1
|
Bao L, Bian X, Ren L, Bao S, Zhang A. Different doses of hydroxychloroquine regulate the structure of intestinal flora and glycosyltransferase activity in rats with IgA nephropathy. Immunobiology 2025; 230:152891. [PMID: 40112730 DOI: 10.1016/j.imbio.2025.152891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/20/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Hydroxychloroquine (HCQ), by virtue of its ability to reduce proteinuria, is an alternative therapy for Immunoglobulin A nephropathy (IgAN). This study investigated the effects of different doses of HCQ on the structure of intestinal flora and glycosyltransferase activity in IgAN rats. METHODS IgAN model rats constructed by treatment of bovine serum albumin, castor oil and lipopolysaccharide were administered with HCQ (18 or 36 mg/kg) by gavage. Then the number of urine erythrocyte and the renal function of rats were evaluated. The levels of galactose-deficient IgA1 (Gd-IgA1), B cell activation factor (BAFF) and C-reactive protein (CRP) in serum and those of inflammatory factors in renal tissue were detected by ELISA. Renal tissue injury and IgA deposition were assessed by histological analysis. The expressions of Core 1 beta1,3-galactosyltransferase (C1GALT1), Core 1 synthase specific molecular chaperone (COSMC) and ST6 N-acetylgalactosaminide alpha-2,6-sialyltransferase 2 (ST6GALNAC2) were quantified by qRT-PCR, Western blot or in vitro enzyme assays. 16 s rDNA sequencing was used to analyze the structure of intestinal flora in rats. RESULTS HCQ dose-dependently decreased the levels of serum creatinine, UREA, Gd-IgA1, BAFF, CRP and urine protein, waned the number of urine erythrocyte, inhibited the expressions of inflammatory factors and IgA deposition in renal tissue, and up-regulated the expressions of C1GALT1, COSMC and down-regulated ST6GALNAC2 expression in peripheral blood mononuclear cells (PBMCs). CONCLUSION HCQ could reduce glomerular swelling in mesangial area and improve the imbalance of intestinal flora in IgAN rats.
Collapse
Affiliation(s)
- Lingling Bao
- Department of Nephrology, The First Affiliated Hospital of Ningbo University, China
| | - Xueyan Bian
- Department of Nephrology, The First Affiliated Hospital of Ningbo University, China.
| | - Liling Ren
- Department of Nephrology, The First Affiliated Hospital of Ningbo University, China
| | - Sizeng Bao
- Department of Nephrology, The First Affiliated Hospital of Ningbo University, China
| | - Aiwei Zhang
- Department of Nephrology, The First Affiliated Hospital of Ningbo University, China
| |
Collapse
|
2
|
Parodi E, Novi M, Bottino P, La Porta E, Merlotti G, Castello LM, Gotta F, Rocchetti A, Quaglia M. The Complex Role of Gut Microbiota in Systemic Lupus Erythematosus and Lupus Nephritis: From Pathogenetic Factor to Therapeutic Target. Microorganisms 2025; 13:445. [PMID: 40005809 PMCID: PMC11858628 DOI: 10.3390/microorganisms13020445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/07/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
The role of gut microbiota (GM) and intestinal dysbiosis in triggering the onset and/or modulating the severity and progression of lupus nephritis (LN) has been the object of intense research over the last few years. Some alterations at the phyla level, such as the abundance of Proteobacteria and reduction in Firmicutes/Bacteroidetes (F/B) ratio and in α-diversity have been consistently reported in systemic lupus erythematosus (SLE), whereas a more specific role has been ascribed to some species (Bacteroides thetaiotaomicron and Ruminococcus gnavus) in LN. Underlying mechanisms include microbial translocation through a "leaky gut" and subsequent molecular mimicry, immune dysregulation (alteration of IFNγ levels and of balance between Treg and Th17 subsets), and epigenetic interactions. Levels of bacterial metabolites, such as butyrate and other short-chain fatty acids (SCFAs), appear to play a key role in modulating LN. Beyond bacterial components of GM, virome and mycobiome are also increasingly recognized as important players in the modulation of an immune response. On the other hand, microbiota-based therapy appears promising and includes diet, prebiotics, probiotics, symbiotics, and fecal microbiota transplantation (FMT). The modulation of microbiota could correct critical alterations, such as F/B ratio and Treg/Th17 imbalance, and blunt production of autoantibodies and renal damage. Despite current limits, GM is emerging as a powerful environmental factor that could be harnessed to interfere with key mechanisms leading to SLE, preventing flares and organ damage, including LN. The aim of this review is to provide a state-of-the-art analysis of the role of GM in triggering and modulating SLE and LN on the one hand, while exploring possible therapeutic manipulation of GM to control the disease on the other hand.
Collapse
Affiliation(s)
- Emanuele Parodi
- Nephrology and Dialysis Unit, SS. Antonio e Biagio e Cesare Arrigo University Hospital, 15121 Alessandria, Italy;
| | - Marialuisa Novi
- Gastroenterology Unit, SS. Antonio e Biagio e Cesare Arrigo University Hospital, 15121 Alessandria, Italy;
| | - Paolo Bottino
- Microbiology Unit, SS. Antonio e Biagio e Cesare Arrigo University Hospital, 15121 Alessandria, Italy; (F.G.); (A.R.)
| | - Edoardo La Porta
- Nephrology and Dialysis Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy;
| | - Guido Merlotti
- Department of Primary Care, Azienda Socio Sanitaria Territoriale (ASST) of Pavia, 27100 Pavia, Italy;
| | - Luigi Mario Castello
- Internal Medicine Unit, SS. Antonio e Biagio e Cesare Arrigo University Hospital, 15121 Alessandria, Italy;
- Department of Translational Medicine, Università del Piemonte Orientale (UPO), 28100 Novara, Italy
| | - Franca Gotta
- Microbiology Unit, SS. Antonio e Biagio e Cesare Arrigo University Hospital, 15121 Alessandria, Italy; (F.G.); (A.R.)
| | - Andrea Rocchetti
- Microbiology Unit, SS. Antonio e Biagio e Cesare Arrigo University Hospital, 15121 Alessandria, Italy; (F.G.); (A.R.)
| | - Marco Quaglia
- Nephrology and Dialysis Unit, SS. Antonio e Biagio e Cesare Arrigo University Hospital, 15121 Alessandria, Italy;
- Department of Translational Medicine, Università del Piemonte Orientale (UPO), 28100 Novara, Italy
| |
Collapse
|
3
|
Lei Y, Liu Q, Li Q, Zhao C, Zhao M, Lu Q. Exploring the Complex Relationship Between Microbiota and Systemic Lupus Erythematosus. Curr Rheumatol Rep 2023; 25:107-116. [PMID: 37083877 DOI: 10.1007/s11926-023-01102-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2023] [Indexed: 04/22/2023]
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by various autoantibodies and multi-organ. Microbiota dysbiosis in the gut, skin, oral, and other surfaces has a significant impact on SLE development. This article summarizes relevant research and provides new microbiome-related strategies for exploring the mechanisms and treating patients with SLE. RECENT FINDINGS SLE patients have disruptions in multiple microbiomes, with the gut microbiota (bacteria, viruses, and fungi) and their metabolites being the most thoroughly researched. This dysbiosis can promote SLE progression through mechanisms such as the leaky gut, molecular mimicry, and epigenetic regulation. Notwithstanding study constraints on the relationship between microbiota and SLE, specific interventions targeting the gut microbiota, such as probiotics, dietary management, and fecal microbiota transplantation, have emerged as promising SLE therapeutics.
Collapse
Affiliation(s)
- Yu Lei
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
- Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qianmei Liu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
| | - Qilin Li
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
- Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Cheng Zhao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
- Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming Zhao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China.
- Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China.
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China.
- Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
4
|
Quoc QL, Cao TBT, Kim SH, Choi Y, Ryu MS, Choi Y, Park HS, Shin YS. Endocrine-disrupting chemical exposure augments neutrophilic inflammation in severe asthma through the autophagy pathway. Food Chem Toxicol 2023; 175:113699. [PMID: 36871881 DOI: 10.1016/j.fct.2023.113699] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/07/2023] [Accepted: 02/27/2023] [Indexed: 03/07/2023]
Abstract
Corticosteroid resistance, progressive lung function decline, and frequent asthma exacerbations are the hallmarks of neutrophilic asthma (NA). However, the potential contributors and their mechanisms of NA aggravation have not yet been fully clarified. This study was conducted to assess the precise mechanism and inflammatory effects of endocrine-disrupting chemicals using mono-n-butyl phthalate (MnBP) on an NA model. BALB/c mice from normal control and LPS/OVA-induced NA groups were treated with or without MnBP. The effects of MnBP on the airway epithelial cells (AECs), macrophages (Mφ), and neutrophils were investigated in vitro and in vivo. NA mice exposed to MnBP had significantly increased airway hyperresponsiveness, total and neutrophil cell counts in the bronchoalveolar lavage fluid, and the percentage of M1Mφ in the lung tissues compared to those non-exposed to MnBP. In in vitro study, MnBP induced the human neutrophil activation to release neutrophil DNA extracellular traps, Mφ polarizing toward M1Mφ, and AEC damage. Treatment with hydroxychloroquine (an autophagy inhibitor) reduced the effects of MnBP in vivo and in vitro. The results of our study suggest that MnBP exposure may increase the risk of neutrophilic inflammation in severe asthma and autophagy pathway-targeted therapeutics can help control MnBP-induced harmful effects in asthma.
Collapse
Affiliation(s)
- Quang Luu Quoc
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea
| | - Thi Bich Tra Cao
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea
| | - Seo-Hee Kim
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea
| | - Yeji Choi
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea
| | - Min Sook Ryu
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea
| | - Youngwoo Choi
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea
| | - Yoo Seob Shin
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea.
| |
Collapse
|
5
|
The Role of Diet in Influencing the Diversity of Gut Microbiome Related to Lupus Disease Activities: A Systematic Review. Int J Microbiol 2022; 2022:6908677. [PMID: 36624775 PMCID: PMC9825236 DOI: 10.1155/2022/6908677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/26/2022] [Accepted: 12/10/2022] [Indexed: 01/02/2023] Open
Abstract
Gut microbiome dysbiosis can affect the host immune system. The balance and activity of the gut microbiome, which are influenced by daily diet, might be associated with disease activity in systemic lupus erythematosus (SLE). Therefore, we conducted a systematic review based on the PRISMA guideline to explore the role and types of diet that affects the gut microbiome related to changes in SLE disease activity. All original and full-text English articles in the last ten years were included using predefined keywords according to PEO (population, exposure, and outcome) design in PubMed. The study subjects were carefully analyzed, including lupus-susceptible mice and humans with SLE on various diets. Children and pregnant women populations were excluded. Of 134 studies found, only seven full-text articles had met the inclusion criteria of which only one study conducted in human. This human study showed that dietary polyphenol as dihydrochalcones and flavanones affected the gut microbiome and ameliorated lupus disease activity. On the contrary, dietary flavones increased Blautia (family: Lachnospiraceae), and that often found in active lupus diseases. Furthermore, six studies in lupus-prone mice models showed that resistant starch (RS), retinoic acid (RA) or all-trans retinoic acid (tRA), and acidic water (AW) had influenced the gut microbiome, leading to an improved lupus development. Conversely, the 2018 commercial rodent diet, vitamin A-retinoic acid (VARA), neutral water (NW), and high tryptophan diet had impacted various microbiomes, resulting in increased lupus activity. Interestingly, several diets have the effect of either increasing or decreasing lupus disease activity depending on the microbiome they affect, such as AW associated with Turicibacter spp., which is frequently found in active lupus disease, and tRA in Bacteroidetes associated with renal pathology. To conclude, diet can influence the gut microbiome, which is related to the disease activity process of SLE.
Collapse
|
6
|
Vakili K, Fathi M, Yaghoobpoor S, Sayehmiri F, Nazerian Y, Nazerian A, Mohamadkhani A, Khodabakhsh P, Réus GZ, Hajibeygi R, Rezaei-Tavirani M. The contribution of gut-brain axis to development of neurological symptoms in COVID-19 recovered patients: A hypothesis and review of literature. Front Cell Infect Microbiol 2022; 12:983089. [PMID: 36619768 PMCID: PMC9815719 DOI: 10.3389/fcimb.2022.983089] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/25/2022] [Indexed: 12/24/2022] Open
Abstract
The gut microbiota undergoes significant alterations in response to viral infections, particularly the novel SARS-CoV-2. As impaired gut microbiota can trigger numerous neurological disorders, we suggest that the long-term neurological symptoms of COVID-19 may be related to intestinal microbiota disorders in these patients. Thus, we have gathered available information on how the virus can affect the microbiota of gastrointestinal systems, both in the acute and the recovery phase of the disease, and described several mechanisms through which this gut dysbiosis can lead to long-term neurological disorders, such as Guillain-Barre syndrome, chronic fatigue, psychiatric disorders such as depression and anxiety, and even neurodegenerative diseases such as Alzheimer's and Parkinson's disease. These mechanisms may be mediated by inflammatory cytokines, as well as certain chemicals such as gastrointestinal hormones (e.g., CCK), neurotransmitters (e.g., 5-HT), etc. (e.g., short-chain fatty acids), and the autonomic nervous system. In addition to the direct influences of the virus, repurposed medications used for COVID-19 patients can also play a role in gut dysbiosis. In conclusion, although there are many dark spots in our current knowledge of the mechanism of COVID-19-related gut-brain axis disturbance, based on available evidence, we can hypothesize that these two phenomena are more than just a coincidence and highly recommend large-scale epidemiologic studies in the future.
Collapse
Affiliation(s)
- Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shirin Yaghoobpoor
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sayehmiri
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yasaman Nazerian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Ashraf Mohamadkhani
- Digestive Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Pariya Khodabakhsh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gislaine Z. Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Ramtin Hajibeygi
- Department of Cardiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Righi E, Lambertenghi L, Gorska A, Sciammarella C, Ivaldi F, Mirandola M, Sartor A, Tacconelli E. Impact of COVID-19 and Antibiotic Treatments on Gut Microbiome: A Role for Enterococcus spp. Biomedicines 2022; 10:2786. [PMID: 36359311 PMCID: PMC9687172 DOI: 10.3390/biomedicines10112786] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 08/29/2023] Open
Abstract
OBJECTIVE Several studies showed the substantial use of antibiotics and increased risk of antimicrobial resistant infections in patients with COVID-19. The impact of COVID-19-related treatments and antibiotics on gut dysbiosis has not been clarified. DESIGN The prospective cohort study included hospitalized COVID-19 patients (April-December 2020). The gut microbiome composition was analysed by 16S sequencing. The gut diversity and changes in opportunistic bacteria (OBs) or symbionts were analysed according to clinical parameters, laboratory markers of disease progression, type of non-antibiotic COVID-19 treatments (NACT) and type, WHO AWaRe group, and duration of antibiotic therapy (AT). RESULTS A total of 82 patients (mean age 66 ± 13 years, 70% males) were enrolled. The relative abundance of Enterococcus was significantly correlated with duration of hospitalization, intensive care unit stay, O2 needs, and D-dimer, ferritin, and IL-6 blood levels. The presence of Enterococcus showed the highest number of correlations with NACT, AT, and AT + NACT (e.g., hydroxychloroquine ± lopinavir/ritonavir) and increased relative abundance with AWaRe Watch/Reserve antibiotics, AT duration, and combinations. Abundance of Dorea, Agathobacter, Roseburia, and Barnesiella was negatively correlated with AT and corticosteroids use. Patients with increased IL-6, D-dimer, and ferritin levels receiving AT were more likely to show dysbiosis with increased abundance of Enterococcus and Bilophila bacteria and decreased abundance of Roseburia compared with those not receiving AT. CONCLUSION Microbiome diversity is affected by COVID-19 severity. In this context, antibiotic treatment may shift the gut microbiome composition towards OBs, particularly Enterococcus. The impact of treatment-driven dysbiosis on OBs infections and long-term consequences needs further study to define the role of gut homeostasis in COVID-19 recovery and inform targeted interventions.
Collapse
Affiliation(s)
- Elda Righi
- Infectious Diseases Unit, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Lorenza Lambertenghi
- Infectious Diseases Unit, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Anna Gorska
- Infectious Diseases Unit, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Concetta Sciammarella
- Infectious Diseases Unit, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Federico Ivaldi
- Infectious Diseases Unit, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Massimo Mirandola
- Infectious Diseases Unit, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Assunta Sartor
- Microbiology Unit, Udine University Hospital, 33100 Udine, Italy
| | - Evelina Tacconelli
- Infectious Diseases Unit, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| |
Collapse
|
8
|
Pan Z, Hu Y, Huang Z, Han N, Li Y, Zhuang X, Yin J, Peng H, Gao Q, Zhang W, Huang Y, Cui Y, Bi Y, Xu ZZ, Yang R. Alterations in gut microbiota and metabolites associated with altitude-induced cardiac hypertrophy in rats during hypobaric hypoxia challenge. SCIENCE CHINA. LIFE SCIENCES 2022; 65:2093-2113. [PMID: 35301705 DOI: 10.1007/s11427-021-2056-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 01/04/2022] [Indexed: 02/08/2023]
Abstract
The gut microbiota is involved in host responses to high altitude. However, the dynamics of intestinal microecology and their association with altitude-related illness are poorly understood. Here, we used a rat model of hypobaric hypoxia challenge to mimic plateau exposure and monitored the gut microbiome, short-chain fatty acids (SCFAs), and bile acids (BAs) over 28 d. We identified weight loss, polycythemia, and pathological cardiac hypertrophy in hypoxic rats, accompanied by a large compositional shift in the gut microbiota, which is mainly driven by the bacterial families of Prevotellaceae, Porphyromonadaceae, and Streptococcaceae. The aberrant gut microbiota was characterized by increased abundance of the Parabacteroides, Alistipes, and Lactococcus genera and a larger Bacteroides to Prevotella ratio. Trans-omics analyses showed that the gut microbiome was significantly correlated with the metabolic abnormalities of SCFAs and BAs in feces, suggesting an interaction network remodeling of the microbiome-metabolome after the hypobaric hypoxia challenge. Interestingly, the transplantation of fecal microbiota significantly increased the diversity of the gut microbiota, partially inhibited the increased abundance of the Bacteroides and Alistipes genera, restored the decrease of plasma propionate, and moderately ameliorated cardiac hypertrophy in hypoxic rats. Our results provide an insight into the longitudinal changes in intestinal microecology during the hypobaric hypoxia challenge. Abnormalities in the gut microbiota and microbial metabolites contribute to the development of high-altitude heart disease in rats.
Collapse
Affiliation(s)
- Zhiyuan Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Yichen Hu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Zongyu Huang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Ni Han
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Yan Li
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Xiaomei Zhuang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Jiye Yin
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Hui Peng
- Tianjin Institute of Environmental & Operational Medicine, Tianjin, 300050, China
| | - Quansheng Gao
- Tianjin Institute of Environmental & Operational Medicine, Tianjin, 300050, China
| | - Wenpeng Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yong Huang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Yujun Cui
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China.
| | - Zhenjiang Zech Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China. .,Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Ruifu Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China.
| |
Collapse
|
9
|
Lyu X, Chen J, Gao X, Yang J. Emerging story of gut dysbiosis in spondyloarthropathy: From gastrointestinal inflammation to spondyloarthritis. Front Cell Infect Microbiol 2022; 12:973563. [PMID: 36072223 PMCID: PMC9441705 DOI: 10.3389/fcimb.2022.973563] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
As a set of inflammatory disorders, spondyloarthritis (SpA) exhibits distinct pathophysiological, clinical, radiological, and genetic characteristics. Due to the extra-articular features of this disorder, early recognition is crucial to limiting disability and improving outcomes. Gut dysbiosis has been linked to SpA development as evidence grows. A pathogenic SpA process is likely to occur when a mucosal immune system interacts with abnormal local microbiota, with subsequent joint involvement. It is largely unknown, however, how microbiota alterations predate the onset of SpA within the “gut-joint axis”. New microbiome therapies, such as probiotics, are used as an adjuvant therapy in the treatment of SpA, suggesting that the modulation of intestinal microbiota and/or intestinal barrier function may contribute to the prevention of SpA. In this review, we highlight the mechanisms of SpA by which the gut microbiota impacts gut inflammation and triggers the activation of immune responses. Additionally, we analyze the regulatory role of therapeutic SpA medication in the gut microbiota and the potential application of probiotics as adjunctive therapy for SpA.
Collapse
Affiliation(s)
- Xing Lyu
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jieli Chen
- Discipline Construction Office, Tianjin Medical University, Tianjin, China
| | - Xingjie Gao
- Department of Biochemistry and Molecular Biology, Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Cellular and Molecular Immunology in Tianjin, Excellent Talent Project, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Jie Yang
- Department of Biochemistry and Molecular Biology, Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Cellular and Molecular Immunology in Tianjin, Excellent Talent Project, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
- *Correspondence: Jie Yang,
| |
Collapse
|
10
|
Chen Y, Lin J, Xiao L, Zhang X, Zhao L, Wang M, Li L. Gut microbiota in systemic lupus erythematosus: A fuse and a solution. J Autoimmun 2022; 132:102867. [PMID: 35932662 DOI: 10.1016/j.jaut.2022.102867] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 07/05/2022] [Indexed: 12/13/2022]
Abstract
Gut commensals help shape and mold host immune system and deeply influence human health. The disease spectrum of mankind that gut microbiome may associate with is ever-growing, but the mechanisms are still enigmas. Characterized by loss of self-tolerance and sustained self-attack, systemic lupus erythematosus (SLE) is labeled with chronic inflammation, production of autoantibodies and multisystem injury, which so far are mostly incurable. Gut microbiota and their metabolites, now known as important environmental triggers of local/systemic immune responses, have been proposed to be involved in SLE development and progression probably through the following mechanisms: translocation beyond their niches; molecular mimicry to cross-activate immune response targeting self-antigens; epitope spreading to expand autoantibodies spectrum; and bystander activation to promote systemic inflammation. Gut microbiota which varies between individuals may also influence the metabolism and bio-transformation of disease-modifying anti-rheumatic drugs, thus associated with the efficacy and toxicity of these drugs, adding another explanation for heterogenic therapeutic responses. Modulation of gut microbiota via diet, probiotics/prebiotics, antibiotics/phages, fecal microbiota transplantation, or helminth to restore immune tolerance and homeostasis is expected to be a promising neoadjuvant therapy for SLE. We reviewed the advances in this territory and discussed the application prospect of modulating gut microbiota in controlling SLE.
Collapse
Affiliation(s)
- Yanfei Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, China; Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Jin Lin
- Department of Rheumatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, China
| | - Lanlan Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, China; Department of Rheumatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences & Peking Union Medical College, NO.1 Da Hua Road, Dong Dan, Beijing, 100730, China
| | - Lidan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, 100730, China
| | - Min Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences & Peking Union Medical College, NO.1 Da Hua Road, Dong Dan, Beijing, 100730, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, China; Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China.
| |
Collapse
|
11
|
Wang Y, Tian L, Sun L, Zhou W, Zhi W, Qing J, Abdi Saed Y, Dong L, Zhang X, Li Y. Gut Microbes in Immunoglobulin A Nephropathy and Their Potential Therapeutic Applications. Front Med (Lausanne) 2022; 9:823267. [PMID: 35655857 PMCID: PMC9152025 DOI: 10.3389/fmed.2022.823267] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 04/15/2022] [Indexed: 11/13/2022] Open
Abstract
Microbial ecosystem consists of a complex community of bacterial interactions and its host microenvironment (tissue, cell, metabolite). Because the interaction between gut microbiota and host involves many diseases and seriously affects human health, the study of the interaction mechanism between gut microbiota and host has attracted great attention. The gut microbiome is made up of 100 trillion bacteria that have both beneficial and adverse effects on human health. The development of IgA Nephropathy results in changes in the intestinal microbial ecosystem that affect host physiology and health. Similarly, changes in intestinal microbiota also affect the development of IgA Nephropathy. Thus, the gut microbiome represents a novel therapeutic target for improving the outcome of IgA Nephropathy, including hematuria symptoms and disease progression. In this review, we summarize the effect of intestinal microbiota on IgA Nephropathy in recent years and it has been clarified that the intestinal microbiota has a great influence on the pathogenesis and treatment of IgA Nephropathy.
Collapse
Affiliation(s)
- Yi Wang
- The Third Clinical College, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Lingling Tian
- The Third Clinical College, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Lin Sun
- College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Wenjing Zhou
- School of Medical Sciences, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Wenqiang Zhi
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Jianbo Qing
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Yasin Abdi Saed
- Department of Nephrology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Lina Dong
- Core Laboratory, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Xiadong Zhang
- Core Laboratory, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Yafeng Li
- Department of Nephrology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China.,Core Laboratory, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China.,Shanxi Provincial Key Laboratory of Kidney Disease, Taiyuan, China.,Academy of Microbial Ecology, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
12
|
Li XB, Chu XJ, Cao NW, Wang H, Fang XY, Fan YG, Li BZ, Ye DQ. Proton pump inhibitors induce changes in the gut microbiome composition of systemic lupus erythematosus patients. BMC Microbiol 2022; 22:117. [PMID: 35477382 PMCID: PMC9043501 DOI: 10.1186/s12866-022-02533-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 04/20/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Currently, few studies focus on the association between gut microbiota and systemic lupus erythematosus (SLE), and much less studies consider the effect of drug usage. Proton pump inhibitors (PPIs) are commonly used to treat drug-related gastrointestinal damage in SLE patients. Therefore, the purpose of this study is to examine the gut microbiota of SLE patients using PPIs. METHODS Fecal samples from 20 SLE patients with PPIs (P-SLE), 20 SLE patients without PPIs (NP-SLE) and 17 healthy controls (HCs) were obtained. The structure of the bacterial community in the fecal samples was analyzed by 16S rRNA gene sequencing. Redundancy analysis (RDA) was performed to observe the relationship between clinical variables and microbiome composition in P-SLE and NP-SLE patients. Based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) database, functional capabilities of microbiota were estimated. Network analysis was performed to analyze the association of metabolic pathway alterations with altered gut microbiota in P-SLE and NP-SLE patients. RESULTS P-SLE patients exhibited increased alpha-diversity and an altered composition of the gut microbiota compared with NP-SLE patients. The alpha-diversity of NP-SLE patients was significantly lower than HCs but also of P-SLE patients, whose alpha-diversity had become similar to HCs. Compared with NP-SLE patients, the relative abundances of Lactobacillus, Roseburia, Oxalobacter, and Desulfovibrio were increased, while those of Veillonella, Escherichia, Morganella, Pseudomonas and Stenotrophomonas were decreased in P-SLE patients. RDA indicated that PPI use was the only significant exploratory variable for the microbiome composition when comparing SLE patients. KEGG analysis showed that 16 metabolic pathways were significantly different between NP-SLE and P-SLE patients. These metabolic pathways were mainly associated with changes in Escherichia, Roseburia, Stenotrophomonas, Morganella and Alipipes as determined by the network analysis. CONCLUSIONS PPI use is associated with an improved microbiome composition of SLE patients as it 1) increases alpha-diversity levels back to normal, 2) increases the abundance of various (beneficial) commensals, and 3) decreases the abundance of certain opportunistic pathogenic genera such as Escherichia. Validation studies with higher patient numbers are however recommended to explore these patterns in more detail.
Collapse
Affiliation(s)
- Xian-Bao Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Xiu-Jie Chu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Nv-Wei Cao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Hua Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Xin-Yu Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Yin-Guang Fan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Bao-Zhu Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China. .,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China.
| | - Dong-Qing Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China. .,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China.
| |
Collapse
|
13
|
Najmi N, Megantara I, Andriani L, Goenawan H, Lesmana R. Importance of gut microbiome regulation for the prevention and recovery process after SARS-CoV-2 respiratory viral infection (Review). Biomed Rep 2022; 16:25. [PMID: 35251612 PMCID: PMC8889546 DOI: 10.3892/br.2022.1508] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/09/2021] [Indexed: 01/08/2023] Open
Abstract
Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been reported to affect organs other than the lungs, including the liver, brain, kidneys and intestine, and gastrointestinal symptoms, such as nausea, vomiting, diarrhea and abdominal discomfort, have also been reported. Thus, SARS-CoV-2 could potentially directly or indirectly regulate the gut microbiome profile and its homeostasis. The abundance of Coprobacillus, Clostridium ramosum and Clostridium are associated with the severity of COVID-19, and Firmicutes, Bacteriodetes, Proteobacteria and Actinobacteria are also related to COVID-19 infection. The four phyla are correlated with the severity of COVID-19 infection in patients. The modulation of factors that control the physiological growth of the gut microbiome will determine the proportionate ratio of microbiome types (profile). Taken together, gut microbiome profile alterations in COVID-19 patients may have a cross effect with the modulation of cytokine levels in COVID-19 infection. With these findings, several factors that regulate gut microbiome homeostasis may support the degree of the clinical symptoms and hasten the recovery process after COVID-19 infection.
Collapse
Affiliation(s)
- Nuroh Najmi
- Department of Oral Biology, Faculty of Dentistry, Universitas Padjadjaran, Bandung, West Java 45363, Indonesia
- Division of Biological Activty Central Laboratory, Universitas Padjadjaran, Bandung, West Java 45363, Indonesia
| | - Imam Megantara
- Division of Biological Activty Central Laboratory, Universitas Padjadjaran, Bandung, West Java 45363, Indonesia
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java 45363, Indonesia
| | - Lovita Andriani
- Faculty of Animal Husbandry, Universitas Padjadjaran, Bandung, West Java 45363, Indonesia
| | - Hanna Goenawan
- Division of Biological Activty Central Laboratory, Universitas Padjadjaran, Bandung, West Java 45363, Indonesia
- Department of Biomedical Sciences, Physiology Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java 45363, Indonesia
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Bandung, West Java 45363, Indonesia
| | - Ronny Lesmana
- Division of Biological Activty Central Laboratory, Universitas Padjadjaran, Bandung, West Java 45363, Indonesia
- Department of Biomedical Sciences, Physiology Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java 45363, Indonesia
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Bandung, West Java 45363, Indonesia
| |
Collapse
|
14
|
Xiang S, Qu Y, Qian S, Wang R, Wang Y, Jin Y, Li J, Ding X. Association between systemic lupus erythematosus and disruption of gut microbiota: a meta-analysis. Lupus Sci Med 2022; 9:e000599. [PMID: 35346981 PMCID: PMC8961174 DOI: 10.1136/lupus-2021-000599] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 03/06/2022] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Recent studies reported that SLE is characterised by altered interactions between the microbiome and immune system. We performed a meta-analysis of publications on this topic. METHODS Case-control studies that compared patients with SLE and healthy controls (HCs) and determined the diversity of the gut microbiota and the abundance of different microbes were examined. Stata/MP V.16 was used for the meta-analysis. A Bonferroni correction for multiple tests was used to reduce the likelihood of false-positive results. RESULTS We included 11 case-control studies that examined 373 patients with SLE and 1288 HCs. These studies were performed in five countries and nine cities. Compared with HCs, patients with SLE had gut microbiota with lower Shannon-Wiener diversity index (weighted mean difference=-0.22, 95% CI -0.32 to -0.13, p<0.001) and lower Chao1 richness (standardised mean difference (SMD)=-0.62, 95% CI -1.04 to -0.21, p=0.003). Patients with SLE had lower abundance of Ruminococcaceae (SMD = -0.49, 95% CI -0.84 to -0.15,p=0.005), but greater abundance of Enterobacteriaceae (SMD=0.45, 95% CI 0.01 to 0.89, p=0.045) and Enterococcaceae (SMD=0.53, 95% CI 0.05 to 1.01, p=0.03). However, only the results for Ruminococcaceae passed the Bonferroni correction (p=0.0071). The two groups had no significant differences in Lachnospiraceae and Bacteroides (both p>0.05). Patients with SLE who used high doses of glucocorticoids had altered gut microbiota based on the Chao1 species diversity estimator, and hydroxychloroquine use appeared to reduce the abundance of Enterobacteriaceae. CONCLUSIONS Patients with SLE have imbalanced gut microbiota, with a decrease in beneficial bacteria and an increase in harmful bacteria. Drugs used to treat SLE may also alter the gut microbiota of these patients.
Collapse
Affiliation(s)
- Shate Xiang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiqian Qu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Suhai Qian
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rongyun Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yao Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yibo Jin
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinghong Ding
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
15
|
Gut Microbiome-Targeted Modulations Regulate Metabolic Profiles and Alleviate Altitude-Related Cardiac Hypertrophy in Rats. Microbiol Spectr 2022; 10:e0105321. [PMID: 35138162 PMCID: PMC8826942 DOI: 10.1128/spectrum.01053-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
It is well known that humans physiologically or pathologically respond to high altitude, with these responses accompanied by alterations in the gut microbiome. To investigate whether gut microbiota modulation can alleviate high-altitude-related diseases, we administered probiotics, prebiotics, and synbiotics in rat model with altitude-related cardiac impairment after hypobaric hypoxia challenge and observed that all three treatments alleviated cardiac hypertrophy as measured by heart weight-to-body weight ratio and gene expression levels of biomarkers in heart tissue. The disruption of gut microbiota induced by hypobaric hypoxia was also ameliorated, especially for microbes of Ruminococcaceae and Lachnospiraceae families. Metabolome revealed that hypobaric hypoxia significantly altered the plasma short-chain fatty acids (SCFAs), bile acids (BAs), amino acids, neurotransmitters, and free fatty acids, but not the overall fecal SCFAs and BAs. The treatments were able to restore homeostasis of plasma amino acids and neurotransmitters to a certain degree, but not for the other measured metabolites. This study paves the way to further investigate the underlying mechanisms of gut microbiome in high-altitude related diseases and opens opportunity to target gut microbiome for therapeutic purpose. IMPORTANCE Evidence suggests that gut microbiome changes upon hypobaric hypoxia exposure; however, it remains elusive whether this microbiome change is a merely derivational reflection of host physiological alteration, or it synergizes to exacerbate high-altitude diseases. We intervened gut microbiome in the rat model of prolonged hypobaric hypoxia challenge and found that the intervention could alleviate the symptoms of pathological cardiac hypertrophy, gut microbial dysbiosis, and metabolic disruptions of certain metabolites in gut and plasma induced by hypobaric hypoxia. Our study suggests that gut microbiome may be a causative factor for high-altitude-related pathogenesis and a target for therapeutic intervention.
Collapse
|
16
|
Abstract
Oscillospira is a class of organism that often appears in high-throughput sequencing data but has not been purely cultured and is widely present in the animal and human intestines. There is a strong association between variation in Oscillospira abundance and obesity, leanness, and human health. In addition, a growing body of studies has shown that Oscillospira is also implicated in other diseases, such as gallstones and chronic constipation, and has shown some correlation with the positive or negative changes in its course. Sequencing data combined with metabolic profiling indicate that Oscillospira is likely to be a genus capable of producing short-chain fatty acids (SCFAs) such as butyrate, which is an important reference indicator for screening "next-generation probiotics ". Considering the positive effects of Oscillospira in some specific diseases, such as obesity-related metabolic diseases, it has already been characterized as one of the next-generation probiotic candidates and therefore has great potential for development and application in the future food, health care, and biopharmaceutical products.
Collapse
Affiliation(s)
- Jingpeng Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China,CONTACT Jingpeng Yang
| | - Yanan Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Zhiqiang Wen
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Wenzheng Liu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Lingtong Meng
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - He Huang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China,He Huang School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing, China
| |
Collapse
|
17
|
Zhang L, Chu CQ. Gut Microbiota-Medication Interaction in Rheumatic Diseases. Front Immunol 2021; 12:796865. [PMID: 34925383 PMCID: PMC8678121 DOI: 10.3389/fimmu.2021.796865] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 11/15/2021] [Indexed: 02/05/2023] Open
Abstract
Besides its contribution to the development of rheumatic diseases, the gut microbiota interact with anti-rheumatic drugs. The intestinal microbiota can directly metabolize many drugs and indirectly change drug metabolism through a complex multi-dimensional interaction with the host, thus affecting individual response to drug therapy and adverse effects. The focus of the current review is to address recent advances and important progress in our understanding of how the gut microbiota interact with anti-rheumatic drugs and provide perspectives on promoting precision treatment, drug discovery, and better therapy for rheumatic diseases.
Collapse
Affiliation(s)
- Lingshu Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.,Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, United States
| | - Cong-Qiu Chu
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, United States.,Section of Rheumatology, Veterans Affairs (VA) Portland Healthcare System, Portland, OR, United States
| |
Collapse
|
18
|
AKTAS B, ASLIM B. Neuropathy in COVID-19 associated with dysbiosis-related inflammation. Turk J Biol 2021; 45:390-403. [PMID: 34803442 PMCID: PMC8573843 DOI: 10.3906/biy-2105-53] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/30/2021] [Indexed: 01/08/2023] Open
Abstract
Although COVID-19 affects mainly lungs with a hyperactive and imbalanced immune response, gastrointestinal and neurological symptoms such as diarrhea and neuropathic pains have been described as well in patients with COVID-19. Studies indicate that gut-lung axis maintains host homeostasis and disease development with the association of immune system, and gut microbiota is involved in the COVID-19 severity in patients with extrapulmonary conditions. Gut microbiota dysbiosis impairs the gut permeability resulting in translocation of gut microbes and their metabolites into the circulatory system and induce systemic inflammation which, in turn, can affect distal organs such as the brain. Moreover, gut microbiota maintains the availability of tryptophan for kynurenine pathway, which is important for both central nervous and gastrointestinal system in regulating inflammation. SARS-CoV-2 infection disturbs the gut microbiota and leads to immune dysfunction with generalized inflammation. It has been known that cytokines and microbial products crossing the blood-brain barrier induce the neuroinflammation, which contributes to the pathophysiology of neurodegenerative diseases including neuropathies. Therefore, we believe that both gut-lung and gut-brain axes are involved in COVID-19 severity and extrapulmonary complications. Furthermore, gut microbial dysbiosis could be the reason of the neurologic complications seen in severe COVID-19 patients with the association of dysbiosis-related neuroinflammation. This review will provide valuable insights into the role of gut microbiota dysbiosis and dysbiosis-related inflammation on the neuropathy in COVID-19 patients and the disease severity.
Collapse
Affiliation(s)
- Busra AKTAS
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Burdur Mehmet Akif Ersoy University, BurdurTurkey
| | - Belma ASLIM
- Department of Biology, Faculty of Sciences, Gazi University, AnkaraTurkey
| |
Collapse
|
19
|
Chen ZR, Liu J, Liao ZG, Zhou J, Peng HW, Gong F, Hu JF, Zhou Y. COVID-19 and gastroenteric manifestations. World J Clin Cases 2021; 9:4990-4997. [PMID: 34307549 PMCID: PMC8283602 DOI: 10.12998/wjcc.v9.i19.4990] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/14/2021] [Accepted: 05/15/2021] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the infection of a novel coronavirus [severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)], has become a pandemic. The infection has resulted in about one hundred million COVID-19 cases and millions of deaths. Although SARS-CoV-2 mainly spreads through the air and impairs the function of the respiratory system, it also attacks the gastrointestinal epithelial cells through the same receptor, angiotensin converting enzyme 2 receptor, which results in gastroenteric symptoms and potential fecal-oral transmission. Besides the infection of SARS-CoV-2, the treatments of COVID-19 also contribute to the gastroenteric manifestations due to the adverse drug reactions of anti-COVID-19 drugs. In this review, we update the clinical features, basic studies, and clinical practices of COVID-19-associated gastroenteric manifestations.
Collapse
Affiliation(s)
- Zhang-Ren Chen
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Jing Liu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Zhi-Guo Liao
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Jian Zhou
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Hong-Wei Peng
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Fei Gong
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Jin-Fang Hu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Ying Zhou
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| |
Collapse
|