1
|
Nouari W, Aribi M. Innate lymphoid cells, immune functional dynamics, epithelial parallels, and therapeutic frontiers in infections. Int Rev Immunol 2025:1-28. [PMID: 40242974 DOI: 10.1080/08830185.2025.2490233] [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: 04/03/2024] [Revised: 02/19/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025]
Abstract
Innate lymphoid cells (ILCs) have emerged as pivotal players in the field of immunology, expanding our understanding of innate immunity beyond conventional paradigms. This comprehensive review delves into the multifaceted world of ILCs, beginning with their serendipitous discovery and traversing their ontogeny and heterogeneity. We explore the distinct subsets of ILCs unraveling their intriguing plasticity, which adds a layer of complexity to their functional repertoire. As we journey through the functional activities of ILCs, we address their role in immune responses against various infections, categorizing their interactions with helminthic parasites, bacterial pathogens, fungal infections, and viral invaders. Notably, this review offers a detailed examination of ILCs in the context of specific infections, such as Mycobacterium tuberculosis, Citrobacter rodentium, Clostridium difficile, Salmonella typhimurium, Helicobacter pylori, Listeria monocytogenes, Staphylococcus aureus, Pseudomonas aeruginosa, Influenza virus, Cytomegalovirus, Herpes simplex virus, and severe acute respiratory syndrome coronavirus 2. This selection aimed for a comprehensive exploration of ILCs in various infectious contexts, opting for microorganisms based on extensive research findings rather than considerations of virulence or emergence. Furthermore, we raise intriguing questions about the potential immune functional resemblances between ILCs and epithelial cells, shedding light on their interconnectedness within the mucosal microenvironment. The review culminates in a critical assessment of the therapeutic prospects of targeting ILCs during infection, emphasizing their promise as novel immunotherapeutic targets. Nevertheless, due to their recent discovery and evolving understanding, effectively manipulating ILCs is challenging. Ensuring specificity and safety while evaluating long-term effects in clinical settings will be crucial.
Collapse
Affiliation(s)
- Wafa Nouari
- Laboratory of Applied Molecular Biology and Immunology, University of Tlemcen, Tlemcen, Algeria
| | - Mourad Aribi
- Laboratory of Applied Molecular Biology and Immunology, University of Tlemcen, Tlemcen, Algeria
| |
Collapse
|
2
|
Verma M, Randhawa S, Bathla M, Teji N, Acharya A. Strategic use of nanomaterials as double-edged therapeutics to control carcinogenesis via regulation of dysbiosis and bacterial infection: current status and future prospects. J Mater Chem B 2025; 13:4770-4790. [PMID: 40192037 DOI: 10.1039/d4tb02409e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The human microbiome plays a crucial role in modulating health and disease susceptibility through a complex network of interactions with the host. When the delicate balance of this microbial ecosystem is disrupted, it often correlates with the onset of systemic diseases. An over-abundance of pathogenic microorganisms within the microbiome has been implicated as a driving factor in the development of disease conditions such as diabetes, obesity, and chronic infections. It has been observed that microbiome dysbiosis perturbs metabolic, inflammatory, and immunological pathways, potentially facilitating carcinogenesis. Furthermore, the metabolites associated with microbial dysbiosis exert multifaceted effects, including metabolic interference, host DNA damage, and tumor promotion, further underscoring the microbiome's significance in several of the cancers. This new exploration of microbiome involvement in carcinogenesis needs additional patient sample analysis, which could provide new insights into cancer diagnosis and treatment. However, treating these diseases using drugs, traditional methods, etc. has resulted in multi-drug resistance, and this has eventually made the situation worrisome. This review highlights the importance of nanotechnology, which may tackle these pathogenic conditions simultaneously by targeting common receptors present in bacteria and cancer. Herein, we have explained how nanotechnology may come to the forefront for these treatments. It explores the potential of non-antibiotic disinfectants, i.e., nanoparticles (NPs) with dual targeting capabilities against microbes and cancer cells, using mechanisms such as ROS generation and DNA damage while minimizing the chances of drug resistance.
Collapse
Affiliation(s)
- Mohini Verma
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur (H.P.) 176061, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Shiwani Randhawa
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur (H.P.) 176061, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Manik Bathla
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur (H.P.) 176061, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Nandini Teji
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur (H.P.) 176061, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Amitabha Acharya
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur (H.P.) 176061, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad-201002, India
| |
Collapse
|
3
|
Yan Y, Satoh-Takayama N. New perspectives on gastric disorders: the relationship between innate lymphoid cells and microbes in the stomach. Cell Mol Life Sci 2025; 82:113. [PMID: 40074935 PMCID: PMC11904066 DOI: 10.1007/s00018-025-05632-w] [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: 12/26/2024] [Revised: 01/28/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025]
Abstract
A growing number of studies in recent years have revealed the changes in the gastric microbiota during the development of gastric diseases, breaking the stereotype that the stomach is hostile to microorganisms beyond H. pylori. After a decade of intensive research, the discovery of innate lymphoid cells (ILCs) has provided a new perspective on the immune response in many diseases. In the context of defense against infectious pathogens, the pre-existing innate defense mechanism of tissue-resident ILCs can rapidly recognize and respond to microbes to eliminate infection at the earliest stages. Here, we outline the basic function of ILCs in the gastric mucosa and in shaping the gastric microbiome. We discuss the interactions between the gastric microbiota and ILCs, explaining how the ILCs actively drive the immune response against bacterial pathogens that can lead to the development of the gastric disease.
Collapse
Affiliation(s)
- Yunzi Yan
- Precision Immune Regulation RIKEN ECL Research Unit, Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Naoko Satoh-Takayama
- Precision Immune Regulation RIKEN ECL Research Unit, Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan.
- Immunobiology Laboratory, Graduate School of Medical Life Sciences, Yokohama City University, Yokohama, Kanagawa, Japan.
| |
Collapse
|
4
|
Palkovsky M, Modrackova N, Neuzil-Bunesova V, Liberko M, Soumarova R. The Bidirectional Impact of Cancer Radiotherapy and Human Microbiome: Microbiome as Potential Anti-tumor Treatment Efficacy and Toxicity Modulator. In Vivo 2025; 39:37-54. [PMID: 39740900 PMCID: PMC11705129 DOI: 10.21873/invivo.13803] [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: 07/29/2024] [Revised: 10/02/2024] [Accepted: 10/14/2024] [Indexed: 01/02/2025]
Abstract
Microbiome and radiotherapy represent bidirectionally interacting entities. The human microbiome has emerged as a pivotal modulator of the efficacy and toxicity of radiotherapy; however, a reciprocal effect of radiotherapy on microbiome composition alterations has also been observed. This review explores the relationship between the microbiome and extracranial solid tumors, particularly focusing on the bidirectional impact of radiotherapy on organ-specific microbiome. This article aims to provide a systematic review on the radiotherapy-induced microbial alteration in-field as well as in distant microbiomes. In this review, particular focus is directed to the oral and gut microbiome, its role in the development and progression of cancer, and how it is altered throughout radiotherapy. This review concludes with recommendations for future research, such as exploring microbiome modification to optimize radiotherapy-induced toxicities or enhance its anti-cancer effects.
Collapse
Affiliation(s)
- Martin Palkovsky
- Department of Oncology, University Hospital Kralovske Vinohrady, Prague, Czech Republic;
- Charles University, Third Faculty of Medicine, Department of Oncology, Prague, Czech Republic
| | - Nikol Modrackova
- Czech University of Life Sciences Prague, Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Prague, Czech Republic
| | - Vera Neuzil-Bunesova
- Czech University of Life Sciences Prague, Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Prague, Czech Republic
| | - Marian Liberko
- Department of Oncology, University Hospital Kralovske Vinohrady, Prague, Czech Republic
- Charles University, Third Faculty of Medicine, Department of Oncology, Prague, Czech Republic
| | - Renata Soumarova
- Department of Oncology, University Hospital Kralovske Vinohrady, Prague, Czech Republic
- Charles University, Third Faculty of Medicine, Department of Oncology, Prague, Czech Republic
| |
Collapse
|
5
|
Lee JG. Microbiota, Gut Health, and Laryngopharyngeal Reflux Disease. Otolaryngol Clin North Am 2024. [DOI: 10.1016/j.otc.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
6
|
Chatterjee A, Kumar S, Roy Sarkar S, Halder R, Kumari R, Banerjee S, Sarkar B. Dietary polyphenols represent a phytotherapeutic alternative for gut dysbiosis associated neurodegeneration: A systematic review. J Nutr Biochem 2024; 129:109622. [PMID: 38490348 DOI: 10.1016/j.jnutbio.2024.109622] [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: 08/07/2023] [Revised: 03/04/2024] [Accepted: 03/09/2024] [Indexed: 03/17/2024]
Abstract
Globally, neurodegeneration and cerebrovascular disease are common and growing causes of morbidity and mortality. Pathophysiology of this group of diseases encompasses various factors from oxidative stress to gut microbial dysbiosis. The study of the etiology and mechanisms of oxidative stress as well as gut dysbiosis-induced neurodegeneration in Alzheimer's disease, Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis, autism spectrum disorder, and Huntington's disease has recently received a lot of attention. Numerous studies lend credence to the notion that changes in the intestinal microbiota and enteric neuroimmune system have an impact on the initiation and severity of these diseases. The prebiotic role of polyphenols can influence the makeup of the gut microbiota in neurodegenerative disorders by modulating intracellular signalling pathways. Metabolites of polyphenols function directly as neurotransmitters by crossing the blood-brain barrier or indirectly via influencing the cerebrovascular system. This assessment aims to bring forth an interlink between the consumption of polyphenols biotransformed by gut microbiota which in turn modulate the gut microbial diversity and biochemical changes in the brain. This systematic review will further augment research towards the association of dietary polyphenols in the management of gut dysbiosis-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Amrita Chatterjee
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Ranchi, Jharkhand, India
| | - Satish Kumar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Ranchi, Jharkhand, India
| | - Suparna Roy Sarkar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Ranchi, Jharkhand, India
| | - Ritabrata Halder
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Ranchi, Jharkhand, India
| | - Rashmi Kumari
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Ranchi, Jharkhand, India
| | - Sugato Banerjee
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Biswatrish Sarkar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Ranchi, Jharkhand, India.
| |
Collapse
|
7
|
Liu Z, Zhang D, Chen S. Unveiling the gastric microbiota: implications for gastric carcinogenesis, immune responses, and clinical prospects. J Exp Clin Cancer Res 2024; 43:118. [PMID: 38641815 PMCID: PMC11027554 DOI: 10.1186/s13046-024-03034-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/29/2024] [Indexed: 04/21/2024] Open
Abstract
High-throughput sequencing has ushered in a paradigm shift in gastric microbiota, breaking the stereotype that the stomach is hostile to microorganisms beyond H. pylori. Recent attention directed toward the composition and functionality of this 'community' has shed light on its potential relevance in cancer. The microbial composition in the stomach of health displays host specificity which changes throughout a person's lifespan and is subject to both external and internal factors. Distinctive alterations in gastric microbiome signature are discernible at different stages of gastric precancerous lesions and malignancy. The robust microbes that dominate in gastric malignant tissue are intricately implicated in gastric cancer susceptibility, carcinogenesis, and the modulation of immunosurveillance and immune escape. These revelations offer fresh avenues for utilizing gastric microbiota as predictive biomarkers in clinical settings. Furthermore, inter-individual microbiota variations partially account for differential responses to cancer immunotherapy. In this review, we summarize current literature on the influence of the gastric microbiota on gastric carcinogenesis, anti-tumor immunity and immunotherapy, providing insights into potential clinical applications.
Collapse
Affiliation(s)
- Zhiyi Liu
- Department of Oncology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Dachuan Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Siyu Chen
- Department of Oncology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China.
| |
Collapse
|
8
|
Fol M, Karpik W, Zablotni A, Kulesza J, Kulesza E, Godkowicz M, Druszczynska M. Innate Lymphoid Cells and Their Role in the Immune Response to Infections. Cells 2024; 13:335. [PMID: 38391948 PMCID: PMC10886880 DOI: 10.3390/cells13040335] [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: 01/05/2024] [Revised: 02/07/2024] [Accepted: 02/10/2024] [Indexed: 02/24/2024] Open
Abstract
Over the past decade, a group of lymphocyte-like cells called innate lymphoid cells (ILCs) has gained considerable attention due to their crucial role in regulating immunity and tissue homeostasis. ILCs, lacking antigen-specific receptors, are a group of functionally differentiated effector cells that act as tissue-resident sentinels against infections. Numerous studies have elucidated the characteristics of ILC subgroups, but the mechanisms controlling protective or pathological responses to pathogens still need to be better understood. This review summarizes the functions of ILCs in the immunology of infections caused by different intracellular and extracellular pathogens and discusses their possible therapeutic potential.
Collapse
Affiliation(s)
- Marek Fol
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (M.F.); (W.K.); (M.G.)
| | - Wojciech Karpik
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (M.F.); (W.K.); (M.G.)
| | - Agnieszka Zablotni
- Department of Bacterial Biology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland;
| | - Jakub Kulesza
- Department of Internal Diseases and Clinical Pharmacology, Medical University of Lodz, 91-347 Lodz, Poland;
| | - Ewelina Kulesza
- Department of Rheumatology and Internal Diseases, Medical University of Lodz, 90-549 Lodz, Poland;
| | - Magdalena Godkowicz
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (M.F.); (W.K.); (M.G.)
- Lodz Institutes of the Polish Academy of Sciences, The Bio-Med-Chem Doctoral School, University of Lodz, 90-237 Lodz, Poland
| | - Magdalena Druszczynska
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (M.F.); (W.K.); (M.G.)
| |
Collapse
|
9
|
Zheng Y, Han F, Ho A, Xue Y, Wu Z, Chen X, Sandberg JK, Ma S, Leeansyah E. Role of MAIT cells in gastrointestinal tract bacterial infections in humans: More than a gut feeling. Mucosal Immunol 2023; 16:740-752. [PMID: 37353006 DOI: 10.1016/j.mucimm.2023.06.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 06/25/2023]
Abstract
Mucosa-associated invariant T (MAIT) cells are the largest population of unconventional T cells in humans. These antimicrobial T cells are poised with rapid effector responses following recognition of the cognate riboflavin (vitamin B2)-like metabolite antigens derived from microbial riboflavin biosynthetic pathway. Presentation of this unique class of small molecule metabolite antigens is mediated by the highly evolutionarily conserved major histocompatibility complex class I-related protein. In humans, MAIT cells are widely found along the upper and lower gastrointestinal tracts owing to their high expression of chemokine receptors and homing molecules directing them to these tissue sites. In this review, we discuss recent findings regarding the roles MAIT cells play in various gastrointestinal bacterial infections, and how their roles appear to differ depending on the etiological agents and the anatomical location. We further discuss the potential mechanisms by which MAIT cells contribute to pathogen control, orchestrate adaptive immunity, as well as their potential contribution to inflammation and tissue damage during gastrointestinal bacterial infections, and the ensuing tissue repair following resolution. Finally, we propose and discuss the use of the emerging three-dimensional organoid technology to test different hypotheses regarding the role of MAIT cells in gastrointestinal bacterial infections, inflammation, and immunity.
Collapse
Affiliation(s)
- Yichao Zheng
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China; Precision Medicine and Healthcare Research Centre, Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, China
| | - Fei Han
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Amanda Ho
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China; Precision Medicine and Healthcare Research Centre, Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, China
| | - Yiting Xue
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China; Precision Medicine and Healthcare Research Centre, Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, China
| | - Zhengyu Wu
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Xingchi Chen
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Johan K Sandberg
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Shaohua Ma
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China; Precision Medicine and Healthcare Research Centre, Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, China
| | - Edwin Leeansyah
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.
| |
Collapse
|
10
|
Wang R, Cui W, Yang H. The interplay between innate lymphoid cells and microbiota. mBio 2023; 14:e0039923. [PMID: 37318214 PMCID: PMC10470585 DOI: 10.1128/mbio.00399-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/21/2023] [Indexed: 06/16/2023] Open
Abstract
Innate lymphoid cells (ILCs) are mainly resident in mucosal tissues such as gastrointestinal tract and respiratory tract, so they are closely linked to the microbiota. ILCs can protect commensals to maintain homeostasis and increase resistance to pathogens. Moreover, ILCs also play an early role in defense against a variety of pathogenic microorganisms including pathogenic bacteria, viruses, fungi and parasites, before the intervention of adaptive immune system. Due to the lack of adaptive antigen receptors expressed on T cells and B cells, ILCs need to use other means to sense the signals of microbiota and play a role in corresponding regulation. In this review, we focus on and summarize three major mechanisms used in the interaction between ILCs and microbiota: the mediation of accessory cells represented by dendritic cells; the metabolic pathways of microbiota or diet; the participation of adaptive immune cells.
Collapse
Affiliation(s)
- Rui Wang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Wenwen Cui
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Huan Yang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
11
|
Zhou S, Li C, Liu L, Yuan Q, Miao J, Wang H, Ding C, Guan W. Gastric microbiota: an emerging player in gastric cancer. Front Microbiol 2023; 14:1130001. [PMID: 37180252 PMCID: PMC10172576 DOI: 10.3389/fmicb.2023.1130001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/04/2023] [Indexed: 05/16/2023] Open
Abstract
Gastric cancer (GC) is a common cancer worldwide with a high mortality rate. Many microbial factors influence GC, of which the most widely accepted one is Helicobacter pylori (H. pylori) infection. H. pylori causes inflammation, immune reactions and activation of multiple signaling pathways, leading to acid deficiency, epithelial atrophy, dysplasia and ultimately GC. It has been proved that complex microbial populations exist in the human stomach. H. pylori can affect the abundance and diversity of other bacteria. The interactions among gastric microbiota are collectively implicated in the onset of GC. Certain intervention strategies may regulate gastric homeostasis and mitigate gastric disorders. Probiotics, dietary fiber, and microbiota transplantation can potentially restore healthy microbiota. In this review, we elucidate the specific role of the gastric microbiota in GC and hope these data can facilitate the development of effective prevention and therapeutic approaches for GC.
Collapse
Affiliation(s)
- Shizhen Zhou
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Chenxi Li
- Laboratory Medicine Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lixiang Liu
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qinggang Yuan
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, Jiangsu, China
| | - Ji Miao
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Hao Wang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Chao Ding
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wenxian Guan
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| |
Collapse
|
12
|
Garvey M. The Association between Dysbiosis and Neurological Conditions Often Manifesting with Chronic Pain. Biomedicines 2023; 11:biomedicines11030748. [PMID: 36979726 PMCID: PMC10045203 DOI: 10.3390/biomedicines11030748] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/21/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
The prevalence of neurological conditions which manifest with chronic pain is increasing globally, where the World Health Organisation has now classified chronic pain as a risk factor for death by suicide. While many chronic pain conditions have a definitive underlying aetiology, non-somatic conditions represent difficult-to-diagnose and difficult-to-treat public health issues. The interaction of the immune system and nervous system has become an important area in understanding the occurrence of neuroinflammation, nociception, peripheral and central sensitisation seen in chronic pain. More recently, however, the role of the resident microbial species in the human gastrointestinal tract has become evident. Dysbiosis, an alteration in the microbial species present in favour of non-beneficial and pathogenic species has emerged as important in many chronic pain conditions, including functional somatic syndromes, autoimmune disease and neurological diseases. In particular, a decreased abundance of small chain fatty acid, e.g., butyrate-producing bacteria, including Faecalibacterium, Firmicutes and some Bacteroides spp., is frequently evident in morbidities associated with long-term pain. Microbes involved in the production of neurotransmitters serotonin, GABA, glutamate and dopamine, which mediate the gut-brain, axis are also important. This review outlines the dysbiosis present in many disease states manifesting with chronic pain, where an overlap in morbidities is also frequently present in patients.
Collapse
Affiliation(s)
- Mary Garvey
- Department of Life Science, Atlantic Technological University, F91 YW50 Sligo, Ireland; ; Tel.: +353-071-9305529
- Centre for Precision Engineering, Materials and Manufacturing Research (PEM), Atlantic Technological University, F91 YW50 Sligo, Ireland
| |
Collapse
|
13
|
Thekkekkara D, Manjula SN, Mishra N, Bhatt S, Shilpi S. Synbiotics in the Management of Breast Cancer. SYNBIOTICS FOR THE MANAGEMENT OF CANCER 2023:289-304. [DOI: 10.1007/978-981-19-7550-9_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Gut Microbiota Host-Gene Interaction. Int J Mol Sci 2022; 23:ijms232213717. [PMID: 36430197 PMCID: PMC9698405 DOI: 10.3390/ijms232213717] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/28/2022] [Accepted: 11/07/2022] [Indexed: 11/09/2022] Open
Abstract
Studies carried out in the last ten years have shown that the metabolites made up from the gut microbiota are essential for multiple functions, such as the correct development of the immune system of newborns, interception of pathogens, and nutritional enrichment of the diet. Therefore, it is not surprising that alteration of the gut microbiota is the starting point of gastrointestinal infection, obesity, type 2 diabetes, inflammatory bowel disease, colorectal cancer, and lung cancer. Diet changes and antibiotics are the major factors damaging the gut microbiota. Early exposure of the newborns to antibiotics may prevent their correct development of the immune system, exposing them to pathogen infections, allergies, and chronic inflammatory diseases. We already know much on how host genes, microbiota, and the environment interact, owing to experiments in several model animals, especially in mice; advances in molecular technology; microbiota transplantation; and comparative metagenomic analysis. However, much more remains to be known. Longitudinal studies on patients undergoing to therapy, along with the identification of bacteria prevalent in responding patients may provide valuable data for improving therapies.
Collapse
|
15
|
Juarez VM, Montalbine AN, Singh A. Microbiome as an immune regulator in health, disease, and therapeutics. Adv Drug Deliv Rev 2022; 188:114400. [PMID: 35718251 PMCID: PMC10751508 DOI: 10.1016/j.addr.2022.114400] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 05/11/2022] [Accepted: 06/12/2022] [Indexed: 11/27/2022]
Abstract
New discoveries in drugs and drug delivery systems are focused on identifying and delivering a pharmacologically effective agent, potentially targeting a specific molecular component. However, current drug discovery and therapeutic delivery approaches do not necessarily exploit the complex regulatory network of an indispensable microbiota that has been engineered through evolutionary processes in humans or has been altered by environmental exposure or diseases. The human microbiome, in all its complexity, plays an integral role in the maintenance of host functions such as metabolism and immunity. However, dysregulation in this intricate ecosystem has been linked with a variety of diseases, ranging from inflammatory bowel disease to cancer. Therapeutics and bacteria have an undeniable effect on each other and understanding the interplay between microbes and drugs could lead to new therapies, or to changes in how existing drugs are delivered. In addition, targeting the human microbiome using engineered therapeutics has the potential to address global health challenges. Here, we present the challenges and cutting-edge developments in microbiome-immune cell interactions and outline novel targeting strategies to advance drug discovery and therapeutics, which are defining a new era of personalized and precision medicine.
Collapse
Affiliation(s)
- Valeria M Juarez
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States
| | - Alyssa N Montalbine
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States
| | - Ankur Singh
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, United States.
| |
Collapse
|
16
|
Fakharian F, Asgari B, Nabavi-Rad A, Sadeghi A, Soleimani N, Yadegar A, Zali MR. The interplay between Helicobacter pylori and the gut microbiota: An emerging driver influencing the immune system homeostasis and gastric carcinogenesis. Front Cell Infect Microbiol 2022; 12:953718. [PMID: 36046747 PMCID: PMC9423097 DOI: 10.3389/fcimb.2022.953718] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/25/2022] [Indexed: 01/06/2023] Open
Abstract
The human gut microbiota are critical for preserving the health status because they are required for digestion and nutrient acquisition, the development of the immune system, and energy metabolism. The gut microbial composition is greatly influenced by the colonization of the recalcitrant pathogen Helicobacter pylori (H. pylori) and the conventional antibiotic regimens that follow. H. pylori is considered to be the main microorganism in gastric carcinogenesis, and it appears to be required for the early stages of the process. However, a non-H. pylori microbiota profile is also suggested, primarily in the later stages of tumorigenesis. On the other hand, specific groups of gut microbes may produce beneficial byproducts such as short-chain fatty acids (acetate, butyrate, and propionate) that can modulate inflammation and tumorigenesis pathways. In this review, we aim to present how H. pylori influences the population of the gut microbiota to modify the host immunity and trigger the development of gastric carcinogenesis. We will also highlight the effect of the gut microbiota on immunotherapeutic approaches such as immune checkpoint blockade in cancer treatment to present a perspective for further development of innovative therapeutic paradigms to prevent the progression of H. pylori-induced stomach cancer.
Collapse
Affiliation(s)
- Farzaneh Fakharian
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnoush Asgari
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Nabavi-Rad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Soleimani
- Department of Microbiology, Faculty of Biological Sciences and Technology, Shahid Beheshti University, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Wang L, Li F, Gu B, Qu P, Liu Q, Wang J, Tang J, Cai S, Zhao Q, Ming Z. Metaomics in Clinical Laboratory: Potential Driving Force for Innovative Disease Diagnosis. Front Microbiol 2022; 13:883734. [PMID: 35783436 PMCID: PMC9247514 DOI: 10.3389/fmicb.2022.883734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Currently, more and more studies suggested that reductionism was lack of holistic and integrative view of biological processes, leading to limited understanding of complex systems like microbiota and the associated diseases. In fact, microbes are rarely present in individuals but normally live in complex multispecies communities. With the recent development of a variety of metaomics techniques, microbes could be dissected dynamically in both temporal and spatial scales. Therefore, in-depth understanding of human microbiome from different aspects such as genomes, transcriptomes, proteomes, and metabolomes could provide novel insights into their functional roles, which also holds the potential in making them diagnostic biomarkers in many human diseases, though there is still a huge gap to fill for the purpose. In this mini-review, we went through the frontlines of the metaomics techniques and explored their potential applications in clinical diagnoses of human diseases, e.g., infectious diseases, through which we concluded that novel diagnostic methods based on human microbiomes shall be achieved in the near future, while the limitations of these techniques such as standard procedures and computational challenges for rapid and accurate analysis of metaomics data in clinical settings were also examined.
Collapse
Affiliation(s)
- Liang Wang
- Department of Bioinformatics, School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, China
| | - Fen Li
- Department of Laboratory Medicine, Huaiyin Hospital, Huai’an, China
| | - Bin Gu
- Department of Bioinformatics, School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, China
| | - Pengfei Qu
- The First School of Clinical Medicine, Xuzhou Medical University, Xuzhou, China
| | - Qinghua Liu
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macao SAR, China
| | - Junjiao Wang
- Department of Bioinformatics, School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, China
| | - Jiawei Tang
- Department of Bioinformatics, School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, China
| | - Shubin Cai
- College of Computer Science and Software Engineering, Shenzhen University, Shenzhen, China
| | - Qi Zhao
- School of Computer Science and Software Engineering, University of Science and Technology Liaoning, Anshan, China
- *Correspondence: Qi Zhao,
| | - Zhong Ming
- College of Computer Science and Software Engineering, Shenzhen University, Shenzhen, China
- Zhong Ming,
| |
Collapse
|
18
|
Safikhani Mahmoodzadeh A, Moazamian E, Shamsdin SA, Kaydani GA. Prevalence of Virulence Genes and Antigen Pattern in Helicobacter pylori-Infected Patients and the Level of Some Inflammatory Cytokines Compared with Non-infected Individuals. Jundishapur J Microbiol 2022; 15. [DOI: 10.5812/jjm-121144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 02/22/2022] [Accepted: 04/05/2022] [Indexed: 01/04/2025] Open
Abstract
Background: The worldwide prevalence of Helicobacter pylori is about 50%. This bacterium needs a number of virulence factors for pathogenesis. Objectives: This study aimed to determine the prevalence of virulence genes (ureB, cytotoxin-associated gene A [cagA], and vacuolating cytotoxin [vacA]), as well as the antigenic profile in H. pylori strains. Methods: Eighty-five patients with abdominal pain, including 46 H. pylori-positive and 39 H. pylori-negative cases, were enrolled in this study. The serum levels of interleukin (IL)-17F, tumor necrosis factor α (TNF-α), and interferon γ (IFN-γ) cytokines were measured by multiplex kits and flow cytometry. After molecular identification by the ureC gene, vacA, cagA, and ureB genes were detected by polymerase chain reaction (PCR). Finally, after antigenic extraction, the whole-cell protein was exhibited by sodium dodecyl sulphate–polyacrylamide gel electrophoresis (SDS-PAGE). Results: The prevalence of vacA, ureB, and cagA genes were 91.3%, 67.39%, and 50%, respectively. The frequency of genes and cell surface antigens were not significantly different based on the gastritis severity (P > 0.05). IL-17F significantly (P = 0.046) increased in the presence of 19.5 kDa (outer membrane protein [OMP]). Moreover, the OMP antigen significantly enhanced immunoglobulin A (IgA; P = 0.013). In the presence of the 66-kDa (ureB) antigen, the serum level of IFN-γ increased (p = 0.041). Finally, the CagA protein led to increased IgG antibody levels (p = 0.027). Conclusions: Early detection of H. pylori infection can play a crucial role in managing it. Our results suggest that IL-17F, TNF-α, and IFN-γ cytokines could be diagnostic markers. However, further studies are required to fully investigate this suggestion.
Collapse
|
19
|
The Implication of Gastric Microbiome in the Treatment of Gastric Cancer. Cancers (Basel) 2022; 14:cancers14082039. [PMID: 35454944 PMCID: PMC9028069 DOI: 10.3390/cancers14082039] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Gastric cancer (GC) represents the fifth most common cancer worldwide. Recent developments in PCR and metagenomics clarify that the stomach contains a powerful microbiota. Conventional treatments for GC that include surgery, chemotherapy, and radiotherapy are not very effective. That’s why new therapeutic strategies are needed. The intestinal microbiota is involved in oncogenesis and cancer prevention, and the effectiveness of chemotherapy. Recent studies have shown that certain bacteria may enhance the effect of some traditional antineoplastic drugs and immunotherapies. Abstract Gastric cancer (GC) is one of the most common and deadly malignancies worldwide. Helicobacter pylori have been documented as a risk factor for GC. The development of sequencing technology has broadened the knowledge of the gastric microbiome, which is essential in maintaining homeostasis. Recent studies have demonstrated the involvement of the gastric microbiome in the development of GC. Therefore, the elucidation of the mechanism by which the gastric microbiome contributes to the development and progression of GC may improve GC’s prevention, diagnosis, and treatment. In this review, we discuss the current knowledge about changes in gastric microbial composition in GC patients, their role in carcinogenesis, the possible therapeutic role of the gastric microbiome, and its implications for current GC therapy.
Collapse
|
20
|
Wang H, Li J, Wu G, Zhang F, Yin J, He Y. The effect of intrinsic factors and mechanisms in shaping human gut microbiota. MEDICINE IN MICROECOLOGY 2022. [DOI: 10.1016/j.medmic.2022.100054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
21
|
Thrastardottir TO, Copeland VJ, Constantinou C. The Association Between the Gut Microbiome, Nutritional Habits, Antibiotics, and Gastric Cancer: a Scoping Review. Curr Nutr Rep 2022; 11:19-38. [PMID: 35020173 DOI: 10.1007/s13668-021-00391-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2021] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW: Gastric cancer (GC) represents one of the leading causes of cancer-related deaths worldwide. The prevalence of GC among the younger population has been increasing in recent years, and the latter is associated with late detection and advanced disease status. The factors involved in the development of GC have been the focus of investigation in the past few years; yet no consistent conclusions or preventative solutions have been suggested. This scoping review aims to explore and report on research studies conducted to investigate the association between GC, the gut microbiome, and nutritional habits. RECENT FINDINGS : Using a predefined protocol in compliance with the PRISMA guidelines, a search was conducted on four separate databases including Google Scholar, Cochrane Library, ProQuest, and PubMed to investigate the association between GC, microbiome, and nutrition. A total of 1219 articles were identified through this process. Forty-three articles met the initial screening criteria and following full-article analysis, and 10 articles met the full inclusion criteria and were included in the current review. The results of the study support that there are differences in the gut microbiota between GC patients and healthy controls and that GC patients may have increased microbiota richness and lower diversity compared to healthy controls. In addition, the detection of pre/early GC may be possible through the analysis of the microbiota using fecal sampling. Nutritional habits, probiotics, and antibiotics may also play an important role in affecting GC development. Preliminary studies support a role for the gut microbiota in developing GC. Yet, the lack of sufficient randomized controlled studies investigating the association between GC, the gut microbiota, and nutritional habits demonstrates the need for further clinical research to develop preventative strategies that will aim to reduce the increased incidence of GC among all age groups including younger populations.
Collapse
Affiliation(s)
- Tinna Osk Thrastardottir
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, 21 Ilia Papakyriakou, 2414 Engomi, P.O. Box 24005, 1700, Nicosia, Cyprus
| | - Viana Jacquline Copeland
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, 21 Ilia Papakyriakou, 2414 Engomi, P.O. Box 24005, 1700, Nicosia, Cyprus
| | - Constantina Constantinou
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, 21 Ilia Papakyriakou, 2414 Engomi, P.O. Box 24005, 1700, Nicosia, Cyprus.
| |
Collapse
|
22
|
Interdisciplinary insights into the link between gut microbiome and gastric carcinogenesis-what is currently known? Gastric Cancer 2022; 25:1-10. [PMID: 34741681 PMCID: PMC8732854 DOI: 10.1007/s10120-021-01260-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/20/2021] [Indexed: 02/07/2023]
Abstract
Currently, gastric cancer is one of the leading death-related cancer globally. The etiopathogenesis of gastric cancer is multifactorial and includes among others dysbiotic alterations of gastric microbiota. Molecular techniques revealed that stomach is not a sterile organ and it is resides with ecosystem of microbes. Due to the fact that the role of Helicobacter pylori infection in development of gastric cancer is established and well-studied, this paper is mainly focused on the role of other bacterial as well as viral and fungal gut microbiota imbalance in gastric carcinogenesis. Notably, not only the composition of gastric microbiota may play an important role in development of gastric cancer, but also its activity. Microbial metabolites, such as short-chain fatty acids, polyamines, N-nitroso compounds, and lactate, may significantly affect gastric carcinogenesis. Therefore, this paper discussed aforementioned aspects with the interdisciplinary insights (regarding also immunological point of view) into the association between gut microbiome and gastric carcinogenesis based on up-to-date studies.
Collapse
|
23
|
Nabavi-Rad A, Sadeghi A, Asadzadeh Aghdaei H, Yadegar A, Smith SM, Zali MR. The double-edged sword of probiotic supplementation on gut microbiota structure in Helicobacter pylori management. Gut Microbes 2022; 14:2108655. [PMID: 35951774 PMCID: PMC9373750 DOI: 10.1080/19490976.2022.2108655] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/13/2022] [Accepted: 07/29/2022] [Indexed: 02/04/2023] Open
Abstract
As Helicobacter pylori management has become more challenging and less efficient over the last decade, the interest in innovative interventions is growing by the day. Probiotic co-supplementation to antibiotic therapies is reported in several studies, presenting a moderate reduction in drug-related side effects and a promotion in positive treatment outcomes. However, the significance of gut microbiota involvement in the competence of probiotic co-supplementation is emphasized by a few researchers, indicating the alteration in the host gastrointestinal microbiota following probiotic and drug uptake. Due to the lack of long-term follow-up studies to determine the efficiency of probiotic intervention in H. pylori eradication, and the delicate interaction of the gut microbiota with the host wellness, this review aims to discuss the gut microbiota alteration by probiotic co-supplementation in H. pylori management to predict the comprehensive effectiveness of probiotic oral administration.Abbreviations: acyl-CoA- acyl-coenzyme A; AMP- antimicrobial peptide; AMPK- AMP-activated protein kinase; AP-1- activator protein 1; BA- bile acid; BAR- bile acid receptor; BCAA- branched-chain amino acid; C2- acetate; C3- propionate; C4- butyrate; C5- valeric acid; CagA- Cytotoxin-associated gene A; cAMP- cyclic adenosine monophosphate; CD- Crohn's disease; CDI- C. difficile infection; COX-2- cyclooxygenase-2; DC- dendritic cell; EMT- epithelial-mesenchymal transition; FMO- flavin monooxygenases; FXR- farnesoid X receptor; GPBAR1- G-protein-coupled bile acid receptor 1; GPR4- G protein-coupled receptor 4; H2O2- hydrogen peroxide; HCC- hepatocellular carcinoma; HSC- hepatic stellate cell; IBD- inflammatory bowel disease; IBS- irritable bowel syndrome; IFN-γ- interferon-gamma; IgA immunoglobulin A; IL- interleukin; iNOS- induced nitric oxide synthase; JAK1- janus kinase 1; JAM-A- junctional adhesion molecule A; LAB- lactic acid bacteria; LPS- lipopolysaccharide; MALT- mucosa-associated lymphoid tissue; MAMP- microbe-associated molecular pattern; MCP-1- monocyte chemoattractant protein-1; MDR- multiple drug resistance; mTOR- mammalian target of rapamycin; MUC- mucin; NAFLD- nonalcoholic fatty liver disease; NF-κB- nuclear factor kappa B; NK- natural killer; NLRP3- NLR family pyrin domain containing 3; NOC- N-nitroso compounds; NOD- nucleotide-binding oligomerization domain; PICRUSt- phylogenetic investigation of communities by reconstruction of unobserved states; PRR- pattern recognition receptor; RA- retinoic acid; RNS- reactive nitrogen species; ROS- reactive oxygen species; rRNA- ribosomal RNA; SCFA- short-chain fatty acids; SDR- single drug resistance; SIgA- secretory immunoglobulin A; STAT3- signal transducer and activator of transcription 3; T1D- type 1 diabetes; T2D- type 2 diabetes; Th17- T helper 17; TLR- toll-like receptor; TMAO- trimethylamine N-oxide; TML- trimethyllysine; TNF-α- tumor necrosis factor-alpha; Tr1- type 1 regulatory T cell; Treg- regulatory T cell; UC- ulcerative colitis; VacA- Vacuolating toxin A.
Collapse
Affiliation(s)
- Ali Nabavi-Rad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sinéad Marian Smith
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Retnakumar R, Nath AN, Nair GB, Chattopadhyay S. Gastrointestinal microbiome in the context of Helicobacter pylori infection in stomach and gastroduodenal diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 192:53-95. [DOI: 10.1016/bs.pmbts.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
25
|
Mao LQ, Zhou YL, Wang SS, Chen L, Hu Y, Yu LM, Xu JM, Lyu B. Impact of Helicobacter pylori eradication on the gastric microbiome. Gut Pathog 2021; 13:60. [PMID: 34645495 PMCID: PMC8513236 DOI: 10.1186/s13099-021-00460-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 10/07/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Helicobacter pylori (Hp) eradication has been used for many years. Yet, the impact of this eradication on the normal gastric microflora is not well understood. In this study, we explored the effect of eradication on the stomach microbial community and its recovery after successful Hp eradication. METHODS Among the 89 included patients, 23, 17, 40, and 9 were included in the Hp-negative, Hp-positive, successful eradication, and failed eradication groups, respectively. Four subgroups were further determined according to disease status (Hp-negative chronic gastritis [N-CG], Hp-negative atrophic gastritis [N-AG], successful-eradication chronic gastritis [SE-CG], and atrophic gastritis with successful eradication [SE-AG]). During the endoscopic examination, one piece of gastric mucosa tissue was obtained from the lesser curvature side of the gastric antrum and gastric corpus, respectively. In addition, 16S rDNA gene sequencing was used to analyze the gastric mucosal microbiome. RESULTS In the Hp-negative group, the gastric microbiota was dominated by five phyla: Firmicutes, Proteobacteria, Actinobacteria, Bacteroidetes, and Fusobacteria. After successfully eradicating Hp, the bacterial flora in the stomach recovered to a considerable extent. In the failed eradication group, the flora was similar to the flora in Hp-positive subjects based on the alpha and beta diversities. Among the groups, Curvibacter and Acinetobacter were enriched in the presence of Hp (i.e., failed eradication and Hp-positive groups), suggesting that these two genera could be used as biomarkers in the symbiotic flora in the presence of Hp. SE-CG was characterized by an increase in Firmicutes taxa and a decrease in Proteobacteria taxa compared with N-CG. SE-AG was characterized by a decrease in Firmicutes relative to N-AG. Finally, no differences were found in the pairwise comparisons of nitrate and nitrite reductase functions of the microflora among the four subgroups. CONCLUSIONS After Hp infection, the diversity and relative abundance of gastric microflora were significantly decreased. Yet, gastric microbiota could be partially restored to the Hp-negative status after eradication. Still, this effect was incomplete and might contribute to the long-term risks.
Collapse
Affiliation(s)
- Li-Qi Mao
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Department of Gastroenterology, The First People's Hospital of Huzhou, The First Affiliated Hospital of Huzhou Teachers College, Huzhou, China
| | - Yan-Lin Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuang-Shuang Wang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Department of Gastroenterology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, China
| | - Lin Chen
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yue Hu
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Lei-Min Yu
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Department of Gastroenterology, Guangxing Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Jing-Ming Xu
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Bin Lyu
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
26
|
Serrano C, Harris PR, Smith PD, Bimczok D. Interactions between H. pylori and the Gastric Microbiome: Impact on Gastric Homeostasis and Disease. CURRENT OPINION IN PHYSIOLOGY 2021; 21:57-64. [PMID: 34113748 PMCID: PMC8186273 DOI: 10.1016/j.cophys.2021.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Like many seemingly inhospitable environments on our planet, the highly acidic human stomach harbors a diverse bacterial microflora. The best-known member of the human gastric flora, Helicobacter pylori, causes a number of gastric diseases, including peptic ulcer disease and gastric adenocarcinoma. In the absence of Helicobacter pylori infection, the gastric microbiota displays some features similar to the oral cavity with Firmicutes the most common phylum, followed by Proteobacteria and Bacteroidetes. When present, H. pylori dominates the gastric microbiome and reduces diversity and composition of other taxa. The composition of the gastric microbiome also is altered in the setting of proton pump inhibitor therapy and gastric neoplasia. This review summarizes foundational and recent studies that have investigated the composition of the human gastric microbiome in a variety of patient groups, with a focus on potential mechanisms involved in regulation of gastric microbial community structure.
Collapse
Affiliation(s)
- Carolina Serrano
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paul R. Harris
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Phillip D. Smith
- Department of Medicine, Division of Gastroenterology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Diane Bimczok
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717
| |
Collapse
|
27
|
Sharma A, Im SH. Special issue on the human microbiome: from symbiosis to therapy. Exp Mol Med 2020; 52:1361-1363. [PMID: 32908210 PMCID: PMC8080605 DOI: 10.1038/s12276-020-00488-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 12/26/2022] Open
Affiliation(s)
- Amit Sharma
- Department of Life Sciences, Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Sin-Hyeog Im
- Department of Life Sciences, Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Gyeongbuk, 37673, Republic of Korea.
- ImmunoBiome Inc. POSTECH Biotech Center, Pohang, 37673, Republic of Korea.
| |
Collapse
|