151
|
Gu M, Samuelson DR, Taylor CM, Molina PE, Luo M, Siggins RW, Shellito JE, Welsh DA. Alcohol-associated intestinal dysbiosis alters mucosal-associated invariant T-cell phenotype and function. Alcohol Clin Exp Res 2021; 45:934-947. [PMID: 33704802 PMCID: PMC8283808 DOI: 10.1111/acer.14589] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Chronic alcohol consumption is associated with a compromised innate and adaptive immune responses to infectious disease. Mucosa-associated invariant T (MAIT) cells play a critical role in antibacterial host defense. However, whether alcohol-associated deficits in innate and adaptive immune responses are mediated by alterations in MAIT cells remains unclear. METHODS To investigate the impact of alcohol on MAIT cells, mice were treated with binge-on-chronic alcohol for 10 days and sacrificed at day 11. MAIT cells in the barrier organs (lung, liver, and intestine) were characterized by flow cytometry. Two additional sets of animals were used to examine the involvement of gut microbiota on alcohol-induced MAIT cell changes: (1) Cecal microbiota from alcohol-fed (AF) mice were adoptive transferred into antibiotic-pretreated mice and (2) AF mice were treated with antibiotics during the experiment. MAIT cells in the barrier organs were measured via flow cytometry. RESULTS Binge-on-chronic alcohol feeding led to a significant reduction in the abundance of MAIT cells in the barrier tissues. However, CD69 expression on tissue-associated MAIT cells was increased in AF mice compared with pair-fed (PF) mice. The expression of Th1 cytokines and the corresponding transcriptional factor was tissue specific, showing downregulation in the intestine and increases in the lung and liver in AF animals. Transplantation of fecal microbiota from AF mice resulted in a MAIT cell profile aligned to that of AF mouse donor. Antibiotic treatment abolished the MAIT cell differences between AF and PF animals. CONCLUSION MAIT cells in the intestine, liver, and lung are perturbed by alcohol use and these changes are partially attributable to alcohol-associated dysbiosis. MAIT cell dysfunction may contribute to alcohol-induced innate and adaptive immunity and consequently end-organ pathophysiology.
Collapse
Affiliation(s)
- Min Gu
- Department of Internal Medicine, Section of Pulmonary/Critical Care & Allergy/Immunology, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - Derrick R. Samuelson
- Department of Internal Medicine, Section of Pulmonary/Critical Care & Allergy/Immunology, Louisiana State University Health Science Center, New Orleans, LA, USA
- Department of Internal Medicine, Division of Pulmonary, Critical Care, & Sleep, University of Nebraska Medical Center, Omaha, NE, USA
| | - Christopher M. Taylor
- Department of Microbiology, Immunology, & Parasitology, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - Patricia E. Molina
- Department of Physiology, Louisiana State University Health Science Center, New Orleans, LA, USA
- Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - Meng Luo
- Department of Microbiology, Immunology, & Parasitology, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - Robert W. Siggins
- Department of Physiology, Louisiana State University Health Science Center, New Orleans, LA, USA
- Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - Judd E. Shellito
- Department of Internal Medicine, Section of Pulmonary/Critical Care & Allergy/Immunology, Louisiana State University Health Science Center, New Orleans, LA, USA
- Department of Microbiology, Immunology, & Parasitology, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - David A. Welsh
- Department of Internal Medicine, Section of Pulmonary/Critical Care & Allergy/Immunology, Louisiana State University Health Science Center, New Orleans, LA, USA
- Department of Microbiology, Immunology, & Parasitology, Louisiana State University Health Science Center, New Orleans, LA, USA
- Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Science Center, New Orleans, LA, USA
| |
Collapse
|
152
|
Jahan D, Peile E, Sheikh MA, Islam S, Parasnath S, Sharma P, Iskandar K, Dhingra S, Charan J, Hardcastle TC, Samad N, Chowdhury TS, Dutta S, Haque M. Is it time to reconsider prophylactic antimicrobial use for hematopoietic stem cell transplantation? a narrative review of antimicrobials in stem cell transplantation. Expert Rev Anti Infect Ther 2021; 19:1259-1280. [PMID: 33711240 DOI: 10.1080/14787210.2021.1902304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Hematopoietic Stem Cell Transplantation (HSCT) is a life-saving procedure for multiple types of hematological cancer, autoimmune diseases, and genetic-linked metabolic diseases in humans. Recipients of HSCT transplant are at high risk of microbial infections that significantly correlate with the presence of graft-versus-host disease (GVHD) and the degree of immunosuppression. Infection in HSCT patients is a leading cause of life-threatening complications and mortality. AREAS COVERED This review covers issues pertinent to infection in the HSCT patient, including bacterial and viral infection; strategies to reduce GVHD; infection patterns; resistance and treatment options; adverse drug reactions to antimicrobials, problems of antimicrobial resistance; perturbation of the microbiome; the role of prebiotics, probiotics, and antimicrobial peptides. We highlight potential strategies to minimize the use of antimicrobials. EXPERT OPINION Measures to control infection and its transmission remain significant HSCT management policy and planning issues. Transplant centers need to consider carefully prophylactic use of antimicrobials for neutropenic patients. The judicious use of appropriate antimicrobials remains a crucial part of the treatment protocol. However, antimicrobials' adverse effects cause microbiome diversity and dysbiosis and have been shown to increase morbidity and mortality.
Collapse
Affiliation(s)
- Dilshad Jahan
- Department of Hematology, Asgar Ali Hospital, 111/1/A Distillery Road, Gandaria Beside Dhupkhola, Dhaka 1204, Bangladesh
| | - Ed Peile
- Department of Medical Education, Warwick Medical School, University of Warwick, Coventry, UK
| | | | - Salequl Islam
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| | - Sharlene Parasnath
- Department of Clinical Hematology, Inkosi Albert Luthuli Central Hospital, 800 Vusi Mzimela Road, Cato Manor, Durban, South Africa
| | - Paras Sharma
- Department of Pharmacognosy, BVM College of Pharmacy, Gwalior, India
| | - Katia Iskandar
- Lebanese University, School of Pharmacy, Beirut, Lebanon.,INSPECT-LB: Institute National de Sante Publique, Epidemiologie Clinique et Toxicologie, Beirut, Lebanon.,Universite Paul Sabatier UT3, INSERM, UMR1027, Toulouse, France
| | - Sameer Dhingra
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Bihar, India
| | - Jaykaran Charan
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Timothy Craig Hardcastle
- Trauma Service, Inkosi Albert Luthuli Central Hospital, Mayville, South Africa.,Department of Surgery, Nelson R Mandela School of Clinical Medicine, UKZN, South Africa
| | - Nandeeta Samad
- Department of Public Health, North South University, Bangladesh
| | | | - Siddhartha Dutta
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Mainul Haque
- The Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kem Perdana Sungai Besi, Kuala Lumpur, Malaysia
| |
Collapse
|
153
|
Rodríguez-Padilla Á, Morales-Martín G, Pérez-Quintero R, Gómez-Salgado J, Rada-Morgades R, Ruiz-Frutos C. Diversion Colitis: Macro and Microscopic Findings after Probiotics Stimulation. BIOLOGY 2021; 10:biology10040303. [PMID: 33917556 PMCID: PMC8067519 DOI: 10.3390/biology10040303] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 12/24/2022]
Abstract
Simple Summary The observations presented in this study conclude that the preoperative stimulation with probiotics of the efferent loop through the dysfunctional bowel, to allow the slow infusion, can have a reducing effect on the endoscopic and histopathological alterations of diversion colitis. This procedure may be an alternative treatment to resolve the inflammation in patients where the surgical option is not feasible or available. Abstract The use of a loop ileostomy as the defunctioning procedure of choice to protect a distal colonic anastomosis causes histological and endoscopic changes in the intestinal mucosal architecture, which have been related to chronic inflammation and changes in the microflora that consequently impact the intestinal structure and function following fecal stream diversion. The aim of this study was to evaluate the histological and endoscopic changes on the colonic mucosa in patients with diversion colitis after stimulation of the efferent loop with probiotics prior to closure of the protective ileostomy. A prospective, randomized, double-blind, controlled study was designed. All patients who underwent surgery for colorectal carcinoma with protective ileostomy between January 2017 and December 2018 were included. These patients were pending reconstructive surgery and were diagnosed with endoscopic and histological diversion colitis. Divided into two groups, a group stimulated with probiotics (SG) and a control group (CG). 34 cases and 35 controls were included in the study. Histological and endoscopic changes were evaluated after stimulation, after restorative surgery and during the short-term follow-up after surgery. A decrease in endoscopic pathological findings (mucosal friability, mucous erosions, polyps, edema, erythema and stenosis) and in histological findings (follicular hyperplasia, eosinophils, cryptic abscesses, lymphocyte infiltration, plasma cell infiltration and architecture distortion) was observed in SG. These results were statistically significant with a p < 0.001. The stimulation of the efferent loop of the ileostomy in patients with diversion colitis produced a decrease of the endoscopic and histological severity of colitis in the short term.
Collapse
Affiliation(s)
- Ángela Rodríguez-Padilla
- Department of General Surgery, Infanta Elena University Clinical Hospital, 21080 Huelva, Spain; (Á.R.-P.); (G.M.-M.)
| | - Germán Morales-Martín
- Department of General Surgery, Infanta Elena University Clinical Hospital, 21080 Huelva, Spain; (Á.R.-P.); (G.M.-M.)
| | - Rocío Pérez-Quintero
- Department of General Surgery, Juan Ramón Jiménez University Clinical Hospital, 21005 Huelva, Spain; (R.P.-Q.); (R.R.-M.)
| | - Juan Gómez-Salgado
- Department of Sociology, Social Work and Public Health, Faculty of Labour Sciences, University of Huelva, 21007 Huelva, Spain;
- Safety and Health Postgraduate Programme, Universidad Espíritu Santo, Guayaquil 092301, Ecuador
- Correspondence: ; Tel.: +34-959219700
| | - Ricardo Rada-Morgades
- Department of General Surgery, Juan Ramón Jiménez University Clinical Hospital, 21005 Huelva, Spain; (R.P.-Q.); (R.R.-M.)
| | - Carlos Ruiz-Frutos
- Department of Sociology, Social Work and Public Health, Faculty of Labour Sciences, University of Huelva, 21007 Huelva, Spain;
- Safety and Health Postgraduate Programme, Universidad Espíritu Santo, Guayaquil 092301, Ecuador
| |
Collapse
|
154
|
Zhang C, Cui X, Feng L, Han Z, Peng D, Fu W, Xing Y. The deficiency of FKBP-5 inhibited hepatocellular progression by increasing the infiltration of distinct immune cells and inhibiting obesity-associated gut microbial metabolite. J Gastrointest Oncol 2021; 12:711-721. [PMID: 34012660 DOI: 10.21037/jgo-21-71] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Gut microbiota has a number of essential roles in nutrition metabolism and immune homeostasis, and is closely related to hepatocellular progression. In recent years, studies have also shown that FK506 binding protein 5 (FKBP-5) plays a crucial role in immune regulation. However, it is not yet clear whether FKBP-5 promotes the development of hepatocellular carcinoma (HCC) by affecting immune function and gut microbiota. Methods FKBP-5 expression was verified by immunochemistry and western blot and reverse transcription polymerase chain reaction (RT-qPCR) assays. After treatment in WT and FKBP-5-/- mice, the histological characteristic of mice liver tissue was assessed by H&E staining, and hepatic leukocytes and hepatic NKT cells were identified by flow cytometer. Meanwhile, primary bile acids (BAs), secondary BAs, serum total cholesterol, and the weight of abdomen adipose tissues were examined, and the gut microbiota was evaluated by 16S ribosomal ribonucleic acid (rRNA) sequencing. Results We discovered that FKBP-5 was highly expressed in HCC tissues. Meanwhile, FKBP-5 deletion inhibited tumor progression by increasing CD8+ T, CD4+ T, NKT and CD4+NKT cells in mice after diethylnitrosamine (DEN) injection. Besides, we proved that FKBP-5 deletion generated rapid and significant reductions in the intestinal BAs, the weight of abdomen adipose tissues and the serum total cholesterol. FKBP-5 deletion also led to a change in the composition of gut microbiota, suggesting that BAs are the main dietary factor regulating gut microbiota, which could be affected by FKBP-5 deletion. Further, we uncovered that anti-CD4 and anti-CD8 treatments facilitated hepatocellular progression by modulating gut microbiota composition in FKBP-5-/- mice. Conclusions Therefore, we demonstrated that FKBP-5 deletion inhibited hepatocellular progression by modulating immune response and gut microbiome-mediated BAs metabolism.
Collapse
Affiliation(s)
- Chuantao Zhang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiang Cui
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Lian Feng
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zhiyi Han
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Deti Peng
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Wenjun Fu
- South China Research Center for Acupuncture and Moxibustion, School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yufeng Xing
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| |
Collapse
|
155
|
Liu AR, Yang SP, Zhang XL. Effects of interaction between mesenchymal stem cells and gut microbiota in treatment of inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2021; 29:312-318. [DOI: 10.11569/wcjd.v29.i6.312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Ai-Ru Liu
- Department of Gastroenterology, the Second Hospital of Hebei Medical University, Shijiazhuang 050035, Hebei Province, China
| | - Shao-Peng Yang
- Department of Gastroenterology, the Second Hospital of Hebei Medical University, Shijiazhuang 050035, Hebei Province, China
| | - Xiao-Lan Zhang
- Department of Gastroenterology, the Second Hospital of Hebei Medical University, Shijiazhuang 050035, Hebei Province, China
| |
Collapse
|
156
|
Kharitonova LA, Osmanov IM, Ploskireva AA, Solodovnikova ON, Milova YE. Digestional diseases in Covid‑19 in children. EXPERIMENTAL AND CLINICAL GASTROENTEROLOGY 2021; 1:53-66. [DOI: 10.31146/1682-8658-ecg-185-1-53-66] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The new coronavirus infection (COVID‑19) caused by the SARS-CoV‑2 coronavirus is rightfully recognized as a pandemic of the 21st century and poses a threat to the health of all human race. According to the World Health Organization, at October 4, 2020, were registered worldwide 34,804,348 confirmed cases of COVID‑19, including 1,030,738 deaths. According to currently available data, children accounted for 1 to 5% of patients diagnosed with COVID‑19 [4]. Children aged between 10–18 years dominated (45.9%), and patients first years of life accounted 26.1%. At the same time, over the period since the beginning of the pandemic, only a few fatal outcomes of the disease in children have been recorded in world statistics.The high contagiousness of the SARS-CoV‑2 coronavirus, the ability for long-term replication, and tropism for the cells of the immune system provide the virus with the possibility of almost simultaneous multiple organ damage with the rapid development of complications of both the respiratory tract and other organs and systems.The gastrointestinal tract is involved in this pathological process, representing one of the “shock organs”. In addition to common respiratory symptoms, some COVID‑19 patients experience dyspeptic disorders such as diarrhea, nausea, and vomiting. Anal swab samples from patients positive for COVID‑19, are positive for SARS-CoV‑2 nucleic acid, and SARS-CoV‑2 were isolated from stool samples from COVID‑19 patients. In addition, in both adults and children, there is a strong association between damage to the digestive system and SARS-CoV‑2 infection. This review summarizes the manifestations and potential mechanisms of gastrointestinal manifestations in COVID‑19 in children.The aim of the work is a review of current data on the lesion of the gastrointestinal tract in children with COVID‑19 infection, and its impact on the management of patients with chronic diseases of the digestive tract.Materials and methods of research. Was carried out the analysis of publications in PubMed, EMBASE and Web of Science systems in the period from December 2019 to June 2020, documenting the clinical course of COVID‑19 disease with gastrointestinal lesions. The following search terms are used: “COVID‑19”, “SARS-CoV‑2”, “coronavirus”, “SARS-CoV‑2 pandemic, epidemic”, “outbreak”, “diarrhea”, “gastrointestinal symptoms”, “stool”, “feces” without any language restrictions. The main source of information was full-text scientific publications of both, foreign and russian authors.In addition, clinical cases of COVID‑19 were analyzed among patients of infectious departments of the Children’s Clinical Hospital named after Z. A. Bashlyaeva, Moscow. Clinical manifestations of the disease, data from laboratory and instrumental studies were evaluated. The most informative of them is given as a clinical example.
Collapse
Affiliation(s)
- L. A. Kharitonova
- Russian National Research Medical University N. I. Pirogov, Ministry of Health of Russia
| | | | - A. A. Ploskireva
- Russian National Research Medical University N. I. Pirogov, Ministry of Health of Russia; Federal Service on Customers’ Rights Protection and Human Well-being Surveillance
| | - O. N. Solodovnikova
- Russian National Research Medical University N. I. Pirogov, Ministry of Health of Russia
| | - Yu. E. Milova
- Children’s Municipal Clinic № 94 Of The Department Of Health of Moscow City
| |
Collapse
|
157
|
|
158
|
Strati F, Pujolassos M, Burrello C, Giuffrè MR, Lattanzi G, Caprioli F, Troisi J, Facciotti F. Antibiotic-associated dysbiosis affects the ability of the gut microbiota to control intestinal inflammation upon fecal microbiota transplantation in experimental colitis models. MICROBIOME 2021; 9:39. [PMID: 33549144 PMCID: PMC7868014 DOI: 10.1186/s40168-020-00991-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/29/2020] [Indexed: 05/12/2023]
Abstract
BACKGROUND The gut microbiota plays a central role in host physiology and in several pathological mechanisms in humans. Antibiotics compromise the composition and functions of the gut microbiota inducing long-lasting detrimental effects on the host. Recent studies suggest that the efficacy of different clinical therapies depends on the action of the gut microbiota. Here, we investigated how different antibiotic treatments affect the ability of the gut microbiota to control intestinal inflammation upon fecal microbiota transplantation in an experimental colitis model and in ex vivo experiments with human intestinal biopsies. RESULTS Murine fecal donors were pre-treated with different antibiotics, i.e., vancomycin, streptomycin, and metronidazole before FMT administration to colitic animals. The analysis of the gut microbiome, fecal metabolome, and the immunophenotyping of colonic lamina propria immune cells revealed that antibiotic pre-treatment significantly influences the capability of the microbiota to control intestinal inflammation. Streptomycin and vancomycin-treated microbiota failed to control intestinal inflammation and were characterized by the blooming of pathobionts previously associated with IBD as well as with metabolites related to the presence of oxidative stress and metabolism of simple sugars. On the contrary, the metronidazole-treated microbiota retained its ability to control inflammation co-occurring with the enrichment of Lactobacillus and of innate immune responses involving iNKT cells. Furthermore, ex vivo cultures of human intestinal lamina propria mononuclear cells and iNKT cell clones from IBD patients with vancomycin pre-treated sterile fecal water showed a Th1/Th17 skewing in CD4+ T-cell populations; metronidazole, on the other hand, induced the polarization of iNKT cells toward the production of IL10. CONCLUSIONS Diverse antibiotic regimens affect the ability of the gut microbiota to control intestinal inflammation in experimental colitis by altering the microbial community structure and microbiota-derived metabolites. Video Abstract.
Collapse
Affiliation(s)
- Francesco Strati
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy.
| | - Meritxell Pujolassos
- Theoreo srl, Spin-off Company of the University of Salerno, Montecorvino Pugliano, Italy
| | - Claudia Burrello
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Maria Rita Giuffrè
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Georgia Lattanzi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Flavio Caprioli
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Jacopo Troisi
- Theoreo srl, Spin-off Company of the University of Salerno, Montecorvino Pugliano, Italy
- European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
| | - Federica Facciotti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy.
| |
Collapse
|
159
|
Battaglia M, Garrett-Sinha LA. Bacterial infections in lupus: Roles in promoting immune activation and in pathogenesis of the disease. J Transl Autoimmun 2020; 4:100078. [PMID: 33490939 PMCID: PMC7804979 DOI: 10.1016/j.jtauto.2020.100078] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Bacterial infections of the lung, skin, bloodstream and other tissues are common in patients with systemic lupus erythematosus (lupus) and are often more severe and invasive than similar infections in control populations. A variety of studies have explored the changes in bacterial abundance in lupus patients, the rates of infection and the influence of particular bacterial species on disease progression, using both human patient samples and mouse models of lupus. OBJECTIVE The aim of this review is to summarize human and mouse studies that describe changes in the bacterial microbiome in lupus, the role of a leaky gut in stimulating inflammation, identification of specific bacterial species associated with lupus, and the potential roles of certain common bacterial infections in promoting lupus progression. METHODS Information was collected using searches of the Pubmed database for articles relevant to bacterial infections in lupus and to microbiome changes associated with lupus. RESULTS The reviewed studies demonstrate significant changes in the bacterial microbiome of lupus patients as compared to control subjects and in lupus-prone mice compared to control mice. Furthermore, there is evidence supporting the existence of a leaky gut in lupus patients and in lupus-prone mice. This leaky gut may allow live bacteria or bacterial components to enter the circulation and cause inflammation. Invasive bacterial infections are more common and often more severe in lupus patients. These include infections caused by Staphylococcus aureus, Salmonella enterica, Escherichia coli, Streptococcus pneumoniae and mycobacteria. These bacterial infections can trigger increased immune activation and inflammation, potentially stimulating activation of autoreactive lymphocytes and leading to worsening of lupus symptoms. CONCLUSIONS Together, the evidence suggests that lupus predisposes to infection, while infection may trigger worsening lupus, leading to a feedback loop that may reinforce autoimmune symptoms.
Collapse
Affiliation(s)
- Michael Battaglia
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Lee Ann Garrett-Sinha
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| |
Collapse
|
160
|
Effects of Antibiotics upon the Gut Microbiome: A Review of the Literature. Biomedicines 2020; 8:biomedicines8110502. [PMID: 33207631 PMCID: PMC7696078 DOI: 10.3390/biomedicines8110502] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/09/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
The human gastrointestinal tract carries a large number of microorganisms associated with complex metabolic processes and interactions. Although antibiotic treatment is crucial for combating infections, its negative effects on the intestinal microbiota and host immunity have been shown to be of the utmost importance. Multiple studies have recognized the adverse consequences of antibiotic use upon the gut microbiome in adults and neonates, causing dysbiosis of the microbiota. Repeated antibiotic treatments in clinical care or low-dosage intake from food could be contributing factors in this issue. Researchers in both human and animal studies have strived to explain this multifaceted relationship. The present review intends to elucidate the axis of the gastrointestinal microbiota and antibiotics resistance and to highlight the main aspects of the issue.
Collapse
|
161
|
Wang F, Zheng S, Zheng C, Sun X. Attaching clinical significance to COVID-19-associated diarrhea. Life Sci 2020; 260:118312. [PMID: 32846165 PMCID: PMC7443214 DOI: 10.1016/j.lfs.2020.118312] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 02/07/2023]
Abstract
The Corona Virus Disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), erupted in 2020 and created severe public health and socioeconomic challenges worldwide. A subset of patients, in addition to presenting with typical features such as fever, cough and dyspnea, was also afflicted with diarrhea. However, the clinical features and prognoses related to COVID-19-associated diarrhea have not attracted sufficient attention. This review of the medical literature examines the incidence, pathogenesis, clinical characteristics, fecal virus changes, prognoses and influencing factors of COVID-19-associated diarrhea. The reported incidence of diarrhea in patients with COVID-19 ranged from 2% to 49.5%. The main cause of diarrhea was found to be invasive by SARS-CoV-2 of ACE-2-expressing epithelial cells of the small intestine, causing local intestinal damage. This cellular invasion may be the key factor for the much longer duration of SARS-CoV-2 positivity observed for feces compared to pharyngeal swabs. The associated diarrhea in these patients upsets the balance of intestinal flora, resulting in more-severe disease intensity and worse prognosis. Clinicians should be vigilant to this kind of COVID-19-associated diarrhea, and design more effective prevention and treatment options for patients with positive fecal nucleic acid tests and intestinal microflora disorders.
Collapse
Affiliation(s)
- Fantao Wang
- Department of Stomatology, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Shiliang Zheng
- Department of General medicine, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Chengbin Zheng
- School of Information Science and Engineering, Qufu Normal University, Rizhao, China
| | - Xiaodong Sun
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China.
| |
Collapse
|
162
|
Resistant Maltodextrin Alleviates Dextran Sulfate Sodium-Induced Intestinal Inflammatory Injury by Increasing Butyric Acid to Inhibit Proinflammatory Cytokine Levels. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7694734. [PMID: 33015180 PMCID: PMC7519446 DOI: 10.1155/2020/7694734] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/28/2020] [Accepted: 09/08/2020] [Indexed: 12/16/2022]
Abstract
Inflammatory bowel disease (IBD), one kind of intestinal chronic inflammatory disease, is characterized by colonic epithelial barrier injury, overproduction of proinflammatory cytokines, and fewer short-chain fatty acids (SCFAs). The present study is aimed at testing the hypothesis that resistant maltodextrin (RM), a soluble dietary fiber produced by starch debranching, alleviated dextran sulfate sodium- (DSS-) induced colitis in mice. Female C57BL/6 mice with or without oral administration of 50 mg/kg RM for 19 days were challenged with 3% DSS in drinking water to induce colitis (from day 14 to day 19). Although RM could not reverse DSS-induced weight loss or colon shortening, it reduced inflammatory cell infiltration and epithelial damage in colon tissue, as well as the transfer of intestinal permeability indicators including serum diamine oxidase (DAO) and D-lactic acid (D-LA). ELISA analysis indicated that RM significantly suppressed the increase of Th1 cytokines induced by DSS in the colon such as tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ). The levels of proinflammatory cytokines interleukin-1β (IL-1β), IL-17, and IL-8 in the DSS group were significantly higher than those in the control group and RM group, but no significant difference was observed in the RM-DSS group compared with the RM group. Interestingly, IL-10 levels of the DSS group were significantly higher than those of the other groups. With respect to SCFAs, DSS administration significantly decreased the concentration of faecal butyric acid while the RM-DSS group showed a tendency to increase (P = 0.08). In general, RM alleviated dextran sulfate sodium-induced intestinal inflammation through increasing the level of butyric acid and subsequently inhibiting the expression of proinflammatory cytokines.
Collapse
|
163
|
Perillo F, Amoroso C, Strati F, Giuffrè MR, Díaz-Basabe A, Lattanzi G, Facciotti F. Gut Microbiota Manipulation as a Tool for Colorectal Cancer Management: Recent Advances in Its Use for Therapeutic Purposes. Int J Mol Sci 2020; 21:E5389. [PMID: 32751239 PMCID: PMC7432108 DOI: 10.3390/ijms21155389] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is a multifaceted disease influenced by both environmental and genetic factors. A large body of literature has demonstrated the role of gut microbes in promoting inflammatory responses, creating a suitable microenvironment for the development of skewed interactions between the host and the gut microbiota and cancer initiation. Even if surgery is the primary therapeutic strategy, patients with advanced disease or cancer recurrence after surgery remain difficult to cure. Therefore, the gut microbiota has been proposed as a novel therapeutic target in light of recent promising data in which it seems to modulate the response to cancer immunotherapy. The use of microbe-targeted therapies, including antibiotics, prebiotics, live biotherapeutics, and fecal microbiota transplantation, is therefore considered to support current therapies in CRC management. In this review, we will discuss the importance of host-microbe interactions in CRC and how promoting homeostatic immune responses through microbe-targeted therapies may be useful in preventing/treating CRC development.
Collapse
Affiliation(s)
- Federica Perillo
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
| | - Chiara Amoroso
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
| | - Francesco Strati
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
| | - Maria Rita Giuffrè
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
| | - Angélica Díaz-Basabe
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20135 Milan, Italy
| | - Georgia Lattanzi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20135 Milan, Italy
| | - Federica Facciotti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
| |
Collapse
|