balanced by age, sex, body weight (BW), and farm of origin and randomly assigned to the following treatment groups: 1.25% BW/d (dry matter basis) custom-formulated concentrate only (CON; n = 9) or concentrate top-dressed with 21 g/d S. cerevisiae fermentation product (SCFP; n = 10) for 98 d. Horses had ad libitum access to Coastal bermudagrass hay. On day 84, one randomly selected radial carpal joint from each horse was injected with 0.5 ng lipopolysaccharide (LPS) solution. The remaining carpal joint was injected with sterile lactated Ringer’s solution as a contralateral control. Synovial fluid obtained before supplementation (day 0) and on day 84 at preinjection hour 0 and 6, 12, 24, 168, and 336 h postinjection was analyzed for prostaglandin E2 (PGE2), carboxypropeptide of type II collagen (CPII), and collagenase cleavage neopeptide (C2C) by commercial assays. Rectal temperature, heart rate, respiration rate, carpal surface temperature, and carpal circumference were recorded prior to each sample collection and for 24 h postinjection. Data were analyzed using linear models with repeated measures. From day 0 to 84, synovial C2C declined (P ≤ 0.01) and the CPII:C2C ratio increased (P ≤ 0.01) in all horses with no effect of diet. In response to intra-articular LPS, synovial PGE2 increased by hour 6 (P ≤ 0.01) and returned to baseline by hour 336; CPII increased by hour 12, remained elevated through hour 168 (P ≤ 0.01), and returned to baseline by hour 336; and C2C increased by hour 6 (P ≤ 0.01) but did not return to baseline through hour 336 (P ≤ 0.01). Post-intra-articular injection, PGE2 levels were lower in SCFP than CON horses (P = 0.01) regardless of injection type. Synovial CPII and the CPII:C2C ratio demonstrated stability during the LPS challenge in SCFP compared with CON horses (P ≤ 0.01). Clinical parameters were not influenced by diet but increased in response to repeated arthrocentesis (P ≤ 0.01). Dietary SCFP may favorably modulate intra-articular inflammation following an acute stressor and influence cartilage turnover in young horses.
Collapse
Affiliation(s)
- Rafael E Martinez
- Department of Animal Science, Texas A&M University and Texas A&M AgriLife Research, College Station, TX 77843, USA
| | - Jessica L Leatherwood
- Department of Animal Science, Texas A&M University and Texas A&M AgriLife Research, College Station, TX 77843, USA
| | - Carolyn E Arnold
- Department of Large Animal Clinical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Kati G Glass
- Department of Large Animal Clinical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Kelly W Walter
- Department of Agricultural Science, Truman State University, Kirksville, MO 63501, USA
| | - Hannah C Valigura
- Department of Animal Science, Texas A&M University and Texas A&M AgriLife Research, College Station, TX 77843, USA
| | | | - Sarah H White-Springer
- Department of Animal Science, Texas A&M University and Texas A&M AgriLife Research, College Station, TX 77843, USA
| |
Collapse
|
103
|
Song Y, Hao D, Jiang H, Huang M, Du Q, Lin Y, Liu F, Chen B. Nrf2 Regulates CHI3L1 to Suppress Inflammation and Improve Post-Traumatic Osteoarthritis. J Inflamm Res 2021; 14:4079-4088. [PMID: 34466014 PMCID: PMC8403022 DOI: 10.2147/jir.s310831] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 08/18/2021] [Indexed: 01/16/2023] Open
Abstract
Introduction Post-traumatic osteoarthritis (PTOA) is an inflammatory condition that occurs following mechanical joint trauma and that results in joint degeneration. This study sought to evaluate the regulatory function of nuclear factor erythroid 2-related factor 2 (Nrf2) in a murine model of anterior cruciate ligament transection (ACLT)-induced PTOA and in an in vitro model of synoviocyte inflammation induced by LPS treatment with the goal of exploring the role of chitinase 3-like-1 (CHI3L1) in this pathogenic context. Methods PTOA model mice were intra-articularly injected with Nrf2 overexpression lentiviral vector, and safranin O-fast green staining as well as the Osteoarthritis Research Society International (OARSI) Scoring System were used to evaluate the severity of cartilage damage. Protein expression in the synovial tissue was evaluated by Western blotting, immunohistochemical staining, and ELISA. Additionally, murine synoviocytes were infected with Nrf2 overexpression lentivirus and stimulated with LPS. The levels of inflammatory cytokines were detected by ELISA. ROS levels were measured using dihydroethidium (DHE) dye. Results We determined that the overexpression of Nrf2 was sufficient to reduce cartilage degradation in the context of PTOA in vivo, and we observed a significant decrease in the expression of matrix metalloproteinase 13 (MMP13) in the articular cartilage of samples from mice overexpressing Nrf2 relative to control mice. Synovial CHI3L1 expression and serum TNF-α, IL-1β, and IL-6 levels were reduced in animals overexpressing this transcription factor relative to PTOA model controls. Consistent with these findings, murine synoviocytes treated with LPS exhibited dose-dependent increases in ROS, TNF-α, IL-1β, IL-6, Nrf2, and CHI3L1 levels, whereas Nrf2 overexpression was sufficient to suppress these increases. Conclusion Our data indicated that Nrf2 negatively regulates CHI3L1, suggesting that this signaling axis may regulate PTOA progression and may thus be a viable therapeutic target in individuals affected by this condition.
Collapse
Affiliation(s)
- Yang Song
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.,Division of Traumatology and Joint, Department of Orthopaedics, Shunde Hospital, Southern Medical University, Foshan, 528308, People's Republic of China
| | - Dake Hao
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Huan Jiang
- Department of Anesthesiology, Shunde Hospital, Southern Medical University, Foshan, 528308, People's Republic of China
| | - Mingguang Huang
- Division of Traumatology and Joint, Department of Orthopaedics, Shunde Hospital, Southern Medical University, Foshan, 528308, People's Republic of China
| | - Qingjun Du
- Division of Traumatology and Joint, Department of Orthopaedics, Shunde Hospital, Southern Medical University, Foshan, 528308, People's Republic of China
| | - Yi Lin
- Division of Traumatology and Joint, Department of Orthopaedics, Shunde Hospital, Southern Medical University, Foshan, 528308, People's Republic of China
| | - Fei Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Bin Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| |
Collapse
|
104
|
Hernandez CJ. Musculoskeletal Microbiology: The Microbiome in Orthopaedic Biomechanics. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021; 19:100290. [PMID: 34151053 PMCID: PMC8211101 DOI: 10.1016/j.cobme.2021.100290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Orthopaedic biomechanics and bioengineering is a founding discipline of biomedical engineering that focuses on the effects of mechanical stress and strain on musculoskeletal tissues during growth, function and repair. In the past decade the gut microbiome has emerged as a contributor to disease processes throughout the body, but only recently has been shown to influence orthopaedic biomechanics. Here I review emergent findings showing that the gut microbiome can regulate important aspects of the musculoskeletal system including growth and development; tissue failure and disease; and orthopaedic surgery. These early findings suggest that the microbiome may help answer questions in orthopaedic biomechanics that are not well addressed by current interventions, and highlights the promise of the emerging field of "Musculoskeletal Microbiology".
Collapse
Affiliation(s)
- Christopher J Hernandez
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
- Hospital for Special Surgery, New York, NY, USA
| |
Collapse
|
105
|
Larder CE, Iskandar MM, Kubow S. Gastrointestinal Digestion Model Assessment of Peptide Diversity and Microbial Fermentation Products of Collagen Hydrolysates. Nutrients 2021; 13:nu13082720. [PMID: 34444880 PMCID: PMC8401164 DOI: 10.3390/nu13082720] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/30/2021] [Accepted: 08/04/2021] [Indexed: 01/11/2023] Open
Abstract
Osteoarthritis (OA), the most common form of arthritis, is associated with metabolic diseases and gut microbiome dysbiosis. OA patients often take supplements of collagen hydrolysates (CHs) with a high peptide content. Following digestion, some peptides escape absorption to induce prebiotic effects via their colonic fermentation to generate short-chain fatty acids (SCFAs), branched-chain fatty acids (BCFAs) and colonic gases (NH4 and H2S). The capacity of CHs to generate microbial metabolites is unknown. Proteomic analysis of two CHs (CH-GL and CH-OPT) demonstrated different native peptide profiles with increased peptide diversity after in vitro gastric and small intestinal digestion. Subsequent 24 h fermentation of the CH digests in a dynamic gastrointestinal (GI) digestion model containing human fecal matter showed that CH-OPT increased (p < 0.05) H2S, SCFAs (propionic, butyric and valeric acids), BCFAs, and decreased NH4 in the ascending colon reactor with no major changes seen with CH-GL. No major effects were observed in the transverse and descending vessels for either CH. These findings signify that CHs can induce prebiotic effects in the ascending colon that are CH dependent. More studies are needed to determine the physiological significance of CH-derived colonic metabolites, in view of emerging evidence connecting the gut to OA and metabolic diseases.
Collapse
|
106
|
Zhao R, Ji Y, Chen X, Hu Q, Zhao L. Polysaccharide from Flammulina velutipes attenuates markers of metabolic syndrome by modulating the gut microbiota and lipid metabolism in high fat diet-fed mice. Food Funct 2021; 12:6964-6980. [PMID: 34137411 DOI: 10.1039/d1fo00534k] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Natural biological macromolecules with putative functions of gut microbiota regulation possess the advantage of improving metabolic syndrome (MS). In this research, we aimed to determine the effects of Flammulina velutipes polysaccharide (FVP) (Expt. 1) and fecal microbiota transplantation (FMT) (Expt. 2) on MS-related disorders, gut microbiota structure changes and their underlying mechanisms in a murine model fed with high-fat diet (HFD). In Expt. 1, six-week-old male C57BL/6J mice were fed with a control diet (10% calories from fat) or a high fat diet (45% calories from fat), administered with saline or FVP (0.4 mg per g b.w.) by gavage over a 12-week period. In Expt. 2, mice were fed with a HFD, administered with fecal supernatants from healthy and FVP-fed donor mice for 12 weeks simultaneously. The body mass, blood lipid levels and blood glucose homeostasis of mice were analyzed, and total RNA from mouse liver and adipose tissue were extracted by TRIzol and the lipid metabolism-related gene expressions were calculated by qRT-PCR. Gut microbiota changes were evaluated by high-throughput sequencing. Results indicated that FVP and FMT supplementations showed an attenuation effect on mouse obesity, hyperlipidemia and insulin resistance. Up-regulated expressions of Ampkα1 and Ppara were found both in FVP and FMT treatment groups. Different changes were found in the gut microbiota caused by FVP and FMT, respectively. PICRUSt analysis indicated that compared with FVP supplementation, FMT showed a significant effect on regulating lipid metabolism in HFD-fed mice. The findings from this study indicated that oral administrations of FVP or FMT could significantly attenuate MS-related obesity, hyperlipidemia and insulin resistance in HFD-fed mice, and the beneficial effects may be mediated through lipid metabolism and gut microbiota regulation in different ways. These results improve the understanding of the functional activity of FVP as prebiotics.
Collapse
Affiliation(s)
- Ruiqiu Zhao
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.
| | | | | | | | | |
Collapse
|
107
|
Karakan T, Tuohy KM, Janssen-van Solingen G. Low-Dose Lactulose as a Prebiotic for Improved Gut Health and Enhanced Mineral Absorption. Front Nutr 2021; 8:672925. [PMID: 34386514 PMCID: PMC8353095 DOI: 10.3389/fnut.2021.672925] [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: 02/26/2021] [Accepted: 06/11/2021] [Indexed: 12/11/2022] Open
Abstract
Although medium and high doses of lactulose are used routinely for the treatment of constipation and hepatic encephalopathy, respectively, a wealth of evidence demonstrates that, at low doses, lactulose can also be used as a prebiotic to stimulate the growth of health-promoting bacteria in the gastrointestinal tract. Indeed, multiple preclinical and clinical studies have shown that low doses of lactulose enhance the proliferation of health-promoting gut bacteria (e.g., Bifidobacterium and Lactobacillus spp.) and increase the production of beneficial metabolites [e.g., short-chain fatty acids (SCFAs)], while inhibiting the growth of potentially pathogenic bacteria (e.g., certain clostridia). SCFAs produced upon microbial fermentation of lactulose, the most abundant of which is acetate, are likely to contribute to immune regulation, which is important not only within the gut itself, but also systemically and for bone health. Low-dose lactulose has also been shown to enhance the absorption of minerals such as calcium and magnesium from the gut, an effect which may have important implications for bone health. This review provides an overview of the preclinical and clinical evidence published to date showing that low-dose lactulose stimulates the growth of health-promoting gut bacteria, inhibits the growth of pathogenic bacteria, increases the production of beneficial metabolites, improves mineral absorption, and has good overall tolerability. Implications of these data for the use of lactulose as a prebiotic are also discussed.
Collapse
Affiliation(s)
- Tarkan Karakan
- Department of Gastroenterology, Gazi University School of Medicine, Ankara, Turkey
| | - Kieran Michael Tuohy
- Department of Food Quality and Nutrition, Research and Innovation Center, Fondazione Edmund Mach, San Michele all'Adige, Italy
| | | |
Collapse
|
108
|
Specific Pathogen-Free Animals for Civilian and Military Trauma: a Cautionary Note in the Translation of New Drug Therapies. Shock 2021; 54:232-236. [PMID: 32665536 DOI: 10.1097/shk.0000000000001495] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Specific-pathogen free (SPF) animals were introduced into biomedical research in the early 1960s to reduce the incidence of disease into experimental design. The goal was to provide animals with selected microbiota compatible with sustained health. Sixty years later, SPF status has become a variable itself in biomedical research. Alterations in the gut microbiome-host relationship can profoundly influence basic physiology, immune/inflammatory function, susceptibility to infection and disease, and behavior. In addition, it can influence the translational success of a drug or technology from animal models to humans. We discuss this aspect of SPF status in animal models used for military or civilian trauma and shock research. Currently, there is a broad spectrum of SPF exclusion and inclusion criteria which vary from one supplier or animal husbandry facility. If translation to humans is the end-game of trauma research, we recommend replicating a gut microbiome similar to the wild-type for optimal success. We further suggest that at the end of each publication a URL access be provided on Animal Microbial/Pathogen Exclusion Status that a study was based upon. This may help address the differences in results within a single laboratory or between laboratories around the world and improve translation success.
Collapse
|
109
|
Collins KH, Schwartz DJ, Lenz KL, Harris CA, Guilak F. Taxonomic changes in the gut microbiota are associated with cartilage damage independent of adiposity, high fat diet, and joint injury. Sci Rep 2021; 11:14560. [PMID: 34267289 PMCID: PMC8282619 DOI: 10.1038/s41598-021-94125-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/30/2021] [Indexed: 12/02/2022] Open
Abstract
Lipodystrophic mice are protected from cartilage damage following joint injury. This protection can be reversed by the implantation of a small adipose tissue graft. The purpose of this study was to evaluate the relationship between the gut microbiota and knee cartilage damage while controlling for adiposity, high fat diet, and joint injury using lipodystrophic (LD) mice. LD and littermate control (WT) mice were fed a high fat diet, chow diet, or were rescued with fat implantation, then challenged with destabilization of the medial meniscus surgery to induce osteoarthritis (OA). 16S rRNA sequencing was conducted on feces. MaAslin2 was used to determine associations between taxonomic relative abundance and OA severity. While serum LPS levels between groups were similar, synovial fluid LPS levels were increased in both limbs of HFD WT mice compared to all groups, except for fat transplanted animals. The Bacteroidetes:Firmicutes ratio of the gut microbiota was significantly reduced in HFD and OA-rescued animals when compared to chow. Nine novel significant associations were found between gut microbiota taxa and OA severity. These findings suggest the presence of causal relationships the gut microbiome and cartilage health, independent of diet or adiposity, providing potential therapeutic targets through manipulation of the microbiome.
Collapse
Affiliation(s)
- Kelsey H Collins
- Department of Orthopaedic Surgery, Washington University, Couch Building Room 3213, 4523 Clayton Avenue, St Louis, MO, 63110, USA
- Shriners Hospitals for Children, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA
| | - Drew J Schwartz
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kristin L Lenz
- Department of Orthopaedic Surgery, Washington University, Couch Building Room 3213, 4523 Clayton Avenue, St Louis, MO, 63110, USA
- Shriners Hospitals for Children, St. Louis, MO, USA
| | - Charles A Harris
- Division of Endocrinology, Washington University, St. Louis, MO, USA
- Early Clinical Development & Experimental Sciences, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, Couch Building Room 3213, 4523 Clayton Avenue, St Louis, MO, 63110, USA.
- Shriners Hospitals for Children, St. Louis, MO, USA.
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA.
| |
Collapse
|
110
|
Kim JW, Ju JH. Gut Microbiome Bridges Over Troubled Joints. JOURNAL OF RHEUMATIC DISEASES 2021; 28:111-112. [PMID: 37475991 PMCID: PMC10324901 DOI: 10.4078/jrd.2021.28.3.111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 06/21/2021] [Indexed: 07/22/2023]
Affiliation(s)
- Ji Won Kim
- Division of Rheumatology, Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Ji Hyeon Ju
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
111
|
Wang J, Ishfaq M, Li J. Baicalin ameliorates Mycoplasma gallisepticum-induced inflammatory injury in the chicken lung through regulating the intestinal microbiota and phenylalanine metabolism. Food Funct 2021; 12:4092-4104. [PMID: 33977979 DOI: 10.1039/d1fo00055a] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Baicalin shows excellent protective effects against Mycoplasma gallisepticum (MG) induced inflammatory injury as discussed in our previous studies. However, the physiological effects of baicalin are notable in contrast to its low bioavailability, and the critical mechanism for the protective effects of baicalin against MG infection is still unclear. The main objective of this study was to investigate whether baicalin alleviates MG-induced lung inflammatory injury through regulating gut microbiota. Using an MG infection model, results showed that baicalin treatment significantly reduced MG colonization and ameliorated the abnormal pathological changes in the lung. Baicalin treatment also reduced the level of proinflammatory cytokines and suppressed proinflammatory protein expression. Notably, MG infection changed the gut microbiota composition, however, the abnormal gut microbiota composition was partially alleviated by baicalin treatment. Baicalin significantly enriched the commensal bacterium Bacteroides fragilis, and gavaged with Bacteroides fragilis alleviating MG infection-induced inflammatory injury in the lung. In addition, baicalin reversed peripheral accumulation of phenylalanine induced by MG infection. Importantly, increased phenylalanine induced excessive necroptosis through the modulation of gga-miR-190a-3p-Fas-associated protein with death domain (FADD) axis in HD11 macrophages. Together, our findings highlighted the role of gut microbiota and phenylalanine metabolism in MG infection and confirmed that baicalin could effectively inhibit MG-induced inflammatory injury in the lung by remodeling the gut microbiota and phenylalanine metabolism.
Collapse
Affiliation(s)
- Jian Wang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, Xiangfang District, P. R. China.
| | - Muhammad Ishfaq
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, Xiangfang District, P. R. China.
| | - Jichang Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, Xiangfang District, P. R. China. and Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, Xiangfang District, P. R. China
| |
Collapse
|
112
|
Chen J, Wang A, Wang Q. Dysbiosis of the gut microbiome is a risk factor for osteoarthritis in older female adults: a case control study. BMC Bioinformatics 2021; 22:299. [PMID: 34082693 PMCID: PMC8173911 DOI: 10.1186/s12859-021-04199-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/12/2021] [Indexed: 11/10/2022] Open
Abstract
Background Osteoarthritis (OA) is a multifactorial joint degenerative disease with low-grade inflammation. The gut microbiome has recently emerged as an pathogenic factor of OA, and prebiotics supplementation could alleviate OA symptoms in animal models. However, the relationship between the gut microbiome and OA in the older female adults is hitherto not clear. Results Here we studied the gut microbiome of 57 OA patients and their healthy controls by metagenome-wide association study based on previously published data. A significant reduction in the richness and diversity of gut microbiome were observed in OA patients. Bifidobacterium longum and Faecalibacterium prausnitzii were decreased while Clostridium spp. was increased in the OA group. The functional modules, particularly the energetic metabolism and acetate production were also decreased in the OA patients. To evaluate the diagnostic value of identified species for elderly patients with OA, we constructed a set of random forest disease classifiers based on species differences between the two groups. Among them, 9 species reached the lowest classification error in the random forest cross validation, and the area under ROC of the model was 0.81. Conclusions Significant alterations in the gut microbial composition and function were observed between the older patients with OA and their controls, and a random forest classifier model for OA were constructed based on the differences in our study. Our study have identified several potential gut microbial targets in the elderly females with OA, which will facilitate the treatment of OA based on gut microbiota, is of great value in alleviating pain and improving the quality of life for them. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-021-04199-0.
Collapse
Affiliation(s)
- Juanjuan Chen
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, People's Republic of China
| | - Anqi Wang
- Institute of Clinical Research and Translational Medicine, Gansu Provincial Hospital, Lanzhou, Gansu, People's Republic of China
| | - Qi Wang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, People's Republic of China.
| |
Collapse
|
113
|
Wang Z, Zhu H, Jiang Q, Zhu YZ. The gut microbiome as non-invasive biomarkers for identifying overweight people at risk for osteoarthritis. Microb Pathog 2021; 157:104976. [PMID: 34023440 DOI: 10.1016/j.micpath.2021.104976] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To evaluate the potential for identifying overweight people at risk for osteoarthritis from a gut microbiome biomarker. BACKGROUND Osteoarthritis (OA) is the most common form of arthritis, affecting millions of people worldwide. Being overweight increases the load placed on the joints such as the knee, which increases stress and could hasten the breakdown of cartilage. Identifying overweight people at risk for osteoarthritis remains a challenge. However, emerging evidence indicates that microbial dysbiosis in the human gut might play an important role in many inflammatory diseases. Considering the role of inflammation in OA development, analysis of the gut microbiome might be a potential non-invasive tool for overweight individuals to evaluate their risk for OA. RESULTS In this prospective study, we collected 182 stool samples from overweight OA patients (n = 86) and overweight normal people (n = 96) (25 kg/m2<BMI<30 kg/m2). 16S ribosomal RNA gene sequencing for V3 and V4 regions on the Illumina MiSeq platform was used to identify bacteria at different levels and it showed that both the diversity and richness of the gut microbiome decreased in overweight OA patients. Correspondingly, 9 phyla and 87 genera had significantly differences between overweight OA patients and overweight normal people. Finally, we identified 7 optimal microbial biomarkers in genus levels as a panel, including Gemmiger, Klebsiella, Akkermansia, Bacteroides, Prevotella, Alistipes and Parabacteroides, to build the random forest model, and achieved a 83.36% area under the curve (AUC) of receiver operating characteristic (ROC). CONCLUSION We present the first 16S rRNA gene sequencing profiling study of stool microbiomes in overweight people to discover and validate microbial biomarkers indicating risk for OA. Our study successfully established a 7 biomarkers prediction panel, moving towards affordable non-invasive early diagnostic biomarkers for OA in stool samples from overweight individuals.
Collapse
Affiliation(s)
- Zhou Wang
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Macau SAR, China
| | - Huaijun Zhu
- Department of Pharmacy, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China; Laboratory for Bone and Joint Diseases, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Yi Zhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Macau SAR, China; Shanghai Key Laboratory of Bioactive Small Molecules & School of Pharmacy, Fudan University, Shanghai, China.
| |
Collapse
|
114
|
Tan TC, Chong TKY, Low AHL, Leung YY. Microbiome and osteoarthritis: New insights from animal and human studies. Int J Rheum Dis 2021; 24:984-1003. [PMID: 33961348 DOI: 10.1111/1756-185x.14123] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 03/15/2021] [Accepted: 04/10/2021] [Indexed: 01/08/2023]
Abstract
Osteoarthritis (OA) is a common cause of disability, especially among the elderly. With an ageing and increasingly obese population, OA will become more prevalent. Obesity and metabolic syndrome are risk factors for OA and have been implicated in its pathogenesis. The gut microbiome may shed light on this possible common pathogenesis. Recent animal and human studies have gained important insights into the relationship between OA, obesity, and the gut microbiome. Animal studies have demonstrated links between obesity and increased severity of OA and altered gut microbial DNA profile. Use of prebiotics and probiotics in animal trials provides proof-of-concept that interventional options to the gut microbiome can modulate the progression of OA favorably. Current evidence in human studies is limited. Shifts in gut microbial profile and reduced gut microbial diversity have been associated with people with OA, as well as blood and synovial fluid lipopolysaccharide endotoxemia. Linkages between microbiome dysbiosis and host responses may help in the understanding of OA pathogenesis and the discovery of therapeutic targets. This narrative review provides a summary of up-to-date animal and human studies on the gut microbiome and its link with OA.
Collapse
Affiliation(s)
- Tze Chin Tan
- Department of Rheumatology & Immunology, Singapore General Hospital, Singapore City, Singapore.,Duke-NUS Medical School, Singapore City, Singapore
| | - Timothy Kit Yeong Chong
- Department of Rheumatology & Immunology, Singapore General Hospital, Singapore City, Singapore
| | - Andrea Hsiu Ling Low
- Department of Rheumatology & Immunology, Singapore General Hospital, Singapore City, Singapore.,Duke-NUS Medical School, Singapore City, Singapore
| | - Ying Ying Leung
- Department of Rheumatology & Immunology, Singapore General Hospital, Singapore City, Singapore.,Duke-NUS Medical School, Singapore City, Singapore
| |
Collapse
|
115
|
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
|
116
|
Luna M, Guss JD, Vasquez-Bolanos LS, Alepuz AJ, Dornevil S, Strong J, Alabi D, Shi Q, Pannellini T, Otero M, Brito IL, van der Meulen MCH, Goldring SR, Hernandez CJ. Obesity and load-induced posttraumatic osteoarthritis in the absence of fracture or surgical trauma. J Orthop Res 2021; 39:1007-1016. [PMID: 32658313 PMCID: PMC7855296 DOI: 10.1002/jor.24799] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/01/2020] [Accepted: 07/09/2020] [Indexed: 02/04/2023]
Abstract
Osteoarthritis is increasingly viewed as a heterogeneous disease with multiple phenotypic subgroups. Obesity enhances joint degeneration in mouse models of posttraumatic osteoarthritis (PTOA). Most models of PTOA involve damage to surrounding tissues caused by surgery/fracture; it is unclear if obesity enhances cartilage degeneration in the absence of surgery/fracture. We used a nonsurgical animal model of load-induced PTOA to determine the effect of obesity on cartilage degeneration 2 weeks after loading. Cartilage degeneration was caused by a single bout of cyclic tibial loading at either a high or moderate load magnitude in adult male mice with severe obesity (C57Bl6/J + high-fat diet), mild obesity (toll-like receptor 5 deficient mouse [TLR5KO]), or normal adiposity (C57Bl6/J mice + normal diet and TLR5KO mice in which obesity was prevented by manipulation of the gut microbiome). Two weeks after loading, cartilage degeneration occurred in limbs loaded at a high magnitude, as determined by OARSI scores (P < .001). However, the severity of cartilage damage did not differ among groups. Osteophyte width and synovitis of loaded limbs did not differ among groups. Furthermore, obesity did not enhance cartilage damage in limbs evaluated 6 weeks after loading. Constituents of the gut microbiota differed among groups. Our findings suggest that, in the absence of surgery/fracture, obesity may not influence cartilage loss after a single mechanical insult, suggesting that either damage to surrounding tissues or repeated mechanical insult is necessary for obesity to influence cartilage degeneration. These findings further illustrate heterogeneity in PTOA phenotypes and complex interactions between mechanical/metabolic factors in cartilage loss.
Collapse
Affiliation(s)
- Marysol Luna
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Jason D. Guss
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | | | - Adrian J. Alepuz
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Sophie Dornevil
- College of Human Ecology, Cornell University, Ithaca, NY, USA
| | - Jasmin Strong
- College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
| | - Denise Alabi
- College of Arts and Sciences, Cornell University, Ithaca, NY, USA
| | - Qiaojuan Shi
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | | | | | - Ilana L. Brito
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Marjolein C. H. van der Meulen
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA,Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA,Hospital for Special Surgery, New York, NY, USA
| | | | - Christopher J. Hernandez
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA,Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA,Hospital for Special Surgery, New York, NY, USA
| |
Collapse
|
117
|
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
|
118
|
Fortuna R, Hart DA, Sharkey KA, Schachar RA, Johnston K, Reimer RA. Effect of a prebiotic supplement on knee joint function, gut microbiota, and inflammation in adults with co-morbid obesity and knee osteoarthritis: study protocol for a randomized controlled trial. Trials 2021; 22:255. [PMID: 33827639 PMCID: PMC8025512 DOI: 10.1186/s13063-021-05212-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 03/20/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a chronic and painful condition where the articular cartilage surfaces progressively degenerate, resulting in loss of function and progressive disability. Obesity is a primary risk factor for the development and progression of knee OA, defined as the "metabolic OA" phenotype. Metabolic OA is associated with increased fat deposits that release inflammatory cytokines/adipokines, thereby resulting in systemic inflammation which can contribute to cartilage degeneration. There is currently no cure for OA. Prebiotics are a type of dietary fiber that can positively influence gut microbiota thereby reducing systemic inflammation and offering protection of joint integrity in rodents. However, no human clinical trials have tested the effects of prebiotics in adults with obesity suffering from knee OA. Therefore, the purpose of this double-blind, placebo-controlled, randomized trial is to determine if prebiotic supplementation can, through positive changes in the gut microbiota, improve knee function and physical performance in adults with obesity and knee OA. METHODS Adults (n = 60) with co-morbid obesity (BMI > 30 kg/m2) and knee OA (Kellgren-Lawrence grade II-III) will be recruited from the Alberta Hip and Knee Clinic and the Rocky Mountain Health Clinic and surrounding community of Calgary, Canada, and randomized (stratified by sex, BMI, and age) to prebiotic (oligofructose-enriched inulin; 16 g/day) or a calorie-matched placebo (maltodextrin) for 6 months. Anthropometrics, performance-based tests, knee pain, serum inflammatory markers and metabolomics, quality of life, and gut microbiota will be assessed at baseline, 3 months, 6 months (end of prebiotic supplementation), and 3 months following the end of the prebiotic supplementation. CLINICAL SIGNIFICANCE There is growing pressure on health care systems for aggressive OA treatment such as total joint replacement. Less aggressive, yet effective, conservative treatment options have the potential to address the growing prevalence of co-morbid obesity and knee OA by delaying the need for joint replacement or ideally preventing its need altogether. The results of this clinical trial will provide the first evidence regarding the efficacy of prebiotic supplementation on knee joint function and pain in adults with obesity and knee OA. If successful, the results may provide a simple, safe, and easy to adhere to intervention to reduce knee joint pain and improve the quality of life of adults with co-morbid knee OA and obesity. TRIAL REGISTRATION Clinical Trials.gov NCT04172688 . Registered on 21 November 2019.
Collapse
Affiliation(s)
- Rafael Fortuna
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, Alberta Canada
| | - David A. Hart
- McCaig Institute for Bone and Joint Health, Department of Surgery, and Faculty of Kinesiology, University of Calgary, Calgary, Alberta Canada
| | - Keith A. Sharkey
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta Canada
| | | | - Kelly Johnston
- Division of Hip and Knee Reconstruction, Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, Alberta Canada
| | - Raylene A. Reimer
- Faculty of Kinesiology and Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 2500 University Dr. NW, Calgary, Alberta Canada
| |
Collapse
|
119
|
Lee YH, Song GG. The Gut Microbiome and Osteoarthritis: A Two-Sample Mendelian Randomization Study. JOURNAL OF RHEUMATIC DISEASES 2021; 28:94-100. [PMID: 37476017 PMCID: PMC10324885 DOI: 10.4078/jrd.2021.28.2.94] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 02/08/2021] [Accepted: 03/04/2021] [Indexed: 07/22/2023]
Abstract
Objective The aim of this study was to examine if the intestinal microbiome is causally correlated with osteoarthritis (OA) incidence. Methods A two-sample Mendelian randomization (MR) study was conducted using inverse variance weighting (IVW), weighted median, and MR-Egger regression techniques Publicly accessible summary statistics dataset of intestinal microbiomes of European descent from genome-wide association studies (GWASs) (a total with 3,326 individuals) was used as an exposure As an outcome, summary data from the GWAS include 3,498 patients with OA of the knee and hip from the arcOGEN sample and 11,009 controls of European descent. Results We identified 29 single-nucleotide polymorphisms from GWAS of intestinal microbiomes as instrumental variables The IVW approach found no evidence to suggest a causal relationship between the intestinal microbiota and OA (beta=-0001, standard error [SE]=0004, p=0748) The regression test of MR-Egger showed that the directional pleiotropy was unlikely to be a bias (intercept=0002, SE=0007, p=0697) and the MR-Egger study showed no causal relation between the intestinal microbiota and the OA (beta=-0002, SE=0005, p=0630) The weighted median analysis also did not have indications of a causal relationship between the intestinal microbiota and OA (beta=-0002, SE=0005, p=0630) The MR results calculated using IVW, the median weighted and the MR-Egger regression approaches were consistent. Conclusion The findings of the MR analysis did not support a causal relationship between intestinal microbiome and OA risk.
Collapse
Affiliation(s)
- Young Ho Lee
- Department of Rheumatology, Korea University College of Medicine, Seoul, Korea
| | - Gwan Gyu Song
- Department of Rheumatology, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
120
|
Kuang X, Chiou J, Lo K, Wen C. Magnesium in joint health and osteoarthritis. Nutr Res 2021; 90:24-35. [PMID: 34023805 DOI: 10.1016/j.nutres.2021.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 01/31/2021] [Accepted: 03/16/2021] [Indexed: 12/28/2022]
Abstract
Osteoarthritis (OA) is a prevalent debilitating age-related skeletal disease. The hallmark of OA is the degradation of articular cartilage that cushions the joint during movement. It is characterized by chronic pain and disability. Magnesium, a critical trace element in the human body, plays a pivotal role in metabolism homeostasis and the energy balance. Humans obtain magnesium mainly from the diet. However, inadequate magnesium intake is not uncommon. Moreover, the magnesium status deteriorates with ageing. There has been a growing body of clinical studies pointing to an intimate relationship between dietary magnesium and OA although the conclusion remains controversial. As reported, the magnesium ion concentration is essential to determine cell fate. Firstly, the low-concentration magnesium ions induced human fibroblasts senescence. Magnesium supplementation was also able to mitigate chondrocyte apoptosis, and to facilitate chondrocyte proliferation and differentiation. In this literature review, we will outline the existing evidence in animals and humans. We will also discuss the controversies on plasma or intracellular level of magnesium as the indicator of magnesium status. In addition, we put forward the interplay between dietary magnesium intake and intestinal microbiome to modulate the inflammatory milieu in the conjecture of OA pathogenesis. This leads to an emerging hypothesis that the synergistic effect of magnesium and probiotics may open a new avenue for the prevention and treatment of OA.
Collapse
Affiliation(s)
- Xiaoqing Kuang
- Department of Biomedical Engineering, Faculty of Engineering, Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Jiachi Chiou
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Kenneth Lo
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Chunyi Wen
- Department of Biomedical Engineering, Faculty of Engineering, Hong Kong Polytechnic University, Kowloon, Hong Kong.
| |
Collapse
|
121
|
Arora V, Singh G, O-Sullivan I, Ma K, Natarajan Anbazhagan A, Votta-Velis EG, Bruce B, Richard R, van Wijnen AJ, Im HJ. Gut-microbiota modulation: The impact of thegut-microbiotaon osteoarthritis. Gene 2021; 785:145619. [PMID: 33781857 DOI: 10.1016/j.gene.2021.145619] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/11/2021] [Accepted: 03/23/2021] [Indexed: 02/08/2023]
Abstract
Osteoarthritis (OA) is one of the most common medical conditions affecting > 300 million people globally which represents the formidable public health challenge. Despite its clinical and financial ramifications, there are currently no approved disease modifying OA drugs available and symptom palliation is the only alternative. Currently, the amount of data on the human intestinal microbiome is growing at a high rate, both in health and in various pathological conditions. With an increase in the amount of the accumulated data, there is an expanded understanding that the microbiome provides compelling evidence of a link between thegut microbiomeand development ofOA. The microbiota management tools of probiotics and/or prebiotics or symbiotic have been developed and indeed, commercialized over the past few decades with the expressed purpose of altering the microbiota within the gastrointestinal tract which could be a potentially novel intervention to tackle or prevent OA. However, the mechanisms how intestinal microbiota affects the OA pathogenesis are still not clear and further research targeting specific gut microbiota or its metabolites is still needed to advance OA treatment strategies from symptomatic management to individualized interventions of OA pathogenesis. This article provides an overview of the various preclinical and clinical studies using probiotics and prebiotics as plausible therapeutic options that can restore the gastrointestinal microbiota and its impact on the OA pathogenesis. May be in the near future the targeted alterations of gut microbiota may pave the way for developing new interventions to prevent and treat OA.
Collapse
Affiliation(s)
- Vipin Arora
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
| | - Gurjit Singh
- Departments of Bioengineering, the University of Illinois at Chicago, Chicago, IL, USA
| | - InSug O-Sullivan
- Departments of Medicine, the University of Illinois at Chicago, Chicago, IL, USA
| | - Kaige Ma
- Departments of Bioengineering, the University of Illinois at Chicago, Chicago, IL, USA
| | | | - E Gina Votta-Velis
- Departments of Anesthesiology, the University of Illinois at Chicago, Chicago, IL, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Benjamin Bruce
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Ripper Richard
- Departments of Anesthesiology, the University of Illinois at Chicago, Chicago, IL, USA
| | | | - Hee-Jeong Im
- Departments of Bioengineering, the University of Illinois at Chicago, Chicago, IL, USA; Jesse Brown Veterans Affairs Medical Center (JBVAMC) at Chicago, IL, USA.
| |
Collapse
|
122
|
Rezende MU, Brito NLR, Farias FES, Silva CAC, Cernigoy CHA, Rodrigues da Silva JM, Moreira MM, Santana OFN, Hissadomi MI, Frucchi R, Pasqualin T, Campos GC, Pailo AF, Ocampos GP, Camargo OP. Improved function and strength in patients with knee osteoarthritis as a result of adding a two-day educational program to usual care. Prospective randomized trial. OSTEOARTHRITIS AND CARTILAGE OPEN 2021; 3:100137. [DOI: 10.1016/j.ocarto.2020.100137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 12/31/2020] [Indexed: 11/28/2022] Open
|
123
|
Hayashi A, Nagao-Kitamoto H, Kitamoto S, Kim CH, Kamada N. The Butyrate-Producing Bacterium Clostridium butyricum Suppresses Clostridioides difficile Infection via Neutrophil- and Antimicrobial Cytokine-Dependent but GPR43/109a-Independent Mechanisms. THE JOURNAL OF IMMUNOLOGY 2021; 206:1576-1585. [PMID: 33597149 DOI: 10.4049/jimmunol.2000353] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 01/14/2021] [Indexed: 02/06/2023]
Abstract
Short-chain fatty acids, such as butyrate, are major gut microbial metabolites that are beneficial for gastrointestinal health. Clostridium butyricum MIYAIRI588 (CBM588) is a bacterium that produces a robust amount of butyrate and therefore has been used as a live biotherapeutic probiotic in clinical settings. Clostridioides difficile causes life-threatening diarrhea and colitis. The gut resident microbiota plays a critical role in the prevention of C. difficile infection (CDI), as the disruption of the healthy microbiota by antibiotics greatly increases the risk for CDI. We report that CBM588 treatment in mice significantly improved clinical symptoms associated with CDI and increased the number of neutrophils and Th1 and Th17 cells in the colonic lamina propria in the early phase of CDI. The protective effect of CBM588 was abolished when neutrophils, IFN-γ, or IL-17A were depleted, suggesting that induction of the immune reactants is required to elicit the protective effect of the probiotic. The administration of tributyrin, which elevates the concentration of butyrate in the colon, also increased the number of neutrophils in the colonic lamina propria, indicating that butyrate is a potent booster of neutrophil activity during infection. However, GPR43 and GPR109a, two G protein-coupled receptors activated by butyrate, were dispensable for the protective effect of CBM588. These results indicate that CBM588 and butyrate suppress CDI, in part by boosting antimicrobial innate and cytokine-mediated immunity.
Collapse
Affiliation(s)
- Atsushi Hayashi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109.,Miyarisan Pharmaceutical, Central Research Institute, Saitama 331-0804, Japan
| | - Hiroko Nagao-Kitamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Sho Kitamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Chang H Kim
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109; and.,Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109
| | - Nobuhiko Kamada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109;
| |
Collapse
|
124
|
Hernandez CJ. Musculoskeletal microbiology: The utility of the microbiome in orthopedics. J Orthop Res 2021; 39:251-257. [PMID: 33245146 PMCID: PMC7855812 DOI: 10.1002/jor.24927] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/23/2020] [Accepted: 11/19/2020] [Indexed: 02/04/2023]
Abstract
The past 15 years have witnessed a renaissance in the study of the microbes that colonize the human body. The vast majority of the human microbiome resides within the gut. Alterations to the gut microbiome have been associated with the pathogenesis and progression of wide-ranging diseases throughout the body-including atherosclerosis, depression, and obesity. Our understanding of the effects of the gut microbiome on the musculoskeletal system remains in its infancy, but preclinical work has demonstrated an effect of the gut microbiome on the success of orthopedic surgical procedures, osteoporosis, osteoarthritis, and muscle mass. In this perspective I review preclinical findings demonstrating that an impaired presurgical gut microbiome can increase the likelihood of developing periprosthetic joint infection and how alterations in the gut microbiome can reduce bone strength by impairing bone tissue material properties. In addition to discussing these examples, I review the hypothesis that many chronic non-communicable diseases have become more prevalent in modern industrialized societies as a result of changes in the composition of the gut microbiome resulting from changes in environment/lifestyle (diet, sanitation, antibiotic use). The most burdensome musculoskeletal disorders are chronic and non-communicable and may therefore be related to generational shifts in the composition of the gut microbiome, a possibility I illustrate by reviewing changes in the prevalence of osteoarthritis over the last century. Microbiome-based therapeutics are potentially innocuous, inexpensive, and have the potential to be effective with only occasional use, making them attractive for addressing the needs of chronic and/or slowly progressing musculoskeletal disorders.
Collapse
Affiliation(s)
- Christopher J Hernandez
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
- Hospital for Special Surgery, New York, New York, USA
| |
Collapse
|
125
|
Hao X, Shang X, Liu J, Chi R, Zhang J, Xu T. The gut microbiota in osteoarthritis: where do we stand and what can we do? Arthritis Res Ther 2021; 23:42. [PMID: 33504365 PMCID: PMC7839300 DOI: 10.1186/s13075-021-02427-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 01/19/2021] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is one of the most frequent musculoskeletal diseases characterized by degeneration of articular cartilage, subchondral bone remodeling, and synovial membrane inflammation, which is a leading cause of global disability, morbidity, and decreased quality of life. Interpreting the potential mechanisms of OA pathogenesis is essential for developing novel prevention and disease-modifying therapeutic interventions. Gut microbiota is responsible for a series of metabolic, immunological, and structural and neurological functions, potentially elucidating the heterogeneity of OA phenotypes and individual features. In this narrative review, we summarized research evidence supporting the hypothesis of a “gut-joint axis” and the interaction between gut microbiota and the OA-relevant factors, including age, gender, genetics, metabolism, central nervous system, and joint injury, elucidating the underlying mechanisms of this intricate interaction. In the context, we also speculated the promising manipulation of gut microbiota in OA management, such as exercise and fecal microbiota transplantation (FMT), highlighting the clinical values of gut microbiota. Additionally, future research directions, such as more convincing studies by the interventions of gut microbiota, the gene regulation of host contributing to or attributed to the specific phenotypes of gut microbiota related to OA, and the relevance of distinct cell subgroups to gut microbiota, are expected. Moreover, gut microbiota is also the potential biomarker related to inflammation and gut dysbiosis that is able to predict OA progression and monitor the efficacy of therapeutic intervention.
Collapse
Affiliation(s)
- Xiaoxia Hao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Xingru Shang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Jiawei Liu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Ruimin Chi
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Jiaming Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China. .,Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Tao Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China.
| |
Collapse
|
126
|
Abstract
Supplemental Digital Content is available in the text Osteoarthritis is a debilitating disease leading to joint degeneration, inflammation, pain, and disability. Despite efforts to develop a disease modifying treatment, the only accepted and available clinical approaches involve palliation. Although many factors contribute to the development of osteoarthritis, the gut microbiome has recently emerged as an important pathogenic factor in osteoarthritis initiation and progression. This review examines the literature to date regarding the link between the gut microbiome and osteoarthritis.
Collapse
|
127
|
Ge Y, Wang X, Guo Y, Yan J, Abuduwaili A, Aximujiang K, Yan J, Wu M. Gut microbiota influence tumor development and Alter interactions with the human immune system. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:42. [PMID: 33494784 PMCID: PMC7829621 DOI: 10.1186/s13046-021-01845-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 01/14/2021] [Indexed: 02/06/2023]
Abstract
Recent scientific advances have greatly enhanced our understanding of the complex link between the gut microbiome and cancer. Gut dysbiosis is an imbalance between commensal and pathogenic bacteria and the production of microbial antigens and metabolites. The immune system and the gut microbiome interact to maintain homeostasis of the gut, and alterations in the microbiome composition lead to immune dysregulation, promoting chronic inflammation and development of tumors. Gut microorganisms and their toxic metabolites may migrate to other parts of the body via the circulatory system, causing an imbalance in the physiological status of the host and secretion of various neuroactive molecules through the gut-brain axis, gut-hepatic axis, and gut-lung axis to affect inflammation and tumorigenesis in specific organs. Thus, gut microbiota can be used as a tumor marker and may provide new insights into the pathogenesis of malignant tumors.
Collapse
Affiliation(s)
- Yanshan Ge
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China.,Basic School of Medicine, Central South University, Changsha, 410078, Hunan, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410008, Hunan, China
| | - Xinhui Wang
- Basic School of Medicine, Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Yali Guo
- Basic School of Medicine, Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Junting Yan
- Basic School of Medicine, Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Aliya Abuduwaili
- Basic School of Medicine, Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | | | - Jie Yan
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Minghua Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China. .,Basic School of Medicine, Central South University, Changsha, 410078, Hunan, China. .,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
128
|
Wang J, Ishfaq M, Li J. Lactobacillus salivarius ameliorated Mycoplasma gallisepticum-induced inflammatory injury and secondary Escherichia coli infection in chickens: Involvement of intestinal microbiota. Vet Immunol Immunopathol 2021; 233:110192. [PMID: 33476924 DOI: 10.1016/j.vetimm.2021.110192] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/17/2020] [Accepted: 01/10/2021] [Indexed: 12/11/2022]
Abstract
Mycoplasma gallisepticum (MG) infection alone or in combination with other pathogens have brought huge economic losses to the poultry industry. The intestinal microbiota plays a critical role in host defence against respiratory infection. To explore the role of intestinal microbiota in MG-induced inflammation-mediated lung injury and secondary Escherichia coli infection, MG infection model and fecal microbiota transplantation model were developed. The results showed that MG infection changed gut microbiota composition along with lung inflammation injury. Fecal microbiota transplantation from chickens infected with MG to antibiotics cocktail treated chickens decreased host defense against Escherichia coli due to impaired intestinal mucosal barrier, downregulated the mRNA expression levels of host defense enzymes and blocked autophagic flux. Lactobacillus salivarius intake alleviated lung inflammation injury caused by MG infection and increased host defense against Escherichia coli by improved gut microbiota composition. These results highlighted the role of gut microbiota in MG-infection induced lung inflammation injury and secondary infection that offered a new strategy for preventive intervention against MG infection.
Collapse
Affiliation(s)
- Jian Wang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China
| | - Muhammad Ishfaq
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China.
| | - Jichang Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China.
| |
Collapse
|
129
|
Hsu YJ, Chiu CC, Lee MC, Huang WC. Combination of Treadmill Aerobic Exercise with Bifidobacterium longum OLP-01 Supplementation for Treatment of High-Fat Diet-Induced Obese Murine Model. Obes Facts 2021; 14:306-319. [PMID: 34077946 PMCID: PMC8255637 DOI: 10.1159/000516865] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 04/12/2021] [Indexed: 01/15/2023] Open
Abstract
INTRODUCTION Obesity, which can result from disease, genetics, nutrition, lifestyle, and insufficient physical activity, substantially increases an individual's risk of complications and comorbidities. Exercise can be an effective strategy for achieving an energy balance and physiological fitness as part of obesity management. Additionally, probiotics, which are isolated from food and the environment, are being rapidly developed and have functional benefits for mitigating various metabolic dysfunctions associated with obesity. The potentially positive physiological and functional effects of exercise, probiotics, and exercise combined with probiotics should be elucidated in a model of diet-induced obesity. METHODS Bifidobacterium longum subsp. longum OLP-01 (OLP-01) was isolated from an elite Olympic-level athlete who exhibited physiological adaptations to peripheral fatigue caused by exercise training. In this current study, ICR strain mice were fed a high-fat diet (HFD) for 4 weeks to replicate an obesity model. The mice were divided into 5 groups according to the diet administered: control with normal diet, only HFD, HFD + exercise, HFD + OLP, and HFD + exercise + OLP groups. They were administered the probiotic and/or treadmill exercise training for 5 weeks, and their growth curve, physical activity, physiological adaptation, biochemical parameters, body composition, and glucose tolerance were assessed. RESULTS Compared with only exercise or only probiotics, a combination of probiotics and exercise significantly improved the weight, glucose tolerance, fat composition, and exercise-related oxidative stress of mice. Regular and programmed exercise with sufficient rest may be crucial to obesity improvement, and a combination of probiotics and exercise may synergistically assist obesity management and health promotion. CONCLUSION OLP-01 probiotics combined with exercise training can be employed as a strategy for treating obesity. However, the exact regulatory mechanisms underlying this effect, possibly involving microbiota and associated metabolites, warrant further investigation.
Collapse
Affiliation(s)
- Yi-Ju Hsu
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan, Taiwan
| | - Chien-Chao Chiu
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan, Taiwan
| | - Mon-Chien Lee
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan, Taiwan
| | - Wen-Ching Huang
- Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
- *Wen-Ching Huang,
| |
Collapse
|
130
|
Guan Z, Jia J, Zhang C, Sun T, Zhang W, Yuan W, Leng H, Song C. Gut microbiome dysbiosis alleviates the progression of osteoarthritis in mice. Clin Sci (Lond) 2020; 134:3159-3174. [PMID: 33215637 DOI: 10.1042/cs20201224] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/12/2020] [Accepted: 11/20/2020] [Indexed: 02/08/2023]
Abstract
Gut microbiota dysbiosis has been studied under the pathological conditions of osteoarthritis (OA). However, the effect of antibiotic-induced gut flora dysbiosis on OA remains incompletely understood at present. Herein, we used a mouse (8 weeks) OA model of destabilization of the medial meniscus (DMM) and gut microbiome dysbiosis induced by antibiotic treatment with ampicillin and neomycin for 8 weeks. The results show that antibiotic-induced intestinal microbiota dysbiosis reduced the serum level of lipopolysaccharide (LPS) and the inflammatory response, such as suppression of the levels of tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6), which can lead to decreased matrix metalloprotease-13 (MMP-13) expression and improvement of OA after joint injury. In addition, trabecular thickness (Tb.Th) and osteophyte scores were increased significantly in antibiotic-induced male mice compared with female mice. We further used network correlation analysis to verify the effect of gut microbiota dysbiosis on OA. Therefore, the present study contributes to our understanding of the gut-joint axis in OA and reveals the relationship between the inflammatory response, sex and gut microbiota, which may provide new strategies to prevent the symptoms and long-term sequelae of OA. Conclusion: Our data showed that gut microbiome dysbiosis alleviates the progression of OA.
Collapse
Affiliation(s)
- Zhiyuan Guan
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Jialin Jia
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Chenggui Zhang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Tiantong Sun
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Wang Zhang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Wanqiong Yuan
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Huijie Leng
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Chunli Song
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| |
Collapse
|
131
|
Identification and Characterization of the Intra-Articular Microbiome in the Osteoarthritic Knee. Int J Mol Sci 2020; 21:ijms21228618. [PMID: 33207573 PMCID: PMC7697780 DOI: 10.3390/ijms21228618] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/04/2020] [Accepted: 11/13/2020] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is the most common joint disorder in the United States, and the gut microbiome has recently emerged as a potential etiologic factor in OA development. Recent studies have shown that a microbiome is present at joint synovia. Therefore, we aimed to characterize the intra-articular microbiome within osteoarthritic synovia and to illustrate its role in OA disease progression. RNA-sequencing data from OA patient synovial tissue was aligned to a library of microbial reference genomes to identify microbial reads indicative of microbial abundance. Microbial abundance data of OA and normal samples was compared to identify differentially abundant microbes. We computationally explored the correlation of differentially abundant microbes to immunological gene signatures, immune signaling pathways, and immune cell infiltration. We found that microbes correlated to OA are related to dysregulation of two main functional pathways: increased inflammation-induced extracellular matrix remodeling and decreased cell signaling pathways crucial for joint and immune function. We also confirmed that the differentially abundant and biologically relevant microbes we had identified were not contaminants. Collectively, our findings contribute to the understanding of the human microbiome, well-known OA risk factors, and the role microbes play in OA pathogenesis. In conclusion, we present previously undiscovered microbes implicated in the OA disease progression that may be useful for future treatment purposes.
Collapse
|
132
|
Dysregulated Autophagy Mediates Sarcopenic Obesity and Its Complications via AMPK and PGC1α Signaling Pathways: Potential Involvement of Gut Dysbiosis as a Pathological Link. Int J Mol Sci 2020; 21:ijms21186887. [PMID: 32961822 PMCID: PMC7555990 DOI: 10.3390/ijms21186887] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/13/2022] Open
Abstract
Sarcopenic obesity (SOB), which is closely related to being elderly as a feature of aging, is recently gaining attention because it is associated with many other age-related diseases that present as altered intercellular communication, dysregulated nutrient sensing, and mitochondrial dysfunction. Along with insulin resistance and inflammation as the core pathogenesis of SOB, autophagy has recently gained attention as a significant mechanism of muscle aging in SOB. Known as important cellular metabolic regulators, the AMP-activated protein kinase (AMPK) and the peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1α) signaling pathways play an important role in autophagy, inflammation, and insulin resistance, as well as mutual communication between skeletal muscle, adipose tissue, and the liver. Furthermore, AMPK and PGC-1α signaling pathways are implicated in the gut microbiome-muscle axis. In this review, we describe the pathological link between SOB and its associated complications such as metabolic, cardiovascular, and liver disease, falls and fractures, osteoarthritis, pulmonary disease, and mental health via dysregulated autophagy controlled by AMPK and/or PGC-1α signaling pathways. Here, we propose potential treatments for SOB by modulating autophagy activity and gut dysbiosis based on plausible pathological links.
Collapse
|
133
|
Moran MM, Wilson BM, Li J, Engen PA, Naqib A, Green SJ, Virdi AS, Plaas A, Forsyth CB, Keshavarzian A, Sumner DR. The gut microbiota may be a novel pathogenic mechanism in loosening of orthopedic implants in rats. FASEB J 2020; 34:14302-14317. [PMID: 32931052 DOI: 10.1096/fj.202001364r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/29/2020] [Accepted: 08/07/2020] [Indexed: 01/08/2023]
Abstract
Particles released from implants cause inflammatory bone loss, which is a key factor in aseptic loosening, the most common reason for joint replacement failure. With the anticipated increased incidence of total joint replacement in the next decade, implant failure will continue to burden patients. The gut microbiome is increasingly recognized as an important factor in bone physiology, however, its role in implant loosening is currently unknown. We tested the hypothesis that implant loosening is associated with changes in the gut microbiota in a preclinical model. When the particle challenge caused local joint inflammation, decreased peri-implant bone volume, and decreased implant fixation, the gut microbiota was affected. When the particle challenge did not cause this triad of local effects, the gut microbiota was not affected. Our results suggest that cross-talk between these compartments is a previously unrecognized mechanism of failure following total joint replacement.
Collapse
Affiliation(s)
- Meghan M Moran
- Department of Cell & Molecular Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Brittany M Wilson
- Department of Cell & Molecular Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Jun Li
- Department of Internal Medicine, Division of Rheumatology, Rush Medical College, Rush University Medical Center, Chicago, IL, USA
| | - Phillip A Engen
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush Medical College, Chicago, IL, USA
| | - Ankur Naqib
- Department of Cell & Molecular Medicine, Rush University Medical Center, Chicago, IL, USA.,Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush Medical College, Chicago, IL, USA
| | - Stefan J Green
- Genome Research Core, Research Resources Center, University of Illinois-Chicago, Chicago, IL, USA
| | - Amarjit S Virdi
- Department of Cell & Molecular Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Anna Plaas
- Department of Internal Medicine, Division of Rheumatology, Rush Medical College, Rush University Medical Center, Chicago, IL, USA
| | - Christopher B Forsyth
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush Medical College, Chicago, IL, USA
| | - Ali Keshavarzian
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush Medical College, Chicago, IL, USA
| | - Dale R Sumner
- Department of Cell & Molecular Medicine, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
134
|
Oliviero F, Spinella P. Benefits of Probiotics in Rheumatic Diseases. Front Nutr 2020; 7:157. [PMID: 33015127 PMCID: PMC7509441 DOI: 10.3389/fnut.2020.00157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/04/2020] [Indexed: 12/21/2022] Open
Affiliation(s)
- Francesca Oliviero
- Rheumatology Unit, Department of Medicine-DIMED, University of Padova, Padua, Italy
| | - Paolo Spinella
- Clinical Nutrition Unit, Department of Medicine-DIMED, University of Padova, Padua, Italy
| |
Collapse
|
135
|
Mendez ME, Murugesh DK, Sebastian A, Hum NR, McCloy SA, Kuhn EA, Christiansen BA, Loots GG. Antibiotic Treatment Prior to Injury Improves Post-Traumatic Osteoarthritis Outcomes in Mice. Int J Mol Sci 2020; 21:E6424. [PMID: 32899361 PMCID: PMC7503363 DOI: 10.3390/ijms21176424] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/28/2020] [Accepted: 08/29/2020] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is a painful and debilitating disease characterized by the chronic and progressive degradation of articular cartilage. Post-traumatic OA (PTOA) is a secondary form of OA that develops in ~50% of cases of severe articular injury. Inflammation and re-occurring injury have been implicated as contributing to the progression of PTOA after the initial injury. However, there is very little known about external factors prior to injury that could affect the risk of PTOA development. To examine how the gut microbiome affects PTOA development we used a chronic antibiotic treatment regimen starting at weaning for six weeks prior to ACL rupture, in mice. A six-weeks post-injury histological examination showed more robust cartilage staining on the antibiotic (AB)-treated mice than the untreated controls (VEH), suggesting slower disease progression in AB cohorts. Injured joints also showed an increase in the presence of anti-inflammatory M2 macrophages in the AB group. Molecularly, the phenotype correlated with a significantly lower expression of inflammatory genes Tlr5, Ccl8, Cxcl13, and Foxo6 in the injured joints of AB-treated animals. Our results indicate that a reduced state of inflammation at the time of injury and a lower expression of Wnt signaling modulatory protein, Rspo1, caused by AB treatment can slow down or improve PTOA outcomes.
Collapse
Affiliation(s)
- Melanie E. Mendez
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Deepa K. Murugesh
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Aimy Sebastian
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Nicholas R. Hum
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
- UC Merced, School of Natural Sciences, Merced, CA 95343, USA
| | - Summer A. McCloy
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Edward A. Kuhn
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | | | - Gabriela G. Loots
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
- UC Merced, School of Natural Sciences, Merced, CA 95343, USA
| |
Collapse
|
136
|
Li K, Liu A, Zong W, Dai L, Liu Y, Luo R, Ge S, Dong G. Moderate exercise ameliorates osteoarthritis by reducing lipopolysaccharides from gut microbiota in mice. Saudi J Biol Sci 2020; 28:40-49. [PMID: 33424281 PMCID: PMC7783636 DOI: 10.1016/j.sjbs.2020.08.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/13/2020] [Accepted: 08/16/2020] [Indexed: 12/27/2022] Open
Abstract
Lipopolysaccharides (LPSs) released by gut microbiota are correlated with the pathophysiology of osteoarthritis (OA). Exercise remodels the composition of gut microbiota. The present study investigated the hypothesis that wheel-running exercise prevents knee OA induced by high-fat diet (HFD) via reducing LPS from intestinal microorganisms. Male C57BL/6 J mice were treated with sedentary or wheel-running exercise, standard diet (13.5% kcal) or HFD (60% kcal), berberine or not according to their grouping. Knee OA severity, blood and synovial fluid LPS, cecal microbiota, and TLR4 and MMP-13 expression levels were determined. Our findings reveal that HFD treatment decreased gut microbial diversity. Increase in endotoxin-producing bacteria, decrease in gut barrier-protecting bacteria, high LPS levels in the blood and synovial fluid, high TLR4 and MMP-13 expression levels, and severe cartilage degeneration were observed. By contrast, voluntary wheel running caused high gut microbial diversity. The gut microbiota were reshaped, LPS levels in the blood and synovial fluid and TLR4 and MMP-13 expression levels were low, and cartilage degeneration was ameliorated. Berberine treatment reduced LPS levels in the samples, but decreased the diversity of intestinal flora with similar changes to that caused by HFD. In conclusion, unlike taking drugs, exercising can remodel gut microbial ecosystems, reduce the circulating levels of LPS, and thereby contribute to the relief of chronic inflammation and OA. Our findings showed that moderate exercise is a potential therapeutic approach for preventing and treating obesity-related OA.
Collapse
Affiliation(s)
- Kefeng Li
- College of Sports and Health, Shandong Sport University, Jinan 250102, China
| | - Anli Liu
- Department of Acupuncture, Rizhao Hospital of Traditional Chinese Medicine, Rizhao 276800, China
| | - Wenhao Zong
- College of Sports and Health, Shandong Sport University, Jinan 250102, China
| | - Lulu Dai
- College of Sports and Health, Shandong Sport University, Jinan 250102, China
| | - Yang Liu
- College of Sports and Health, Shandong Sport University, Jinan 250102, China
| | - Renping Luo
- College of Sports and Health, Shandong Sport University, Jinan 250102, China
| | - Shulin Ge
- School of Physical Education, Shandong Normal University, Jinan 250014, China
| | - Guijun Dong
- College of Sports and Health, Shandong Sport University, Jinan 250102, China
| |
Collapse
|
137
|
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: 20.8] [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
|
138
|
Probiotic-enriched milk and dairy products increase gut microbiota diversity: a comparative study. Nutr Res 2020; 82:25-33. [PMID: 32949953 DOI: 10.1016/j.nutres.2020.06.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 06/14/2020] [Accepted: 06/26/2020] [Indexed: 02/07/2023]
Abstract
Targeting gut microbiota with probiotics has emerged as a promising nutritional approach for the prevention of obesity and metabolic syndrome. Cultured dairy products can be effectively employed for the delivery of probiotics to the gut as well as for the support of growth and survival of probiotic bacteria. The purpose of this study was to characterize the effects of probiotic-enriched pasteurized milk and dairy products (Greek-style yogurt and cottage cheese) of different origins (cow, goat, and camel) on taxonomic composition of the mouse gut microbiota. We hypothesized that cultured dairy products can be an effective vector for the delivery of probiotics to the gut because of its nutritional value, acidic nature, and long shelf-life. Mice were fed a standard low fat, plant polysaccharide-rich (LF/PP) diet supplemented with the probiotic-enriched milk and dairy products for 5 weeks. Next generation sequencing of DNA from mouse fecal samples was used to characterize the bacterial relative abundance. Mice fed a diet supplemented with camel milk demonstrated characteristic changes in the gut microbiota, which included an increase in relative abundance of order Clostridiales and genus Anaerostipes. Mice fed a diet supplemented with the probiotic-enriched cow cheese exhibited an increase in the relative abundance of order Clostridiales, family Ruminococcaceae, and family Lachnospiraceae. The results obtained and their bioinformatics analysis support the conclusion that camel milk and the probiotic cow cheese induce changes in the mouse gut microbiota, which can be characterized as potentially beneficial to health compared to the changes associated with a standard diet. These findings imply that probiotic-enriched milk and dairy products can be highly effective for the delivery and support of probiotic bacteria of the gut.
Collapse
|
139
|
Electroacupuncture Prevents Osteoarthritis of High-Fat Diet-Induced Obese Rats. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9380965. [PMID: 32724821 PMCID: PMC7366230 DOI: 10.1155/2020/9380965] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/22/2020] [Accepted: 06/16/2020] [Indexed: 01/09/2023]
Abstract
The effects of acupuncture on osteoarthritis (OA) pathogenesis have been demonstrated in vitro and in animal models. However, the potential for acupuncture to mediate protective effects on obese-induced OA has not been examined. Here, we investigated the effects of different acupuncture patterns on OA pathogenesis in high-fat diet- (HFD-) induced obese rats. After 12-week diet-induced obesity, obese rats were treated with three acupuncture protocols for 2 weeks, including ST36, GB34, and ST36+GB34. The results showed that the three acupuncture protocols both prevented obesity-induced cartilage matrix degradation and MMP expression and mitigated obesity-induced systemic and local inflammation but had different regulatory effects on lipid metabolism and gut microbiota disorder of obese-induced OA rats. Furthermore, the three acupuncture protocols increased the microbial diversity and altered the structure of community of feces in obese rats. We found that ST36 and GB34 could inhibit proinflammatory shift in the gut microbiome with an increase in the ratio of Bacteroidetes/Firmicutes and promote the recovery of relative abundance of Clostridium, Akkermansia, Butyricimonas, and Lactococcus. Although both ST36 and GB34 had an anti-inflammatory effect on serum inflammatory mediators, only the acupuncture protocol with both ST36 and GB34 could effectively inhibit LPS-mediated joint inflammation in obesity rats. Therefore, relieving obesity-related chronic inflammation, lipid metabolism disorder, and gut microbiota disorder may be an important mechanism for acupuncture with ST36 and GB34 to promote OA recovery.
Collapse
|
140
|
Dunn CM, Velasco C, Rivas A, Andrews M, Garman C, Jacob PB, Jeffries MA. Identification of Cartilage Microbial DNA Signatures and Associations With Knee and Hip Osteoarthritis. Arthritis Rheumatol 2020; 72:1111-1122. [PMID: 31961065 PMCID: PMC7336391 DOI: 10.1002/art.41210] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/14/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Alterations of the gut microbiota have been implicated in many forms of arthritis, but an examination of cartilage microbial patterns has not been performed. This study was undertaken to characterize the microbial DNA profile of articular cartilage and determine changes associated with osteoarthritis (OA). METHODS We performed 16S ribosomal RNA gene deep sequencing on eroded and intact cartilage samples from knee OA patients (n = 21 eroded and 21 intact samples) and hip OA patients (n = 34 eroded and 33 intact samples) and cadaver controls (n = 10 knee samples and 10 hip samples). Microbial DNA diversity was assessed, groups were compared, and metagenomic profiles were reconstructed. Confirmation was performed in an independent cohort by clade-specific quantitative polymerase chain reaction. Findings in human cartilage were compared to those in cartilage from OA-susceptible C57BL/6 (B6) mice and OA-resistant MRL/MpJ (MRL) mice. Germ-free B6 mouse cartilage was analyzed as a methodologic control. RESULTS Alpha diversity was reduced in human OA versus control samples (P < 0.0001), and in hip versus knee samples (P < 0.0001). Numerous clades were different in human OA versus control samples, and similar findings were noted in comparisons of murine B6 versus MRL mice. Hip samples were microbiologically distinct from knee samples. OA microbial DNA demonstrated increased gram-negative constituents (P = 0.02). Functional analysis demonstrated increases in lipopolysaccharide production (P = 9.9 × 10-3 ), phosphatidylinositol signaling (P = 4.2 × 10-4 ), and nitrogen metabolism (P = 8 × 10-3 ) and decreases in sphingolipid metabolism (P = 7.7 × 10-4 ) associated with OA. CONCLUSION Our study reveals a microbial DNA signature in human and mouse cartilage. Alterations in this signature, including increases in gram-negative constituents, occur during the development and progression of human OA. Furthermore, our findings indicate that strain-specific signatures exist within mouse cartilage that mirror human patterns. Further study of the establishment and potential pathogenic role of these DNA signatures is needed.
Collapse
MESH Headings
- Aged
- Animals
- Arthroplasty, Replacement, Hip
- Arthroplasty, Replacement, Knee
- Cartilage, Articular/metabolism
- Cartilage, Articular/microbiology
- Cartilage, Articular/pathology
- Classification
- DNA, Bacterial/analysis
- Disease Susceptibility
- Female
- Genetic Variation
- Humans
- Male
- Metagenome/genetics
- Mice
- Microbiota/genetics
- Middle Aged
- Osteoarthritis, Hip/microbiology
- Osteoarthritis, Hip/surgery
- Osteoarthritis, Knee/microbiology
- Osteoarthritis, Knee/surgery
- Polymerase Chain Reaction
- RNA, Ribosomal, 16S/genetics
Collapse
Affiliation(s)
- Christopher M. Dunn
- University of Oklahoma Health Sciences Center, Department of Internal Medicine, Division of Rheumatology, Immunology, and Allergy, Oklahoma City, OK
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program, Oklahoma City, OK
| | - Cassandra Velasco
- University of Oklahoma Health Sciences Center, Department of Internal Medicine, Division of Rheumatology, Immunology, and Allergy, Oklahoma City, OK
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program, Oklahoma City, OK
| | - Alexander Rivas
- University of Arkansas for Medical Sciences, Little Rock, AR
| | - Madison Andrews
- University of Oklahoma Health Sciences Center, Department of Internal Medicine, Division of Rheumatology, Immunology, and Allergy, Oklahoma City, OK
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program, Oklahoma City, OK
| | - Cassandra Garman
- University of Oklahoma Health Sciences Center, Department of Internal Medicine, Division of Rheumatology, Immunology, and Allergy, Oklahoma City, OK
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program, Oklahoma City, OK
| | - Paul B. Jacob
- Oklahoma Joint Reconstruction Institute, Oklahoma City, OK
| | - Matlock A. Jeffries
- University of Oklahoma Health Sciences Center, Department of Internal Medicine, Division of Rheumatology, Immunology, and Allergy, Oklahoma City, OK
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program, Oklahoma City, OK
| |
Collapse
|
141
|
Na HS, Park JS, Cho KH, Kwon JY, Choi J, Jhun J, Kim SJ, Park SH, Cho ML. Interleukin-1-Interleukin-17 Signaling Axis Induces Cartilage Destruction and Promotes Experimental Osteoarthritis. Front Immunol 2020; 11:730. [PMID: 32431699 PMCID: PMC7214841 DOI: 10.3389/fimmu.2020.00730] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 03/31/2020] [Indexed: 12/19/2022] Open
Abstract
Osteoarthritis (OA), which is the most common degenerative joint disorder, has been considered a non-inflammatory disease with abnormal mechanics. Interleukin (IL)-17 is a pleiotropic cytokine involved in inflammatory diseases and their production is driven by the cytokine including IL-1 and IL-23. However, little is known about the mechanism of IL-17 in the development of OA. Here, we investigated the role of IL-17 in the pathogenesis of OA using monosodium iodoacetate (MIA)-injected IL-17 and IL-1 receptor antagonist (IL-1Ra) double-deficient mice. In MIA-injected IL-1Ra KO mice, nociceptive properties, degree of cartilage damage, and the level of inflammatory factors in articular cartilage were increased compared to MIA-injected wild-type mice. Interestingly, the intestinal architecture was impaired in IL-1Ra KO mice compared to wild-type mice and the damage was further exacerbated by MIA injection. Deficiency of IL-17 reduced nociceptive properties and cartilage destruction, as well as inflammation-related factors in MIA-injected IL-1Ra KO mice compared to MIA-injected wild-type mice. Furthermore, IL-17-treated chondrocytes from OA patients showed enhanced expression of catabolic factors that are involved in the destruction of cartilage in OA. IL-17 accelerates the destruction of cartilage and small intestine via regulation of several inflammatory mediators in an OA murine model. These results suggest that IL-17 plays a critical role in the development of OA.
Collapse
Affiliation(s)
- Hyun Sik Na
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jin-Sil Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Keun-Hyung Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ji Ye Kwon
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - JeongWon Choi
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jooyeon Jhun
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Seok Jung Kim
- Department of Orthopedic Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Hwan Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Medical Lifescience, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
142
|
Huang Z, Chen J, Li B, Zeng B, Chou CH, Zheng X, Xie J, Li H, Hao Y, Chen G, Pei F, Shen B, Kraus VB, Wei H, Zhou X, Cheng L. Faecal microbiota transplantation from metabolically compromised human donors accelerates osteoarthritis in mice. Ann Rheum Dis 2020; 79:646-656. [PMID: 32205337 PMCID: PMC7384301 DOI: 10.1136/annrheumdis-2019-216471] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 02/17/2020] [Accepted: 03/03/2020] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Emerging evidence suggests that the microbiome plays an important role in the pathogenesis of osteoarthritis (OA). We aimed to test the two-hit model of OA pathogenesis and potentiation in which one 'hit' is provided by an adverse gut microbiome that activates innate immunity; the other 'hit' is underlying joint damage. METHODS Medical history, faecal and blood samples were collected from human healthy controls (OA-METS-, n=4), knee OA without metabolic syndrome (OA+METS-, n=7) and knee OA with metabolic syndrome (OA+METS+, n=9). Each group of human faecal samples, whose microbial composition was identified by 16S rRNA sequencing, was pooled and transplanted into germ-free mice 2 weeks prior to meniscal/ligamentous injury (MLI) (n≥6 per group). Eight weeks after MLI, mice were evaluated for histological OA severity and synovitis, systemic inflammation and gut permeability. RESULTS Histological OA severity following MLI was minimal in germ-free mice. Compared with the other groups, transplantation with the OA+METS+ microbiome was associated with higher mean systemic concentrations of inflammatory biomarkers (interleukin-1β, interleukin-6 and macrophage inflammatory protein-1α), higher gut permeability and worse OA severity. A greater abundance of Fusobacterium and Faecalibaterium and lesser abundance of Ruminococcaceae in transplanted mice were consistently correlated with OA severity and systemic biomarkers concentrations. CONCLUSION The study clearly establishes a direct gut microbiome-OA connection that sets the stage for a new means of exploring OA pathogenesis and potentially new OA therapeutics. Alterations of Fusobacterium, Faecalibaterium and Ruminococcaceae suggest a role of these particular microbes in exacerbating OA.
Collapse
Affiliation(s)
- ZeYu Huang
- State Key Laboratory of Oral Diseases and National Clinical Research for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Jing Chen
- State Key Laboratory of Oral Diseases and National Clinical Research for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - BoLei Li
- State Key Laboratory of Oral Diseases and National Clinical Research for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Benhua Zeng
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Ching-Heng Chou
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, North Carolina, USA
| | - Xin Zheng
- State Key Laboratory of Oral Diseases and National Clinical Research for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - JingWei Xie
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Hao Li
- State Key Laboratory of Oral Diseases and National Clinical Research for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yu Hao
- State Key Laboratory of Oral Diseases and National Clinical Research for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Guo Chen
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - FuXing Pei
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Bin Shen
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Virginia B Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, North Carolina, USA
- Division of Rheumatology, Department of Medicine, Duke University School of Medicine, Duke University, Durham, North Carolina, USA
| | - Hong Wei
- Central Laboratory, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases and National Clinical Research for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
143
|
|
144
|
Oral microbiome: possible harbinger for children's health. Int J Oral Sci 2020; 12:12. [PMID: 32350240 PMCID: PMC7190716 DOI: 10.1038/s41368-020-0082-x] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
The human microbiome functions as an intricate and coordinated microbial network, residing throughout the mucosal surfaces of the skin, oral cavity, gastrointestinal tract, respiratory tract, and reproductive system. The oral microbiome encompasses a highly diverse microbiota, consisting of over 700 microorganisms, including bacteria, fungi, and viruses. As our understanding of the relationship between the oral microbiome and human health has evolved, we have identified a diverse array of oral and systemic diseases associated with this microbial community, including but not limited to caries, periodontal diseases, oral cancer, colorectal cancer, pancreatic cancer, and inflammatory bowel syndrome. The potential predictive relationship between the oral microbiota and these human diseases suggests that the oral cavity is an ideal site for disease diagnosis and development of rapid point-of-care tests. The oral cavity is easily accessible with a non-invasive collection of biological samples. We can envision a future where early life salivary diagnostic tools will be used to predict and prevent future disease via analyzing and shaping the infant’s oral microbiome. In this review, we present evidence for the establishment of the oral microbiome during early childhood, the capability of using childhood oral microbiome to predict future oral and systemic diseases, and the limitations of the current evidence.
Collapse
|
145
|
Lyu JL, Wang TM, Chen YH, Chang ST, Wu MS, Lin YH, Lin YH, Kuan CM. Oral intake of Streptococcus thermophil us improves knee osteoarthritis degeneration: A randomized, double-blind, placebo-controlled clinical study. Heliyon 2020; 6:e03757. [PMID: 32368640 PMCID: PMC7184258 DOI: 10.1016/j.heliyon.2020.e03757] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/05/2020] [Accepted: 03/31/2020] [Indexed: 01/15/2023] Open
Abstract
This preliminary clinical study demonstrates the possibility of a new species of probiotic for improvement of the degeneration of knee osteoarthritis (KOA). TCI633 (Streptococcus thermophil us) is a newly founded bacterium from human breast milk, and it is able to produce hyaluronate (HA) in gastrointestinal (GI) tract. A recent study has proved that TCI633 can substantially alleviate synovial tissue inflammation and cartilage damage in the animal models, but so far it has never been applied in clinical intervention. In this study, we recruited 80 subjects and conducted 12 weeks clinical trial to validate the efficacy of TCI633 for improvement of the progression of KOA. TCI633 could improve serum collagen type II C-telopeptide (sCTX-II) and serum C-reactive protein (sCRP) by 41.58% and 39.58%, respectively, after the study. The improvement rates for sCTX-II and sCRP in TCI633 group were 54% and 57%, respectively, at 12 weeks. Compared to the results of placebo, the indistinct improvement progresses of sCTX-II and sCRP might be caused by the uneventful distribution of K/L populations between the TCI633 and placebo groups, a short term of study period, and few recruited subjects. Moreover, the results of Western Ontario and McMaster Universities (WOMAC) questionnaires show that TCI633 might retard the progression and development of KOA after the trial. In brief, this preliminary research may provide an alternative approach to the improvement of KOA by probiotics although more detailed investigations should be conducted for solid conclusions.
Collapse
Affiliation(s)
- Jia-Ling Lyu
- Research & Design Center, TCI CO., Ltd., Taipei, 114, Taiwan
- Department of Cosmeceutics, China Medical University, Taichung, 404, Taiwan
| | - Ting-Ming Wang
- Department of Orthopaedic Surgery, School of Medicine, National Taiwan University, Taipei, 100, Taiwan
- Department of Orthopaedic Surgery, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Yen-Hao Chen
- Research & Design Center, TCI Gene Inc., Taipei, 114, Taiwan
| | - Shu-Ting Chang
- Research & Design Center, TCI CO., Ltd., Taipei, 114, Taiwan
| | - Ming-Shiang Wu
- Departments of Internal Medicine, School of Medicine, National Taiwan University, Taipei, 100, Taiwan
- Departments of Internal Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Yung-Hao Lin
- Global Business Center, TCI CO., Ltd., Taipei, 114, Taiwan
| | - Yung-Hsiang Lin
- Research & Design Center, TCI CO., Ltd., Taipei, 114, Taiwan
| | - Chen-Meng Kuan
- Research & Design Center, TCI CO., Ltd., Taipei, 114, Taiwan
| |
Collapse
|
146
|
Li H, Liu F, Lu J, Shi J, Guan J, Yan F, Li B, Huo G. Probiotic Mixture of Lactobacillus plantarum Strains Improves Lipid Metabolism and Gut Microbiota Structure in High Fat Diet-Fed Mice. Front Microbiol 2020; 11:512. [PMID: 32273874 PMCID: PMC7113563 DOI: 10.3389/fmicb.2020.00512] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/10/2020] [Indexed: 12/13/2022] Open
Abstract
The global prevalence of obesity is rising year by year, which has become a public health problem worldwide. In recent years, animal studies and clinical studies have shown that some lactic acid bacteria possess an anti-obesity effect. In our previous study, mixed lactobacilli (Lactobacillus plantarum KLDS1.0344 and Lactobacillus plantarum KLDS1.0386) exhibited anti-obesity effects in vivo by significantly reducing body weight gain, Lee's index and body fat rate; however, its underlying mechanisms of action remain unclear. Therefore, the present study aims to explore the possible mechanisms for the inhibitory effect of mixed lactobacilli on obesity. C57BL/6J mice were randomly divided into three groups including control group (Control), high fat diet group (HFD) and mixed lactobacilli group (MX), and fed daily for eight consecutive weeks. The results showed that mixed lactobacilli supplementation significantly improved blood lipid levels and liver function, and alleviated liver oxidative stress. Moreover, the mixed lactobacilli supplementation significantly inhibited lipid accumulation in the liver and regulated lipid metabolism in epididymal fat pads. Notably, the mixed lactobacilli treatment modulated the gut microbiota, resulting in a significant increase in acetic acid and butyric acid. Additionally, Spearman's correlation analysis found that several specific genera were significantly correlated with obesity-related indicators. These results indicated that the mixed lactobacilli supplementation could manipulate the gut microbiota and its metabolites (acetic acid and butyric acid), resulting in reduced liver lipid accumulation and improved lipid metabolism of adipose tissue, which inhibited obesity.
Collapse
Affiliation(s)
- Huizhen Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
- College of Food, Northeast Agricultural University, Harbin, China
| | - Fei Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Jingjing Lu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
- College of Food, Northeast Agricultural University, Harbin, China
| | - Jialu Shi
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
- College of Food, Northeast Agricultural University, Harbin, China
| | - Jiaqi Guan
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
- College of Food, Northeast Agricultural University, Harbin, China
| | - Fenfen Yan
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
- College of Food, Northeast Agricultural University, Harbin, China
| | - Bailiang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
- College of Food, Northeast Agricultural University, Harbin, China
| | - Guicheng Huo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
- College of Food, Northeast Agricultural University, Harbin, China
| |
Collapse
|
147
|
Harasymowicz NS, Choi YR, Wu CL, Iannucci L, Tang R, Guilak F. Intergenerational Transmission of Diet-Induced Obesity, Metabolic Imbalance, and Osteoarthritis in Mice. Arthritis Rheumatol 2020; 72:632-644. [PMID: 31646754 DOI: 10.1002/art.41147] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 10/17/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Obesity and osteoarthritis (OA) are 2 major public health issues affecting millions of people worldwide. Whereas parental obesity affects the predisposition to diseases such as cancer or diabetes in children, transgenerational influences on musculoskeletal conditions such as OA are poorly understood. This study was undertaken to assess the intergenerational effects of a parental/grandparental high-fat diet on the metabolic and skeletal phenotype, systemic inflammation, and predisposition to OA in 2 generations of offspring in mice. METHODS Metabolic phenotype and predisposition to OA were investigated in the first and second (F1 and F2) generations of offspring (n = 10-16 mice per sex per diet) bred from mice fed a high-fat diet (HFD) or a low-fat control diet. OA was induced by destabilizing the medial meniscus. OA, synovitis, and adipose tissue inflammation were determined histologically, while bone changes were measured using micro-computed tomography. Serum and synovial cytokines were measured by multiplex assay. RESULTS Parental high-fat feeding showed an intergenerational effect, with inheritance of increased weight gain (up to 19% in the F1 generation and 9% in F2), metabolic imbalance, and injury-induced OA in at least 2 generations of mice, despite the fact that the offspring were fed the low-fat diet. Strikingly, both F1 and F2 female mice showed an increased predisposition to injury-induced OA (48% higher predisposition in F1 and 19% in F2 female mice fed the HFD) and developed bone microarchitectural changes that were attributable to parental and grandparental high-fat feeding. CONCLUSION The results of this study reveal a detrimental effect of parental HFD and obesity on the musculoskeletal integrity of 2 generations of offspring, indicating the importance of further investigation of these effects. An improved understanding of the mechanisms involved in the transmissibility of diet-induced changes through multiple generations may help in the development of future therapies that would target the effects of obesity on OA and related conditions.
Collapse
Affiliation(s)
- Natalia S Harasymowicz
- Washington University in St. Louis and Shriners Hospitals for Children, St. Louis, Missouri
| | - Yun-Rak Choi
- Washington University in St. Louis and Shriners Hospitals for Children, St. Louis, Missouri, and Yonsei University College of Medicine, Seoul, South Korea
| | - Chia-Lung Wu
- Washington University in St. Louis and Shriners Hospitals for Children, St. Louis, Missouri
| | - Leanne Iannucci
- Washington University in St. Louis and Shriners Hospitals for Children, St. Louis, Missouri
| | - Ruhang Tang
- Washington University in St. Louis and Shriners Hospitals for Children, St. Louis, Missouri
| | - Farshid Guilak
- Washington University in St. Louis and Shriners Hospitals for Children, St. Louis, Missouri
| |
Collapse
|
148
|
Klancic T, Reimer RA. Gut microbiota and obesity: Impact of antibiotics and prebiotics and potential for musculoskeletal health. JOURNAL OF SPORT AND HEALTH SCIENCE 2020; 9:110-118. [PMID: 32099719 PMCID: PMC7031774 DOI: 10.1016/j.jshs.2019.04.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 03/06/2019] [Accepted: 03/12/2019] [Indexed: 06/10/2023]
Abstract
Obesity is a complex disease with multiple contributing factors. One of the most intensely studied factors during the past decade has been the gut microbiota, which is the community of all microbes in the intestinal tract. The gut microbiota, via energy extraction, inflammation, and other actions, is now recognized as an important player in the pathogenesis of obesity. Dysbiosis, or an imbalance in the microbial community, can initiate a cascade of metabolic disturbances in the host. Early life is a particularly important period for the development of the gut microbiota, and perturbations such as with antibiotic exposure can have long-lasting consequences for host health. In early life and throughout the life span, diet is one of the most important factors that shape the gut microbiota. Although diets high in fat and sugar have been shown to contribute to dysbiosis and disease, dietary fiber is recognized as an important fermentative fuel for the gut microbiota and results in the production of short-chain fatty acids that can act as signaling molecules in the host. One particular type of fiber, prebiotic fiber, contributes to changes in the gut microbiota, the most notable of which is an increase in the abundance of Bifidobacterium. This review highlights our current understanding of the role of gut microbiota in obesity development and the ways in which manipulating the microbiota through dietary means, specifically prebiotics, could contribute to improved health in the host, including musculoskeletal health.
Collapse
Affiliation(s)
- Teja Klancic
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Raylene A Reimer
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada; Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
149
|
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: 2.4] [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
|
150
|
How can parental obesity promote OA across generations? Nat Rev Rheumatol 2020; 16:129-130. [PMID: 31949286 DOI: 10.1038/s41584-020-0369-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|