1
|
Freire de Carvalho J, Skare T. Coenzyme Q10 supplementation in rheumatic diseases: A systematic review. Clin Nutr ESPEN 2024; 59:63-69. [PMID: 38220408 DOI: 10.1016/j.clnesp.2023.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 11/13/2023] [Accepted: 11/18/2023] [Indexed: 01/16/2024]
Abstract
Coenzyme Q10 (CoQ10) is a potent antioxidant and anti-inflammatory substance used to treat some rheumatic diseases. Our objective was to review the use of CoQ10 in rheumatic diseases. PubMed/Medline, Embase, Scopus, and Web of Science databases were searched for articles on CoQ10 and rheumatic diseases between 1966 and April 2023. Twenty articles were found, including 483 patients. The investigated conditions were Fibromyalgia (FM) with 15 studies, Rheumatoid Arthritis (RA) with 3 studies, and Antiphospholipid Syndrome (APS) with 2 studies. After CoQ10 supplementation, RA patients observed improvements in disease activity index, inflammatory biomarkers (erythrocyte sedimentation rate), cytokine levels, and a decrease in malondialdehyde. In APS, CoQ10 improved endothelial function and decreased prothrombotic and proinflammatory mediators. Regarding FM, in most of the studies, the patients observed improvements in pain, fatigue, sleep, tender points count, mood disorders, and scores on the Fibromyalgia Impact Questionnaire (FIQ). The drug was well tolerated, with reports of minor side effects in two studies. CoQ10 supplementation seems to be efficacious as a complementary treatment for RA and FM. Upcoming studies with larger samples and including other rheumatic diseases are welcome.
Collapse
Affiliation(s)
- Jozélio Freire de Carvalho
- Núcleo de Pesquisa em Doenças Crônicas não Transmissíveis (NUPEN), School of Nutrition from the Federal University of Bahia, Salvador, Bahia, Brazil.
| | - Thelma Skare
- Unit of Rheumatology, Hospital Evangélico Mackenzie, Curitiba, PR, Brazil
| |
Collapse
|
2
|
Gu S, Yang D, Liu C, Xue W. The role of probiotics in prevention and treatment of food allergy. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
3
|
Qin H, Fu Y, Zhou K, Song H, Fang G, Chen Q, Pang Y. Toddalia asiatica extract attenuates adjuvant-induced arthritis by modulating colon Th17/Treg balance and colony homeostasis. JOURNAL OF ETHNOPHARMACOLOGY 2023; 313:116542. [PMID: 37127142 DOI: 10.1016/j.jep.2023.116542] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/27/2023] [Accepted: 04/21/2023] [Indexed: 05/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Given the adverse effects of the current principal treatments, there is still a great need for effective cures for rheumatoid arthritis (RA), an immune-mediated disease. Toddalia asiatica (L.) Lam is a traditional medicinal herb that can be used for RA treatment because of its anti-inflammatory and analgesic properties. AIM OF THE STUDY To investigate the possible effects of Toddalia asiatica extract (TAE) on intestinal immunity and the intestinal bacterial flora in a rat model of RA. MATERIALS AND METHODS The anti-arthritis effect of TAE was evaluated in arthritis rats induced by complete Freund's adjuvant-induced arthritis (AIA). Arthritis index (AI) scores, systemic inflammation scores, histopathologic changes in the colon and ankle were detected by hematoxylin and eosin staining. Western blot analysis was performed to assess the protein expression of IL-17A, RORC, IL-1β, IL-6, FOXP3, IL-10 in the colon. RT-PCR was performed to assess the expression of the colon's mRNA. Finally, changes to the gut microbiome by sequencing 16S rDNA. Microbial function prediction was performed using PICRUSt with the KEGG databases and correlation analysis was carried out by computing Spearman's rank correlations. RESULTS demonstrated that TAE administration at a dose of 3 g/kg dramatically decreased AI scores, systemic inflammation scores, and histopathologic lesions of the ankle and colon in AIA rats. TAE was found to significantly reduce the expression levels of Th17-related proteins and mRNAs (IL-17A, RORC, IL-1β and IL-6) in the colon, while increasing the expression levels of Treg-related proteins and mRNA (IL-10 and FOXP3), which helped restore the balance of Th17/Treg immune cells in the colon. Meanwhile, TAE was also found to be capable of remodeling the gut microbiota in AIA rats. Depleting RA-associated genera and thereby increasing α-diversity enriched the gut microbiota's diversity and shifted the community composition dramatically, leading to the increase of Firmicutes_unclassified, Ruminococcaceae_unclassified, Muribaculum, Subdoligranulum, Lachnospira, Marvinbryantia, and the reduction of RA-related bacteria Ligilactobacillus, Streptococcus and Eubacterium-eligens-group. Furthermore, PICRUSt analysis revealed that metabolic pathways were associated with TAE treatment, with metabolic pathways dominating. Among them, metabolic pathways were predominant. Correlation studies showed that a total of 9 microorganisms, including Ligilactobacillus, Eubacterium-eligens-group and Subdoligranulum, were significantly associated with Th17/Treg expression. CONCLUSIONS This study demonstrates that TAE is a low-toxicity poly alkaline drug that can rapidly and effectively improve joint symptoms in RA rats and increases beneficial intestinal bacteria and decreases harmful ones, which is associated with modulating Th17/Treg interactions in intestinal T cells and reversing microbial disorders.
Collapse
Affiliation(s)
- Huangguan Qin
- College of Zhuang Medicine, Guangxi University of Traditional Chinese Medicine, Nanning, 530022, PR China; Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, Nanning, 530200, PR China
| | - Yulei Fu
- College of Zhuang Medicine, Guangxi University of Traditional Chinese Medicine, Nanning, 530022, PR China; Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, Nanning, 530200, PR China
| | - Kan Zhou
- College of Zhuang Medicine, Guangxi University of Traditional Chinese Medicine, Nanning, 530022, PR China; Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, Nanning, 530200, PR China
| | - Huanhuan Song
- College of Zhuang Medicine, Guangxi University of Traditional Chinese Medicine, Nanning, 530022, PR China; Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, Nanning, 530200, PR China
| | - Gang Fang
- College of Zhuang Medicine, Guangxi University of Traditional Chinese Medicine, Nanning, 530022, PR China; Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, Nanning, 530200, PR China
| | - Qing Chen
- Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, Nanning, 530200, PR China.
| | - Yuzhou Pang
- College of Zhuang Medicine, Guangxi University of Traditional Chinese Medicine, Nanning, 530022, PR China; Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, Nanning, 530200, PR China.
| |
Collapse
|
4
|
Na HS, Woo JS, Kim JH, Lee JS, Um IG, Cho KH, Kim GH, Cho ML, Chung SJ, Park SH. Coenzyme Q10 encapsulated in micelles ameliorates osteoarthritis by inhibiting inflammatory cell death. PLoS One 2022; 17:e0270351. [PMID: 35749420 PMCID: PMC9231733 DOI: 10.1371/journal.pone.0270351] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/08/2022] [Indexed: 01/15/2023] Open
Abstract
Background Osteoarthritis (OA) is the most common degenerative joint disease and is characterized by breakdown of joint cartilage. Coenzyme Q10 (CoQ10) exerts diverse biological effects on bone and cartilage; observational studies have suggested that CoQ10 may slow OA progression and inflammation. However, any effect of CoQ10 on OA remains unclear. Here, we investigated the therapeutic utility of CoQ10-micelles. Methods Seven-week-old male Wistar rats were injected with monosodium iodoacetate (MIA) to induce OA. CoQ10-micelles were administered orally to MIA-induced OA rats; celecoxib served as the positive control. Pain, tissue destruction, and inflammation were measured. The expression levels of catabolic and inflammatory cell death markers were assayed in CoQ10-micelle-treated chondrocytes. Results Oral supplementation with CoQ10-micelles attenuated OA symptoms remarkably, including pain, tissue destruction, and inflammation. The expression levels of the inflammatory cytokines IL-1β, IL-6, and MMP-13, and of the inflammatory cell death markers RIP1, RIP3, and pMLKL in synovial tissues were significantly reduced by CoQ10-micelle supplementation, suggesting that CoQ10-micelles might attenuate the synovitis of OA. CoQ10-micelle addition to cultured OA chondrocytes reduced the expression levels of catabolic and inflammatory cell death markers. Conclusions CoQ10-micelles might usefully treat 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, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jin Seok Woo
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | - Jeong Su Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - In Gyu Um
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Keun-Hyung Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Medical Lifesciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
- * E-mail: (MLC); (SJC); (SHP)
| | - Sang J. Chung
- Department of Biopharmaceutical Convergence, School of Pharmacy, Sungkyunkwankwan University, Suwon, Korea
- * E-mail: (MLC); (SJC); (SHP)
| | - Sung-Hwan Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- * E-mail: (MLC); (SJC); (SHP)
| |
Collapse
|
5
|
Research status of biodegradable metals designed for oral and maxillofacial applications: A review. Bioact Mater 2021; 6:4186-4208. [PMID: 33997502 PMCID: PMC8099919 DOI: 10.1016/j.bioactmat.2021.01.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 01/10/2021] [Accepted: 01/10/2021] [Indexed: 01/08/2023] Open
Abstract
The oral and maxillofacial regions have complex anatomical structures and different tissue types, which have vital health and aesthetic functions. Biodegradable metals (BMs) is a promising bioactive materials to treat oral and maxillofacial diseases. This review summarizes the research status and future research directions of BMs for oral and maxillofacial applications. Mg-based BMs and Zn-based BMs for bone fracture fixation systems, and guided bone regeneration (GBR) membranes, are discussed in detail. Zn-based BMs with a moderate degradation rate and superior mechanical properties for GBR membranes show great potential for clinical translation. Fe-based BMs have a relatively low degradation rate and insoluble degradation products, which greatly limit their application and clinical translation. Furthermore, we proposed potential future research directions for BMs in the oral and maxillofacial regions, including 3D printed BM bone scaffolds, surface modification for BMs GBR membranes, and BMs containing hydrogels for cartilage regeneration, soft tissue regeneration, and nerve regeneration. Taken together, the progress made in the development of BMs in oral and maxillofacial regions has laid a foundation for further clinical translation.
Collapse
|
6
|
López-Pedrera C, Villalba JM, Patiño-Trives AM, Luque-Tévar M, Barbarroja N, Aguirre MÁ, Escudero-Contreras A, Pérez-Sánchez C. Therapeutic Potential and Immunomodulatory Role of Coenzyme Q 10 and Its Analogues in Systemic Autoimmune Diseases. Antioxidants (Basel) 2021; 10:antiox10040600. [PMID: 33924642 PMCID: PMC8069673 DOI: 10.3390/antiox10040600] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 12/14/2022] Open
Abstract
Coenzyme Q10 (CoQ10) is a mitochondrial electron carrier and a powerful lipophilic antioxidant located in membranes and plasma lipoproteins. CoQ10 is endogenously synthesized and obtained from the diet, which has raised interest in its therapeutic potential against pathologies related to mitochondrial dysfunction and enhanced oxidative stress. Novel formulations of solubilized CoQ10 and the stabilization of reduced CoQ10 (ubiquinol) have improved its bioavailability and efficacy. Synthetic analogues with increased solubility, such as idebenone, or accumulated selectively in mitochondria, such as MitoQ, have also demonstrated promising properties. CoQ10 has shown beneficial effects in autoimmune diseases. Leukocytes from antiphospholipid syndrome (APS) patients exhibit an oxidative perturbation closely related to the prothrombotic status. In vivo ubiquinol supplementation in APS modulated the overexpression of inflammatory and thrombotic risk-markers. Mitochondrial abnormalities also contribute to immune dysregulation and organ damage in systemic lupus erythematosus (SLE). Idebenone and MitoQ improved clinical and immunological features of lupus-like disease in mice. Clinical trials and experimental models have further demonstrated a therapeutic role for CoQ10 in Rheumatoid Arthritis, multiple sclerosis and type 1 diabetes. This review summarizes the effects of CoQ10 and its analogs in modulating processes involved in autoimmune disorders, highlighting the potential of these therapeutic approaches for patients with immune-mediated diseases.
Collapse
Affiliation(s)
- Chary López-Pedrera
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
- Correspondence: ; Tel.: +34-957-213795
| | - José Manuel Villalba
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, 14014 Córdoba, Spain; (J.M.V.); (C.P.-S.)
| | - Alejandra Mª Patiño-Trives
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
| | - Maria Luque-Tévar
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
| | - Nuria Barbarroja
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
| | - Mª Ángeles Aguirre
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
| | - Alejandro Escudero-Contreras
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
| | - Carlos Pérez-Sánchez
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, 14014 Córdoba, Spain; (J.M.V.); (C.P.-S.)
| |
Collapse
|
7
|
K NK, Patil P, Bhandary SK, Haridas V, N SK, E S, Shetty P. Is butyrate a natural alternative to dexamethasone in the management of CoVID-19? F1000Res 2021; 10:273. [PMID: 34046165 PMCID: PMC8108555 DOI: 10.12688/f1000research.51786.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/22/2021] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (CoVID-19) caused by Severe Acute Respiratory Syndrome Coronavirus 2 has affected more than 100 million lives. Severe CoVID-19 infection may lead to acute respiratory distress syndrome and death of the patient, and is associated with hyperinflammation and cytokine storm. The broad spectrum immunosuppressant corticosteroid, dexamethasone, is being used to manage the cytokine storm and hyperinflammation in CoVID-19 patients. However, the extensive use of corticosteroids leads to serious adverse events and disruption of the gut-lung axis. Various micronutrients and probiotic supplementations are known to aid in the reduction of hyperinflammation and restoration of gut microbiota. The attenuation of the deleterious immune response and hyperinflammation could be mediated by short chain fatty acids produced by the gut microbiota. Butyric acid, the most extensively studied short chain fatty acid, is known for its anti-inflammatory properties. Additionally, butyric acid has been shown to ameliorate hyperinflammation and reduce oxidative stress in various pathologies, including respiratory viral infections. In this review, the potential anti-inflammatory effects of butyric acid that aid in cytokine storm depletion, and its usefulness in effective management of critical illness related to CoVID-19 have been discussed.
Collapse
Affiliation(s)
- Nithin K. K
- Division of Proteomics and Cancer Biology, Nitte University Center for Science Education and Research, Nitte (Deemed to be University), Mangaluru, Karnataka, 575018, India
| | - Prakash Patil
- Central Research Laboratory, K S Hegde Medical Academy, Nitte (Deemed to be University), Mangaluru, Karnataka, 575018, India
| | - Satheesh Kumar Bhandary
- Department of ENT, Justice K S Hegde Charitable Hospital, Nitte (Deemed to be University), Mangaluru, Karnataka, 575018, India
| | - Vikram Haridas
- Arthritis Superspeciality Center, Hublic, Karnataka, 580020, India
| | - Suchetha Kumari N
- Department of Biochemistry/Central Research Laboratory, K S Hegde Medical Academy, Nitte (Deemed to be University), Mangaluru, Karnataka, 575018, India
| | - Sarathkumar E
- Division of Proteomics and Cancer Biology, Nitte University Center for Science Education and Research, Nitte (Deemed to be University), Mangaluru, Karnataka, 575018, India
| | - Praveenkumar Shetty
- Department of Biochemistry/Central Research Laboratory, K S Hegde Medical Academy, Nitte (Deemed to be University), Mangaluru, Karnataka, 575018, India
| |
Collapse
|
8
|
Delay L, Gonçalves Dos Santos G, Dias EV, Yaksh TL, Corr M. Sexual Dimorphism in the Expression of Pain Phenotype in Preclinical Models of Rheumatoid Arthritis. Rheum Dis Clin North Am 2021; 47:245-264. [PMID: 33781493 DOI: 10.1016/j.rdc.2020.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Rheumatoid arthritis is one of most frequent rheumatic diseases, affecting around 1% of the population worldwide. Pain impacting the quality of life for the patient with rheumatoid arthritis, is often the primary factor leading them to seek medical care. Although sex-related differences in humans and animal models of rheumatoid arthritis are described, the correlation between pain and sex in rheumatoid arthritis has only recently been directly examined. Here we review the literature and explore the mechanisms underlying the expression of the pain phenotype in females and males in preclinical models of rheumatoid arthritis.
Collapse
Affiliation(s)
- Lauriane Delay
- Department of Anesthesiology, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA.
| | | | - Elayne Vieira Dias
- Department of Anesthesiology, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Tony L Yaksh
- Department of Anesthesiology, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Maripat Corr
- Division of Rheumatology, Allergy and Immunology, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| |
Collapse
|
9
|
Dai X, Yang D, Bao J, Zhang Q, Ding J, Liu M, Ding M, Liu M, Liang J, Jia X. Er Miao San, a traditional Chinese herbal formula, attenuates complete Freund's adjuvant-induced arthritis in rats by regulating Th17/Treg cells. PHARMACEUTICAL BIOLOGY 2020; 58:157-164. [PMID: 32037930 PMCID: PMC7034067 DOI: 10.1080/13880209.2020.1720745] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Context: Er Miao San (EMS) is a traditional Chinese medicine composed of Atractylodis Rhizoma and Phellodendri Cortex in a 1:1 weight ratio. EMS has been used to treat rheumatism in China for many years.Objective: To evaluate the anti-arthritic activity of EMS extract on adjuvant-induced arthritis (AA) in Sprague-Dawley rats and to clarify its mechanisms of action.Materials and methods: EMS (0.75, 1.5 and 3 g/kg, once daily) was orally administered from day 18 after immunization to day 31. The effects of EMS on AA rats were evaluated by histopathological examination, paw swelling and polyarthritis index. The proliferation of fibroblast-like synoviocyte (FLS) and T cells was detected by CCK-8. The percentages of Th17 cells and Treg cells in splenocytes were determined by flow cytometry. Levels of cytokines in serum were detected by ELISA.Results: EMS treatment significantly decreased the paw volume (from 1.20 to 0.81), polyarthritis index (from 9.56 to 4.46) and alleviated ankle joint histopathology in AA rats. EMS inhibited the proliferation of FLS and T cells. Furthermore, EMS treatment decreased Th17 cells (from 4.62 to 2.08%) and increased Treg cells (from 2.77 to 4.75%) in splenocytes. The levels of IL-17A, TNF-α and IL-6 were remarkably decreased in the serum of EMS-treated rats, whereas the levels of IL-10 and TGF-β1 were significantly increased.Conclusions: EMS exhibits anti-arthritic activity in the AA model by regulating the balance of cytokines and the ratio of Th17 and Treg cells. These insights may provide an experimental basis for the clinical treatment of RA.
Collapse
Affiliation(s)
- Xing Dai
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- The First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Dongping Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Jinping Bao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Qiying Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Jiemin Ding
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Min Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Meihuizi Ding
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Mengli Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Juan Liang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Xiaoyi Jia
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- CONTACT Xiaoyi Jia School of Pharmacy, Anhui University of Chinese Medicine, Hefei230012, China
| |
Collapse
|
10
|
Lactobacillus sakei suppresses collagen-induced arthritis and modulates the differentiation of T helper 17 cells and regulatory B cells. J Transl Med 2020; 18:317. [PMID: 32799896 PMCID: PMC7429687 DOI: 10.1186/s12967-020-02477-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 08/02/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND To evaluate the immunomodulatory effect of Lactobacillus sakei in a mouse model of rheumatoid arthritis (RA) and in human immune cells. METHODS We evaluated whether L. sakei reduced the severity of collagen-induced arthritis (CIA) and modulated interleukin (IL)-17 and IL-10 levels, as well as whether it affected the differentiation of CD4+ T cells and regulatory B cells. We evaluated osteoclastogenesis after culturing bone marrow-derived mononuclear cells with L. sakei. RESULTS The differentiation of T helper 17 cells and the serum level of IL-17 were suppressed by L. sakei in both human peripheral blood mononuclear cells and mouse splenocytes. The serum level of IL-10 was significantly increased in the L. sakei-treated group, whereas the regulatory T cell population was unchanged. The population of regulatory B cells significantly increased the in L. sakei-treated group. Oral administration of L. sakei reduced the arthritis incidence and score in mice with CIA. Finally, osteoclastogenesis and the mRNA levels of osteoclast-related genes were suppressed in the L. sakei-treated group. CONCLUSION L. sakei exerted an anti-inflammatory effect in an animal model of RA, regulated Th17 and regulatory B cell differentiation, and suppressed osteoclastogenesis. Our findings suggest that L. sakei has therapeutic potential for RA.
Collapse
|
11
|
Cannas D, Loi E, Serra M, Firinu D, Valera P, Zavattari P. Relevance of Essential Trace Elements in Nutrition and Drinking Water for Human Health and Autoimmune Disease Risk. Nutrients 2020; 12:2074. [PMID: 32668647 PMCID: PMC7400883 DOI: 10.3390/nu12072074] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023] Open
Abstract
Trace elements produce double-edged effects on the lives of animals and particularly of humans. On one hand, these elements represent potentially toxic agents; on the other hand, they are essentially needed to support growth and development and confer protection against disease. Certain trace elements and metals are particularly involved in humoral and cellular immune responses, playing the roles of cofactors for essential enzymes and antioxidant molecules. The amount taken up and the accumulation in human tissues decisively control whether the exerted effects are toxic or beneficial. For these reasons, there is an urgent need to re-consider, harmonize and update current legislative regulations regarding the concentrations of trace elements in food and in drinking water. This review aims to provide information on the interrelation of certain trace elements with risk of autoimmune disease, with a particular focus on type 1 diabetes and multiple sclerosis. In addition, an overview of the current regulations and regulatory gaps is provided in order to highlight the importance of this issue for everyday nutrition and human health.
Collapse
Affiliation(s)
- Daniela Cannas
- Department of Biomedical Sciences, Unit of Biology and Genetics, University of Cagliari, 09042 Cagliari, Italy; (D.C.); (E.L.)
| | - Eleonora Loi
- Department of Biomedical Sciences, Unit of Biology and Genetics, University of Cagliari, 09042 Cagliari, Italy; (D.C.); (E.L.)
| | - Matteo Serra
- Department of Civil, Environmental Engineering and Architecture, University of Cagliari, 09123 Cagliari, Italy;
| | - Davide Firinu
- Department of Medical Sciences and Public Health, Monserrato Campus, University of Cagliari, 09042 Cagliari, Italy;
| | - Paolo Valera
- Department of Civil, Environmental Engineering and Architecture, University of Cagliari, 09123 Cagliari, Italy;
| | - Patrizia Zavattari
- Department of Biomedical Sciences, Unit of Biology and Genetics, University of Cagliari, 09042 Cagliari, Italy; (D.C.); (E.L.)
| |
Collapse
|
12
|
Cutting Edge: Probiotics and Fecal Microbiota Transplantation in Immunomodulation. J Immunol Res 2019; 2019:1603758. [PMID: 31143780 PMCID: PMC6501133 DOI: 10.1155/2019/1603758] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/01/2019] [Indexed: 12/19/2022] Open
Abstract
Probiotics are commensal or nonpathogenic microbes that confer beneficial effects on the host through several mechanisms such as competitive exclusion, antibacterial effects, and modulation of immune responses. Some probiotics have been found to regulate immune responses via immune regulatory mechanisms. T regulatory (Treg) cells, T helper cell balances, dendritic cells, macrophages, B cells, and natural killer (NK) cells can be considered as the most determinant dysregulated mediators in immunomodulatory status. Recently, fecal microbiota transplantation (FMT) has been defined as the transfer of distal gut microbial communities from a healthy individual to a patient's intestinal tract to cure some immune disorders (mainly inflammatory bowel diseases). The aim of this review was followed through the recent literature survey on immunomodulatory effects and mechanisms of probiotics and FMT and also efficacy and safety of probiotics and FMT in clinical trials and applications.
Collapse
|
13
|
Probiotics in Extraintestinal Diseases: Current Trends and New Directions. Nutrients 2019; 11:nu11040788. [PMID: 30959761 PMCID: PMC6521300 DOI: 10.3390/nu11040788] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 03/26/2019] [Accepted: 04/02/2019] [Indexed: 12/25/2022] Open
Abstract
Probiotics are defined as live microorganisms that when administered in adequate amounts confer a health benefit to the host. Their positive supplementation outcomes on several gastrointestinal disorders are well defined. Nevertheless, their actions are not limited to the gut, but may also impart their beneficial effects at distant sites and organs. In this regard, in this review article we: (i) comprehensively describe the main mechanisms of action of probiotics at distant sites, including bones, skin, and brain; (ii) critically present their therapeutic potential against bone, skin, and neuronal diseases (e.g., osteoporosis, non-healing wounds and autoimmune skin illnesses, mood, behavior, memory, and cognitive impairments); (iii) address the current gaps in the preclinical and clinical research; and (iv) indicate new research directions and suggest future investigations.
Collapse
|
14
|
Immunomodulatory effects of probiotics: Can they be used to treat allergies and autoimmune diseases? Maturitas 2018; 119:25-38. [PMID: 30502748 DOI: 10.1016/j.maturitas.2018.11.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 11/06/2018] [Accepted: 11/08/2018] [Indexed: 12/12/2022]
Abstract
As a person ages, physiological, immunological and gut microbiome changes collectively result in an array of chronic conditions. According to the 'hygiene hypothesis' the increasing prevalence of immune-mediated disorders may be related to intestinal dysbiosis, leading to immune dysfunction and associated conditions such as eczema, asthma, allergies and autoimmune diseases. Beneficial probiotic bacteria can be utilized by increasing their abundance within the gastrointestinal lumen, which in turn will modulate immune cells, such as, T helper (Th)-1, Th2, Th17, regulatory T (Treg) cells and B cells, which have direct relevance to human health and the pathogenesis of immune disorders. Here, we describe the cross-talk between probiotics and the gastrointestinal immune system, and their effects in relation to inflammatory bowel disease, multiple sclerosis, allergies and atopic dermatitis.
Collapse
|
15
|
Suzuki S, Gotoda T, Kusano C, Ikehara H, Miyakoshi Y, Fujii K. Effect of Ubiquinol Intake on Defecation Frequency and Stool Form: A Prospective, Double-Blinded, Randomized Control Study. J Med Food 2018; 22:81-86. [PMID: 30192695 DOI: 10.1089/jmf.2018.4233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Bowel habits affect the quality of life (QOL) of patients with functional gastrointestinal disorders. This study evaluated the effects of reduced form coenzyme Q 10 (ubiquinol) intake on defecation frequency and stool form in patients with daily abdominal symptoms. This was a single-center, prospective, double-blind, randomized control study. Forty-one patients who had the daily symptom of constipation or diarrhea were randomly assigned at a 1:1 ratio to receive either ubiquinol (150 mg/day) or placebo for 12 weeks. Patients completed a daily diary to collect information regarding their numbers of defecations and stool forms according to the Bristol Stool Form (BSF) Scale for 7 days at baseline and 12 weeks. QOL was assessed using the 36-item short-form (SF-36) at baseline and 12 weeks. Twenty-one patients were assigned to the ubiquinol group, and 20 were assigned to the placebo group. At 12 weeks, the mean defecation frequency, compared to baseline, significantly decreased in the ubiquinol group (-0.1 times/day, P = .034) and increased in the placebo group (+0.3 times/day, P = .004). There was no significant change in the 12-week BSF Scale score of the ubiquinol group (+0.2, P = .123), whereas that of the placebo group was increased (+0.5, P < .001). The 12-week general health perception SF-36 score was significantly increased in the ubiquinol group (+3.5, P = .045), whereas there was no significant difference in that score in the placebo group (+1.2, P = .178). In conclusion, taking ubiquinol for 12 weeks decreased defecation frequencies and increased the QOL score, suggesting that ubiquinol may change the bowel habits and improve QOL in patients with abdominal distress.
Collapse
Affiliation(s)
- Sho Suzuki
- 1 Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Takuji Gotoda
- 1 Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Chika Kusano
- 1 Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Hisatomo Ikehara
- 1 Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan.,2 Department of Gastroenterology, Yuri Kumiai General Hospital, Akita, Japan
| | - Yo Miyakoshi
- 3 Supplemental Nutrition Division, Kaneka Corporation, Tokyo, Japan
| | - Kenji Fujii
- 3 Supplemental Nutrition Division, Kaneka Corporation, Tokyo, Japan
| |
Collapse
|