1
|
You X, Niu L, Fu J, Ge S, Shi J, Zhang Y, Zhuang P. Bidirectional regulation of the brain-gut-microbiota axis following traumatic brain injury. Neural Regen Res 2025; 20:2153-2168. [PMID: 39359076 PMCID: PMC11759007 DOI: 10.4103/nrr.nrr-d-24-00088] [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: 01/23/2023] [Revised: 03/20/2024] [Accepted: 05/11/2024] [Indexed: 10/04/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202508000-00002/figure1/v/2024-09-30T120553Z/r/image-tiff Traumatic brain injury is a prevalent disorder of the central nervous system. In addition to primary brain parenchymal damage, the enduring biological consequences of traumatic brain injury pose long-term risks for patients with traumatic brain injury; however, the underlying pathogenesis remains unclear, and effective intervention methods are lacking. Intestinal dysfunction is a significant consequence of traumatic brain injury. Being the most densely innervated peripheral tissue in the body, the gut possesses multiple pathways for the establishment of a bidirectional "brain-gut axis" with the central nervous system. The gut harbors a vast microbial community, and alterations of the gut niche contribute to the progression of traumatic brain injury and its unfavorable prognosis through neuronal, hormonal, and immune pathways. A comprehensive understanding of microbiota-mediated peripheral neuroimmunomodulation mechanisms is needed to enhance treatment strategies for traumatic brain injury and its associated complications. We comprehensively reviewed alterations in the gut microecological environment following traumatic brain injury, with a specific focus on the complex biological processes of peripheral nerves, immunity, and microbes triggered by traumatic brain injury, encompassing autonomic dysfunction, neuroendocrine disturbances, peripheral immunosuppression, increased intestinal barrier permeability, compromised responses of sensory nerves to microorganisms, and potential effector nuclei in the central nervous system influenced by gut microbiota. Additionally, we reviewed the mechanisms underlying secondary biological injury and the dynamic pathological responses that occur following injury to enhance our current understanding of how peripheral pathways impact the outcome of patients with traumatic brain injury. This review aimed to propose a conceptual model for future risk assessment of central nervous system-related diseases while elucidating novel insights into the bidirectional effects of the "brain-gut-microbiota axis."
Collapse
Affiliation(s)
- Xinyu You
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Niu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiafeng Fu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shining Ge
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiangwei Shi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yanjun Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Pengwei Zhuang
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
2
|
Yang S, Duan H, Yan Z, Xue C, Niu T, Cheng W, Zhang Y, Zhao X, Hu J, Zhang L. Luteolin Alleviates Ulcerative Colitis in Mice by Modulating Gut Microbiota and Plasma Metabolism. Nutrients 2025; 17:203. [PMID: 39861331 DOI: 10.3390/nu17020203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Ulcerative colitis (UC) is a chronic and easily recurrent inflammatory bowel disease. The gut microbiota and plasma metabolites play pivotal roles in the development and progression of UC. Therefore, therapeutic strategies targeting the intestinal flora or plasma metabolites offer promising avenues for the treatment of UC. Luteolin (Lut), originating from a variety of vegetables and fruits, has attracted attention for its potent anti-inflammatory properties and potential to modulate intestinal flora. METHODS The therapeutic efficacy of Lut was evaluated in an established dextran sodium sulfate (DSS)-induced colitis mice model. The clinical symptoms were analyzed, and biological samples were collected for microscopic examination and the evaluation of the epithelial barrier function, microbiome, and metabolomics. RESULTS The findings revealed that Lut administration at a dose of 25 mg/kg significantly ameliorated systemic UC symptoms in mice, effectively reduced the systemic inflammatory response, and significantly repaired colonic barrier function. Furthermore, Lut supplementation mitigated gut microbiota dysbiosis in a UC murine model, increasing the abundance of Muribaculaceae, Rikenella, and Prevotellaceae while decreasing Escherichia_Shigella and Bacteroides levels. These alterations in gut microbiota also influenced plasma metabolism, significantly increasing phosphatidylcholine (PC), 6'-Deamino- 6'-hydroxyneomycin C, and gamma-L-glutamyl-butyrosine B levels and decreasing Motapizone and Arachidoyl-Ethanolamide (AEA) levels. CONCLUSIONS This study reveals that Lut supplementation modulates intestinal inflammation by restoring the gut microbiota community structure, thereby altering the synthesis of inflammation-related metabolites. Lut is a potential nutritional supplement with anti-inflammatory properties and offers a novel alternative for UC intervention and mitigation. In addition, further studies are needed to ascertain whether specific microbial communities or metabolites can mediate the recovery from UC.
Collapse
Affiliation(s)
- Shuai Yang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Hongwei Duan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Zhenxing Yan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Chen Xue
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Tian Niu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Wenjing Cheng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Yong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Xingxu Zhao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Junjie Hu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Lihong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| |
Collapse
|
3
|
Chen Y, Ding H, Wang Q, Huang Z, Zhang C, Li W, Lin Y, Guo Y, Fang X, Zhang W. Can "LITE" Procedure Combined With a Short Course Antibiotic Treatment Be Effective in Treating the Chronic PJI?-A Prospective Randomized Controlled Trial. Orthop Surg 2025; 17:94-104. [PMID: 39428209 PMCID: PMC11735367 DOI: 10.1111/os.14262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 09/08/2024] [Accepted: 09/18/2024] [Indexed: 10/22/2024] Open
Abstract
OBJECTIVE The prevailing treatment for chronic periprosthetic joint infection (PJI) is a two-stage exchange, yet the optimal duration of antibiotic therapy following this procedure remains a topic of debate. This study aimed to determine whether a short course of postoperative antibiotic therapy can maintain infection control rates following a long interval two-stage exchange (LITE) for PJI. METHODS We conducted a prospective study enrolling patients with chronic PJI who underwent the LITE procedure at our institution from April 2018 to November 2021. Patients were randomly assigned to receive either a long course (12 weeks) or short course (2 weeks) of postoperative antibiotics. The pathogens, antibiotics, inflammatory markers, antibiotic-related complications, cases of reinfection, or re-operation were recorded. Continuous variables were analyzed using the two-sample t-test or Mann-Whitney U test, and categorical variables were analyzed using Fisher's exact tests. Kaplan-Meier survival analysis was used to compare infection control rates. RESULTS A total of 60 patients with chronic PJI who completed the LITE procedure were included in the study (30 patients per group). All patients were followed for a minimum of 24 months (mean 39.2 ± 13.0 months). We observed that the infection control rate in the short-course group was not inferior to that in the long-course group (96.7% vs. 96.7%, p = 1.000). CONCLUSIONS For patients with chronic PJI undergoing the LITE procedure, a 2-week course of postoperative antibiotics suffices to maintain infection control rates. TRIAL REGISTRATION Chinese Clinical Trial Registry: ChiCTR1900027089.
Collapse
Affiliation(s)
- Yang Chen
- Department of Orthopaedic SurgeryNational Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhouChina
- Fujian Provincial Institute of OrthopedicsThe First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
| | - Haiqi Ding
- Department of Orthopaedic SurgeryNational Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhouChina
- Fujian Provincial Institute of OrthopedicsThe First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
| | - Qijin Wang
- Department of Orthopaedic SurgeryNational Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhouChina
- Fujian Provincial Institute of OrthopedicsThe First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
| | - Zida Huang
- Department of Orthopaedic SurgeryNational Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhouChina
- Fujian Provincial Institute of OrthopedicsThe First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
| | - Chaofan Zhang
- Department of Orthopaedic SurgeryNational Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhouChina
- Fujian Provincial Institute of OrthopedicsThe First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
| | - Wenbo Li
- Department of Orthopaedic SurgeryNational Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhouChina
- Fujian Provincial Institute of OrthopedicsThe First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
| | - Yansheng Lin
- Department of Orthopedic SurgeryChangtai County HospitalZhangzhouChina
| | - Yufeng Guo
- Department of Orthopedic SurgeryChangtai County HospitalZhangzhouChina
| | - Xinyu Fang
- Department of Orthopaedic SurgeryNational Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhouChina
- Fujian Provincial Institute of OrthopedicsThe First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
| | - Wenming Zhang
- Department of Orthopaedic SurgeryNational Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhouChina
- Fujian Provincial Institute of OrthopedicsThe First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
| |
Collapse
|
4
|
Hsu FC, Weng TH, Pu TW, Chang PK, Lin TC, Jao SW, Chen CY, Hu JM, Chien WC. Infectious intestinal diseases elevate neurodegenerative disease risk based on a nationwide population-based cohort study. Sci Rep 2024; 14:30968. [PMID: 39730770 DOI: 10.1038/s41598-024-81950-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 12/02/2024] [Indexed: 12/29/2024] Open
Abstract
Infectious intestinal diseases (IIDs) pose a significant health and economic burden worldwide. Recent observations at the Tri-Service General Hospital, Taiwan, suggest a potential association between IIDs and neurodegenerative diseases, prompting an investigation into this relationship. This study explored interactions between IIDs and neurodegenerative diseases. We conducted a population-based retrospective cohort analysis using data from the National Health Insurance Research Database (NHIRD) of Taiwan. Patients diagnosed with IIDs between 2000 and 2015 were identified along with a matched control group. Covariates, including demographics, comorbidities, and healthcare utilization were considered. The hazard ratios (HRs) of neurodegenerative diseases were assessed using a Cox proportional regression analysis. This study included 297,438 patients: 99,146 and 198,292 patients in the IID and control groups, respectively. Patients with IIDs showed a significantly higher overall risk of neurodegenerative diseases (adjusted hazard ratio [aHR] = 1.144, P < 0.001). Subgroup analyses revealed an elevated risk of Parkinson's disease, multiple sclerosis, and other neurodegeneration-associated disorders in the study group. Additionally, a positive correlation was observed between the frequency of medical visits for IIDs and neurodegenerative disease risk. This study provides evidence for a significant association between IIDs and the neurodegenerative disease risk. Early detection and management of IIDs may have implications for long-term neurological health outcomes. Further research is required to elucidate underlying mechanisms and develop targeted interventions and preventive strategies.
Collapse
Affiliation(s)
- Fang-Chin Hsu
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Tzu-Hsuan Weng
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ta-Wei Pu
- Division of Colon and Rectal Surgery, Department of Surgery, Songshan Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Pi-Kai Chang
- Division of Colorectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Tzu-Chiao Lin
- Division of Colorectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Wen Jao
- Division of Colorectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chao-Yang Chen
- Division of Colorectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Je-Ming Hu
- Division of Colorectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Wu-Chien Chien
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
- Taiwanese Injury Prevention and Safety Promotion Association, Taipei, Taiwan.
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, Republic of China.
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
5
|
Seo H, Kim S, Beck S, Song HY. Perspectives on Microbiome Therapeutics in Infectious Diseases: A Comprehensive Approach Beyond Immunology and Microbiology. Cells 2024; 13:2003. [PMID: 39682751 DOI: 10.3390/cells13232003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Although global life expectancy has increased over the past 20 years due to advancements in managing infectious diseases, one-fifth of people still die from infections. In response to this ongoing threat, significant efforts are underway to develop vaccines and antimicrobial agents. However, pathogens evolve resistance mechanisms, complicating their control. The COVID-19 pandemic has underscored the limitations of focusing solely on the pathogen-killing strategies of immunology and microbiology to address complex, multisystemic infectious diseases. This highlights the urgent need for practical advancements, such as microbiome therapeutics, that address these limitations while complementing traditional approaches. Our review emphasizes key outcomes in the field, including evidence of probiotics reducing disease severity and insights into host-microbiome crosstalk that have informed novel therapeutic strategies. These findings underscore the potential of microbiome-based interventions to promote physiological function alongside existing strategies aimed at enhancing host immune responses and pathogen destruction. This narrative review explores microbiome therapeutics as next-generation treatments for infectious diseases, focusing on the application of probiotics and their role in host-microbiome interactions. While offering a novel perspective grounded in a cooperative defense system, this review also addresses the practical challenges and limitations in translating these advancements into clinical settings.
Collapse
Affiliation(s)
- Hoonhee Seo
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Chungnam-do, Republic of Korea
| | - Sukyung Kim
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Chungnam-do, Republic of Korea
| | - Samuel Beck
- Center for Aging Research, Department of Dermatology, Chobanian & Avedisian School of Medicine, Boston University, J-607, 609 Albany, Boston, MA 02118, USA
| | - Ho-Yeon Song
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Chungnam-do, Republic of Korea
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Cheonan-si 31151, Chungnam-do, Republic of Korea
| |
Collapse
|
6
|
Beldie LA, Dica CC, Moța M, Pirvu BF, Burticală MA, Mitrea A, Clenciu D, Efrem IC, Vladu BE, Timofticiuc DCP, Roșu MM, Gheonea TC, Amzolini AM, Moța E, Vladu IM. The Interactions Between Diet and Gut Microbiota in Preventing Gestational Diabetes Mellitus: A Narrative Review. Nutrients 2024; 16:4131. [PMID: 39683525 DOI: 10.3390/nu16234131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Recent studies have revealed that dysbiosis, defined as alterations in gut microbiota, plays an important role in the development and the progression of many non-communicable diseases, including metabolic disorders, such as type 2 diabetes mellitus and gestational diabetes mellitus (GDM). The high frequency of GDM makes this disorder an important public health issue, which needs to be addressed in order to reduce both the maternal and fetal complications that are frequently associated with this disease. The studies regarding the connections between gut dysbiosis and GDM are still in their early days, with new research continuously emerging. This narrative review seeks to outline the mechanisms through which a healthy diet that protects the gut microbiota is able to prevent the occurrence of GDM, thus providing medical nutritional therapeutic perspectives for the management of GDM.
Collapse
Affiliation(s)
- Luiza-Andreea Beldie
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Cristina-Camelia Dica
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Maria Moța
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Bianca-Florentina Pirvu
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Marilena-Alexandra Burticală
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Adina Mitrea
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Diana Clenciu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ion Cristian Efrem
- Department of Medical Semiology, Faculty of Dentistry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Beatrice Elena Vladu
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Diana Cristina Protasiewicz Timofticiuc
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Midwives and Nursing, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Maria Magdalena Roșu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Midwives and Nursing, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Theodora Claudia Gheonea
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Anca Maria Amzolini
- Department of Medical Semiology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Eugen Moța
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ionela Mihaela Vladu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
7
|
Wu Q, Kan J, Fu C, Liu X, Cui Z, Wang S, Le Y, Li Z, Liu Q, Zhang Y, Du J. Insights into the unique roles of extracellular vesicles for gut health modulation: Mechanisms, challenges, and perspectives. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 7:100301. [PMID: 39525958 PMCID: PMC11550031 DOI: 10.1016/j.crmicr.2024.100301] [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] [Indexed: 11/16/2024] Open
Abstract
Extracellular vesicles (EVs), which play significant regulatory roles in maintaining homeostasis and influencing immune responses, significantly impact gut microbiota composition and function, affecting overall gut health. Despite considerable progress, there are still knowledge gaps regarding the mechanisms by which EVs, including plant-derived EVs (PDEVs), animal-derived EVs (ADEVs), and microbiota-derived EVs (MDEVs), modulate gut health. This review delves into the roles and mechanisms of EVs from diverse sources in regulating gut health, focusing on their contributions to maintaining epithelial barrier integrity, facilitating tissue healing, eliciting immune responses, controlling pathogens, and shaping microbiota. We emphasize open challenges and future perspectives for harnessing EVs in the modulation of gut health to gain a deeper understanding of their roles and impact. Importantly, a comprehensive research framework is presented to steer future investigations into the roles and implications of EVs on gut health, facilitating a more profound comprehension of this emerging field.
Collapse
Affiliation(s)
- Qiming Wu
- Nutrilite Health Institute, Shanghai 200031, China
| | - Juntao Kan
- Nutrilite Health Institute, Shanghai 200031, China
| | - Caili Fu
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Xin Liu
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Zhengying Cui
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Sixu Wang
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Yi Le
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Zhanming Li
- Department of Food Quality and Safety, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Qin Liu
- Centre for Chinese Medicine Drug Development Limited, Hong Kong Baptist University, 999077, Hong Kong Special Administrative Region of China
| | - Yuyu Zhang
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Jun Du
- Nutrilite Health Institute, Shanghai 200031, China
| |
Collapse
|
8
|
Choudhury D, Alanbari R, Saveliev P, Sokurenko E, Fuzi M, Tchesnokova V. Clonal and resistance profiles of fluoroquinolone-resistant uropathogenic Escherichia coli in countries with different practices of antibiotic prescription. Front Microbiol 2024; 15:1446818. [PMID: 39417079 PMCID: PMC11479919 DOI: 10.3389/fmicb.2024.1446818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Background Antibiotic prescription practices differ between countries, influencing regional antimicrobial resistance prevalence. However, comparisons of clonal diversity among resistant bacteria in countries with different prescribing practices are rare. The rise of fluoroquinolone-resistant Escherichia coli (FQREC), often multidrug-resistant, exacerbates global antibiotic resistance. Unlike in the USA, antibiotics are commonly dispensed in Iraq without prescriptions, leading to widespread overuse and misuse. This study aimed to assess the impact of varying antibiotic use practices on FQREC diversity. Methods We compared FQREC prevalence, multidrug resistance, and clonality of FQREC among E. coli isolated from urine submitted between 2017 and 2018 to three US hospitals and two Iraqi hospitals. All FQREC isolates were analyzed for QRDR mutations and the presence of PMQR genes. A subset of FQREC strains from the ST131-H30R/Rx subgroups underwent whole-genome sequencing (WGS) and phylogenetic analysis. Results E. coli from Iraq showed significantly higher resistance to all tested antibiotics compared to those from the USA, with 76.2% being FQREC versus 31.2% in the USA (p < 0.01). Iraqi FQREC strains were more frequently multidrug resistant. The predominant subgroup in both countries was ST131-H30, with the notable absence of ST1193 among Iraqi FQREC. Iraqi-origin ST131-H30 strains exhibited higher minimum inhibitory concentrations (MICs) for ciprofloxacin and greater resistance to third-generation cephalosporins (3GC), trimethoprim/sulfamethoxazole (TMP/STX), and imipenem (IMI) than those from the USA. Increased 3GC resistance in Iraqi strains was linked to a higher proportion of bla CTX-M-15-carrying H30Rx subclade isolates. Additionally, Iraqi H30 strains exhibited higher MICs for fluoroquinolones due to more frequent carriage of PMQR determinants compared to US strains. Whole-genome sequencing was performed on 46 Iraqi and 63 US H30 isolates. Phylogenetic analysis revealed two clades-H30R and H30Rx-present in both countries, with isolates from both regions distributed throughout, without the emergence of distinct new major subclones. However, Iraqi isolates tended to cluster in separate subclades, indicating endemic circulation of the strain groups. Conclusion In regions like Iraq, where antibiotics are overused and misused, resistance among uropathogenic E. coli to various antibiotics is significantly higher. Most Iraqi resistant strains belong to well-known international groups, and no new highly successful strains have emerged. The absence of ST1193 in Iraq may reflect regional, socioeconomic, demographic, or cultural factors that hinder the success of certain strain groups in the country.
Collapse
Affiliation(s)
- Debarati Choudhury
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA, United States
| | - Rawan Alanbari
- Department of Microbiology, Al-Mustansiriyah University, College of Medicine, Baghdad, Iraq
| | - Pauline Saveliev
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, United States
| | - Evgeni Sokurenko
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA, United States
| | - Miklos Fuzi
- Independent Researcher, Seattle, WA, United States
| | - Veronika Tchesnokova
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
9
|
Tonog G, Yu H, Moon SK, Lee S, Jeong H, Kim HS, Kim KB, Suh HJ, Kim H. Garlic Bioconverted by Bacillus subtilis Stimulates the Intestinal Immune System and Modulates Gut Microbiota Composition. Mol Nutr Food Res 2024; 68:e2400504. [PMID: 39358948 DOI: 10.1002/mnfr.202400504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/30/2024] [Indexed: 10/04/2024]
Abstract
SCOPE This study evaluates the potential of bioconverted garlic ferments (BGFs) to stimulate the intestinal immune system and modulate cecal microbiota composition. METHODS AND RESULTS In vitro, BGF significantly enhances Peyer's patch (PP)-mediated bone marrow cell proliferation and increases the production of interferon-gamma (IFN-γ), granulocyte macrophage-colony stimulating factor (GM-CSF), interleukin (IL)-6, and immunoglobulin A (IgA) but not IL-4, IL-5, and immunoglobulin E (IgE). Oral administration of BGF to C3H/HeN mice for 4 weeks significantly increases the GM-CSF (42.1-45.8 pg mL-1) and IFN-γ (6.5-12.1 pg mL-1) levels in PP cells. BGF also significantly elevates the levels of tumor necrosis factor-alpha (TNF-α, 165.0-236.3 pg mg-1), GM-CSF (2.4-3.0 ng mg-1), and IFN-γ (1.5-3.2 ng mg-1) in the small intestinal fluid, and TNF-α (2.2-3.1 pg mL-1) and IFN-γ (10.3-0.21.5 pg mL-1) in the mouse serum. Cecal microbial analysis reveals that BGF increases Bacteroidota and Verrucomicrobiota and decreases Actinobacteria and Bacillota at the phylum level in mice. At the genus level, BGF significantly increases the abundance of Fusimonas (250 mg kg-1 BW-1 day-1), Bacteroides (125 and 250 mg kg-1 BW-1 day-1), and Akkermansia (125 mg kg-1 BW-1 day-1) and decreases that of Bifidobacterium (62.5 and 250 mg kg-1 BW-1 day-1) and Limosilactobacillus (125 and 250 mg kg-1 BW-1 day-1). CONCLUSION This study provides the first evidence of BGF's ability to modulate the intestinal immune system and gut microbiota, supporting its potential as a novel functional material to enhance gut immunity.
Collapse
Affiliation(s)
- Genevieve Tonog
- Department of Food and Nutrition, Chung-Ang University, Anseong, 17546, South Korea
| | - Hyeonjun Yu
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, 02841, South Korea
| | - Sung-Kwon Moon
- Department of Food and Nutrition, Chung-Ang University, Anseong, 17546, South Korea
| | - Sanghyun Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong, 17546, South Korea
| | | | | | | | - Hyung Joo Suh
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, 02841, South Korea
| | - Hoon Kim
- Department of Food and Nutrition, Chung-Ang University, Anseong, 17546, South Korea
| |
Collapse
|
10
|
Munteanu C, Schwartz B. Interactions between Dietary Antioxidants, Dietary Fiber and the Gut Microbiome: Their Putative Role in Inflammation and Cancer. Int J Mol Sci 2024; 25:8250. [PMID: 39125822 PMCID: PMC11311432 DOI: 10.3390/ijms25158250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
The intricate relationship between the gastrointestinal (GI) microbiome and the progression of chronic non-communicable diseases underscores the significance of developing strategies to modulate the GI microbiota for promoting human health. The administration of probiotics and prebiotics represents a good strategy that enhances the population of beneficial bacteria in the intestinal lumen post-consumption, which has a positive impact on human health. In addition, dietary fibers serve as a significant energy source for bacteria inhabiting the cecum and colon. Research articles and reviews sourced from various global databases were systematically analyzed using specific phrases and keywords to investigate these relationships. There is a clear association between dietary fiber intake and improved colon function, gut motility, and reduced colorectal cancer (CRC) risk. Moreover, the state of health is reflected in the reciprocal and bidirectional relationships among food, dietary antioxidants, inflammation, and body composition. They are known for their antioxidant properties and their ability to inhibit angiogenesis, metastasis, and cell proliferation. Additionally, they promote cell survival, modulate immune and inflammatory responses, and inactivate pro-carcinogens. These actions collectively contribute to their role in cancer prevention. In different investigations, antioxidant supplements containing vitamins have been shown to lower the risk of specific cancer types. In contrast, some evidence suggests that taking antioxidant supplements can increase the risk of developing cancer. Ultimately, collaborative efforts among immunologists, clinicians, nutritionists, and dietitians are imperative for designing well-structured nutritional trials to corroborate the clinical efficacy of dietary therapy in managing inflammation and preventing carcinogenesis. This review seeks to explore the interrelationships among dietary antioxidants, dietary fiber, and the gut microbiome, with a particular focus on their potential implications in inflammation and cancer.
Collapse
Affiliation(s)
- Camelia Munteanu
- Department of Plant Culture, Faculty of Agriculture, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| | - Betty Schwartz
- The Institute of Biochemistry, Food Science and Nutrition, The School of Nutritional Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| |
Collapse
|
11
|
Wielgosz-Grochowska JP, Domanski N, Drywień ME. Identification of SIBO Subtypes along with Nutritional Status and Diet as Key Elements of SIBO Therapy. Int J Mol Sci 2024; 25:7341. [PMID: 39000446 PMCID: PMC11242202 DOI: 10.3390/ijms25137341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
Small intestinal bacterial overgrowth (SIBO) is a pathology of the small intestine and may predispose individuals to various nutritional deficiencies. Little is known about whether specific subtypes of SIBO, such as the hydrogen-dominant (H+), methane-dominant (M+), or hydrogen/methane-dominant (H+/M+), impact nutritional status and dietary intake in SIBO patients. The aim of this study was to investigate possible correlations between biochemical parameters, dietary nutrient intake, and distinct SIBO subtypes. This observational study included 67 patients who were newly diagnosed with SIBO. Biochemical parameters and diet were studied utilizing laboratory tests and food records, respectively. The H+/M+ group was associated with low serum vitamin D (p < 0.001), low serum ferritin (p = 0.001) and low fiber intake (p = 0.001). The M+ group was correlated with high serum folic acid (p = 0.002) and low intakes of fiber (p = 0.001) and lactose (p = 0.002). The H+ group was associated with low lactose intake (p = 0.027). These results suggest that the subtype of SIBO may have varying effects on dietary intake, leading to a range of biochemical deficiencies. Conversely, specific dietary patterns may predispose one to the development of a SIBO subtype. The assessment of nutritional status and diet, along with the diagnosis of SIBO subtypes, are believed to be key components of SIBO therapy.
Collapse
Affiliation(s)
| | - Nicole Domanski
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada;
| | - Małgorzata Ewa Drywień
- Department of Human Nutrition, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences, 02-776 Warsaw, Poland;
| |
Collapse
|
12
|
Goldfarb M, Gollin G. The Impact of Antibiotic Strategy on Outcomes in Surgically Managed Necrotizing Enterocolitis. J Pediatr Surg 2024; 59:1266-1270. [PMID: 38561306 DOI: 10.1016/j.jpedsurg.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/01/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND We sought to evaluate postoperative antibiotic practices in a large population of patients with necrotizing enterocolitis (NEC) and determine whether any regimens were associated with better outcomes. METHODS The Pediatric Health Information Systems (PHIS) database was queried to identify patients who underwent an intestinal resection for acute NEC between July, 2016 and June, 2021. Data regarding post-resection antibiotic therapy, cutaneous or intraabdominal infection, and fungal or antibiotic-resistant infection were collected. RESULTS 130 infants at 38 children's hospitals met inclusion criteria. Postoperative antibiotics were administered for a median of 13 days. The most frequently used antibiotics were vancomycin and piperacillin/tazobactam. Antibiotic duration greater than five days was not associated with a lower incidence of infection. No antibiotic was associated with a lower incidence of any of the complications assessed, although ampicillin was associated with more infections, overall. The incidence of fungal infection and treatment with a parenteral anti-fungal medication was greater with vancomycin. No antibiotic combination was used enough to be assessed. CONCLUSIONS Administration of antibiotics for more than five days after resection for NEC was not associated with better infectious outcomes and no single antibiotic demonstrated superior efficacy. Consistent with prior studies, fungal infections were more frequent with vancomycin. TYPE OF STUDY Retrospective database study, level 3B. LEVEL OF EVIDENCE II.
Collapse
Affiliation(s)
- Madeline Goldfarb
- Texas Tech Health Sciences Center El Paso, Paul L. Foster School of Medicine, El Paso, TX, USA
| | - Gerald Gollin
- Rady Children's Hospital, San Diego and University of California San Diego School of Medicine, San Diego, CA, USA.
| |
Collapse
|
13
|
Partipilo G, Bowman EK, Palmer EJ, Gao Y, Ridley RS, Alper HS, Keitz BK. Single-Cell Phenotyping of Extracellular Electron Transfer via Microdroplet Encapsulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.13.598847. [PMID: 38915652 PMCID: PMC11195189 DOI: 10.1101/2024.06.13.598847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Electroactive organisms contribute to metal cycling, pollutant removal, and other redox-driven environmental processes. Studying this phenomenon in high-throughput is challenging since extracellular reduction cannot easily be traced back to its cell of origin within a mixed population. Here, we describe the development of a microdroplet emulsion system to enrich EET-capable organisms. We validated our system using the model electroactive organism S. oneidensis and describe the tooling of a benchtop microfluidic system for oxygen-limited processes. We demonstrated enrichment of EET-capable phenotypes from a mixed wild-type and EET-knockout population. As a proof-of-concept application, bacteria were collected from iron sedimentation from Town Lake (Austin, TX) and subjected to microdroplet enrichment. We observed an increase in EET-capable organisms in the sorted population that was distinct when compared to a population enriched in a bulk culture more closely akin to traditional techniques for discovering EET-capable bacteria. Finally, two bacterial species, C. sakazakii and V. fessus not previously shown to be electroactive, were further cultured and characterized for their ability to reduce channel conductance in an organic electrochemical transistor (OECT) and to reduce soluble Fe(III). We characterized two bacterial species not previously shown to exhibit electrogenic behavior. Our results demonstrate the utility of a microdroplet emulsions for identifying putative EET-capable bacteria and how this technology can be leveraged in tandem with existing methods.
Collapse
Affiliation(s)
- Gina Partipilo
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712
| | - Emily K. Bowman
- Interdisciplinary Life Sciences Graduate Program, University of Texas at Austin, Austin, TX, 78712
| | - Emma J. Palmer
- Civil, Architectural, and Environmental Engineering, University of Texas at Austin, Austin, TX, 78712
| | - Yang Gao
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712
| | - Rodney S. Ridley
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712
| | - Hal S. Alper
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712
| | - Benjamin K. Keitz
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712
| |
Collapse
|
14
|
Nasreen S, Ali S, Andleeb S, Summer M, Hussain T, Imdad K, Ara C, Tahir HM. Mechanisms of medicinal, pharmaceutical, and immunomodulatory action of probiotics bacteria and their secondary metabolites against disease management: an overview. Folia Microbiol (Praha) 2024; 69:549-565. [PMID: 38532057 DOI: 10.1007/s12223-024-01155-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 03/05/2024] [Indexed: 03/28/2024]
Abstract
Probiotics or bacteriotherapy is today's hot issue for public entities (Food and Agriculture Organization, and World Health Organization) as well as health and food industries since Metchnikoff and his colleagues hypothesized the correlation between probiotic consumption and human's health. They contribute to the newest and highly efficient arena of promising biotherapeutics. These are usually attractive in biomedical applications such as gut-related diseases like irritable bowel disease, diarrhea, gastrointestinal disorders, fungal infections, various allergies, parasitic and bacterial infections, viral diseases, and intestinal inflammation, and are also worth immunomodulation. The useful impact of probiotics is not limited to gut-related diseases alone. Still, these have proven benefits in various acute and chronic infectious diseases, like cancer, human immunodeficiency virus (HIV) diseases, and high serum cholesterol. Recently, different researchers have paid special attention to investigating biomedical applications of probiotics, but consolidated data regarding bacteriotherapy with a detailed mechanistically applied approach is scarce and controversial. The present article reviews the bio-interface of probiotic strains, mainly (i) why the demand for probiotics?, (ii) the current status of probiotics, (iii) an alternative to antibiotics, (iv) the potential applications towards disease management, (v) probiotics and industrialization, and (vi) futuristic approach.
Collapse
Affiliation(s)
- Sundas Nasreen
- Department of Zoology, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| | - Shaukat Ali
- Department of Zoology, Government College University, Lahore, 54000, Pakistan.
| | - Saiqa Andleeb
- Department of Zoology, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| | - Muhammad Summer
- Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Tauqeer Hussain
- Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Kaleem Imdad
- Department of Bioscience, COMSATS Institute of Information Technology (CIIT), Islamabad, 45550, Pakistan
| | - Chaman Ara
- Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | | |
Collapse
|
15
|
Jurek JM, Castro-Marrero J. A Narrative Review on Gut Microbiome Disturbances and Microbial Preparations in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Implications for Long COVID. Nutrients 2024; 16:1545. [PMID: 38892479 PMCID: PMC11173566 DOI: 10.3390/nu16111545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/10/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Myalgic encephalomyelitis, also known as chronic fatigue syndrome (ME/CFS), and long COVID are complex, multisystemic and long-term disabling conditions characterized by debilitating post-exertional malaise and other core symptoms related to immune dysregulation resultant from post-viral infection, including mitochondrial dysfunction, chronic neuroinflammation and gut dysbiosis. The reported associations between altered microbiota composition and cardinal symptoms of ME/CFS and long COVID suggest that the use of microbial preparations, such as probiotics, by restoring the homeostasis of the brain-immune-gut axis, may help in the management of symptoms in both conditions. Therefore, this review aims to investigate the implications of alerted gut microbiome and assess the evidence supporting use of microbial-based preparations, including probiotics, synbiotics, postbiotics alone and/or in combination with other nutraceuticals in the management of fatigue, inflammation and neuropsychiatric and gastrointestinal symptoms among patients with ME/CFS and long COVID.
Collapse
Affiliation(s)
- Joanna Michalina Jurek
- Unit of Research in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome and Long COVID, Rheumatology Research Division, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
- Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43005 Tarragona, Spain
| | - Jesus Castro-Marrero
- Unit of Research in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome and Long COVID, Rheumatology Research Division, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
| |
Collapse
|
16
|
Ruggles A, Benakis C. Exposure to Environmental Toxins: Potential Implications for Stroke Risk via the Gut- and Lung-Brain Axis. Cells 2024; 13:803. [PMID: 38786027 PMCID: PMC11119296 DOI: 10.3390/cells13100803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/24/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
Recent evidence indicates that exposure to environmental toxins, both short-term and long-term, can increase the risk of developing neurological disorders, including neurodegenerative diseases (i.e., Alzheimer's disease and other dementias) and acute brain injury (i.e., stroke). For stroke, the latest systematic analysis revealed that exposure to ambient particulate matter is the second most frequent stroke risk after high blood pressure. However, preclinical and clinical stroke investigations on the deleterious consequences of environmental pollutants are scarce. This review examines recent evidence of how environmental toxins, absorbed along the digestive tract or inhaled through the lungs, affect the host cellular response. We particularly address the consequences of environmental toxins on the immune response and the microbiome at the gut and lung barrier sites. Additionally, this review highlights findings showing the potential contribution of environmental toxins to an increased risk of stroke. A better understanding of the biological mechanisms underlying exposure to environmental toxins has the potential to mitigate stroke risk and other neurological disorders.
Collapse
Affiliation(s)
| | - Corinne Benakis
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, 81337 Munich, Germany;
| |
Collapse
|
17
|
Santiago MSA, Avellar MCW, Perobelli JE. Could the gut microbiota be capable of making individuals more or less susceptible to environmental toxicants? Toxicology 2024; 503:153751. [PMID: 38354972 DOI: 10.1016/j.tox.2024.153751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/16/2024]
Abstract
Environmental toxicants are chemical substances capable to impair environmental quality and exert adverse effects on humans and other animals. The main routes of exposure to these pollutants are through the respiratory tract, skin, and oral ingestion. When ingested orally, they will encounter trillions of microorganisms that live in a community - the gut microbiota (GM). While pollutants can disrupt the GM balance, GM plays an essential role in the metabolism and bioavailability of these chemical compounds. Under physiological conditions, strategies used by the GM for metabolism and/or excretion of xenobiotics include reductive and hydrolytic transformations, lyase and functional group transfer reactions, and enzyme-mediated functional transformations. Simultaneously, the host performs metabolic processes based mainly on conjugation, oxidation, and hydrolysis reactions. Thus, due to the broad variety of bacterial enzymes present in GM, the repertoire of microbial transformations of chemicals is considered a key component of the machinery involved in the metabolism of pollutants in humans and other mammals. Among pollutants, metals deserve special attention once contamination by metals is a worldwide problem, and their adverse effects can be observed even at very low concentrations due to their toxic properties. In this review, bidirectional interaction between lead, arsenic, cadmium, and mercury and the host organism and its GM will be discussed given the most recent literature, presenting an analysis of the ability of GM to alter the host organism's susceptibility to the toxic effects of heavy metals, as well as evaluating the extent to which interventions targeting the microbiota could be potential initiatives to mitigate the adverse effects resulting from poisoning by heavy metals. This study is the first to highlight the overlap between some of the bacteria found to be altered by metal exposure and the bacteria that also aid the host organism in the metabolism of these metals. This could be a key factor to determine the beneficial species able to minimize the toxicity of metals in future therapeutic approaches.
Collapse
Affiliation(s)
- Marcella S A Santiago
- Laboratory of Experimental Toxicology - LATOEX, Universidade Federal de São Paulo, Instituto do Mar, Carvalho de Mendonça, 144, Santos, SP 11070-100, Brazil
| | - Maria Christina W Avellar
- Department of Pharmacology, Universidade Federal de São Paulo - Escola Paulista de Medicina, Três de Maio, 100, São Paulo, SP 04044-020, Brazil
| | - Juliana E Perobelli
- Laboratory of Experimental Toxicology - LATOEX, Universidade Federal de São Paulo, Instituto do Mar, Carvalho de Mendonça, 144, Santos, SP 11070-100, Brazil.
| |
Collapse
|
18
|
Jernfors T, Lavrinienko A, Vareniuk I, Landberg R, Fristedt R, Tkachenko O, Taskinen S, Tukalenko E, Mappes T, Watts PC. Association between gut health and gut microbiota in a polluted environment. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 914:169804. [PMID: 38184263 DOI: 10.1016/j.scitotenv.2023.169804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/28/2023] [Accepted: 12/29/2023] [Indexed: 01/08/2024]
Abstract
Animals host complex bacterial communities in their gastrointestinal tracts, with which they share a mutualistic interaction. The numerous effects these interactions grant to the host include regulation of the immune system, defense against pathogen invasion, digestion of otherwise undigestible foodstuffs, and impacts on host behaviour. Exposure to stressors, such as environmental pollution, parasites, and/or predators, can alter the composition of the gut microbiome, potentially affecting host-microbiome interactions that can be manifest in the host as, for example, metabolic dysfunction or inflammation. However, whether a change in gut microbiota in wild animals associates with a change in host condition is seldom examined. Thus, we quantified whether wild bank voles inhabiting a polluted environment, areas where there are environmental radionuclides, exhibited a change in gut microbiota (using 16S amplicon sequencing) and concomitant change in host health using a combined approach of transcriptomics, histological staining analyses of colon tissue, and quantification of short-chain fatty acids in faeces and blood. Concomitant with a change in gut microbiota in animals inhabiting contaminated areas, we found evidence of poor gut health in the host, such as hypotrophy of goblet cells and likely weakened mucus layer and related changes in Clca1 and Agr2 gene expression, but no visible inflammation in colon tissue. Through this case study we show that inhabiting a polluted environment can have wide reaching effects on the gut health of affected animals, and that gut health and other host health parameters should be examined together with gut microbiota in ecotoxicological studies.
Collapse
Affiliation(s)
- Toni Jernfors
- Department of Biological and Environmental Science, University of Jyväskylä, FI-40014, Finland.
| | - Anton Lavrinienko
- Department of Biological and Environmental Science, University of Jyväskylä, FI-40014, Finland; Laboratory of Food Systems Biotechnology, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Igor Vareniuk
- Department of Cytology, Histology and Reproductive Medicine, Taras Shevchenko National University of Kyiv, 01033, Ukraine
| | - Rikard Landberg
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
| | - Rikard Fristedt
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
| | - Olena Tkachenko
- Department of Cytology, Histology and Reproductive Medicine, Taras Shevchenko National University of Kyiv, 01033, Ukraine
| | - Sara Taskinen
- Department of Mathematics and Statistics, University of Jyväskylä, FI-40014, Finland
| | - Eugene Tukalenko
- Department of Radiobiology and Radioecology, Institute for Nuclear Research of NAS of Ukraine, 020000, Ukraine
| | - Tapio Mappes
- Department of Biological and Environmental Science, University of Jyväskylä, FI-40014, Finland
| | - Phillip C Watts
- Department of Biological and Environmental Science, University of Jyväskylä, FI-40014, Finland
| |
Collapse
|
19
|
Kumar A, Sivamaruthi BS, Dey S, Kumar Y, Malviya R, Prajapati BG, Chaiyasut C. Probiotics as modulators of gut-brain axis for cognitive development. Front Pharmacol 2024; 15:1348297. [PMID: 38444940 PMCID: PMC10912297 DOI: 10.3389/fphar.2024.1348297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/05/2024] [Indexed: 03/07/2024] Open
Abstract
Various microbial communities reside in the gastrointestinal tract of humans and play an important role in immunity, digestion, drug metabolism, intestinal integrity, and protection from pathogens. Recent studies have revealed that the gut microbiota (GM) is involved in communication with the brain, through a bidirectional communication network known as the gut-brain axis. This communication involves humoral, immunological, endocrine, and neural pathways. Gut dysbiosis negatively impacts these communication pathways, leading to neurological complications and cognitive deficits. Both pre-clinical and clinical studies have demonstrated that probiotics can restore healthy GM, reduce intestinal pH, and reduce inflammation and pathogenic microbes in the gut. Additionally, probiotics improve cell-to-cell signaling and increase blood-brain-derived neurotrophic factors. Probiotics emerge as a potential approach for preventing and managing neurological complications and cognitive deficits. Despite these promising findings, the safety concerns and possible risks of probiotic usage must be closely monitored and addressed. This review article provides a brief overview of the role and significance of probiotics in cognitive health.
Collapse
Affiliation(s)
- Akash Kumar
- Department of Food Technology, SRM University, Sonipat, Delhi, India
| | - Bhagavathi Sundaram Sivamaruthi
- Office of Research Administration, Chiang Mai University, Chiang Mai, Thailand
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
| | - Swarnima Dey
- Department of Food Technology, SRM University, Sonipat, Delhi, India
- Amity Institute of Food Technology, Amity University, Noida, Uttar Pradesh, India
| | - Yogesh Kumar
- Department of Food Technology, SRM University, Sonipat, Delhi, India
| | - Rishabha Malviya
- Department of Paramedical and Allied Sciences, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Bhupendra G. Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Mehsana, India
| | - Chaiyavat Chaiyasut
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
20
|
Borewicz K, Brück WM. Supplemented Infant Formula and Human Breast Milk Show Similar Patterns in Modulating Infant Microbiota Composition and Function In Vitro. Int J Mol Sci 2024; 25:1806. [PMID: 38339084 PMCID: PMC10855883 DOI: 10.3390/ijms25031806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 01/10/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
The gut microbiota of healthy breastfed infants is often dominated by bifidobacteria. In an effort to mimic the microbiota of breastfed infants, modern formulas are fortified with bioactive and bifidogenic ingredients. These ingredients promote the optimal health and development of infants as well as the development of the infant microbiota. Here, we used INFOGEST and an in vitro batch fermentation model to investigate the gut health-promoting effects of a commercial infant formula supplemented with a blend containing docosahexaenoic acid (DHA) (20 mg/100 kcal), polydextrose and galactooligosaccharides (PDX/GOS) (4 g/L, 1:1 ratio), milk fat globule membrane (MFGM) (5 g/L), lactoferrin (0.6 g/L), and Bifidobacterium animalis subsp. lactis, BB-12 (BB-12) (106 CFU/g). Using fecal inoculates from three healthy infants, we assessed microbiota changes, the bifidogenic effect, and the short-chain fatty acid (SCFA) production of the supplemented test formula and compared those with data obtained from an unsupplemented base formula and from the breast milk control. Our results show that even after INFOGEST digestion of the formula, the supplemented formula can still maintain its bioactivity and modulate infants' microbiota composition, promote faster bifidobacterial growth, and stimulate production of SCFAs. Thus, it may be concluded that the test formula containing a bioactive blend promotes infant gut microbiota and SCFA profile to something similar, but not identical to those of breastfed infants.
Collapse
Affiliation(s)
- Klaudyna Borewicz
- Mead Johnson B.V., Middenkampweg 2, 6545 CJ Nijmegen, The Netherlands;
| | - Wolfram Manuel Brück
- Institute for Life Technologies, University of Applied Sciences Western Switzerland Valais-Wallis, 1950 Sion, Switzerland
| |
Collapse
|
21
|
Reuter S, Raspe J, Taube C. Microbes little helpers and suppliers for therapeutic asthma approaches. Respir Res 2024; 25:29. [PMID: 38218816 PMCID: PMC10787474 DOI: 10.1186/s12931-023-02660-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/28/2023] [Indexed: 01/15/2024] Open
Abstract
Bronchial asthma is a prevalent and increasingly chronic inflammatory lung disease affecting over 300 million people globally. Initially considered an allergic disorder driven by mast cells and eosinophils, asthma is now recognized as a complex syndrome with various clinical phenotypes and immunological endotypes. These encompass type 2 inflammatory endotypes characterized by interleukin (IL)-4, IL-5, and IL-13 dominance, alongside others featuring mixed or non-eosinophilic inflammation. Therapeutic success varies significantly based on asthma phenotypes, with inhaled corticosteroids and beta-2 agonists effective for milder forms, but limited in severe cases. Novel antibody-based therapies have shown promise, primarily for severe allergic and type 2-high asthma. To address this gap, novel treatment strategies are essential for better control of asthma pathology, prevention, and exacerbation reduction. One promising approach involves stimulating endogenous anti-inflammatory responses through regulatory T cells (Tregs). Tregs play a vital role in maintaining immune homeostasis, preventing autoimmunity, and mitigating excessive inflammation after pathogenic encounters. Tregs have demonstrated their ability to control both type 2-high and type 2-low inflammation in murine models and dampen human cell-dependent allergic airway inflammation. Furthermore, microbes, typically associated with disease development, have shown immune-dampening properties that could be harnessed for therapeutic benefits. Both commensal microbiota and pathogenic microbes have demonstrated potential in bacterial-host interactions for therapeutic purposes. This review explores microbe-associated approaches as potential treatments for inflammatory diseases, shedding light on current and future therapeutics.
Collapse
Affiliation(s)
- Sebastian Reuter
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, Tüschener Weg 40, 45239, Essen, Germany.
| | - Jonas Raspe
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, Tüschener Weg 40, 45239, Essen, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, Tüschener Weg 40, 45239, Essen, Germany
| |
Collapse
|
22
|
Ren Z, Zheng Z, Feng X. Role of gut microbes in acute lung injury/acute respiratory distress syndrome. Gut Microbes 2024; 16:2440125. [PMID: 39658851 PMCID: PMC11639474 DOI: 10.1080/19490976.2024.2440125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/31/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024] Open
Abstract
Acute lung injury (ALI) is an acute, diffuse inflammatory lung condition triggered by factors of severe infections, trauma, shock, burns, ischemia-reperfusion, and mechanical ventilation. It is primarily characterized by refractory hypoxemia and respiratory distress. The more severe form, acute respiratory distress syndrome (ARDS), can progress to multi-organ failure and has a high mortality rate. Despite extensive research, the exact pathogenesis of ALI and ARDS remains complex and not fully understood. Recent advancements in studying the gut microecology of patients have revealed the critical role that gut microbes play in ALI/ARDS onset and progression. While the exact mechanisms are still under investigation, evidence increasingly points to the influence of gut microbes and their metabolites on ALI/ARDS. This review aims to summarize the role of gut microbes and their metabolites in ALI/ARDS caused by various triggers. Moreover, it explores potential mechanisms and discusses how gut microbe-targeting interventions might offer new clinical strategies for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Zixuan Ren
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhihuan Zheng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiujing Feng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
23
|
Portincasa P, Khalil M, Graziani A, Frühbeck G, Baffy G, Garruti G, Di Ciaula A, Bonfrate L. Gut microbes in metabolic disturbances. Promising role for therapeutic manipulations? Eur J Intern Med 2024; 119:13-30. [PMID: 37802720 DOI: 10.1016/j.ejim.2023.10.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/30/2023] [Accepted: 10/02/2023] [Indexed: 10/08/2023]
Abstract
The prevalence of overweight, obesity, type 2 diabetes, metabolic syndrome and steatotic liver disease is rapidly increasing worldwide with a huge economic burden in terms of morbidity and mortality. Several genetic and environmental factors are involved in the onset and development of metabolic disorders and related complications. A critical role also exists for the gut microbiota, a complex polymicrobial ecology at the interface of the internal and external environment. The gut microbiota contributes to food digestion and transformation, caloric intake, and immune response of the host, keeping the homeostatic control in health. Mechanisms of disease include enhanced energy extraction from the non-digestible dietary carbohydrates, increased gut permeability and translocation of bacterial metabolites which activate a chronic low-grade systemic inflammation and insulin resistance, as precursors of tangible metabolic disorders involving glucose and lipid homeostasis. The ultimate causative role of gut microbiota in this respect remains to be elucidated, as well as the therapeutic value of manipulating the gut microbiota by diet, pre- and pro- synbiotics, or fecal microbial transplantation.
Collapse
Affiliation(s)
- Piero Portincasa
- Clinica Medica "A. Murri", Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro" Medical School, Policlinico Hospital, Piazza G. Cesare 11, Bari 70124, Italy.
| | - Mohamad Khalil
- Clinica Medica "A. Murri", Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro" Medical School, Policlinico Hospital, Piazza G. Cesare 11, Bari 70124, Italy
| | - Annarita Graziani
- Institut AllergoSan Pharmazeutische Produkte Forschungs- und Vertriebs GmbH, Graz, Austria
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, Pamplona, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gyorgy Baffy
- Department of Medicine, VA Boston Healthcare System and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02130, USA
| | - Gabriella Garruti
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, Bari 70124, Italy
| | - Agostino Di Ciaula
- Clinica Medica "A. Murri", Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro" Medical School, Policlinico Hospital, Piazza G. Cesare 11, Bari 70124, Italy.
| | - Leonilde Bonfrate
- Clinica Medica "A. Murri", Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro" Medical School, Policlinico Hospital, Piazza G. Cesare 11, Bari 70124, Italy
| |
Collapse
|
24
|
Chen B, Yang X, Zhan M, Chen Y, Xu J, Xiao J, Xiao H, Song M. Dietary tangeretin improved antibiotic-associated diarrhea in mice by enhancing the intestinal barrier function, regulating the gut microbiota, and metabolic homeostasis. Food Funct 2023; 14:10731-10746. [PMID: 37933488 DOI: 10.1039/d3fo02998k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Antibiotic-associated diarrhea is mediated by antibiotic treatment and is usually caused by the disruption of the intestinal barrier, gut microbiota, and metabolic balance. To identify a dietary strategy that can mitigate the side effects of antibiotics, this study investigated the effect of tangeretin on antibiotic-associated diarrhea in C57BL/6 mice. The results revealed that dietary tangeretin significantly ameliorated symptoms of antibiotic-associated diarrhea, as evidenced by the decreased diarrhea status scores, the reduced fecal water content, the decreased caecum/body weight ratio, and the alleviated colonic tissue damage. Dietary tangeretin also exhibited a protective effect on the intestinal barrier function by upregulating the mRNA and protein expression of claudin-1 and ZO-1. Furthermore, analysis of the gut microbiota using 16S rRNA gene sequencing indicated that dietary tangeretin modulated the gut microbiota of mice with antibiotic-associated diarrhea via increasing the gut microbiota diversity and the abundance of beneficial bacteria, e.g., Lactobacillaceae and Ruminococcaceae, and decreasing the abundance of harmful bacteria, e.g., Enterococcus and Terrisporobacter. Additionally, dietary tangeretin restored the levels of short-chain fatty acids and modulated metabolic pathways by enriching purine metabolism, bile acid metabolism, ABC transporters, and choline metabolism in cancer. Collectively, these findings provide a solid scientific basis for the rational use of tangeretin as a preventive and therapeutic agent for antibiotic-associated diarrhea.
Collapse
Affiliation(s)
- Bin Chen
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, USA.
| | - Xun Yang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Minmin Zhan
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Yilu Chen
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, USA.
| | - Jingyi Xu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, USA.
| | - Mingyue Song
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| |
Collapse
|
25
|
Su Y, Ding T. Targeting microbial quorum sensing: the next frontier to hinder bacterial driven gastrointestinal infections. Gut Microbes 2023; 15:2252780. [PMID: 37680117 PMCID: PMC10486307 DOI: 10.1080/19490976.2023.2252780] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
Bacteria synchronize social behaviors via a cell-cell communication and interaction mechanism termed as quorum sensing (QS). QS has been extensively studied in monocultures and proved to be intensively involved in bacterial virulence and infection. Despite the role QS plays in pathogens during laboratory engineered infections has been proved, the potential functions of QS related to pathogenesis in context of microbial consortia remain poorly understood. In this review, we summarize the basic molecular mechanisms of QS, primarily focusing on pathogenic microbes driving gastrointestinal (GI) infections. We further discuss how GI pathogens disequilibrate the homeostasis of the indigenous microbial consortia, rebuild a realm dominated by pathogens, and interact with host under worsening infectious conditions via pathogen-biased QS signaling. Additionally, we present recent applications and main challenges of manipulating QS network in microbial consortia with the goal of better understanding GI bacterial sociality and facilitating novel therapies targeting bacterial infections.
Collapse
Affiliation(s)
- Ying Su
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Ministry of Education, Key Laboratory of Tropical Diseases Control (Sun Yat-Sen University), Guangzhou, China
| | - Tao Ding
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Ministry of Education, Key Laboratory of Tropical Diseases Control (Sun Yat-Sen University), Guangzhou, China
| |
Collapse
|
26
|
Gallardo-Becerra L, Cervantes-Echeverría M, Cornejo-Granados F, Vazquez-Morado LE, Ochoa-Leyva A. Perspectives in Searching Antimicrobial Peptides (AMPs) Produced by the Microbiota. MICROBIAL ECOLOGY 2023; 87:8. [PMID: 38036921 PMCID: PMC10689560 DOI: 10.1007/s00248-023-02313-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 10/24/2023] [Indexed: 12/02/2023]
Abstract
Changes in the structure and function of the microbiota are associated with various human diseases. These microbial changes can be mediated by antimicrobial peptides (AMPs), small peptides produced by the host and their microbiota, which play a crucial role in host-bacteria co-evolution. Thus, by studying AMPs produced by the microbiota (microbial AMPs), we can better understand the interactions between host and bacteria in microbiome homeostasis. Additionally, microbial AMPs are a new source of compounds against pathogenic and multi-resistant bacteria. Further, the growing accessibility to metagenomic and metatranscriptomic datasets presents an opportunity to discover new microbial AMPs. This review examines the structural properties of microbiota-derived AMPs, their molecular action mechanisms, genomic organization, and strategies for their identification in any microbiome data as well as experimental testing. Overall, we provided a comprehensive overview of this important topic from the microbial perspective.
Collapse
Affiliation(s)
- Luigui Gallardo-Becerra
- Departamento de Microbiologia Molecular, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico (UNAM), Avenida Universidad 2001, C.P. 62210, Cuernavaca, Morelos, Mexico
| | - Melany Cervantes-Echeverría
- Departamento de Microbiologia Molecular, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico (UNAM), Avenida Universidad 2001, C.P. 62210, Cuernavaca, Morelos, Mexico
| | - Fernanda Cornejo-Granados
- Departamento de Microbiologia Molecular, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico (UNAM), Avenida Universidad 2001, C.P. 62210, Cuernavaca, Morelos, Mexico
| | - Luis E Vazquez-Morado
- Departamento de Microbiologia Molecular, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico (UNAM), Avenida Universidad 2001, C.P. 62210, Cuernavaca, Morelos, Mexico
| | - Adrian Ochoa-Leyva
- Departamento de Microbiologia Molecular, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico (UNAM), Avenida Universidad 2001, C.P. 62210, Cuernavaca, Morelos, Mexico.
| |
Collapse
|
27
|
Li ML, Sun SP, Sun K, Lv B, Fan YH. Role of tryptophan metabolism in inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2023; 31:896-903. [DOI: 10.11569/wcjd.v31.i21.896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023] Open
Abstract
Inflammatory bowel disease (IBD) is comprised of ulcerative colitis and Crohn's disease, the pathogenesis of which is closely related to intestinal flora disorders. Abnormalities in the intestinal microenvironment caused by intestinal flora disorders affect amino acid metabolism. Tryptophan is an essential amino acid, and its metabolites are involved in the regulation of immunity, neuronal function, intestinal homeostasis, etc. The development of IBD disease is accompanied by tryptophan deficiency or metabolic abnormalities. This review focuses on the relationship between the intestinal flora metabolite tryptophan and its metabolites and the occurrence and development of IBD disease, and provides new ideas for future diagnostic methods for predicting IBD disease activity and protocols for treating IBD.
Collapse
Affiliation(s)
- Meng-Lin Li
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Shao-Peng Sun
- Zhejiang Provincial Key Laboratory of Pathophysiology of Gastrointestinal Diseases, Hangzhou 310053, Zhejiang Province, China
| | - Ke Sun
- Department of Nephrology, The First Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Bin Lv
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Yi-Hong Fan
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
28
|
Hai Q, Wang J, Kang W, Cheng S, Li J, Lyu N, Li Y, Luo Z, Liu Z. Metagenomic and metabolomic analysis of changes in intestinal contents of rainbow trout ( Oncorhynchus mykiss) infected with infectious hematopoietic necrosis virus at different culture water temperatures. Front Microbiol 2023; 14:1275649. [PMID: 37908544 PMCID: PMC10614001 DOI: 10.3389/fmicb.2023.1275649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/02/2023] [Indexed: 11/02/2023] Open
Abstract
Infectious hematopoietic necrosis (IHN) is a major disease that limits the culture of rainbow trout. In practical production, it has been found that the temperature of the culture water is a crucial factor affecting its mortality. Currently, little is known about how temperature affects the immune response of rainbow trout gut microbiota and metabolites to IHNV. In this study, our main objective is to analyze the changes in gut microorganisms of rainbow trout (juvenile fish with a consistent genetic background) after 14 days of infection with IHNV (5 × 105 pfu/fish) at 12-13°C (C: injected with saline, A: injected with IHNV) and 16-17°C (D: injected with saline, B: injected with IHNV) using metagenomic and metabolomic analyses, and to screen for probiotics that are effective against IHNV. The results showed that infection with IHNV at 12-13°C caused Eukaryote loss. Compared to Group C, Group A showed a significant increase in harmful pathogens, such as Yersiniaceae, and a significant alteration of 4,087 gut metabolites. Compared to group D, group B showed a significant increase in the abundance of Streptococcaceae and Lactococcus lactis, along with significant changes in 4,259 intestinal metabolites. Compared with their respective groups, the levels of two immune-related metabolites, 1-Octadecanoyl-glycero-3-phosphoethanolamine and L-Glutamate, were significantly upregulated in groups A and B. Compared to group B, Group A showed significantly higher pathogenic bacteria including Aeromonas, Pseudomonas, and Yersiniaceae, while group B showed a significant increase in Streptococcaceae and Lactococcus lactis. Additionally, there were 4,018 significantly different metabolites between the two groups. Interestingly, 1-Octadecanoyl-sn-glycero-3-phosphoethanolamine and L-Glutamate were significantly higher in group A than in group B. Some of the different metabolites in C vs. A are correlated with Fomitopsis pinicola, while in D vs. B they were correlated with Lactococcus raffinolactis, and in A vs. B they were correlated with Hypsizygus marmoreus. This study exposed how rainbow trout gut microbiota and metabolites respond to IHNV at different temperatures, and screens beneficial bacteria with potential resistance to IHN, providing new insights and scientific basis for the prevention and treatment of IHN.
Collapse
Affiliation(s)
| | - Jianfu Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Wang Q, He L, Tan H, Huang G, Liu J. Causal relationship between gut microbiota and otitis media: a two-sample Mendelian randomized study. Eur Arch Otorhinolaryngol 2023; 280:4715-4717. [PMID: 37405450 DOI: 10.1007/s00405-023-08102-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/28/2023] [Indexed: 07/06/2023]
Affiliation(s)
- Qiuyang Wang
- Department of ENT, First Affiliated Hospital of Soochow University, No.188 Shizi Street, Suzhou, 215000, Jiangsu, China
- Department of Otolaryngology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Lisha He
- School of Public Health and Management, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Teaching Office, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Huazhang Tan
- Department of Otolaryngology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Guoxin Huang
- Department of Evidence-Based Medicine Center, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, 15 Jiefang Road, Fancheng District, Xiangyang, 441000, China.
- School of Public Health and Management, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Jisheng Liu
- Department of ENT, First Affiliated Hospital of Soochow University, No.188 Shizi Street, Suzhou, 215000, Jiangsu, China.
| |
Collapse
|
30
|
Di Ciaula A, Bonfrate L, Khalil M, Garruti G, Portincasa P. Contribution of the microbiome for better phenotyping of people living with obesity. Rev Endocr Metab Disord 2023; 24:839-870. [PMID: 37119391 PMCID: PMC10148591 DOI: 10.1007/s11154-023-09798-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2023] [Indexed: 05/01/2023]
Abstract
Obesity has reached epidemic proportion worldwide and in all ages. Available evidence points to a multifactorial pathogenesis involving gene predisposition and environmental factors. Gut microbiota plays a critical role as a major interface between external factors, i.e., diet, lifestyle, toxic chemicals, and internal mechanisms regulating energy and metabolic homeostasis, fat production and storage. A shift in microbiota composition is linked with overweight and obesity, with pathogenic mechanisms involving bacterial products and metabolites (mainly endocannabinoid-related mediators, short-chain fatty acids, bile acids, catabolites of tryptophan, lipopolysaccharides) and subsequent alterations in gut barrier, altered metabolic homeostasis, insulin resistance and chronic, low-grade inflammation. Although animal studies point to the links between an "obesogenic" microbiota and the development of different obesity phenotypes, the translational value of these results in humans is still limited by the heterogeneity among studies, the high variation of gut microbiota over time and the lack of robust longitudinal studies adequately considering inter-individual confounders. Nevertheless, available evidence underscores the existence of several genera predisposing to obesity or, conversely, to lean and metabolically health phenotype (e.g., Akkermansia muciniphila, species from genera Faecalibacterium, Alistipes, Roseburia). Further longitudinal studies using metagenomics, transcriptomics, proteomics, and metabolomics with exact characterization of confounders are needed in this field. Results must confirm that distinct genera and specific microbial-derived metabolites represent effective and precision interventions against overweight and obesity in the long-term.
Collapse
Affiliation(s)
- Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| | - Leonilde Bonfrate
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| | - Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| | - Gabriella Garruti
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| |
Collapse
|
31
|
Mehta JP, Ayakar S, Singhal RS. The potential of paraprobiotics and postbiotics to modulate the immune system: A Review. Microbiol Res 2023; 275:127449. [PMID: 37454427 DOI: 10.1016/j.micres.2023.127449] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
Probiotics are viable microorganisms that provide beneficial health effects when consumed in adequate quantity by the host. Immunomodulation is one of the major beneficial effects of probiotics that is a result of the colonization of probiotic microorganisms in the gut, their interaction with the intestinal cells, production of various metabolites and by-products. The last few years have displayed an increasing number of studies on non-viable probiotics (paraprobiotics) and microbial by-products (postbiotics) that prove beneficial to human health by providing positive immune responses even in the inactivated form. The increasing number of research studies compare the effects of viable and non-viable probiotics, their by-products, and metabolites. This review focuses on the ability of different types of paraprobiotics and postbiotics to modulate the immune system. A majority of paraprobiotics are developed from Lactobacillus and Bifidobacterium strains. The postbiotic components that modulate the biological reactions include lipoteichoic acids, bacteriocins, short-chain fatty acids, peptidoglycan, and exopolysaccharides have been reported. We have reviewed paraprobiotics and postbiotics that are commercial as well as in research. Paraprobiotics and postbiotics can be a possible replacement for live probiotics for immunocompromised people. Paraprobiotics display an active role in maintaining T-cell mediated immunity and have been shown to treat colitis. Postbiotic components exhibit properties of pro and anti-immune, anti-tumor, anti-microbial, antioxidant, and anti-biofilm. More research is required on the efficient conversion of probiotics to paraprobiotics, the isolation and purification of different postbiotics, and stability studies during the shelf life. The majority of the articles report the effects of direct ingestion of different '-biotics' without blending in any food product.
Collapse
Affiliation(s)
- Jeet P Mehta
- Department of Biotechnology, Institute of Chemical Technology, Indian Oil Odisha Campus, Bhubaneswar, Odisha 751013, India
| | - Sonal Ayakar
- Department of Biotechnology, Institute of Chemical Technology, Indian Oil Odisha Campus, Bhubaneswar, Odisha 751013, India; Department of Biological Science, Birla Institute of Technology & Science, Pilani, K K Birla Goa Campus, Goa 403726, India.
| | - Rekha S Singhal
- Department of Food Engineering and Technology, Institute of Chemical Technology, Mumbai, Maharashtra 400019, India
| |
Collapse
|
32
|
Kearns PJ, Winter AS, Woodhams DC, Northup DE. The Mycobiome of Bats in the American Southwest Is Structured by Geography, Bat Species, and Behavior. MICROBIAL ECOLOGY 2023; 86:1565-1574. [PMID: 37126126 DOI: 10.1007/s00248-023-02230-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/23/2023] [Indexed: 06/19/2023]
Abstract
Bats are widespread mammals that play key roles in ecosystems as pollinators and insectivores. However, there is a paucity of information about bat-associated microbes, in particular their fungal communities, despite the important role microbes play in host health and overall host function. The emerging fungal disease, white-nose syndrome, presents a potential challenge to the bat microbiome and understanding healthy bat-associated taxa will provide valuable information about potential microbiome-pathogen interactions. To address this knowledge gap, we collected 174 bat fur/skin swabs from 14 species of bats captured in five locations in New Mexico and Arizona and used high-throughput sequencing of the fungal internal transcribed (ITS) region to characterize bat-associated fungal communities. Our results revealed a highly heterogeneous bat mycobiome that was structured by geography and bat species. Furthermore, our data suggest that bat-associated fungal communities are affected by bat foraging, indicating the bat skin microbiota is dynamic on short time scales. Finally, despite the strong effects of site and species, we found widespread and abundant taxa from several taxonomic groups including the genera Alternaria and Metschnikowia that have the potential to be inhibitory towards fungal and bacterial pathogens.
Collapse
Affiliation(s)
- Patrick J Kearns
- Department of Biology, University of Massachusetts Boston, Boston, MA, 02125, USA.
| | - Ara S Winter
- Department of Biology, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Douglas C Woodhams
- Department of Biology, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Diana E Northup
- Department of Biology, University of New Mexico, Albuquerque, NM, 87131, USA
| |
Collapse
|
33
|
Lücke J, Heinrich F, Malsy J, Meins N, Schnell J, Böttcher M, Nawrocki M, Zhang T, Bertram F, Sabihi M, Kempski J, Blankenburg T, Duprée A, Reeh M, Wolter S, Mann O, Izbicki JR, Lohse AW, Gagliani N, Lütgehetmann M, Bunders MJ, Altfeld M, Sauter G, Giannou AD, Krasemann S, Ondruschka B, Huber S. Intestinal IL-1β Plays a Role in Protecting against SARS-CoV-2 Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1052-1061. [PMID: 37556130 PMCID: PMC10476162 DOI: 10.4049/jimmunol.2200844] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 07/11/2023] [Indexed: 08/10/2023]
Abstract
The intestine is constantly balancing the maintenance of a homeostatic microbiome and the protection of the host against pathogens such as viruses. Many cytokines mediate protective inflammatory responses in the intestine, among them IL-1β. IL-1β is a proinflammatory cytokine typically activated upon specific danger signals sensed by the inflammasome. SARS-CoV-2 is capable of infecting multiple organs, including the intestinal tract. Severe cases of COVID-19 were shown to be associated with a dysregulated immune response, and blocking of proinflammatory pathways was demonstrated to improve patient survival. Indeed, anakinra, an Ab against the receptor of IL-1β, has recently been approved to treat patients with severe COVID-19. However, the role of IL-1β during intestinal SARS-CoV-2 infection has not yet been investigated. Here, we analyzed postmortem intestinal and blood samples from patients who died of COVID-19. We demonstrated that high levels of intestinal IL-1β were associated with longer survival time and lower intestinal SARS-CoV-2 RNA loads. Concurrently, type I IFN expression positively correlated with IL-1β levels in the intestine. Using human intestinal organoids, we showed that autocrine IL-1β sustains RNA expression of IFN type I by the intestinal epithelial layer. These results outline a previously unrecognized key role of intestinal IL-1β during SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jöran Lücke
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabian Heinrich
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Microbiology, Virology, and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob Malsy
- I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Leibniz Institute of Virology, Hamburg, Germany
- German Center for Infection Research, Hamburg-Lubeck-Borstel-Riems, Germany
| | - Nicholas Meins
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Josa Schnell
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marius Böttcher
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mikolaj Nawrocki
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tao Zhang
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franziska Bertram
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Morsal Sabihi
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Kempski
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tom Blankenburg
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Duprée
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Reeh
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Wolter
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Oliver Mann
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob R. Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ansgar W. Lohse
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Gagliani
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marc Lütgehetmann
- Institute of Medical Microbiology, Virology, and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Madeleine J. Bunders
- Leibniz Institute of Virology, Hamburg, Germany
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anastasios D. Giannou
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Susanne Krasemann
- Institute for Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Huber
- Section of Molecular Immunology and Gastroenterology, I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
34
|
Qin C, Tang N, Gan Y, Zhao H, Li Y, Tian GB, Yang YY, Yuan P, Ding X. Liposomes Co-Delivering Curcumin and Colistin to Overcome Colistin Resistance in Bacterial Infections. Adv Healthc Mater 2023; 12:e2202903. [PMID: 37523195 DOI: 10.1002/adhm.202202903] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 07/21/2023] [Indexed: 08/01/2023]
Abstract
Antibiotic colistin is the last line of defense against multidrug-resistant (MDR) Gram-negative bacterial infections. Emergence of colistin resistance in microbes is a critical challenge. Herein, curcumin is discovered, for the first time, to reverse the resistance phenotype of colistin-resistant bacteria via a checkerboard assay. For the co-delivery of curcumin and colistin, negatively charged poly(ethylene glycol)-functionalized liposomes encapsulating both drugs (Lipo-cc) are prepared. Killing kinetics and live/dead assays confirm the antibacterial activity of Lipo-cc against colistin-resistant bacteria, which is more potent than that of the free curcumin and colistin combination. Mechanistical studies reveal that Lipo-cc restores the affinity of colistin for the bacterial membrane and improves the uptake of curcumin, which leads to reduced efflux pump activity, achieving a synergistic effect of colistin and curcumin. At the effective antibacterial dose, Lipo-cc does not exhibit any toxicity. The therapeutic efficacy of Lipo-cc is further demonstrated in an intestinal bacterial infection model induced with colistin-resistant Escherichia coli. Lipo-cc reduces the bacterial burden with over 6-log reduction and alleviated inflammation caused by infection. Importantly, unlike colistin, Lipo-cc does not affect the homeostasis of the intestinal flora. Taken together, Lipo-cc successfully overcame colistin resistance, indicating its potential for the treatment of colistin-resistant bacterial infections.
Collapse
Affiliation(s)
- Chengyuan Qin
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, 518107, Shenzhen, P. R. China
| | - Ning Tang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, 518107, Shenzhen, P. R. China
| | - Yingying Gan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, 518107, Shenzhen, P. R. China
| | - Huimin Zhao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, 518107, Shenzhen, P. R. China
| | - Yuzhen Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, 518107, Shenzhen, P. R. China
| | - Guo-Bao Tian
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, P. R. China
| | - Yi Yan Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Singapore, 138668, Singapore
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, 518107, Shenzhen, P. R. China
| | - Xin Ding
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, 518107, Shenzhen, P. R. China
| |
Collapse
|
35
|
Yadav S, Shah D, Dalai P, Agrawal-Rajput R. The tale of antibiotics beyond antimicrobials: Expanding horizons. Cytokine 2023; 169:156285. [PMID: 37393846 DOI: 10.1016/j.cyto.2023.156285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/02/2023] [Accepted: 06/24/2023] [Indexed: 07/04/2023]
Abstract
Antibiotics had proved to be a godsend for mankind since their discovery. They were once the magical solution to the vexing problem of infection-related deaths. German scientist Paul Ehrlich had termed salvarsan as the silver bullet to treatsyphilis.As time passed, the magic of newly discovered silver bullets got tarnished with raging antibiotic resistance among bacteria and associated side-effects. Still, antibiotics remain the primary line of treatment for bacterial infections. Our understanding of their chemical and biological activities has increased immensely with advancement in the research field. Non-antibacterial effects of antibiotics are studied extensively to optimise their safer, broad-range use. These non-antibacterial effects could be both useful and harmful to us. Various researchers across the globe including our lab are studying the direct/indirect effects and molecular mechanisms behind these non-antibacterial effects of antibiotics. So, it is interesting for us to sum up the available literature. In this review, we have briefed the possible reason behind the non-antibacterial effects of antibiotics, owing to the endosymbiotic origin of host mitochondria. We further discuss the physiological and immunomodulatory effects of antibiotics. We then extend the review to discuss molecular mechanisms behind the plausible use of antibiotics as anticancer agents.
Collapse
Affiliation(s)
- Shivani Yadav
- Immunology Lab, Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India
| | - Dhruvi Shah
- Immunology Lab, Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India
| | - Parmeswar Dalai
- Immunology Lab, Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India
| | - Reena Agrawal-Rajput
- Immunology Lab, Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India.
| |
Collapse
|
36
|
Hughey MC, Warne R, Dulmage A, Reeve RE, Curtis GH, Whitfield K, Schock DM, Crespi E. Diet- and salinity-induced modifications of the gut microbiota are associated with differential physiological responses to ranavirus infection in Rana sylvatica. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220121. [PMID: 37305908 PMCID: PMC10258663 DOI: 10.1098/rstb.2022.0121] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 03/26/2023] [Indexed: 06/13/2023] Open
Abstract
Greater knowledge of how host-microbiome interactions vary with anthropogenic environmental change and influence pathogenic infections is needed to better understand stress-mediated disease outcomes. We investigated how increasing salinization in freshwaters (e.g. due to road de-icing salt runoff) and associated increases in growth of nutritional algae influenced gut bacterial assembly, host physiology and responses to ranavirus exposure in larval wood frogs (Rana sylvatica). Elevating salinity and supplementing a basic larval diet with algae increased larval growth and also increased ranavirus loads. However, larvae given algae did not exhibit elevated kidney corticosterone levels, accelerated development or weight loss post-infection, whereas larvae fed a basic diet did. Thus, algal supplementation reversed a potentially maladaptive stress response to infection observed in prior studies in this system. Algae supplementation also reduced gut bacterial diversity. Notably, we observed higher relative abundances of Firmicutes in treatments with algae-a pattern consistent with increased growth and fat deposition in mammals-that may contribute to the diminished stress responses to infection via regulation of host metabolism and endocrine function. Our study informs mechanistic hypotheses about the role of microbiome mediation of host responses to infection that can be tested in future experiments in this host-pathogen system. This article is part of the theme issue 'Amphibian immunity: stress, disease and ecoimmunology'.
Collapse
Affiliation(s)
- Myra C. Hughey
- Department of Biology, Vassar College, Poughkeepsie, NY 12604, USA
| | - Robin Warne
- School of Biological Sciences, Southern Illinois University, Carbondale, IL 62901, USA
| | - Alexa Dulmage
- School of Biological Sciences, Center for Reproductive Biology, Washington State University, Pullman, WA 99164, USA
| | - Robyn E. Reeve
- School of Biological Sciences, Center for Reproductive Biology, Washington State University, Pullman, WA 99164, USA
| | - Grace H. Curtis
- School of Biological Sciences, Center for Reproductive Biology, Washington State University, Pullman, WA 99164, USA
| | - Kourtnie Whitfield
- School of Biological Sciences, Center for Reproductive Biology, Washington State University, Pullman, WA 99164, USA
| | | | - Erica Crespi
- School of Biological Sciences, Center for Reproductive Biology, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
37
|
Yang Q, Qin B, Hou W, Qin H, Yin F. Pathogenesis and therapy of radiation enteritis with gut microbiota. Front Pharmacol 2023; 14:1116558. [PMID: 37063268 PMCID: PMC10102376 DOI: 10.3389/fphar.2023.1116558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/07/2023] [Indexed: 04/03/2023] Open
Abstract
Radiotherapy is widely used in clinic due to its good effect for cancer treatment. But radiotherapy of malignant tumors in the abdomen and pelvis is easy to cause radiation enteritis complications. Gastrointestinal tract contains numerous microbes, most of which are mutualistic relationship with the host. Abdominal radiation results in gut microbiota dysbiosis. Microbial therapy can directly target gut microbiota to reverse microbiota dysbiosis, hence relieving intestinal inflammation. In this review, we mainly summarized pathogenesis and novel therapy of the radiation-induced intestinal injury with gut microbiota dysbiosis and envision the opportunities and challenges of radiation enteritis therapy.
Collapse
Affiliation(s)
- Qilin Yang
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- School of Clinical Medicine of Nanjing Medical University, Nanjing, China
| | - Bingzhi Qin
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Weiliang Hou
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Shanghai Cancer Institute, Renji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Weiliang Hou, ; Huanlong Qin, ; Fang Yin,
| | - Huanlong Qin
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- *Correspondence: Weiliang Hou, ; Huanlong Qin, ; Fang Yin,
| | - Fang Yin
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- *Correspondence: Weiliang Hou, ; Huanlong Qin, ; Fang Yin,
| |
Collapse
|
38
|
Kirundi J, Moghadamrad S, Urbaniak C. Microbiome-liver crosstalk: A multihit therapeutic target for liver disease. World J Gastroenterol 2023; 29:1651-1668. [PMID: 37077519 PMCID: PMC10107210 DOI: 10.3748/wjg.v29.i11.1651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/05/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023] Open
Abstract
Liver disease has become a leading cause of death, particularly in the West, where it is attributed to more than two million deaths annually. The correlation between gut microbiota and liver disease is still not fully understood. However, it is well known that gut dysbiosis accompanied by a leaky gut causes an increase in lipopolysaccharides in circulation, which in turn evoke massive hepatic inflammation promoting liver cirrhosis. Microbial dysbiosis also leads to poor bile acid metabolism and low short-chain fatty acids, all of which exacerbate the inflammatory response of liver cells. Gut microbial homeostasis is maintained through intricate processes that ensure that commensal microbes adapt to the low oxygen potential of the gut and that they rapidly occupy all the intestinal niches, thus outcompeting any potential pathogens for available nutrients. The crosstalk between the gut microbiota and its metabolites also guarantee an intact gut barrier. These processes that protect against destabilization of gut microbes by potential entry of pathogenic bacteria are collectively called colonization resistance and are equally essential for liver health. In this review, we shall investigate how the mechanisms of colonization resistance influence the liver in health and disease and the microbial-liver crosstalk potential as therapeutic target areas.
Collapse
Affiliation(s)
- Jorum Kirundi
- Department of Biomedical Research, University of Bern, Bern 3014, Switzerland
| | - Sheida Moghadamrad
- Department of Gastroenterology/Hepatology, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona and Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano 6900, Switzerland
| | | |
Collapse
|
39
|
Kotronarou K, Charalambous A, Evangelou A, Georgiou O, Demetriou A, Apidianakis Y. Dietary Stimuli, Intestinal Bacteria and Peptide Hormones Regulate Female Drosophila Defecation Rate. Metabolites 2023; 13:metabo13020264. [PMID: 36837883 PMCID: PMC9965912 DOI: 10.3390/metabo13020264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/09/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Peptide hormones control Drosophila gut motility, but the intestinal stimuli and the gene networks coordinating this trait remain poorly defined. Here, we customized an assay to quantify female Drosophila defecation rate as a proxy of intestinal motility. We found that bacterial infection with the human opportunistic bacterial pathogen Pseudomonas aeruginosa (strain PA14) increases defecation rate in wild-type female flies, and we identified specific bacteria of the fly microbiota able to increase defecation rate. In contrast, dietary stress, imposed by either water-only feeding or high ethanol consumption, decreased defecation rate and the expression of enteroendocrine-produced hormones in the fly midgut, such as Diuretic hormone 31 (Dh31). The decrease in defecation due to dietary stress was proportional to the impact of each stressor on fly survival. Furthermore, we exploited the Drosophila Genetic Reference Panel wild type strain collection and identified strains displaying high and low defecation rates. We calculated the narrow-sense heritability of defecation rate to be 91%, indicating that the genetic variance observed using our assay is mostly additive and polygenic in nature. Accordingly, we performed a genome-wide association (GWA) analysis revealing 17 candidate genes linked to defecation rate. Downregulation of four of them (Pmp70, CG11307, meso18E and mub) in either the midgut enteroendocrine cells or in neurons reduced defecation rate and altered the midgut expression of Dh31, that in turn regulates defecation rate via signaling to the visceral muscle. Hence, microbial and dietary stimuli, and Dh31-controlling genes, regulate defecation rate involving signaling within and among neuronal, enteroendocrine, and visceral muscle cells.
Collapse
|
40
|
Sharma S, Hegde P, Panda S, Orimoloye MO, Aldrich CC. Drugging the microbiome: targeting small microbiome molecules. Curr Opin Microbiol 2023; 71:102234. [PMID: 36399893 DOI: 10.1016/j.mib.2022.102234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022]
Abstract
The human microbiome represents a large and diverse collection of microbes that plays an integral role in human physiology and pathophysiology through interactions with the host and within the microbial community. While early work exploring links between microbiome signatures and diseases states has been associative, emerging evidence demonstrates the metabolic products of the human microbiome have more proximal causal effects on disease phenotypes. The therapeutic implications of this shift are profound as manipulation of the microbiome by the administration of live biotherapeutics, ongoing, can now be pursued alongside research efforts toward describing inhibitors of key microbiome enzymes involved in the biosynthesis of metabolites implicated in various disease states and processing of host-derived metabolites. With growing interest in 'drugging the microbiome', we review few notable microbial metabolites for which traditional drug-development campaigns have yielded compounds with therapeutic promise.
Collapse
Affiliation(s)
- Sachin Sharma
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, USA
| | - Pooja Hegde
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, USA
| | - Subhankar Panda
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, USA
| | - Moyosore O Orimoloye
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, USA
| | - Courtney C Aldrich
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
41
|
Elfiky SA, Mahmoud Ahmed S, Elmenshawy AM, Sultan GM, Asser SL. Study of the gut microbiome as a novel target for prevention of hospital-associated infections in intensive care unit patients. Acute Crit Care 2023; 38:76-85. [PMID: 36935537 PMCID: PMC10030239 DOI: 10.4266/acc.2022.01116] [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: 08/25/2022] [Accepted: 11/23/2022] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Hospital-acquired infections (HAIs) are increasing due to the spread of multi-drugresistant organisms. Gut dysbiosis in an intensive care unit (ICU) patients at admission showed an altered abundance of some bacterial genera associated with the occurrence of HAIs and mortality. In the present study, we investigated the pattern of the gut microbiome in ICU patients at admission to correlate it with the development of HAIs during ICU stay. METHODS Twenty patients admitted to an ICU with a cross-matched control group of 30 healthy subjects of matched age and sex. Quantitative SYBR green real-time polymerase chain reaction was done for the identification and quantitation of selected bacteria. RESULTS Out of those twenty patients, 35% developed ventilator-associated pneumonia during their ICU stay. Gut microbiome analysis showed a significant decrease in Firmicutes and Firmicutes to Bacteroidetes ratio in ICU patients in comparison to the control and in patients who developed HAIs in comparison to the control group and patients who did not develop HAIs. There was a statistically significant increase in Bacteroides in comparison to the control group. There was a statistically significant decrease in Bifidobacterium and Faecalibacterium prausnitzii and an increase in Lactobacilli in comparison to the control group with a negative correlation between Acute Physiology and Chronic Health Evaluation (APACHE) II score and Firmicutes to Bacteroidetes and Prevotella to Bacteroides ratios. CONCLUSIONS Gut dysbiosis of patients at the time of admission highlights the importance of identification of the microbiome of patients admitted to the ICU as a target for preventing of HAIs.
Collapse
Affiliation(s)
- Suzan Ahmed Elfiky
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Alexandria University, Alexandria Governorate, Egypt
| | - Shwikar Mahmoud Ahmed
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Alexandria University, Alexandria Governorate, Egypt
| | - Ahmed Mostafa Elmenshawy
- Department of Critical Care Medicine, Faculty of Medicine, Alexandria University, Alexandria Governorate, Egypt
| | - Gehad Mahmoud Sultan
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Alexandria University, Alexandria Governorate, Egypt
| | - Sara Lotfy Asser
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Alexandria University, Alexandria Governorate, Egypt
| |
Collapse
|
42
|
Juthi RT, Sazed SA, Sarmin M, Haque R, Alam MS. COVID-19 and diarrhea: putative mechanisms and management. Int J Infect Dis 2023; 126:125-131. [PMID: 36403817 PMCID: PMC9672967 DOI: 10.1016/j.ijid.2022.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/08/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative agent of the coronavirus disease 2019 (COVID-19), has recently posed a threat to global health by spreading at a high rate and taking millions of lives worldwide. Along with the respiratory symptoms, there are gastrointestinal manifestations and one of the most common gastrointestinal symptoms is diarrhea which is seen in a significant percentage of COVID-19 patients. LITERATURE REVIEW Several studies have shown the plausible correlation between overexpressed angiotensin converting enzyme 2 (ACE2) in enterocytes and SARS-CoV-2, as ACE2 is the only known receptor for the virus entry. Along with the dysregulated ACE2, there are other contributing factors such as gut microbiome dysbiosis, adverse effects of antiviral and antibiotics for treating infections and inflammatory response to SARS-CoV-2 which bring about increased permeability of gut cells and subsequent occurrence of diarrhea. Few studies found that the SARS-CoV-2 is capable of damaging liver cells too. No single effective treatment option is available. LIMITATIONS Confirmed pathophysiology is still unavailable. Studies regarding global population are also insufficient. CONCLUSION In this review, based on the previous works and literature, we summarized the putative molecular pathophysiology of COVID-19 associated diarrhea, concomitant complications and the standard practices of management of diarrhea and hepatic manifestations in international setups.
Collapse
Affiliation(s)
- Rifat Tasnim Juthi
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Saiful Arefeen Sazed
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Monira Sarmin
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Rashidul Haque
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Mohammad Shafiul Alam
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh,Corresponding author. Mohammad Shafiul Alam, Scientist, Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka-1212, Bangladesh. Tel: +8801711-469232
| |
Collapse
|
43
|
Lu J, Jin X, Yang S, Li Y, Wang X, Wu M. Immune mechanism of gut microbiota and its metabolites in the occurrence and development of cardiovascular diseases. Front Microbiol 2022; 13:1034537. [PMID: 36590426 PMCID: PMC9794627 DOI: 10.3389/fmicb.2022.1034537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/24/2022] [Indexed: 12/15/2022] Open
Abstract
The risk of cardiovascular disease (CVD) is associated with unusual changes in the human gut microbiota, most commonly coronary atherosclerotic heart disease, hypertension, and heart failure. Immune mechanisms maintain a dynamic balance between the gut microbiota and the host immune system. When one side changes and the balance is disrupted, different degrees of damage are inflicted on the host and a diseased state gradually develops over time. This review summarizes the immune mechanism of the gut microbiota and its metabolites in the occurrence of common CVDs, discusses the relationship between gut-heart axis dysfunction and the progression of CVD, and lists the currently effective methods of regulating the gut microbiota for the treatment of CVDs.
Collapse
|
44
|
Cardoso MH, Meneguetti BT, Oliveira-Júnior NG, Macedo MLR, Franco OL. Antimicrobial peptide production in response to gut microbiota imbalance. Peptides 2022; 157:170865. [PMID: 36038014 DOI: 10.1016/j.peptides.2022.170865] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/18/2022]
Abstract
The gut microbiota presents essential functions in the immune response. The gut epithelium acts as a protective barrier and, therefore, can produce several antimicrobial peptides (AMPs) that can act against pathogenic microorganisms, including bacteria. Several factors cause a disturbance in gut microbiota, including the exacerbated and erroneous use of antibiotics. Antibiotic therapy has been closely related to bacterial resistance and is also correlated with undesired side-effects to the host, including the eradication of commensal bacteria. Consequently, this results in gut microbiota imbalance and inflammatory bowel diseases (IBD) development. In this context, AMPs in the gut epithelium play a restructuring role for gut microbiota. Some naturally occurring AMPs are selective for pathogenic bacteria, thus preserving the health microbiota. Therefore, AMPs produced by the host's epithelial cells represent effective molecules in treating gut bacterial infections. Bearing this in mind, this review focused on describing the importance of the host's AMPs in gut microbiota modulation and their role as anti-infective agents against pathogenic bacteria.
Collapse
Affiliation(s)
- Marlon H Cardoso
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS 79117900, Brazil; Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF 70790160, Brazil; Laboratório de Purificação de Proteínas e suas Funções Biológicas, Universidade Federal de Mato Grosso do Sul, Cidade Universitária, 79070900 Campo Grande, Mato Grosso do Sul, Brazil.
| | - Beatriz T Meneguetti
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS 79117900, Brazil
| | - Nelson G Oliveira-Júnior
- Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF 70790160, Brazil
| | - Maria L R Macedo
- Laboratório de Purificação de Proteínas e suas Funções Biológicas, Universidade Federal de Mato Grosso do Sul, Cidade Universitária, 79070900 Campo Grande, Mato Grosso do Sul, Brazil
| | - Octávio L Franco
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS 79117900, Brazil; Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF 70790160, Brazil.
| |
Collapse
|
45
|
An in vitro fermentation model to study the impact of bacteriophages targeting Shiga toxin-encoding Escherichia coli on the colonic microbiota. NPJ Biofilms Microbiomes 2022; 8:74. [PMID: 36163472 PMCID: PMC9512901 DOI: 10.1038/s41522-022-00334-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Lytic bacteriophages are considered safe for human consumption as biocontrol agents against foodborne pathogens, in particular in ready-to-eat foodstuffs. Phages could, however, evolve to infect different hosts when passing through the gastrointestinal tract (GIT). This underlines the importance of understanding the impact of phages towards colonic microbiota, particularly towards bacterial families usually found in the colon such as the Enterobacteriaceae. Here we propose in vitro batch fermentation as model for initial safety screening of lytic phages targeting Shiga toxin-producing Escherichia coli (STEC). As inoculum we used faecal material of three healthy donors. To assess phage safety, we monitored fermentation parameters, including short chain fatty acid production and gas production/intake by colonic microbiota. We performed shotgun metagenomic analysis to evaluate the outcome of phage interference with colonic microbiota composition and functional potential. During the 24 h incubation, concentrations of phage and its host were also evaluated. We found the phage used in this study, named E. coli phage vB_EcoS_Ace (Ace), to be safe towards human colonic microbiota, independently of the donors’ faecal content used. This suggests that individuality of donor faecal microbiota did not interfere with phage effect on the fermentations. However, the model revealed that the attenuated STEC strain used as phage host perturbed the faecal microbiota as based on metagenomic analysis, with potential differences in metabolic output. We conclude that the in vitro batch fermentation model used in this study is a reliable safety screening for lytic phages intended to be used as biocontrol agents.
Collapse
|
46
|
Jiang H, Bao J, Xing Y, Cao G, Li X, Chen Q. Metabolomic and metagenomic analyses of the Chinese mitten crab Eriocheir sinensis after challenge with Metschnikowia bicuspidata. Front Microbiol 2022; 13:990737. [PMID: 36212869 PMCID: PMC9538530 DOI: 10.3389/fmicb.2022.990737] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
Milky disease caused by Metschnikowia bicuspidata fungus has significantly harmed the Chinese mitten crab Eriocheir sinensis aquaculture industry. However, the effect of M. bicuspidata infection on the metabolism and intestinal flora of the crab remains unclear. In this study, we aimed to explore the changes in the metabolism and intestinal flora E. sinensis after 48 h of infection with M. bicuspidata, using metabolomic and metagenomic analyses. Metabolomic analysis results revealed 420 significantly different metabolites between the infected and control groups, and these metabolites were enriched in 58 metabolic pathways. M. bicuspidata infection decreased the levels of metabolites related to amino acid biosynthesis, the tricarboxylic acid cycle, as well as lysine, histidine, linolenic, arachidonic, and linoleic acid metabolism. These results indicated that M. bicuspidata infection significantly affected the energy metabolism, growth, and immunity of E. sinensis. The results of metagenomic analysis showed that the anaerobes and ascomycetes populations significantly increased and decreased, respectively, after M. bicuspidata infection. These changes in intestinal flora significantly upregulated metabolic and synthetic pathways while downregulating immunity-related pathways. The results of integrated metabolomic and metagenomic analyses showed that 55 differentially expressed genes and 28 operational taxonomic units were correlated with 420 differential metabolites. Thus, the intestinal flora changes caused by M. bicuspidata infection also affected the metabolites. This study provides novel insights into the metabolic-and intestinal microflora-based effects of M. bicuspidata infection in E. sinensis, as well as a theoretical basis for the interaction between fungi and crustaceans.
Collapse
Affiliation(s)
- Hongbo Jiang
- Aquaculture Department, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Shenyang Agricultural University, Shenyang, China
| | - Jie Bao
- Aquaculture Department, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Yuenan Xing
- Aquaculture Department, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Gangnan Cao
- Aquaculture Department, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Xiaodong Li
- Aquaculture Department, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Qijun Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Shenyang Agricultural University, Shenyang, China
- *Correspondence: Qijun Chen,
| |
Collapse
|
47
|
Shi S, Zhou D, Xu Y, Dong J, Han Y, He G, Li W, Hu J, Liu Y, Zhao K. Effect of Lactobacillus reuteri S5 Intervention on Intestinal Microbiota Composition of Chickens Challenged with Salmonella enteritidis. Animals (Basel) 2022; 12:ani12192528. [PMID: 36230269 PMCID: PMC9559494 DOI: 10.3390/ani12192528] [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: 08/19/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
To understand the mechanism of lactic acid bacteria against Salmonella enteritidis infection; we examined how lactic acid bacteria regulated the intestinal microbiota to resist infection by pathogenic bacteria. The probiotic strain Lactobacillus reuteri S5 was used to construct an animal model of S. enteritidis infected broilers. A high-throughput sequencing technology was used to analyze the regulatory effects of L. reuteri S5 on the structure of the intestinal microbiota of broilers infected with S. enteritidis; and to examine the possible defense mechanism they used. Our results showed that the administration of L. reuteri S5 reduced colonization of S. enteritidis (p < 0.05), decreased intestinal permeability (p < 0.05), and reduced the bacterial displacement likely due by S. enteritidis colonization (p < 0.05), suggesting some enhancement of the intestinal barrier function. Furthermore, L. reuteri S5 increased the number of operational taxonomic units (OTUs) in the chicken cecal microflora and the relative abundance of Lactobacillaceae and decreased the relative abundance of Enterobacteriaceae. These results suggest that the lactic acid bacterium L. reuteri S5 protected the intestinal microbiota of chickens against S. enteritidis infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Kai Zhao
- Correspondence: ; Tel.: +86-138-6578-6710; Fax: +86-0556-5708061
| |
Collapse
|
48
|
Ferreira-Junior AS, Borgonovi TF, De Salis LVV, Leite AZ, Dantas AS, De Salis GVV, Cruz GNF, De Oliveira LFV, Gomes E, Penna ALB, De Oliveira GLV. Detection of Intestinal Dysbiosis in Post-COVID-19 Patients One to Eight Months after Acute Disease Resolution. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph191610189. [PMID: 36011823 PMCID: PMC9408204 DOI: 10.3390/ijerph191610189] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/13/2022] [Accepted: 08/14/2022] [Indexed: 06/09/2023]
Abstract
The intestinal microbiota plays an important role in the immune response against viral infections, modulating both innate and adaptive immune responses. The cytokine storm is associated with COVID-19 severity, and the patient's immune status is influenced by the intestinal microbiota in a gut-lung bidirectional interaction. In this study, we evaluate the intestinal microbiota of Brazilian patients in different post-COVID-19 periods, and correlate this with clinical data and the antibiotic therapy used during the acute phase. DNA extracted from stool samples was sequenced and total anti-SARS-CoV-2 antibodies and C-reactive protein were quantified. Compared with controls, there were significant differences in the microbiota diversity in post-COVID-19 patients, suggesting an intestinal dysbiosis even several months after acute disease resolution. Additionally, we detected some genera possibly associated with the post-COVID-19 dysbiosis, including Desulfovibrio, Haemophillus, Dialister, and Prevotella, in addition to decreased beneficial microbes, associated with antibiotic-induced dysbiosis, such as Bifidobacterium and Akkermansia. Therefore, our hypothesis is that dysbiosis and the indiscriminate use of antibiotics during the pandemic may be associated with post-COVID-19 clinical manifestations. In our study, 39% (n = 58) of patients reported symptoms, including fatigue, dyspnea, myalgia, alopecia, anxiety, memory loss, and depression. These data suggest that microbiota modulation may represent a target for recovery from acute COVID-19 and a therapeutic approach for post-COVID-19 sequelae.
Collapse
Affiliation(s)
| | - Tais Fernanda Borgonovi
- Food Engineering and Technology Department, Sao Paulo State University (UNESP), Sao Jose do Rio Preto 15054-000, Brazil
| | | | - Aline Zazeri Leite
- Microbiology Program, Institute of Biosciences, Humanities and Exact Sciences (IBILCE), Sao Jose do Rio Preto 15054-000, Brazil
| | | | | | | | | | - Eleni Gomes
- Microbiology Program, Institute of Biosciences, Humanities and Exact Sciences (IBILCE), Sao Jose do Rio Preto 15054-000, Brazil
| | - Ana Lúcia Barretto Penna
- Microbiology Program, Institute of Biosciences, Humanities and Exact Sciences (IBILCE), Sao Jose do Rio Preto 15054-000, Brazil
- Food Engineering and Technology Department, Sao Paulo State University (UNESP), Sao Jose do Rio Preto 15054-000, Brazil
| | - Gislane Lelis Vilela De Oliveira
- Microbiology Program, Institute of Biosciences, Humanities and Exact Sciences (IBILCE), Sao Jose do Rio Preto 15054-000, Brazil
- Food Engineering and Technology Department, Sao Paulo State University (UNESP), Sao Jose do Rio Preto 15054-000, Brazil
| |
Collapse
|
49
|
Singh P, Ali SA. Multifunctional Role of S100 Protein Family in the Immune System: An Update. Cells 2022; 11:cells11152274. [PMID: 35892571 PMCID: PMC9332480 DOI: 10.3390/cells11152274] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/12/2022] [Accepted: 07/21/2022] [Indexed: 12/04/2022] Open
Abstract
S100 is a broad subfamily of low-molecular weight calcium-binding proteins (9–14 kDa) with structural similarity and functional discrepancy. It is required for inflammation and cellular homeostasis, and can work extracellularly, intracellularly, or both. S100 members participate in a variety of activities in a healthy cell, including calcium storage and transport (calcium homeostasis). S100 isoforms that have previously been shown to play important roles in the immune system as alarmins (DAMPs), antimicrobial peptides, pro-inflammation stimulators, chemo-attractants, and metal scavengers during an innate immune response. Currently, during the pandemic, it was found that several members of the S100 family are implicated in the pathophysiology of COVID-19. Further, S100 family protein members were proposed to be used as a prognostic marker for COVID-19 infection identification using a nasal swab. In the present review, we compiled the vast majority of recent studies that focused on the multifunctionality of S100 proteins in the complex immune system and its associated activities. Furthermore, we shed light on the numerous molecular approaches and signaling cascades regulated by S100 proteins during immune response. In addition, we discussed the involvement of S100 protein members in abnormal defense systems during the pathogenesis of COVID-19.
Collapse
Affiliation(s)
- Parul Singh
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, Karnal 132001, India;
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, Karnal 132001, India;
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +91-8708591790
| |
Collapse
|
50
|
Sharon I, Quijada NM, Pasolli E, Fabbrini M, Vitali F, Agamennone V, Dötsch A, Selberherr E, Grau JH, Meixner M, Liere K, Ercolini D, de Filippo C, Caderni G, Brigidi P, Turroni S. The Core Human Microbiome: Does It Exist and How Can We Find It? A Critical Review of the Concept. Nutrients 2022; 14:nu14142872. [PMID: 35889831 PMCID: PMC9323970 DOI: 10.3390/nu14142872] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 11/16/2022] Open
Abstract
The core microbiome, which refers to a set of consistent microbial features across populations, is of major interest in microbiome research and has been addressed by numerous studies. Understanding the core microbiome can help identify elements that lead to dysbiosis, and lead to treatments for microbiome-related health states. However, defining the core microbiome is a complex task at several levels. In this review, we consider the current state of core human microbiome research. We consider the knowledge that has been gained, the factors limiting our ability to achieve a reliable description of the core human microbiome, and the fields most likely to improve that ability. DNA sequencing technologies and the methods for analyzing metagenomics and amplicon data will most likely facilitate higher accuracy and resolution in describing the microbiome. However, more effort should be invested in characterizing the microbiome’s interactions with its human host, including the immune system and nutrition. Other components of this holobiontic system should also be emphasized, such as fungi, protists, lower eukaryotes, viruses, and phages. Most importantly, a collaborative effort of experts in microbiology, nutrition, immunology, medicine, systems biology, bioinformatics, and machine learning is probably required to identify the traits of the core human microbiome.
Collapse
Affiliation(s)
- Itai Sharon
- Migal-Galilee Research Institute, P.O. Box 831, Kiryat Shmona 11016, Israel
- Faculty of Sciences and Technology, Tel-Hai Academic College, Upper Galilee 1220800, Israel
- Correspondence:
| | - Narciso Martín Quijada
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, A-1210 Vienna, Austria; (N.M.Q.); (E.S.)
- Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, FFoQSI GmbH, A-3430 Tulln an der Donau, Austria
| | - Edoardo Pasolli
- Department of Agricultural Sciences, Division of Microbiology, University of Naples Federico II, 80055 Portici, Italy; (E.P.); (D.E.)
- Task Force on Microbiome Studies, University of Naples Federico II, 80055 Portici, Italy
| | - Marco Fabbrini
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (M.F.); (S.T.)
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy;
| | - Francesco Vitali
- Institute of Agricultural Biology and Biotechnology (IBBA), National Research Council (CNR), Via Moruzzi 1, 56124 Pisa, Italy; (F.V.); (C.d.F.)
| | - Valeria Agamennone
- Microbiology and Systems Biology, Netherlands Organization for Applied Scientific Research (TNO), Utrechtseweg 48, 3704 HE Zeist, The Netherlands;
| | - Andreas Dötsch
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut (MRI)-Federal Research Institute of Nutrition and Food, 76131 Karlsruhe, Germany;
| | - Evelyne Selberherr
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, A-1210 Vienna, Austria; (N.M.Q.); (E.S.)
| | - José Horacio Grau
- Amedes Genetics, Amedes Medizinische Dienstleistungen GmbH, 10117 Berlin, Germany; (J.H.G.); (M.M.); (K.L.)
- Center for Species Survival, Smithsonian Conservation Biology Institute, Washington, DC 20008, USA
| | - Martin Meixner
- Amedes Genetics, Amedes Medizinische Dienstleistungen GmbH, 10117 Berlin, Germany; (J.H.G.); (M.M.); (K.L.)
| | - Karsten Liere
- Amedes Genetics, Amedes Medizinische Dienstleistungen GmbH, 10117 Berlin, Germany; (J.H.G.); (M.M.); (K.L.)
| | - Danilo Ercolini
- Department of Agricultural Sciences, Division of Microbiology, University of Naples Federico II, 80055 Portici, Italy; (E.P.); (D.E.)
- Task Force on Microbiome Studies, University of Naples Federico II, 80055 Portici, Italy
| | - Carlotta de Filippo
- Institute of Agricultural Biology and Biotechnology (IBBA), National Research Council (CNR), Via Moruzzi 1, 56124 Pisa, Italy; (F.V.); (C.d.F.)
| | - Giovanna Caderni
- NEUROFARBA Department, Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy;
| | - Patrizia Brigidi
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy;
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (M.F.); (S.T.)
| |
Collapse
|