1
|
Kou R, Mi F, Peng C, Ding X, Meng C, Liu F, Xiong L. Structural characterization and immunomodulatory activity of polysaccharides from the lateral roots of Aconitum carmichaelii. Int J Biol Macromol 2024; 282:136935. [PMID: 39490860 DOI: 10.1016/j.ijbiomac.2024.136935] [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: 08/03/2024] [Revised: 09/27/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Two polysaccharides, named FPS1-1 and FPS1-2, were separated from the neutral polysaccharides of the lateral roots of Aconitum carmichaelii, a widely used traditional Chinese medicine (Fuzi in Chinese). The monosaccharide composition analysis indicated that both FPS1-1 and FPS1-2 were glucans. However, further physicochemical analysis of FPS1-1 and FPS1-2 revealed distinct properties between the two glucans. FPS1-1 had a molecular weight (Mw) of 106.23 kDa with a spherical conformation, while FPS1-2 had a lower Mw of 19.23 kDa with a random coil conformation. The structure of FPS1-2 was further determined as a glucan whose backbone structure was composed of →4)-α-D-Glcp-(1→. The immunological activities of two polysaccharides were evaluated by a cyclophosphamide (CTX)-induced immunodeficiency model in mice. The result showed that FPS1-2 could restore CTX-induced immunosuppression by modulating CD4+ T cells differentiation and promoting cytokine secretion. Notably, FPS1-2 could modulate the colonic short-chain fatty acid (SCFA) levels and reverse the gut microbial dysbiosis induced by CTX. These findings reveal the potential benefits of Fuzi polysaccharides and provide evidences for developing immunologically functional products from Fuzi polysaccharides.
Collapse
Affiliation(s)
- Renbo Kou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Fuxin Mi
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Xingjie Ding
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chunwang Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Fei Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Liang Xiong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; School of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
2
|
Luesma MJ, López-Marco L, Monzón M, Santander S. Enteric Nervous System and Its Relationship with Neurological Diseases. J Clin Med 2024; 13:5579. [PMID: 39337066 PMCID: PMC11433641 DOI: 10.3390/jcm13185579] [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: 07/26/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
The enteric nervous system (ENS) is a fundamental component of the gastrointestinal system, composed of a vast network of neurons and glial cells. It operates autonomously but is interconnected with the central nervous system (CNS) through the vagus nerve. This communication, known as the gut-brain axis, influences the bidirectional communication between the brain and the gut. Background/Objectives: This study aimed to review neurological pathologies related to the ENS. Methods: To this end, a comprehensive literature search was conducted in the "PubMed" database. Articles available in "free format" were selected, applying the filters "Humans" and limiting the search to publications from the last ten years. Results: The ENS has been linked to various neurological diseases, from autism spectrum disorder to Parkinson's disease including neurological infection with the varicella zoster virus (VZV), even sharing pathologies with the CNS. This finding suggests that the ENS could serve as an early diagnostic marker or therapeutic target for neurological diseases. Gastrointestinal symptoms often precede CNS symptoms, and the ENS's accessibility aids in diagnosis and treatment. Parkinson's patients may show intestinal lesions up to twenty years before CNS symptoms, underscoring the potential for early diagnosis. However, challenges include developing standardized diagnostic protocols and the uneven distribution of dopaminergic neurons in the ENS. Continued research is needed to explore the ENS's potential in improving disease prognosis. Conclusions: The ENS is a promising area for early diagnosis and therapeutic development. Nevertheless, it is essential to continue research in this area, especially to gain a deeper understanding of its organization, function, and regenerative capacity.
Collapse
Affiliation(s)
- María José Luesma
- Department of Human Anatomy and Histology, University of Zaragoza, 50009 Zaragoza, Spain
| | - Liberto López-Marco
- Department of Human Anatomy and Histology, University of Zaragoza, 50009 Zaragoza, Spain
| | - Marta Monzón
- Department of Human Anatomy and Histology, University of Zaragoza, 50009 Zaragoza, Spain
| | - Sonia Santander
- Department of Pharmacology, Physiology, Legal and Forensic Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| |
Collapse
|
3
|
Tewari N, Dey P. Navigating commensal dysbiosis: Gastrointestinal host-pathogen interplay orchestrating opportunistic infections. Microbiol Res 2024; 286:127832. [PMID: 39013300 DOI: 10.1016/j.micres.2024.127832] [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: 04/22/2024] [Revised: 06/23/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024]
Abstract
The gut commensals, which are usually symbiotic or non-harmful bacteria that live in the gastrointestinal tract, have a positive impact on the health of the host. This review, however, specifically discuss distinct conditions where commensals aid in the development of pathogenic opportunistic infections. We discuss that the categorization of gut bacteria as either pathogens or non-pathogens depends on certain circumstances, which are significantly affected by the tissue microenvironment and the dynamic host-microbe interaction. Under favorable circumstances, commensals have the ability to transform into opportunistic pathobionts by undergoing overgrowth. These conditions include changes in the host's physiology, simultaneous infection with other pathogens, effective utilization of nutrients, interactions between different species of bacteria, the formation of protective biofilms, genetic mutations that enhance pathogenicity, acquisition of genes associated with virulence, and the ability to avoid the host's immune response. These processes allow commensals to both initiate infections themselves and aid other pathogens in populating the host. This review highlights the need of having a detailed and sophisticated knowledge of the two-sided nature of gut commensals. Although commensals mostly promote health, they may also become harmful in certain changes in the environment or the body's functioning. This highlights the need of acknowledging the intricate equilibrium in interactions between hosts and microbes, which is crucial for preserving intestinal homeostasis and averting diseases. Finally, we also emphasize the further need of research to better understand and anticipate the behavior of gut commensals in different situations, since they play a crucial and varied role in human health and disease.
Collapse
Affiliation(s)
- Nisha Tewari
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab 147004, India
| | - Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab 147004, India.
| |
Collapse
|
4
|
Galeana-Cadena D, Gómez-García IA, Lopez-Salinas KG, Irineo-Moreno V, Jiménez-Juárez F, Tapia-García AR, Boyzo-Cortes CA, Matías-Martínez MB, Jiménez-Alvarez L, Zúñiga J, Camarena A. Winds of change a tale of: asthma and microbiome. Front Microbiol 2023; 14:1295215. [PMID: 38146448 PMCID: PMC10749662 DOI: 10.3389/fmicb.2023.1295215] [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: 09/15/2023] [Accepted: 11/15/2023] [Indexed: 12/27/2023] Open
Abstract
The role of the microbiome in asthma is highlighted, considering its influence on immune responses and its connection to alterations in asthmatic patients. In this context, we review the variables influencing asthma phenotypes from a microbiome perspective and provide insights into the microbiome's role in asthma pathogenesis. Previous cohort studies in patients with asthma have shown that the presence of genera such as Bifidobacterium, Lactobacillus, Faecalibacterium, and Bacteroides in the gut microbiome has been associated with protection against the disease. While, the presence of other genera such as Haemophilus, Streptococcus, Staphylococcus, and Moraxella in the respiratory microbiome has been implicated in asthma pathogenesis, indicating a potential link between microbial dysbiosis and the development of asthma. Furthermore, respiratory infections have been demonstrated to impact the composition of the upper respiratory tract microbiota, increasing susceptibility to bacterial diseases and potentially triggering asthma exacerbations. By understanding the interplay between the microbiome and asthma, valuable insights into disease mechanisms can be gained, potentially leading to the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- David Galeana-Cadena
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | - Itzel Alejandra Gómez-García
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Karen Gabriel Lopez-Salinas
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Valeria Irineo-Moreno
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Fabiola Jiménez-Juárez
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Alan Rodrigo Tapia-García
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Red de Medicina para la Educación, el Desarrollo y la Investigación Científica de Iztacala, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Carlos Alberto Boyzo-Cortes
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | - Melvin Barish Matías-Martínez
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Luis Jiménez-Alvarez
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | - Joaquín Zúñiga
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Angel Camarena
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| |
Collapse
|
5
|
Guha L, Agnihotri TG, Jain A, Kumar H. Gut microbiota and traumatic central nervous system injuries: Insights into pathophysiology and therapeutic approaches. Life Sci 2023; 334:122193. [PMID: 37865177 DOI: 10.1016/j.lfs.2023.122193] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/12/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
Traumatic brain injury and spinal cord injury are two distinct but fundamentally similar types of acute insults to the central nervous system (CNS) that often culminate in death or cognitive and motor impairment. Over the past decade, researchers have tapped into research to discover the potential role being played by gut bacteria in CNS. After an acute CNS injury, the altered composition of the gut microbiota disturbs the balance of the bidirectional gut-brain axis, aggravating secondary CNS injury, motor dysfunctions, and cognitive deficits, which worsens the patient's prognosis. Some of the well-known therapeutic interventions which can also be used as adjuvant therapy for alleviating CNS injuries include, the use of pro and prebiotics, fecal microbiota transplantation, and microbial engineering. In this review, we aim to discuss the importance of gut microbes in our nervous system, anatomy, and signaling pathways involved in regulating the gut-brain axis, the alteration of the gut microbiome in CNS injuries, and the therapeutic strategies to target gut microbiomes in traumatic CNS injuries.
Collapse
Affiliation(s)
- Lahanya Guha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Tejas Girish Agnihotri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Aakanchha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
6
|
Gouda NA, Alshammari SO, Abourehab MAS, Alshammari QA, Elkamhawy A. Therapeutic potential of natural products in inflammation: underlying molecular mechanisms, clinical outcomes, technological advances, and future perspectives. Inflammopharmacology 2023; 31:2857-2883. [PMID: 37950803 DOI: 10.1007/s10787-023-01366-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 10/06/2023] [Indexed: 11/13/2023]
Abstract
Chronic inflammation is a common underlying factor in many major diseases, including heart disease, diabetes, cancer, and autoimmune disorders, and is responsible for up to 60% of all deaths worldwide. Metformin, statins, and corticosteroids, and NSAIDs (non-steroidal anti-inflammatory drugs) are often given as anti-inflammatory pharmaceuticals, however, often have even more debilitating side effects than the illness itself. The natural product-based therapy of inflammation-related diseases has no adverse effects and good beneficial results compared to substitute conventional anti-inflammatory medications. In this review article, we provide a concise overview of present pharmacological treatments, the pathophysiology of inflammation, and the signaling pathways that underlie it. In addition, we focus on the most promising natural products identified as potential anti-inflammatory therapeutic agents. Moreover, preclinical studies and clinical trials evaluating the efficacy of natural products as anti-inflammatory therapeutic agents and their pragmatic applications with promising outcomes are reviewed. In addition, the safety, side effects and technical barriers of natural products are discussed. Furthermore, we also summarized the latest technological advances in the discovery and scientific development of natural products-based medicine.
Collapse
Affiliation(s)
- Noha A Gouda
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi, 10326, Republic of Korea
| | - Saud O Alshammari
- Department of Pharmacognosy and Alternative Medicine, Faculty of Pharmacy, Northern Border University, Rafha, 76321, Saudi Arabia
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Qamar A Alshammari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Northern Border University, Rafha, 76321, Saudi Arabia
| | - Ahmed Elkamhawy
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi, 10326, Republic of Korea.
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
7
|
Dey P, Ray Chaudhuri S. The opportunistic nature of gut commensal microbiota. Crit Rev Microbiol 2023; 49:739-763. [PMID: 36256871 DOI: 10.1080/1040841x.2022.2133987] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 07/30/2022] [Accepted: 10/05/2022] [Indexed: 11/03/2022]
Abstract
The abundance of gut commensals has historically been associated with health-promoting effects despite the fact that the definition of good or bad microbiota remains condition-specific. The beneficial or pathogenic nature of microbiota is generally dictated by the dimensions of host-microbiota and microbe-microbe interactions. With the increasing popularity of gut microbiota in human health and disease, emerging evidence suggests opportunistic infections promoted by those gut bacteria that are generally considered beneficial. Therefore, the current review deals with the opportunistic nature of the gut commensals and aims to summarise the concepts behind the occasional commensal-to-pathogenic transformation of the gut microbes. Specifically, relevant clinical and experimental studies have been discussed on the overgrowth and bacteraemia caused by commensals. Three key processes and their underlying mechanisms have been summarised to be responsible for the opportunistic nature of commensals, viz. improved colonisation fitness that is dictated by commensal-pathogen interactions and availability of preferred nutrients; pathoadaptive mutations that can trigger the commensal-to-pathogen transformation; and evasion of host immune response as a survival and proliferation strategy of the microbes. Collectively, this review provides an updated concept summary on the underlying mechanisms of disease causative events driven by gut commensal bacteria.
Collapse
Affiliation(s)
- Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Saumya Ray Chaudhuri
- Council of Scientific and Industrial Research (CSIR), Institute of Microbial Technology, Chandigarh, India
| |
Collapse
|
8
|
Zhou JY, Glendenning LM, Cavanaugh JM, McNeer SK, Goodman WA, Cobb BA. Intestinal Tr1 Cells Confer Protection against Colitis in the Absence of Foxp3+ Regulatory T Cell-Derived IL-10. Immunohorizons 2023; 7:456-466. [PMID: 37314833 PMCID: PMC10580124 DOI: 10.4049/immunohorizons.2200071] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 06/15/2023] Open
Abstract
The intestinal mucosa is continually exposed to diverse microbial and dietary Ags, requiring coordinated efforts by specialized populations of regulatory T cells (Tregs) to maintain homeostasis. Suppressive mechanisms used by intestinal Tregs include the secretion of anti-inflammatory cytokines such as IL-10 and TGF-β. Defects in IL-10 signaling are associated with severe infantile enterocolitis in humans, and mice deficient in IL-10 or its receptors develop spontaneous colitis. To determine the requirement of Foxp3+ Treg-specific IL-10 for protection against colitis, we generated Foxp3-specific IL-10 knockout (KO) mice (IL-10 conditional KO [cKO] mice). Colonic Foxp3+ Tregs isolated from IL-10cKO mice showed impaired ex vivo suppressive function, although IL-10cKO mice maintained normal body weights and developed only mild inflammation over 30 wk of age (in contrast to severe colitis in global IL-10KO mice). Protection from colitis in IL-10cKO mice was associated with an expanded population of IL-10-producing type 1 Tregs (Tr1, CD4+Foxp3-) in the colonic lamina propria that produced more IL-10 on a per-cell basis compared with wild-type intestinal Tr1 cells. Collectively, our findings reveal a role for Tr1 cells in the gut, as they expand to fill a tolerogenic niche in conditions of suboptimal Foxp3+ Treg-mediated suppression and provide functional protection against experimental colitis.
Collapse
Affiliation(s)
- Julie Y. Zhou
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Leandre M. Glendenning
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Jill M. Cavanaugh
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Sarah K. McNeer
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Wendy A. Goodman
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Brian A. Cobb
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
| |
Collapse
|
9
|
Panwar S, Kumari S, Verma J, Bakshi S, Narendrakumar L, Paul D, Das B. Toxin-linked mobile genetic elements in major enteric bacterial pathogens. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2023; 4:e5. [PMID: 39295911 PMCID: PMC11406385 DOI: 10.1017/gmb.2023.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 12/31/2022] [Accepted: 02/24/2023] [Indexed: 09/21/2024]
Abstract
One of the fascinating outcomes of human microbiome studies adopting multi-omics technology is its ability to decipher millions of microbial encoded functions in the most complex and crowded microbial ecosystem, including the human gastrointestinal (GI) tract without cultivating the microbes. It is well established that several functions that modulate the human metabolism, nutrient assimilation, immunity, infections, disease severity and therapeutic efficacy of drugs are mostly of microbial origins. In addition, these microbial functions are dynamic and can disseminate between microbial taxa residing in the same ecosystem or other microbial ecosystems through horizontal gene transfer. For clinicians and researchers alike, understanding the toxins, virulence factors and drug resistance traits encoded by the microbes associated with the human body is of utmost importance. Nevertheless, when such traits are genetically linked with mobile genetic elements (MGEs) that make them transmissible, it creates an additional burden to public health. This review mainly focuses on the functions of gut commensals and the dynamics and crosstalk between commensal and pathogenic bacteria in the gut. Also, the review summarises the plethora of MGEs linked with virulence genes present in the genomes of various enteric bacterial pathogens, which are transmissible among other pathogens and commensals.
Collapse
Affiliation(s)
- Shruti Panwar
- Functional Genomics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Shashi Kumari
- Functional Genomics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Jyoti Verma
- Functional Genomics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Susmita Bakshi
- Functional Genomics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Lekshmi Narendrakumar
- Functional Genomics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Deepjyoti Paul
- Functional Genomics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Bhabatosh Das
- Functional Genomics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
10
|
Gao L, Xia X, Shuai Y, Zhang H, Jin W, Zhang X, Zhang Y. Gut microbiota, a hidden protagonist of traditional Chinese medicine for acute ischemic stroke. Front Pharmacol 2023; 14:1164150. [PMID: 37124192 PMCID: PMC10133705 DOI: 10.3389/fphar.2023.1164150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 04/04/2023] [Indexed: 05/02/2023] Open
Abstract
Acute ischemic stroke (AIS) is one of the leading diseases causing death and disability worldwide, and treatment options remain very limited. Traditional Chinese Medicine (TCM) has been used for thousands of years to treat ischemic stroke and has been proven to have significant efficacy, but its mechanism of action is still unclear. As research related to the brain-gut-microbe axis progresses, there is increasing evidence that the gut microbiota plays an important role during AIS. The interaction between TCM and the gut microbiota has been suggested as a possible key link to the therapeutic effects of TCM. We have compiled and reviewed recent studies on the relationship between AIS, TCM, and gut microbiota, with the expectation of providing more ideas to elucidate the mechanism of action of TCM in the treatment of AIS.
Collapse
Affiliation(s)
- Lin Gao
- Emergency Department, Chengdu University of Traditional Chinese Medicine Affiliated Hospital, Chengdu, Sichuan, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiuwen Xia
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yinqi Shuai
- Emergency Department, Chengdu University of Traditional Chinese Medicine Affiliated Hospital, Chengdu, Sichuan, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Hong Zhang
- Emergency Department, Chengdu University of Traditional Chinese Medicine Affiliated Hospital, Chengdu, Sichuan, China
| | - Wei Jin
- Emergency Department, Chengdu University of Traditional Chinese Medicine Affiliated Hospital, Chengdu, Sichuan, China
| | - Xiaoyun Zhang
- Emergency Department, Chengdu University of Traditional Chinese Medicine Affiliated Hospital, Chengdu, Sichuan, China
- *Correspondence: Yi Zhang, ; Xiaoyun Zhang,
| | - Yi Zhang
- Geriatric Department, Chengdu University of Traditional Chinese Medicine Affiliated Hospital, Chengdu, Sichuan, China
- *Correspondence: Yi Zhang, ; Xiaoyun Zhang,
| |
Collapse
|
11
|
Wang L, Xu H, Yang H, Zhou J, Zhao L, Zhang F. Glucose metabolism and glycosylation link the gut microbiota to autoimmune diseases. Front Immunol 2022; 13:952398. [PMID: 36203617 PMCID: PMC9530352 DOI: 10.3389/fimmu.2022.952398] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/19/2022] [Indexed: 11/21/2022] Open
Abstract
Carbohydrates serve as important energy sources and structural substances for human body as well as for gut microbes. As evidenced by the advances in immunometabolism, glucose metabolism and adenosine triphosphate (ATP) generation are deeply involved in immune cell activation, proliferation, and signaling transduction as well as trafficking and effector functions, thus contributing to immune response programming and assisting in host adaption to microenvironment changes. Increased glucose uptake, aberrant expression of glucose transporter 1 (e.g., GLU1), and abnormal glycosylation patterns have been identified in autoimmunity and are suggested as partially responsible for the dysregulated immune response and the modification of gut microbiome composition in the autoimmune pathogenesis. The interaction between gut microbiota and host carbohydrate metabolism is complex and bidirectional. Their impact on host immune homeostasis and the development of autoimmune diseases remains to be elucidated. This review summarized the current knowledge on the crosstalk of glucose metabolism and glycosylation in the host with intestinal microbiota and discussed their possible role in the development and progression of autoimmune diseases. Potential therapeutic strategies targeting glucose metabolism and glycosylation in modulating gut ecosystem and treating autoimmune diseases were discussed as well.
Collapse
Affiliation(s)
- Lu Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
| | - Haojie Xu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
| | - Huaxia Yang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
| | - Jiaxin Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- *Correspondence: Jiaxin Zhou, ; Lidan Zhao,
| | - Lidan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- *Correspondence: Jiaxin Zhou, ; Lidan Zhao,
| | - Fengchun Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
12
|
Madella AM, Van Bergenhenegouwen J, Garssen J, Masereeuw R, Overbeek SA. Microbial-Derived Tryptophan Catabolites, Kidney Disease and Gut Inflammation. Toxins (Basel) 2022; 14:toxins14090645. [PMID: 36136583 PMCID: PMC9505404 DOI: 10.3390/toxins14090645] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Uremic metabolites, molecules either produced by the host or from the microbiota population existing in the gastrointestinal tract that gets excreted by the kidneys into urine, have significant effects on both health and disease. Tryptophan-derived catabolites are an important group of bacteria-produced metabolites with an extensive contribution to intestinal health and, eventually, chronic kidney disease (CKD) progression. The end-metabolite, indoxyl sulfate, is a key contributor to the exacerbation of CKD via the induction of an inflammatory state and oxidative stress affecting various organ systems. Contrastingly, other tryptophan catabolites positively contribute to maintaining intestinal homeostasis and preventing intestinal inflammation—activities signaled through nuclear receptors in particular—the aryl hydrocarbon receptor (AhR) and the pregnane X receptor (PXR). This review discusses the origins of these catabolites, their effect on organ systems, and how these can be manipulated therapeutically in the future as a strategy to treat CKD progression and gut inflammation management. Furthermore, the use of biotics (prebiotics, probiotics, synbiotics) as a means to increase the presence of beneficial short-chain fatty acids (SCFAs) to achieve intestinal homeostasis is discussed.
Collapse
Affiliation(s)
- Avra Melina Madella
- Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
- Correspondence: (A.M.M.); or (S.A.O.); Tel.: +31-30-209-5000 (S.A.O.)
| | - Jeroen Van Bergenhenegouwen
- Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
- Danone Nutricia Research, Uppsalalaan 12, Utrecht Science Park, 3584 CT Utrecht, The Netherlands
| | - Johan Garssen
- Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
- Danone Nutricia Research, Uppsalalaan 12, Utrecht Science Park, 3584 CT Utrecht, The Netherlands
| | - Rosalinde Masereeuw
- Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Saskia Adriana Overbeek
- Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
- Danone Nutricia Research, Uppsalalaan 12, Utrecht Science Park, 3584 CT Utrecht, The Netherlands
- Correspondence: (A.M.M.); or (S.A.O.); Tel.: +31-30-209-5000 (S.A.O.)
| |
Collapse
|
13
|
Wang N, Chen L, Yi K, Zhang B, Li C, Zhou X. The effects of microbiota on reproductive health: A review. Crit Rev Food Sci Nutr 2022; 64:1486-1507. [PMID: 36066460 DOI: 10.1080/10408398.2022.2117784] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Reproductive issues are becoming an increasing global problem. There is increasing interest in the relationship between microbiota and reproductive health. Stable microbiota communities exist in the gut, reproductive tract, uterus, testes, and semen. Various effects (e.g., epigenetic modifications, nervous system, metabolism) of dysbiosis in the microbiota can impair gamete quality; interfere with zygote formation, embryo implantation, and embryo development; and increase disease susceptibility, thus adversely impacting reproductive capacity and pregnancy. The maintenance of a healthy microbiota can protect the host from pathogens, increase reproductive potential, and reduce the rates of adverse pregnancy outcomes. In conclusion, this review discusses microbiota in the male and female reproductive systems of multiple animal species. It explores the effects and mechanisms of microbiota on reproduction, factors that influence microbiota composition, and applications of microbiota in reproductive disorder treatment and detection. The findings support novel approaches for managing reproductive diseases through microbiota improvement and monitoring. In addition, it will stimulate further systematic explorations of microbiota-mediated effects on reproduction.
Collapse
Affiliation(s)
- Nan Wang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Lu Chen
- College of Animal Sciences, Jilin University, Changchun, China
| | - Kangle Yi
- Hunan Institute of Animal and Veterinary Science, Changsha, China
| | - Baizhong Zhang
- Hunan Institute of Animal and Veterinary Science, Changsha, China
| | - Chunjin Li
- College of Animal Sciences, Jilin University, Changchun, China
| | - Xu Zhou
- College of Animal Sciences, Jilin University, Changchun, China
| |
Collapse
|
14
|
Patrick S. A tale of two habitats: Bacteroides fragilis, a lethal pathogen and resident in the human gastrointestinal microbiome. Microbiology (Reading) 2022; 168. [DOI: 10.1099/mic.0.001156] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Bacteroides fragilis
is an obligately anaerobic Gram-negative bacterium and a major colonizer of the human large colon where
Bacteroides
is a predominant genus. During the growth of an individual clonal population, an astonishing number of reversible DNA inversion events occur, driving within-strain diversity. Additionally, the
B. fragilis
pan-genome contains a large pool of diverse polysaccharide biosynthesis loci, DNA restriction/modification systems and polysaccharide utilization loci, which generates remarkable between-strain diversity. Diversity clearly contributes to the success of
B. fragilis
within its normal habitat of the gastrointestinal (GI) tract and during infection in the extra-intestinal host environment. Within the GI tract,
B. fragilis
is usually symbiotic, for example providing localized nutrients for the gut epithelium, but
B. fragilis
within the GI tract may not always be benign. Metalloprotease toxin production is strongly associated with colorectal cancer.
B. fragilis
is unique amongst bacteria; some strains export a protein >99 % structurally similar to human ubiquitin and antigenically cross-reactive, which suggests a link to autoimmune diseases.
B. fragilis
is not a primary invasive enteric pathogen; however, if colonic contents contaminate the extra-intestinal host environment, it successfully adapts to this new habitat and causes infection; classically peritoneal infection arising from rupture of an inflamed appendix or GI surgery, which if untreated, can progress to bacteraemia and death. In this review selected aspects of
B. fragilis
adaptation to the different habitats of the GI tract and the extra-intestinal host environment are considered, along with the considerable challenges faced when studying this highly variable bacterium.
Collapse
Affiliation(s)
- Sheila Patrick
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences Queen’s University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, UK
| |
Collapse
|
15
|
Zhou JY, Zhou D, Telfer K, Reynero K, Jones MB, Hambor J, Cobb BA. Antigen presenting cell response to polysaccharide A is characterized by the generation of anti-inflammatory macrophages. Glycobiology 2022; 32:136-147. [PMID: 34939104 PMCID: PMC8934142 DOI: 10.1093/glycob/cwab111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/01/2021] [Accepted: 10/16/2021] [Indexed: 03/09/2024] Open
Abstract
Polysaccharide A (PSA) is the immunodominant capsular carbohydrate from the gram negative commensal microbe Bacteroides fragilis that has shown remarkable potency in ameliorating many rodent models of inflammatory disease by eliciting downstream suppressive CD4+ T cells. PSA is composed of a zwitterionic repeating unit that allows it to be processed by antigen presenting cells (APCs) and presented by MHCII in a glycosylation-dependent manner. While previous work has uncovered much about the interactions between MHCII and PSA, as well as the downstream T cell response, little is known about how PSA affects the phenotype of MHCII+ APCs, including macrophages. Here, we utilized an unbiased systems approach consisting of RNAseq transcriptomics, high-throughput flow cytometry, Luminex analysis and targeted validation experiments to characterize the impact of PSA-mediated stimulation of splenic MHCII+ cells. The data revealed that PSA potently elicited the upregulation of an alternatively activated M2 macrophage transcriptomic and cell surface signature. Cell-type-specific validation experiments further demonstrated that PSA-exposed bone marrow-derived macrophages (BMDMs) induced cell surface and intracellular markers associated with M2 macrophages compared with conventional peptide ovalbumin (ova)-exposed BMDMs. In contrast to macrophages, we also found that CD11c+ dendritic cells (DCs) upregulated the pro-T cell activation costimulatory molecule CD86 following PSA stimulation. Consistent with the divergent BMDM and DC changes, PSA-exposed DCs elicited an antigen-experienced T cell phenotype in co-cultures, whereas macrophages did not. These findings collectively demonstrate that the PSA-induced immune response is characterized by both T cell stimulation via presentation by DCs, and a previously unrecognized anti-inflammatory polarization of macrophages.
Collapse
Affiliation(s)
- Julie Y Zhou
- Department of Pathology, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106-7288, USA
| | - David Zhou
- Department of Computer Science, Arizona State University, 1151 S. Forest Avenue, Tempe, AZ 85281, USA
| | - Kevin Telfer
- Department of Pathology, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106-7288, USA
| | - Kalob Reynero
- Department of Pathology, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106-7288, USA
| | - Mark B Jones
- Department of Pathology, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106-7288, USA
| | - John Hambor
- Research Beyond Borders, Boehringer Ingelheim Pharmaceuticals, 900 Ridgebury Road, Ridgefield, CT 06877, USA
| | - Brian A Cobb
- Department of Pathology, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106-7288, USA
| |
Collapse
|
16
|
Du J, Qiao H, Xie D. A prognostic model based on 10 gene signatures associated with intestinal microbiota predicts survival prognosis of esophageal squamous cell adenocarcinoma. ALL LIFE 2022. [DOI: 10.1080/26895293.2022.2046653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Jiang Du
- Department of Thoracic Surgery, Chinese Medical University Affiliated No. 1 Hospital, Shenyang, People’s Republic of China
| | - Han Qiao
- Clinical Medicine, Chinese Medical University, Shenyang, People’s Republic of China
| | - Dalong Xie
- Department of Anatomy, College of Basic Medicine, China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
17
|
Yuan B, Lu XJ, Wu Q. Gut Microbiota and Acute Central Nervous System Injury: A New Target for Therapeutic Intervention. Front Immunol 2022; 12:800796. [PMID: 35003127 PMCID: PMC8740048 DOI: 10.3389/fimmu.2021.800796] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 12/09/2021] [Indexed: 12/15/2022] Open
Abstract
Acute central nervous system (CNS) injuries, including stroke, traumatic brain injury (TBI), and spinal cord injury (SCI), are the common causes of death or lifelong disabilities. Research into the role of the gut microbiota in modulating CNS function has been rapidly increasing in the past few decades, particularly in animal models. Growing preclinical and clinical evidence suggests that gut microbiota is involved in the modulation of multiple cellular and molecular mechanisms fundamental to the progression of acute CNS injury-induced pathophysiological processes. The altered composition of gut microbiota after acute CNS injury damages the equilibrium of the bidirectional gut-brain axis, aggravating secondary brain injury, cognitive impairments, and motor dysfunctions, which leads to poor prognosis by triggering pro-inflammatory responses in both peripheral circulation and CNS. This review summarizes the studies concerning gut microbiota and acute CNS injuries. Experimental models identify a bidirectional communication between the gut and CNS in post-injury gut dysbiosis, intestinal lymphatic tissue-mediated neuroinflammation, and bacterial-metabolite-associated neurotransmission. Additionally, fecal microbiota transplantation, probiotics, and prebiotics manipulating the gut microbiota can be used as effective therapeutic agents to alleviate secondary brain injury and facilitate functional outcomes. The role of gut microbiota in acute CNS injury would be an exciting frontier in clinical and experimental medicine.
Collapse
Affiliation(s)
- Bin Yuan
- Department of Neurosurgery, The Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Xiao-Jie Lu
- Department of Neurosurgery, The Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China.,Department of Neurosurgery, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Qi Wu
- Department of Neurosurgery, Jinling Hospital, Nanjing University, School of Medicine, Nanjing, China
| |
Collapse
|
18
|
Abstract
Environmental chemicals can alter gut microbial community composition, known as dysbiosis. However, the gut microbiota is a highly dynamic system and its functions are still largely underexplored. Likewise, it is unclear whether xenobiotic exposure affects host health through impairing host-microbiota interactions. Answers to this question not only can lead to a more precise understanding of the toxic effects of xenobiotics but also can provide new targets for the development of new therapeutic strategies. Here, we aim to identify the major challenges in the field of microbiota-exposure research and highlight the need to exam the health effects of xenobiotic-induced gut microbiota dysbiosis in host bodies. Although the changes of gut microbiota frequently co-occur with the xenobiotic exposure, the causal relationship of xenobiotic-induced microbiota dysbiosis and diseases is rarely established. The high dynamics of the gut microbiota and the complex interactions among exposure, microbiota, and host, are the major challenges to decipher the specific health effects of microbiota dysbiosis. The next stage of study needs to combine various technologies to precisely assess the xenobiotic-induced gut microbiota perturbation and the subsequent health effects in host bodies. The exposure, gut microbiota dysbiosis, and disease outcomes have to be causally linked. Many microbiota-host interactions are established by previous studies, including signaling metabolites and response pathways in the host, which may use as start points for future research to examine the mechanistic interactions of exposure, gut microbiota, and host health. In conclusion, to precisely understand the toxicity of xenobiotics and develop microbiota-based therapies, the causal and mechanistic links of exposure and microbiota dysbiosis have to be established in the next stage study.
Collapse
Affiliation(s)
- Liang Chi
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC, United States
| | - Pengcheng Tu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC, United States
| | - Hongyu Ru
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC, United States
| | - Kun Lu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC, United States,CONTACT Kun Lu Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC27599, United States
| |
Collapse
|
19
|
Ashrafian F, Keshavarz Azizi Raftar S, Lari A, Shahryari A, Abdollahiyan S, Moradi HR, Masoumi M, Davari M, Khatami S, Omrani MD, Vaziri F, Masotti A, Siadat SD. Extracellular vesicles and pasteurized cells derived from Akkermansia muciniphila protect against high-fat induced obesity in mice. Microb Cell Fact 2021; 20:219. [PMID: 34863163 PMCID: PMC8645101 DOI: 10.1186/s12934-021-01709-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 11/16/2021] [Indexed: 12/15/2022] Open
Abstract
Background Several studies have shown that probiotics have beneficial effects on weight control and metabolic health. In addition to probiotics, recent studies have investigated the effects of paraprobiotics and postbiotics. Therefore, we evaluated the preventive effects of live and pasteurized Akkermansia muciniphila MucT (A. muciniphila) and its extracellular vesicles (EVs) on HFD-induced obesity. Results The results showed that body weight, metabolic tissues weight, food consumption, and plasma metabolic parameters were increased in the HFD group, whereas A. muciniphila preventive treatments inhibited these HFD. The effects of pasteurized A. muciniphila and its extracellular vesicles were more noticeable than its active form. The HFD led to an increase in the colonic, adipose tissue, and liver inflammations and increased the expression of genes involved in lipid metabolism and homeostasis. Nevertheless, these effects were inhibited in mice that were administered A. muciniphila and its EVs. The assessment of the gut microbiota revealed significant differences in the microbiota composition after feeding with HFD. However, all treatments restored the alterations in some bacterial genera and closely resemble the control group. Also, the correlation analysis indicated that some gut microbiota might be associated with obesity-related indices. Conclusions Pasteurized A. muciniphila and its EVs, as paraprobiotic and postbiotic agents, were found to play a key role in the regulation of metabolic functions to prevent obesity, probably by affecting the gut-adipose-liver axis. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12934-021-01709-w.
Collapse
Affiliation(s)
- Fatemeh Ashrafian
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.,Clinical Research Department, Pasteur Institute of Iran, Tehran, Iran
| | | | - Arezou Lari
- Systems Biomedicine Unit, Pasteur Institute of Iran, Tehran, Iran
| | - Arefeh Shahryari
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Sara Abdollahiyan
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.,Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Moradi
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Morteza Masoumi
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.,Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | - Mehdi Davari
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.,Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | - Shohreh Khatami
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Mir Davood Omrani
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzam Vaziri
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.,Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | - Andrea Masotti
- Research Laboratories, Children's Hospital Bambino Gesù-IRCCS, Rome, Italy
| | - Seyed Davar Siadat
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran. .,Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
20
|
THE INTESTINAL COMMENSAL, Bacteroides fragilis, MODULATES HOST RESPONSES TO VIRAL INFECTION AND THERAPY: LESSONS FOR EXPLORATION DURING Mycobacterium tuberculosis INFECTION. Infect Immun 2021; 90:e0032121. [PMID: 34606367 DOI: 10.1128/iai.00321-21] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The gut microbiota has emerged as a critical player in host health. Bacteroides fragilis is a prominent member of the gut microbiota within the phyla Bacteroidetes. This commensal bacterium produces unique capsular polysaccharides processed by antigen-presenting cells and activates CD4+ T cells to secrete inflammatory cytokines. Indeed, due to their immunomodulatory functions, B. fragilis and its capsular polysaccharide-A (PSA) are arguably the most explored single commensal microbiota/symbiotic factor. B. fragilis/PSA has been shown to protect against colitis, encephalomyelitis, colorectal cancer, pulmonary inflammation, and asthma. Here, we review (1) recent data on the immunomodulatory role of B. fragilis/PSA during viral infections and therapy, (2) B. fragilis PSA's dual ability to mediate pro-and anti-inflammatory processes, and the potential for exploring this unique characteristic during intracellular bacterial infections such as with Mycobacterium tuberculosis (3) discuss the protective roles of single commensal-derived probiotic species including B. fragilis in lung inflammation and respiratory infections that may provide essential cues for possible exploration of microbiota based/augmented therapies in tuberculosis (TB). Available data on the relationship between B. fragilis/PSA, the immune system, and disease suggest clinical relevance for developing B. fragilis into a next-generation probiotic or, possibly, the engineering of PSA into a potent carbohydrate-based vaccine.
Collapse
|
21
|
Sperk M, Ambikan AT, Ray S, Singh K, Mikaeloff F, Diez RC, Narayanan A, Vesterbacka J, Nowak P, Sönnerborg A, Neogi U. Fecal Metabolome Signature in the HIV-1 Elite Control Phenotype: Enrichment of Dipeptides Acts as an HIV-1 Antagonist but a Prevotella Agonist. J Virol 2021; 95:e0047921. [PMID: 34232744 PMCID: PMC8387056 DOI: 10.1128/jvi.00479-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/30/2021] [Indexed: 12/21/2022] Open
Abstract
HIV-1 elite controllers (EC) are a rare group among HIV-1-infected individuals who can naturally control viral replication for a prolonged period. Due to their heterogeneous nature, no universal mechanism could be attributed to the EC status; instead, several host and viral factors have been discussed as playing a role. In this study, we investigated the fecal metabolome and microbiome in a Swedish cohort of EC (n = 14), treatment-naive viremic progressors (VP; n = 16), and HIV-negative individuals (HC; n = 12). Fecal untargeted metabolomics was performed by four ultra-high-performance liquid chromatography tandem mass spectrometry (UHPLC-MS/MS). Molecular docking and biochemical microscale thermophoresis (MST) were used to describe the peptide-metabolite interactions. Single-cycle infectivity assays were performed in TZM-Bl cell lines using CCR5- and CXCR4-tropic HIV-1 strains. The microbiome analysis was performed using 16S rRNA sequencing. Th effects of metabolites on bacterial species viability were determined using several clinical isolates. We observed an enrichment of dipeptides in EC compared to VP and HC (adjusted P < 0.05). In silico analysis by molecular docking, in vitro biochemical assays, and ex vivo infection assays identified anti-HIV-1 properties for two dipeptides (WG and VQ) that could bind to the HIV-1 gp120, of which WG was more potent. The microbiome analysis identified enrichment of the genus Prevotella in EC, and these dipeptides supported bacterial growth of the genus Prevotella in vitro. The enrichments of the dipeptides and higher abundance of Prevotella have a distinct mechanism of elite control status in HIV-1 infection that influences host metabolism. IMPORTANCE HIV-1 elite controllers (EC) are a rare group among HIV-1-infected individuals who can naturally control viral replication for a prolonged period. Due to their heterogeneous nature, no universal mechanism could be attributed to the EC status; instead, several host and viral factors have been discussed as playing a role. In this study, we investigated the fecal metabolome and microbiome in a Swedish cohort of EC, treatment-naive viremic progressors (VP), and HIV-negative individuals (HC). We observed an enrichment of dipeptides in EC compared to the other two study groups. In silico and in vitro analyses identified anti-HIV-1 properties for two dipeptides that could bind to the HIV-1 gp120 and act as an HIV-1 antagonist. Furthermore, these dipeptides supported bacterial growth of the genus Prevotella in vitro that was enriched in EC, which influences host metabolism. Thus, increased levels of both dipeptides and Prevotella could provide beneficial effects for EC.
Collapse
Affiliation(s)
- Maike Sperk
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, ANA Futura, Stockholm, Sweden
| | - Anoop T. Ambikan
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, ANA Futura, Stockholm, Sweden
| | - Shilpa Ray
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, ANA Futura, Stockholm, Sweden
| | - Kamal Singh
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA, 65211, USA
| | - Flora Mikaeloff
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, ANA Futura, Stockholm, Sweden
| | - Rafael Ceña Diez
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institute, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Ashwathy Narayanan
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, ANA Futura, Stockholm, Sweden
| | - Jan Vesterbacka
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institute, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Piotr Nowak
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institute, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Anders Sönnerborg
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, ANA Futura, Stockholm, Sweden
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institute, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Ujjwal Neogi
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, ANA Futura, Stockholm, Sweden
- Manipal Institute of Virology (MIV), Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
22
|
Phillips-Farfán B, Gómez-Chávez F, Medina-Torres EA, Vargas-Villavicencio JA, Carvajal-Aguilera K, Camacho L. Microbiota Signals during the Neonatal Period Forge Life-Long Immune Responses. Int J Mol Sci 2021; 22:ijms22158162. [PMID: 34360926 PMCID: PMC8348731 DOI: 10.3390/ijms22158162] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 12/27/2022] Open
Abstract
The microbiota regulates immunological development during early human life, with long-term effects on health and disease. Microbial products include short-chain fatty acids (SCFAs), formyl peptides (FPs), polysaccharide A (PSA), polyamines (PAs), sphingolipids (SLPs) and aryl hydrocarbon receptor (AhR) ligands. Anti-inflammatory SCFAs are produced by Actinobacteria, Bacteroidetes, Firmicutes, Spirochaetes and Verrucomicrobia by undigested-carbohydrate fermentation. Thus, fiber amount and type determine their occurrence. FPs bind receptors from the pattern recognition family, those from commensal bacteria induce a different response than those from pathogens. PSA is a capsular polysaccharide from B. fragilis stimulating immunoregulatory protein expression, promoting IL-2, STAT1 and STAT4 gene expression, affecting cytokine production and response modulation. PAs interact with neonatal immunity, contribute to gut maturation, modulate the gut–brain axis and regulate host immunity. SLPs are composed of a sphingoid attached to a fatty acid. Prokaryotic SLPs are mostly found in anaerobes. SLPs are involved in proliferation, apoptosis and immune regulation as signaling molecules. The AhR is a transcription factor regulating development, reproduction and metabolism. AhR binds many ligands due to its promiscuous binding site. It participates in immune tolerance, involving lymphocytes and antigen-presenting cells during early development in exposed humans.
Collapse
Affiliation(s)
- Bryan Phillips-Farfán
- Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría, México City 04530, Mexico; (B.P.-F.); (K.C.-A.)
| | - Fernando Gómez-Chávez
- Laboratorio de Inmunología Experimental, Instituto Nacional de Pediatría, México City 04530, Mexico; (F.G.-C.); (J.A.V.-V.)
- Cátedras CONACyT-Instituto Nacional de Pediatría, México City 04530, Mexico
- Departamento de Formación Básica Disciplinaria, Escuela Nacional de Medicina y Homeopatía del Instituto Politécnico Nacional, Mexico City 07320, Mexico
| | | | | | - Karla Carvajal-Aguilera
- Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría, México City 04530, Mexico; (B.P.-F.); (K.C.-A.)
| | - Luz Camacho
- Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría, México City 04530, Mexico; (B.P.-F.); (K.C.-A.)
- Correspondence:
| |
Collapse
|
23
|
Ghezzi L, Cantoni C, Pinget GV, Zhou Y, Piccio L. Targeting the gut to treat multiple sclerosis. J Clin Invest 2021; 131:e143774. [PMID: 34196310 DOI: 10.1172/jci143774] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The gut-brain axis (GBA) refers to the complex interactions between the gut microbiota and the nervous, immune, and endocrine systems, together linking brain and gut functions. Perturbations of the GBA have been reported in people with multiple sclerosis (pwMS), suggesting a possible role in disease pathogenesis and making it a potential therapeutic target. While research in the area is still in its infancy, a number of studies revealed that pwMS are more likely to exhibit altered microbiota, altered levels of short chain fatty acids and secondary bile products, and increased intestinal permeability. However, specific microbes and metabolites identified across studies and cohorts vary greatly. Small clinical and preclinical trials in pwMS and mouse models, in which microbial composition was manipulated through the use of antibiotics, fecal microbiota transplantation, and probiotic supplements, have provided promising outcomes in preventing CNS inflammation. However, results are not always consistent, and large-scale randomized controlled trials are lacking. Herein, we give an overview of how the GBA could contribute to MS pathogenesis, examine the different approaches tested to modulate the GBA, and discuss how they may impact neuroinflammation and demyelination in the CNS.
Collapse
Affiliation(s)
- Laura Ghezzi
- Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA.,University of Milan, Milan, Italy
| | - Claudia Cantoni
- Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Gabriela V Pinget
- Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Yanjiao Zhou
- Department of Medicine, School of Medicine, UConn Health, Farmington, Connecticut, USA
| | - Laura Piccio
- Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA.,Brain and Mind Centre, School of Medical Sciences, University of Sydney, Sydney, New South Wales, Australia.,Hope Center for Neurological Disorders, Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
24
|
Wu Q, Yu X, Li J, Sun S, Tu Y. Metabolic regulation in the immune response to cancer. Cancer Commun (Lond) 2021; 41:661-694. [PMID: 34145990 PMCID: PMC8360644 DOI: 10.1002/cac2.12182] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/25/2021] [Accepted: 06/11/2021] [Indexed: 02/06/2023] Open
Abstract
Metabolic reprogramming in tumor‐immune interactions is emerging as a key factor affecting pro‐inflammatory carcinogenic effects and anticancer immune responses. Therefore, dysregulated metabolites and their regulators affect both cancer progression and therapeutic response. Here, we describe the molecular mechanisms through which microenvironmental, systemic, and microbial metabolites potentially influence the host immune response to mediate malignant progression and therapeutic intervention. We summarized the primary interplaying factors that constitute metabolism, immunological reactions, and cancer with a focus on mechanistic aspects. Finally, we discussed the possibility of metabolic interventions at multiple levels to enhance the efficacy of immunotherapeutic and conventional approaches for future anticancer treatments.
Collapse
Affiliation(s)
- Qi Wu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Xin Yu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Juanjuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Yi Tu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| |
Collapse
|
25
|
Zhou JY, Alvarez CA, Cobb BA. Integration of IL-2 and IL-4 signals coordinates divergent regulatory T cell responses and drives therapeutic efficacy. eLife 2021; 10:e57417. [PMID: 33617447 PMCID: PMC7899647 DOI: 10.7554/elife.57417] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 01/20/2021] [Indexed: 12/14/2022] Open
Abstract
Cells exist within complex milieus of communicating factors, such as cytokines, that combine to generate context-specific responses, yet nearly all knowledge about the function of each cytokine and the signaling propagated downstream of their recognition is based on the response to individual cytokines. Here, we found that regulatory T cells (Tregs) integrate concurrent signaling initiated by IL-2 and IL-4 to generate a response divergent from the sum of the two pathways in isolation. IL-4 stimulation of STAT6 phosphorylation was blocked by IL-2, while IL-2 and IL-4 synergized to enhance STAT5 phosphorylation, IL-10 production, and the selective proliferation of IL-10-producing Tregs, leading to increased inhibition of conventional T cell activation and the reversal of asthma and multiple sclerosis in mice. These data define a mechanism of combinatorial cytokine signaling and lay the foundation upon which to better understand the origins of cytokine pleiotropy while informing improved the clinical use of cytokines.
Collapse
Affiliation(s)
- Julie Y Zhou
- Department of Pathology, Case Western Reserve University School of MedicineClevelandUnited States
| | - Carlos A Alvarez
- Department of Pathology, Case Western Reserve University School of MedicineClevelandUnited States
| | - Brian A Cobb
- Department of Pathology, Case Western Reserve University School of MedicineClevelandUnited States
| |
Collapse
|
26
|
The Gut Microbiota: How Does It Influence the Development and Progression of Liver Diseases. Biomedicines 2020; 8:biomedicines8110501. [PMID: 33207562 PMCID: PMC7697996 DOI: 10.3390/biomedicines8110501] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/06/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
The gut–liver axis plays important roles in both the maintenance of a healthy liver and the pathogenesis of liver diseases, where the gut microbiota acts as a major determinant of this relationship. Gut bacteria-derived metabolites and cellular components are key molecules that affect the function of the liver and modulate the pathology of liver diseases. Accumulating evidence showed that gut microbiota produces a myriad of molecules, including lipopolysaccharide, lipoteichoic acid, peptidoglycan, and DNA, as well as short-chain fatty acids, bile acids, trimethylamine, and indole derivatives. The translocation of these components to the liver exerts beneficial or pathogenic effects by interacting with liver immune cells. This is a bidirectional relationship. Therefore, the existence of crosstalk between the gut and liver and its implications on host health and diseases are essential for the etiology and treatment of diseases. Several mechanisms have been proposed for the pathogenesis of liver diseases, but still, the mechanisms behind the pathogenic role of gut-derived components on liver pathogenesis remain elusive and not understandable. This review discusses the current progress on the gut microbiota and its components in terms of the progression of liver diseases, and in turn, how liver diseases indirectly affect the intestinal function and induce intestinal inflammation. Moreover, this paper highlights the current therapeutic and preventive strategies used to restore the gut microbiota composition and improve host health.
Collapse
|
27
|
Alvarez CA, Jones MB, Hambor J, Cobb BA. Characterization of Polysaccharide A Response Reveals Interferon Responsive Gene Signature and Immunomodulatory Marker Expression. Front Immunol 2020; 11:556813. [PMID: 33193325 PMCID: PMC7649347 DOI: 10.3389/fimmu.2020.556813] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/24/2020] [Indexed: 12/15/2022] Open
Abstract
Polysaccharide A (PSA), a capsular carbohydrate from the commensal gut bacteria Bacteroides fragilis, has been shown to possess both potent T cell-dependent pro- and anti-inflammatory properties. PSA is able to induce abscess and adhesion formation in sepsis models, but can also inhibit asthma, inflammatory bowel disease (IBD) and experimental autoimmune encephalomyelitis (EAE) through MHCII-dependent activation of CD4+ T cells. Yet, despite decades of study, the ability of PSA to balance both these pro- and anti-inflammatory responses remains poorly understood. Here, we utilized an unbiased systems immunology approach consisting of RNAseq transcriptomics, high-throughput flow cytometry, and Luminex analysis to characterize the full impact of PSA-mediated stimulation of CD4+ T cells. We found that exposure to PSA resulted in the upregulation and secretion of IFNγ, TNFα, IL-6, and CXCL10, consistent with an interferon responsive gene (IRG) signature. Importantly, PSA stimulation also led to expression of immune checkpoint markers Lag3, Tim3, and, especially, PD1, which were also enriched and sustained in the gut associated lymphoid tissue of PSA-exposed mice. Taken together, PSA responding cells display an unusual mixture of pro-inflammatory cytokines and anti-inflammatory surface receptors, consistent with the ability to both cause and inhibit inflammatory disease.
Collapse
Affiliation(s)
- Carlos A. Alvarez
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Mark B. Jones
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - John Hambor
- Research Beyond Borders, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, United States
| | - Brian A. Cobb
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
28
|
Ghosh S, Trabbic KR, Shi M, Nishat S, Eradi P, Kleski KA, Andreana PR. Chemical synthesis and immunological evaluation of entirely carbohydrate conjugate Globo H-PS A1. Chem Sci 2020; 11:13052-13059. [PMID: 34123241 PMCID: PMC8163331 DOI: 10.1039/d0sc04595k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 10/15/2020] [Indexed: 12/19/2022] Open
Abstract
An anticancer, entirely carbohydrate conjugate, Globo H-polysaccharide A1 (Globo H-PS A1), was chemically prepared and immunologically evaluated in C57BL/6 mice. Tumor associated carbohydrate antigen Globo H hexasaccharide was synthesized in an overall 7.8% yield employing a convergent [3 + 3] strategy that revealed an anomeric aminooxy group used for conjugation to oxidized PS A1 via an oxime linkage. Globo H-PS A1, formulated with adjuvants monophosphoryl lipid A and TiterMax® Gold. After immunization an antigen specific immune response was observed in ELISA with anti-Globo H IgG/IgM antibodies. Specificity of the corresponding antibodies was determined by FACS showing cell surface binding to Globo H-positive cancer cell lines MCF-7 and OVCAR-5. The anti-Globo H antibodies also exhibited complement-dependent cellular cytotoxicity against MCF-7 and OVCAR-5 cells.
Collapse
Affiliation(s)
- Samir Ghosh
- The University of Toledo, Department of Chemistry and Biochemistry 2801 West Bancroft Street Toledo Ohio USA 43606
| | - Kevin R Trabbic
- The University of Toledo, Department of Chemistry and Biochemistry 2801 West Bancroft Street Toledo Ohio USA 43606
| | - Mengchao Shi
- The University of Toledo, Department of Chemistry and Biochemistry 2801 West Bancroft Street Toledo Ohio USA 43606
| | - Sharmeen Nishat
- The University of Toledo, Department of Chemistry and Biochemistry 2801 West Bancroft Street Toledo Ohio USA 43606
| | - Pradheep Eradi
- The University of Toledo, Department of Chemistry and Biochemistry 2801 West Bancroft Street Toledo Ohio USA 43606
| | - Kristopher A Kleski
- The University of Toledo, Department of Chemistry and Biochemistry 2801 West Bancroft Street Toledo Ohio USA 43606
| | - Peter R Andreana
- The University of Toledo, Department of Chemistry and Biochemistry 2801 West Bancroft Street Toledo Ohio USA 43606
| |
Collapse
|
29
|
Yekani M, Baghi HB, Naghili B, Vahed SZ, Sóki J, Memar MY. To resist and persist: Important factors in the pathogenesis of Bacteroides fragilis. Microb Pathog 2020; 149:104506. [PMID: 32950639 DOI: 10.1016/j.micpath.2020.104506] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/15/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023]
Abstract
Bacteroides fragilis is a most frequent anaerobic pathogen isolated from human infections, particularly found in the abdominal cavity. Different factors contribute to the pathogenesis and persistence of B. fragilis at infection sites. The knowledge of the virulence factors can provide applicable information for finding alternative options for the antibiotic therapy and treatment of B. fragilis caused infections. Herein, a comprehensive review of the important B. fragilis virulence factors was prepared. In addition to B. fragilis toxin (BFT) and its potential role in the diarrhea and cancer development, some other important virulence factors and characteristics of B. fragilis are described including capsular polysaccharides, iron acquisition, resistance to antimicrobial agents, and survival during the prolonged oxidative stress, quorum sensing, and secretion systems.
Collapse
Affiliation(s)
- Mina Yekani
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee,Kashan University of Medical Sciences, Kashan, Iran
| | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behrooz Naghili
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - József Sóki
- Institute of Clinical Microbiology, Faculty of Medicine, University of Szeged, Szeged, Hungary.
| | - Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Microbiology Department, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
30
|
Khatun F, Toth I, Stephenson RJ. Immunology of carbohydrate-based vaccines. Adv Drug Deliv Rev 2020; 165-166:117-126. [PMID: 32320714 DOI: 10.1016/j.addr.2020.04.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/13/2020] [Accepted: 04/17/2020] [Indexed: 11/29/2022]
Abstract
Carbohydrates are considered as promising targets for vaccine development against infectious diseases where cell surface glycan's on many infectious agents are attributed to playing an important role in pathogenesis. Understanding the relationship between carbohydrates and immune components at a molecular level is crucial for the development of well-defined vaccines. Recently, carbohydrate immunology research has been accelerated by the development of new technologies that contribute to the design of optimum antigens, synthesis of antigens and the studies of antigen-antibody interactions, and as a result, several promising carbohydrate-based vaccine candidates have been prepared in recent years. This article briefly presents the mechanistic pathways of polysaccharide, glycoconjugate, glycolipid and zwitterionic vaccines and the interplay between carbohydrate antigen and immune response.
Collapse
Affiliation(s)
- Farjana Khatun
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; East West University, Dhaka, 1212, Bangladesh
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia; Institute for Molecular Biosciences, The University of Queensland, Woolloongabba, QLD 4072, Australia.
| | - Rachel J Stephenson
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia.
| |
Collapse
|
31
|
MacCalman TE, Phillips-Jones MK, Harding SE. Glycoconjugate vaccines: some observations on carrier and production methods. Biotechnol Genet Eng Rev 2020; 35:93-125. [PMID: 32048549 DOI: 10.1080/02648725.2019.1703614] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Glycoconjugate vaccines use protein carriers to improve the immune response to polysaccharide antigens. The protein component allows the vaccine to interact with T cells, providing a stronger and longer-lasting immune response than a polysaccharide interacting with B cells alone. Whilst in theory the mere presence of a protein component in a vaccine should be sufficient to improve vaccine efficacy, the extent of improvement varies. In the present review, a comparison of the performances of vaccines developed with and without a protein carrier are presented. The usefulness of analytical tools for macromolecular integrity assays, in particular nuclear magnetic resonance, circular dichroism, analytical ultracentrifugation and SEC coupled to multi-angle light scattering (MALS) is indicated. Although we focus mainly on bacterial capsular polysaccharide-protein vaccines, some consideration is also given to research on experimental cancer vaccines using zwitterionic polysaccharides which, unusually for polysaccharides, are able to invoke T-cell responses and have been used in the development of potential all-polysaccharide-based cancer vaccines.A general trend of improved immunogenicity for glycoconjugate vaccines is described. Since the immunogenicity of a vaccine will also depend on carrier protein type and the way in which it has been linked to polysaccharide, the effects of different carrier proteins and production methods are also reviewed. We suggest that, in general, there is no single best carrier for use in glycoconjugate vaccines. This indicates that the choice of carrier protein is optimally made on a case-by-case basis, based on what generates the best immune response and can be produced safely in each individual case.Abbreviations: AUC: analytical ultracentrifugation; BSA: bovine serum albumin; CD: circular dichroism spectroscopy; CPS: capsular polysaccharide; CRM197: Cross Reactive Material 197; DT: diphtheria toxoid; Hib: Haemophilius influenzae type b; MALS: multi-angle light scattering; Men: Neisseria menigitidis; MHC-II: major histocompatibility complex class II; NMR: nuclear magnetic resonance spectroscopy; OMP: outer membrane protein; PRP: polyribosyl ribitol phosphate; PSA: Polysaccharide A1; Sa: Salmonella; St.: Streptococcus; SEC: size exclusion chromatography; Sta: Staphylococcus; TT: tetanus toxoid; ZPS: zwitterionic polysaccharide(s).
Collapse
Affiliation(s)
- Thomas E MacCalman
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, Nottingham, UK
| | - Mary K Phillips-Jones
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, Nottingham, UK
| | - Stephen E Harding
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, Nottingham, UK.,Kulturhistorisk Museum, University of Oslo, Oslo, Norway
| |
Collapse
|
32
|
Sterrett S, Peng BJ, Burton RL, LaFon DC, Westfall AO, Singh S, Pride M, Anderson AS, Ippolito GC, Schroeder HW, Nahm MH, Krishna Prasad A, Goepfert P, Bansal A. Peripheral CD4 T follicular cells induced by a conjugated pneumococcal vaccine correlate with enhanced opsonophagocytic antibody responses in younger individuals. Vaccine 2020; 38:1778-1786. [PMID: 31911030 DOI: 10.1016/j.vaccine.2019.12.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/06/2019] [Accepted: 12/12/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND PCV13 (conjugated polysaccharide) and PPSV23 (polysaccharide only) are two licensed vaccines targeting S. pneumoniae. The role of CD4 T-cell responses in pneumococcal vaccines among healthy participants and their impact on antibodies is not yet known. METHODS Ten adults (5 old and 5 young) received PCV13 (prime) and a year later PPSV23 (boost). Blood samples were collected prior to and multiple time points after vaccination. CD4 T cells responding to CRM197, polysaccharide (PS), CRM197 conjugated polysaccharide (CPS), PCV13 and PPSV23 vaccines were measured by flow cytometry. Serum antibodies were analyzed via multiplex opsonophagocytosis (MOPA) and pneumococcal IgG assays. RESULTS Vaccine-specific CD4 T cells were induced in all ten vaccinees post PCV13. Older vaccinees mounted higher peak responses and those specific for PCV13 and conjugated PS-1 were more polyfunctional compared to the younger group. Vaccine-elicited peripheral T follicular helper (Tfh) cells were only detected in the younger group who also exhibited a higher fold change in OPA titers post both vaccines. Importantly, Tfh cells following PCV13 correlated only with PCV13 serotype specific OPA titers after PPSV23 vaccination. CONCLUSIONS These findings demonstrate age related differences in immune response and the potential importance of Tfh in modulating functional antibody responses following pneumococcal vaccination.
Collapse
Affiliation(s)
- Sarah Sterrett
- University of Alabama at Birmingham, Department of Medicine, Birmingham, AL, United States
| | - Binghao J Peng
- University of Alabama at Birmingham, Department of Medicine, Birmingham, AL, United States
| | - Robert L Burton
- University of Alabama at Birmingham, Department of Medicine, Birmingham, AL, United States
| | - David C LaFon
- University of Alabama at Birmingham, Department of Medicine, Birmingham, AL, United States
| | - Andrew O Westfall
- University of Alabama at Birmingham, Department of Biostatistics Birmingham, Birmingham, AL, United States
| | - Suddham Singh
- Pfizer Vaccine Research & Development, Pearl River, New York, United States
| | - Michael Pride
- Pfizer Vaccine Research & Development, Pearl River, New York, United States
| | | | | | - Harry W Schroeder
- University of Alabama at Birmingham, Department of Medicine, Birmingham, AL, United States; University of Alabama at Birmingham, Department of Microbiology Birmingham, Birmingham, AL, United States
| | - Moon H Nahm
- University of Alabama at Birmingham, Department of Medicine, Birmingham, AL, United States; University of Alabama at Birmingham, Department of Microbiology Birmingham, Birmingham, AL, United States
| | - A Krishna Prasad
- Pfizer Vaccine Research & Development, Pearl River, New York, United States
| | - Paul Goepfert
- University of Alabama at Birmingham, Department of Medicine, Birmingham, AL, United States; University of Alabama at Birmingham, Department of Microbiology Birmingham, Birmingham, AL, United States.
| | - Anju Bansal
- University of Alabama at Birmingham, Department of Medicine, Birmingham, AL, United States.
| |
Collapse
|
33
|
Advances in Gut Microbiota of Viral Hepatitis Cirrhosis. BIOMED RESEARCH INTERNATIONAL 2019; 2019:9726786. [PMID: 31886272 PMCID: PMC6893240 DOI: 10.1155/2019/9726786] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/28/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023]
Abstract
Although gut dysbiosis appears in 20%-75% of cirrhotic patients, there are limited data on microbiota profiles in viral hepatitis cirrhotics and its role in progression to cirrhosis. Further understanding on the relationship between gut dysbiosis and cirrhosis presents a unique opportunity in not only predicting the development of cirrhosis but also discovering new therapies. Recent advances have been made on identifying unique microbiota in viral hepatitis cirrhotics and adopting the microbiota index to predict cirrhosis. Therapeutic intervention with microbiome-modulating has been explored. Cirrhosis from viral infection has unique bacterial or fungal profiles, which include increased numbers of Prevotella, Streptococcus, Staphylococcaceae, and Enterococcus, as well as decreased Ruminococcus and Clostridium. In addition, the gut microbiota can stimulate liver immunity, effectively helping hepatitis virus clearance. In clinical settings, CDR, GDI, Basidiomycota/Ascomycota, specific POD, and so forth are efficient microbiota indexes to diagnose or prognosticate cirrhosis from viral hepatitis. FMT, probiotics, and prebiotics can restore microbial diversity in cirrhotic patients with viral hepatitis, decrease ammonia serum or endotoxemia levels, prevent complications, reduce rehospitalization rate, and improve prognosis. Cirrhotics from viral hepatitis had unique bacterial or fungal profiles, associated with specific metabolic, immune, and endocrinological statuses. Such profiles are modifiable with medical treatment. The role of gut archaea and virome, implementation of FMT, microbiota metabolites as adjuvant immunotherapy, and microbiota indexes for prognostication deserve attention.
Collapse
|
34
|
Inda ME, Broset E, Lu TK, de la Fuente-Nunez C. Emerging Frontiers in Microbiome Engineering. Trends Immunol 2019; 40:952-973. [PMID: 31601521 DOI: 10.1016/j.it.2019.08.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 08/15/2019] [Accepted: 08/15/2019] [Indexed: 02/07/2023]
Abstract
The gut microbiome has a significant impact on health and disease and can actively contribute to obesity, diabetes, inflammatory bowel disease, cardiovascular disease, and neurological disorders. We do not yet have the necessary tools to fine-tune the microbial communities that constitute the microbiome, though such tools could unlock extensive benefits to human health. Here, we provide an overview of the current state of technological tools that may be used for microbiome engineering. These tools can enable investigators to define the parameters of a healthy microbiome and to determine how gut bacteria may contribute to the etiology of a variety of diseases. These tools may also allow us to explore the exciting prospect of developing targeted therapies and personalized treatments for microbiome-linked diseases.
Collapse
Affiliation(s)
- María Eugenia Inda
- Synthetic Biology Group, MIT Synthetic Biology Center, Department of Biological Engineering and Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Esther Broset
- Machine Biology Group, Departments of Psychiatry and Microbiology, Perelman School of Medicine, and Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Grupo de Genética de Micobacterias, Departamento de Microbiología y Medicina Preventiva, Facultad de Medicina, Universidad de Zaragoza, 50009, Spain
| | - Timothy K Lu
- Synthetic Biology Group, MIT Synthetic Biology Center, Department of Biological Engineering and Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Perelman School of Medicine, and Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
35
|
Singh RP. Glycan utilisation system in Bacteroides and Bifidobacteria and their roles in gut stability and health. Appl Microbiol Biotechnol 2019; 103:7287-7315. [PMID: 31332487 DOI: 10.1007/s00253-019-10012-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 02/07/2023]
Abstract
Gut residential hundred trillion microbial cells are indispensable for maintaining gut homeostasis and impact on host physiology, development and immune systems. Many of them have displayed excellence in utilising dietary- and host-derived complex glycans and are producing useful postbiotics including short-chain fatty acids to primarily fuel different organs of the host. Therefore, employing individual microbiota is nowadays becoming a propitious target in biomedical for improving gut dysbiosis conditions of the host. Among other gut microbial communities, Bacteroides and Bifidobacteria are coevolved to utilise diverse ranges of diet- and host-derived glycans through harmonising distinct glycan utilisation systems. These gut symbionts frequently share digested oligosaccharides, carbohydrate-active enzymes and fermentable intermediate molecules for sustaining gut microbial symbiosis and improving fitness of own or other communities. Genomics approaches have provided unprecedented insights into these functions, but their precise mechanisms of action have poorly known. Sympathetic glycan-utilising strategy of each gut commensal will provide overview of mechanistic dynamic nature of the gut environment and will then assist in applying aptly personalised nutritional therapy. Thus, the review critically summarises cutting edge understanding of major plant- and host-derived glycan-utilising systems of Bacteroides and Bifidobacteria. Their evolutionary adaptation to gut environment and roles of postbiotics in human health are also highlighted.
Collapse
Affiliation(s)
- Ravindra Pal Singh
- Food and Nutritional Biotechnology Division, National Agri-Food Biotechnology Institute (NABI), SAS, Nagar, Punjab, 140306, India.
| |
Collapse
|
36
|
Jones MB, Alvarez CA, Johnson JL, Zhou JY, Morris N, Cobb BA. CD45Rb-low effector T cells require IL-4 to induce IL-10 in FoxP3 Tregs and to protect mice from inflammation. PLoS One 2019; 14:e0216893. [PMID: 31120919 PMCID: PMC6533033 DOI: 10.1371/journal.pone.0216893] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 04/30/2019] [Indexed: 01/04/2023] Open
Abstract
CD4+ effector/memory T cells (Tem) represent a leading edge of the adaptive immune system responsible for protecting the body from infection, cancer, and other damaging processes. However, a subset of Tem cells with low expression of CD45Rb (RbLoTem) has been shown to suppress inflammation despite their effector surface phenotype and the lack of FoxP3 expression, the canonical transcription factor found in most regulatory T cells. In this report, we show that RbLoTem cells can suppress inflammation by influencing Treg behavior. Co-culturing activated RbLoTem and Tregs induced high expression of IL-10 in vitro, and conditioned media from RbLoTem cells induced IL-10 expression in FoxP3+ Tregs in vitro and in vivo, indicating that RbLoTem cells communicate with Tregs in a cell-contact independent fashion. Transcriptomic and multi-analyte Luminex data identified both IL-2 and IL-4 as potential mediators of RbLoTem-Treg communication, and antibody-mediated neutralization of either IL-4 or CD124 (IL-4Rα) prevented IL-10 induction in Tregs. Moreover, isolated Tregs cultured with recombinant IL-2 and IL-4 strongly induced IL-10 production. Using house dust mite (HDM)-induced airway inflammation as a model, we confirmed that the in vivo suppressive activity of RbLoTem cells was lost in IL-4-ablated RbLoTem cells. These data support a model in which RbLoTem cells communicate with Tregs using a combination of IL-2 and IL-4 to induce robust expression of IL-10 and suppression of inflammation.
Collapse
Affiliation(s)
- Mark B. Jones
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Carlos A. Alvarez
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Jenny L. Johnson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Julie Y. Zhou
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Nathan Morris
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Brian A. Cobb
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
37
|
Nicolas GR, Chang PV. Deciphering the Chemical Lexicon of Host-Gut Microbiota Interactions. Trends Pharmacol Sci 2019; 40:430-445. [PMID: 31079848 DOI: 10.1016/j.tips.2019.04.006] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/03/2019] [Accepted: 04/12/2019] [Indexed: 02/06/2023]
Abstract
The human intestine harbors an immense, diverse, and critical population of bacteria that has effects on numerous aspects of host physiology, immunity, and disease. Emerging evidence suggests that many of the interactions between the host and the gut microbiota are mediated via the microbial metabolome, or the collection of small-molecule metabolites produced by intestinal bacteria. This review summarizes findings from recent work by focusing on different classes of metabolites produced by the gut microbiota and their effects in modulating host health and disease. These metabolites ultimately serve as a form of communication between the gut microbiome and the host, and a better understanding of this chemical language could potentially lead to novel strategies for treating a wide variety of human disorders.
Collapse
Affiliation(s)
- Gael R Nicolas
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Pamela V Chang
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA; Center for Infection and Pathobiology, Cornell University, Ithaca, NY 14853, USA; Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
38
|
Kulkarni DH, Newberry RD. Intestinal Macromolecular Transport Supporting Adaptive Immunity. Cell Mol Gastroenterol Hepatol 2019; 7:729-737. [PMID: 30710727 PMCID: PMC6463120 DOI: 10.1016/j.jcmgh.2019.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 01/11/2019] [Accepted: 01/11/2019] [Indexed: 12/13/2022]
Abstract
The gastrointestinal tract performs opposing functions of nutrient absorption, barrier maintenance, and the delivery of luminal substances for the appropriate induction of tolerogenic or protective adaptive immunity. The single-layer epithelium lining the gastrointestinal tract is central to each of these functions by facilitating the uptake and processing of nutrients, providing a physical and chemical barrier to potential pathogens, and delivering macromolecular substances to the immune system to initiate adaptive immune responses. Specific transport mechanisms allow nutrient uptake and the delivery of macromolecules to the immune system while maintaining the epithelial barrier. This review examines historical observations supporting macromolecular transport by the intestinal epithelium, recent insights into the transport of luminal macromolecules to promote adaptive immunity, and how this process is regulated to promote appropriate immune responses. Understanding how luminal macromolecules are delivered to the immune system and how this is regulated may provide insight into the pathophysiology of inflammatory diseases of the gastrointestinal tract and potential preventative or therapeutic strategies.
Collapse
Affiliation(s)
- Devesha H Kulkarni
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Rodney D Newberry
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
39
|
Benner M, Ferwerda G, Joosten I, van der Molen RG. How uterine microbiota might be responsible for a receptive, fertile endometrium. Hum Reprod Update 2019; 24:393-415. [PMID: 29668899 DOI: 10.1093/humupd/dmy012] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 03/27/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Fertility depends on a receptive state of the endometrium, influenced by hormonal and anatomical adaptations, as well as the immune system. Local and systemic immunity is greatly influenced by microbiota. Recent discoveries of 16S rRNA in the endometrium and the ability to detect low-biomass microbiota fueled the notion that the uterus may be indeed a non-sterile compartment. To date, the concept of the 'sterile womb' focuses on in utero effects of microbiota on offspring and neonatal immunity. However, little awareness has been raised regarding the importance of uterine microbiota for endometrial physiology in reproductive health; manifested in fertility and placentation. OBJECTIVE AND RATIONALE Commensal colonization of the uterus has been widely discussed in the literature. The objective of this review is to outline the possible importance of this uterine colonization for a healthy, fertile uterus. We present the available evidence regarding uterine microbiota, focusing on recent findings based on 16S rRNA, and depict the possible importance of uterine colonization for a receptive endometrium. We highlight a possible role of uterine microbiota for host immunity and tissue adaptation, as well as conferring protection against pathogens. Based on knowledge of the interaction of the mucosal immune cells of the gut with the local microbiome, we want to investigate the potential implications of commensal colonization for uterine health. SEARCH METHODS PubMed and Google Scholar were searched for articles in English indexed from 1 January 2008 to 1 March 2018 for '16S rRNA', 'uterus' and related search terms to assess available evidence on uterine microbiome analysis. A manual search of the references within the resulting articles was performed. To investigate possible functional contributions of uterine microbiota to health, studies on microbiota of other body sites were additionally assessed. OUTCOMES Challenging the view of a sterile uterus is in its infancy and, to date, no conclusions on a 'core uterine microbiome' can be drawn. Nevertheless, evidence for certain microbiota and/or associated compounds in the uterus accumulates. The presence of microbiota or their constituent molecules, such as polysaccharide A of the Bacteroides fragilis capsule, go together with healthy physiological function. Lessons learned from the gut microbiome suggest that the microbiota of the uterus may potentially modulate immune cell subsets needed for implantation and have implications for tissue morphology. Microbiota can also be crucial in protection against uterine infections by defending their niche and competing with pathogens. Our review highlights the need for well-designed studies on a 'baseline' microbial state of the uterus representing the optimal starting point for implantation and subsequent placenta formation. WIDER IMPLICATIONS The complex interplay of processes and cells involved in healthy pregnancy is still poorly understood. The correct receptive endometrial state, including the local immune environment, is crucial not only for fertility but also placenta formation since initiation of placentation highly depends on interaction with immune cells. Implantation failure, recurrent pregnancy loss, and other pathologies of endometrium and placenta, such as pre-eclampsia, represent an increasing societal burden. More robust studies are needed to investigate uterine colonization. Based on current data, future research needs to include the uterine microbiome as a relevant factor in order to understand the players needed for healthy pregnancy.
Collapse
Affiliation(s)
- Marilen Benner
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Geert Grooteplein 10, PO Box 9101, Internal mail 469, 6500 HB Nijmegen, The Netherlands
| | - Gerben Ferwerda
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Geert Grooteplein 10, PO Box 9101, Internal mail 469, 6500 HB Nijmegen, The Netherlands
| | - Irma Joosten
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Geert Grooteplein 10, PO Box 9101, Internal mail 469, 6500 HB Nijmegen, The Netherlands
| | - Renate G van der Molen
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Geert Grooteplein 10, PO Box 9101, Internal mail 469, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
40
|
Abstract
Capsular polysaccharides are a dominant class of antigens from bacteria, both pathogenic and symbiotic or commensal. With the rise of awareness for the influence of the microbiota over immune system development and immune homeostasis, analysis of the antigens is more important than ever. Here we describe a method for the isolation of capsular polysaccharide from gram-negative bacteria, with the purification of polysaccharide from the commensal bacterium Bacteroides fragilis serving as an example. The method efficiently removes all detectable endotoxins and other lipid components, proteins, and nucleic acids, providing a source of capsular polysaccharide for immunologic study.
Collapse
|
41
|
Johnson JL, Jones MB, Cobb BA. Polysaccharide-experienced effector T cells induce IL-10 in FoxP3+ regulatory T cells to prevent pulmonary inflammation. Glycobiology 2018; 28:50-58. [PMID: 29087497 DOI: 10.1093/glycob/cwx093] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 10/23/2017] [Indexed: 01/07/2023] Open
Abstract
Inhibition of peripheral inflammatory disease by carbohydrate antigens derived from normal gut microbiota has been demonstrated for the GI tract, brain, peritoneum, and most recently the airway. We have demonstrated that polysaccharide A (PSA) from the commensal organism Bacteroides fragilis activates CD4+ T cells upon presentation by the class II major histocompatibility complex, and that these PSA-experienced T cells prevent the development of lung inflammation in murine models. While the PSA-responding T cells themselves are not canonical FoxP3+ regulatory T cells (Tregs), their ability to prevent inflammation is dependent upon the suppressive cytokine IL-10. Using an adoptive T cell transfer approach, we have discovered that PSA-experienced T cells require IL-10 expression by PSA-naïve recipient animals in order to prevent inflammation. A cooperative relationship was found between PSA-activated effector/memory T cells and tissue-resident FoxP3+ Tregs both in vivo and in vitro, and it is this cooperation that enables the suppressive activity of PSA outside of the gut environment where exposure takes place. These findings suggest that carbohydrate antigens from the normal microbiota communicate with peripheral tissues to maintain homeostasis through T cell-to-T cell cooperation.
Collapse
Affiliation(s)
- Jenny L Johnson
- Case Western Reserve University School of Medicine, Department of Pathology, 10900 Euclid Avenue, Cleveland, OH 44106-7288, USA
| | - Mark B Jones
- Case Western Reserve University School of Medicine, Department of Pathology, 10900 Euclid Avenue, Cleveland, OH 44106-7288, USA
| | - Brian A Cobb
- Case Western Reserve University School of Medicine, Department of Pathology, 10900 Euclid Avenue, Cleveland, OH 44106-7288, USA
| |
Collapse
|
42
|
Emerging glycobiology tools: A renaissance in accessibility. Cell Immunol 2018; 333:2-8. [PMID: 29759530 DOI: 10.1016/j.cellimm.2018.04.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/23/2018] [Accepted: 04/24/2018] [Indexed: 01/01/2023]
Abstract
The glycobiology of the immune response is a topic that has garnered increased attention due to a number of key discoveries surrounding IgG function, the specificity of some broadly neutralizing anti-HIV antibodies, cancer immunoregulation by galectin molecules and others. This review is the opening article in a Special Edition of Cellular Immunology focused on glycoimmunology, and has the goal of setting the context for these articles by providing a mini-review of how glycans impact immunity. We also focus on some of the technological and methodological advances in the field of glycobiology that are being deployed to lower the barrier of entry into the glycosciences, and to more fully interrogate the glycome and its function.
Collapse
|
43
|
Martín R, Chain F, Miquel S, Motta JP, Vergnolle N, Sokol H, Langella P. Using murine colitis models to analyze probiotics-host interactions. FEMS Microbiol Rev 2018; 41:S49-S70. [PMID: 28830096 DOI: 10.1093/femsre/fux035] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/08/2017] [Indexed: 02/07/2023] Open
Abstract
Probiotics are defined as 'live microorganisms which when administered in adequate amounts confer a health benefit on the host'. So, to consider a microorganism as a probiotic, a demonstrable beneficial effect on the health host should be shown as well as an adequate defined safety status and the capacity to survive transit through the gastrointestinal tract and to storage conditions. In this review, we present an overview of the murine colitis models currently employed to test the beneficial effect of the probiotic strains as well as an overview of the probiotics already tested. Our aim is to highlight both the importance of the adequate selection of the animal model to test the potential probiotic strains and of the value of the knowledge generated by these in vivo tests.
Collapse
Affiliation(s)
- Rebeca Martín
- INRA, Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Florian Chain
- INRA, Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Sylvie Miquel
- Laboratoire Microorganismes: Génome et Environnement (LMGE), UMR CNRS 6023, Université Clermont-Auvergne, 63000 Clermont-Ferrand, France
| | - Jean-Paul Motta
- Department of Biological Science, Inflammation Research Network, University of Calgary, AB T3E 4N1, Canada.,IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, F-31300 Toulouse, France
| | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, F-31300 Toulouse, France
| | - Harry Sokol
- INRA, Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France.,Sorbonne University - Université Pierre et Marie Curie (UPMC), 75252 Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Equipe de Recherche Labélisée (ERL) 1157, Avenir Team Gut Microbiota and Immunity, 75012 Paris, France.,Department of Gastroenterology, Saint Antoine Hospital, Assistance Publique - Hopitaux de Paris, UPMC, 75012 Paris, France
| | - Philippe Langella
- INRA, Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| |
Collapse
|
44
|
Erturk-Hasdemir D, Kasper DL. Finding a needle in a haystack: Bacteroides fragilis polysaccharide A as the archetypical symbiosis factor. Ann N Y Acad Sci 2018. [PMID: 29528123 DOI: 10.1111/nyas.13660] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Starting from birth, all animals develop a symbiotic relationship with their resident microorganisms that benefits both the microbe and the host. Recent advances in technology have substantially improved our ability to direct research toward the identification of important microbial species that affect host physiology. The identification of specific commensal molecules from these microbes and their mechanisms of action is still in its early stages. Polysaccharide A (PSA) of Bacteroides fragilis is the archetypical example of a commensal molecule that can modulate the host immune system in health and disease. This zwitterionic polysaccharide has a critical impact on the development of the mammalian immune system and also on the stimulation of interleukin 10-producing CD4+ T cells; consequently, PSA confers benefits to the host with regard to experimental autoimmune, inflammatory, and infectious diseases. In this review, we summarize the current understanding of the immunomodulatory effects of B. fragilis PSA and discuss these effects as a novel immunological paradigm. In particular, we discuss recent advances in our understanding of the unique functional mechanisms of this molecule and its therapeutic potential, and we review the recent literature in the field of microbiome research aimed at discovering new commensal products and their immunomodulatory potential.
Collapse
Affiliation(s)
- Deniz Erturk-Hasdemir
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts
| | - Dennis L Kasper
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
45
|
Interplay of Carbohydrate and Carrier in Antibacterial Glycoconjugate Vaccines. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2018; 175:355-378. [PMID: 30143807 DOI: 10.1007/10_2018_71] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bacterial infections are a serious health concern and are responsible for millions of illnesses and deaths each year in communities around the world. Vaccination is an important public health measure for reducing and eliminating this burden, and regions with comprehensive vaccination programs have achieved significant reductions in infection and mortality. This is often accomplished by immunization with bacteria-derived carbohydrates, typically in conjunction with other biomolecules, which induce immunological memory and durable protection against bacterial human pathogens. For many species, however, vaccines are currently unavailable or have suboptimal efficacy characterized by short-lived memory and incomplete protection, especially among at-risk populations. To address this challenge, new tools and techniques have emerged for engineering carbohydrates and conjugating them to carrier molecules in a tractable and scalable manner. Collectively, these approaches are yielding carbohydrate-based vaccine designs with increased immunogenicity and protective efficacy, thereby opening up new opportunities for this important class of antigens. In this chapter we detail the current understanding of how carbohydrates interact with the immune system to provide immunity; how glycoengineering, especially in the context of glycoconjugate vaccines, can be used to modify and enhance immune responses; and current trends and strategies being pursued for the rational design of next-generation glycosylated antibacterial vaccines. Graphical Abstract.
Collapse
|
46
|
Spiljar M, Merkler D, Trajkovski M. The Immune System Bridges the Gut Microbiota with Systemic Energy Homeostasis: Focus on TLRs, Mucosal Barrier, and SCFAs. Front Immunol 2017; 8:1353. [PMID: 29163467 PMCID: PMC5670327 DOI: 10.3389/fimmu.2017.01353] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/03/2017] [Indexed: 12/17/2022] Open
Abstract
The gut microbiota is essential for the development and regulation of the immune system and the metabolism of the host. Germ-free animals have altered immunity with increased susceptibility to immunologic diseases and show metabolic alterations. Here, we focus on two of the major immune-mediated microbiota-influenced components that signal far beyond their local environment. First, the activation or suppression of the toll-like receptors (TLRs) by microbial signals can dictate the tone of the immune response, and they are implicated in regulation of the energy homeostasis. Second, we discuss the intestinal mucosal surface is an immunologic component that protects the host from pathogenic invasion, is tightly regulated with regard to its permeability and can influence the systemic energy balance. The short chain fatty acids are a group of molecules that can both modulate the intestinal barrier and escape the gut to influence systemic health. As modulators of the immune response, the microbiota-derived signals influence functions of distant organs and can change susceptibility to metabolic diseases.
Collapse
Affiliation(s)
- Martina Spiljar
- Faculty of Medicine, Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland.,Diabetes Center, Faculty of Medicine, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland
| | - Doron Merkler
- Faculty of Medicine, Department of Pathology and Immunology, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland
| | - Mirko Trajkovski
- Faculty of Medicine, Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland.,Diabetes Center, Faculty of Medicine, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland.,Institute of Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland
| |
Collapse
|
47
|
Greenhalgh K, Meyer KM, Aagaard KM, Wilmes P. The human gut microbiome in health: establishment and resilience of microbiota over a lifetime. Environ Microbiol 2017; 18:2103-16. [PMID: 27059297 PMCID: PMC7387106 DOI: 10.1111/1462-2920.13318] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
With technological advances in culture-independent molecular methods, we are uncovering a new facet of our natural history by accounting for the vast diversity of microbial life which colonizes the human body. The human microbiome contributes functional genes and metabolites which affect human physiology and are, therefore, considered an important factor for maintaining health. Much has been described in the past decade based primarily on 16S rRNA gene amplicon sequencing regarding the diversity, structure, stability and dynamics of human microbiota in their various body habitats, most notably within the gastrointestinal tract (GIT). Relatively high levels of variation have been described across different stages of life and geographical locations for the GIT microbiome. These observations may prove helpful for the future contextualization of patterns in other body habitats especially in relation to identifying generalizable trends over human lifetime. Given the large degree of complexity and variability, a key challenge will be how to define baseline healthy microbiomes and how to identify features which reflect deviations therefrom in the future. In this context, metagenomics and functional omics will likely play a central role as they will allow resolution of microbiome-conferred functionalities associated with health. Such information will be vital for formulating therapeutic interventions aimed at managing microbiota-mediated health particularly in the GIT over the course of a human lifetime.
Collapse
Affiliation(s)
- Kacy Greenhalgh
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Kristen M Meyer
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Kjersti M Aagaard
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| |
Collapse
|
48
|
Jun JC, Jones MB, Oswald DM, Sim ES, Jonnalagadda AR, Kreisman LSC, Cobb BA. T cell-intrinsic TLR2 stimulation promotes IL-10 expression and suppressive activity by CD45RbHi T cells. PLoS One 2017; 12:e0180688. [PMID: 28742882 PMCID: PMC5526543 DOI: 10.1371/journal.pone.0180688] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/20/2017] [Indexed: 12/20/2022] Open
Abstract
While Toll-like receptors (TLRs) represent one of the best characterized innate immune pathways, evidence suggests that TLRs are not restricted to innate leukocytes and some epithelial cells, but are also expressed in T cells. Specifically, published evidence focusing on FoxP3+ regulatory T cells demonstrate that they express functional TLR2, which is already known among the TLR family for its association with immune suppression; however, little is known about the relationship between T cell-intrinsic TLR2 binding and cytokine production, T cell differentiation, or T cell receptor (TCR) stimulation. Here, we demonstrate that TCR and TLR2 co-stimulation provides a T cell-intrinsic signal which generates a dramatic, synergistic cytokine response dominated by IL-10. Importantly, the response was not seen in either CD4+CD25+ or CD4+FoxP3+ Tregs, yet resulted in the expansion of a suppressive CD4+CD25+CD62L-CD44+CD45Rbhi effector/memory T cell subset not typically associated with immune inhibition. This study reveals the striking ability of a prototypical innate immune receptor to trigger a potent and suppressive IL-10 response in effector/memory T cells, supporting the notion that TLR2 is a co-regulatory receptor on T cells.
Collapse
Affiliation(s)
- Janice C. Jun
- Case Western Reserve University School of Medicine, Department of Pathology, Cleveland, OH, United States of America
- Case Western Reserve University School of Dental Medicine, Cleveland, OH, United States of America
| | - Mark B. Jones
- Case Western Reserve University School of Medicine, Department of Pathology, Cleveland, OH, United States of America
| | - Douglas M. Oswald
- Case Western Reserve University School of Medicine, Department of Pathology, Cleveland, OH, United States of America
| | - Edward S. Sim
- Case Western Reserve University School of Medicine, Department of Pathology, Cleveland, OH, United States of America
| | - Amruth R. Jonnalagadda
- Case Western Reserve University School of Medicine, Department of Pathology, Cleveland, OH, United States of America
| | - Lori S. C. Kreisman
- Case Western Reserve University School of Medicine, Department of Pathology, Cleveland, OH, United States of America
| | - Brian A. Cobb
- Case Western Reserve University School of Medicine, Department of Pathology, Cleveland, OH, United States of America
- * E-mail:
| |
Collapse
|
49
|
Abstract
The human gastrointestinal tract is populated by a diverse, highly mutualistic microbial flora, which is known as the microbiome. Disruptions to the microbiome have been shown to be associated with severe pathologies of the host, including metabolic disease, cancer, and inflammatory bowel disease. Mood and behavior are also susceptible to alterations in the gut microbiota. A particularly striking example of the symbiotic effects of the microbiome is the immune system, whose cells depend critically on a diverse array of microbial metabolites for normal development and behavior. This includes metabolites that are produced by bacteria from dietary components, metabolites that are produced by the host and biochemically modified by gut bacteria, and metabolites that are synthesized de novo by gut microbes. In this review, we highlight the role of the intestinal microbiome in human metabolic and inflammatory diseases and focus in particular on the molecular mechanisms that govern the gut-immune axis.
Collapse
Affiliation(s)
- Thomas Siegmund Postler
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Sankar Ghosh
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
50
|
Jalanka J, Mattila E, Jouhten H, Hartman J, de Vos WM, Arkkila P, Satokari R. Long-term effects on luminal and mucosal microbiota and commonly acquired taxa in faecal microbiota transplantation for recurrent Clostridium difficile infection. BMC Med 2016; 14:155. [PMID: 27724956 PMCID: PMC5057499 DOI: 10.1186/s12916-016-0698-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 09/16/2016] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Faecal microbiota transplantation (FMT) is an effective treatment for recurrent Clostridium difficile infection (rCDI). It restores the disrupted intestinal microbiota and subsequently suppresses C. difficile. The long-term stability of the intestinal microbiota and the recovery of mucosal microbiota, both of which have not been previously studied, are assessed herein. Further, the specific bacteria behind the treatment efficacy are also investigated. METHODS We performed a high-throughput microbiota profiling using a phylogenetic microarray analysis of 131 faecal and mucosal samples from 14 rCDI patients pre- and post-FMT during a 1-year follow-up and 23 samples from the three universal donors over the same period. RESULTS The FMT treatment was successful in all patients. FMT reverted the patients' bacterial community to become dominated by Clostridium clusters IV and XIVa, the major anaerobic bacterial groups of the healthy gut. In the mucosa, the amount of facultative anaerobes decreased, whereas Bacteroidetes increased. Post-FMT, the patients' microbiota profiles were more similar to their own donors than what is generally observed for unrelated subjects and this striking similarity was retained throughout the 1-year follow-up. Furthermore, the universal donor approach allowed us to identify bacteria commonly established in all CDI patients and revealed a commonly acquired core microbiota consisting of 24 bacterial taxa. CONCLUSIONS FMT induces profound microbiota changes, therefore explaining the high clinical efficacy for rCDI. The identification of commonly acquired bacteria could lead to effective bacteriotherapeutic formulations. FMT can affect microbiota in the long-term and offers a means to modify it relatively permanently for the treatment of microbiota-associated diseases.
Collapse
Affiliation(s)
- Jonna Jalanka
- Immunobiology Research Program and Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Eero Mattila
- Department of Infectious Disease, Helsinki University Central Hospital, Helsinki, Finland
| | - Hanne Jouhten
- Immunobiology Research Program and Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jorn Hartman
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Willem M de Vos
- Immunobiology Research Program and Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Perttu Arkkila
- Department of Gastroenterology, Helsinki University Central Hospital, Helsinki, Finland
| | - Reetta Satokari
- Immunobiology Research Program and Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, Helsinki, Finland. .,, PO Box 21, Haartmaninkatu 3, 00290, Helsinki, Finland.
| |
Collapse
|