1
|
Ibrahim OM, Kalinski P. Breaking Barriers: Modulation of Tumor Microenvironment to Enhance Bacillus Calmette-Guérin Immunotherapy of Bladder Cancer. Cells 2024; 13:699. [PMID: 38667314 PMCID: PMC11049012 DOI: 10.3390/cells13080699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/13/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
The clinical management of bladder cancer continues to present significant challenges. Bacillus Calmette-Guérin (BCG) immunotherapy remains the gold standard of treatment for non-muscle invasive bladder cancer (NMIBC), but many patients develop recurrence and progression to muscle-invasive disease (MIBC), which is resistant to BCG. This review focuses on the immune mechanisms mobilized by BCG in bladder cancer tumor microenvironments (TME), mechanisms of BCG resistance, the dual role of the BCG-triggered NFkB/TNFα/PGE2 axis in the regulation of anti-tumor and tumor-promoting aspects of inflammation, and emerging strategies to modulate their balance. A better understanding of BCG resistance will help develop new treatments and predictive biomarkers, paving the way for improved clinical outcomes in bladder cancer patients.
Collapse
Affiliation(s)
- Omar M. Ibrahim
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Pawel Kalinski
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
2
|
Dabbaghipour R, Ahmadi E, Entezam M, Farzam OR, Sohrabi S, Jamali S, Sichani AS, Paydar H, Baradaran B. Concise review: The heterogenous roles of BATF3 in cancer oncogenesis and dendritic cells and T cells differentiation and function considering the importance of BATF3-dependent dendritic cells. Immunogenetics 2024; 76:75-91. [PMID: 38358555 DOI: 10.1007/s00251-024-01335-x] [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: 09/26/2023] [Accepted: 12/23/2023] [Indexed: 02/16/2024]
Abstract
The transcription factor, known as basic leucine zipper ATF-like 3 (BATF3), is a crucial contributor to the development of conventional type 1 dendritic cells (cDC1), which is definitely required for priming CD8 + T cell-mediated immunity against intracellular pathogens and malignancies. In this respect, BATF3-dependent cDC1 can bring about immunological tolerance, an autoimmune response, graft immunity, and defense against infectious agents such as viruses, microbes, parasites, and fungi. Moreover, the important function of cDC1 in stimulating CD8 + T cells creates an excellent opportunity to develop a highly effective target for vaccination against intracellular pathogens and diseases. BATF3 has been clarified to control the development of CD8α+ and CD103+ DCs. The presence of BATF3-dependent cDC1 in the tumor microenvironment (TME) reinforces immunosurveillance and improves immunotherapy approaches, which can be beneficial for cancer immunotherapy. Additionally, BATF3 acts as a transcriptional inhibitor of Treg development by decreasing the expression of the transcription factor FOXP3. However, when overexpressed in CD8 + T cells, it can enhance their survival and facilitate their transition to a memory state. BATF3 induces Th9 cell differentiation by binding to the IL-9 promoter through a BATF3/IRF4 complex. One of the latest research findings is the oncogenic function of BATF3, which has been approved and illustrated in several biological processes of proliferation and invasion.
Collapse
Affiliation(s)
- Reza Dabbaghipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ahmadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mona Entezam
- Department of Medical Genetics, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omid Rahbar Farzam
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Sohrabi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sajjad Jamali
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Saber Sichani
- Department of Medical Genetics, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Hadi Paydar
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Xu J, Hissong R, Bareis R, Creech A, Goughenour KD, Freeman CM, Olszewski MA. Batf3-dependent orchestration of the robust Th1 responses and fungal control during cryptococcal infection, the role of cDC1. mBio 2024; 15:e0285323. [PMID: 38349130 PMCID: PMC10936214 DOI: 10.1128/mbio.02853-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/22/2024] [Indexed: 03/14/2024] Open
Abstract
While type I conventional dendritic cells (cDC1s) are vital for generating adaptive immunity against intracellular pathogens and tumors, their role in defense against fungal pathogen Cryptococcus neoformans remains unclear. We investigated the role of the cDC1 subset in a fungus-restricting mouse model of cryptococcal infection. The cDC1 subset displayed a unique transcriptional signature with highly upregulated T-cell recruitment, polarization, and activation pathways compared to other DC subsets. Using Batf3-/- mice, which lack the cDC1 population, our results support that Batf3-dependent cDC1s are pivotal for the development of the effective immune response against cryptococcal infection, particularly within the lung and brain. Deficiency in Batf3 cDC1 led to diminished CD4 accumulation and decreased IFNγ production across multiple organs, supporting that cDC1s are a major driver of potent Th1 responses during cryptococcal infection. Consistently, mice lacking Batf3-cDC1 demonstrated markedly diminished fungicidal activity and weaker containment of the fungal pathogen. In conclusion, Batf3-dependent cDC1 can function as a linchpin in mounting Th1 response, ensuring effective fungal control during cryptococcal infection. Harnessing cDC1 pathways may present a promising strategy for interventions against this pathogen.IMPORTANCECryptococcus neoformans causes severe meningoencephalitis, accounting for an estimated 200,000 deaths each year. Central to mounting an effective defense against these infections is T-cell-mediated immunity, which is orchestrated by dendritic cells (DCs). The knowledge about the role of specific DC subsets in shaping anti-cryptococcal immunity is limited. Here, we demonstrate that Batf3 cDC1s are important drivers of protective Th1 CD4 T-cell responses required for clearance of cryptococcal infection. Deficiency of Batf3 cDC1 in the infected mice leads to significantly reduced Th1 response and exacerbated fungal growth to the point where depleting the remaining CD4 T cells no longer affects fungal burden. Unveiling this pivotal role of cDC1 in antifungal defense is likely to be important for the development of vaccines and therapies against life-threatening fungal pathogens.
Collapse
Affiliation(s)
- Jintao Xu
- Research Service, Department of Veterans Affairs Health System, Ann Arbor VA Health System, Ann Arbor, Michigan, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Rylan Hissong
- Research Service, Department of Veterans Affairs Health System, Ann Arbor VA Health System, Ann Arbor, Michigan, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Rachel Bareis
- Research Service, Department of Veterans Affairs Health System, Ann Arbor VA Health System, Ann Arbor, Michigan, USA
| | - Arianna Creech
- Research Service, Department of Veterans Affairs Health System, Ann Arbor VA Health System, Ann Arbor, Michigan, USA
| | - Kristie D. Goughenour
- Research Service, Department of Veterans Affairs Health System, Ann Arbor VA Health System, Ann Arbor, Michigan, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Christine M. Freeman
- Research Service, Department of Veterans Affairs Health System, Ann Arbor VA Health System, Ann Arbor, Michigan, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Michal A. Olszewski
- Research Service, Department of Veterans Affairs Health System, Ann Arbor VA Health System, Ann Arbor, Michigan, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
Kustrimovic N, Bilato G, Mortara L, Baci D. The Urinary Microbiome in Health and Disease: Relevance for Bladder Cancer. Int J Mol Sci 2024; 25:1732. [PMID: 38339010 PMCID: PMC10855347 DOI: 10.3390/ijms25031732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/24/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
Bladder cancer (BC) constitutes one of the most diagnosed types of cancer worldwide. Advancements in and new methodologies for DNA sequencing, leading to high-throughput microbiota testing, have pinpointed discrepancies in urinary microbial fingerprints between healthy individuals and patients with BC. Although several studies suggest an involvement of microbiota dysbiosis in the pathogenesis, progression, and therapeutic response to bladder cancer, an established direct causal relationship remains to be elucidated due to the lack of standardized methodologies associated with such studies. This review compiles an overview of the microbiota of the human urinary tract in healthy and diseased individuals and discusses the evidence to date on microbiome involvement and potential mechanisms by which the microbiota may contribute to the development of BC. We also explore the potential profiling of urinary microbiota as a biomarker for risk stratification, as well as the prediction of the response to intravesical therapies and immunotherapy in BC patients. Further investigation into the urinary microbiome of BC patients is imperative to unravel the complexities of the role played by host-microbe interactions in shaping wellness or disease and yield valuable insights into and strategies for the prevention and personalized treatment of BC.
Collapse
Affiliation(s)
- Natasa Kustrimovic
- Center for Translational Research on Autoimmune and Allergic Disease—CAAD, Università del Piemonte Orientale, 28100 Novara, Italy;
| | - Giorgia Bilato
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy;
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy;
| | - Denisa Baci
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy;
- Molecular Cardiology Laboratory, IRCCS—Policlinico San Donato, 20097 Milan, Italy
| |
Collapse
|
5
|
Jiménez-Cortegana C, Palomares F, Alba G, Santa-María C, de la Cruz-Merino L, Sánchez-Margalet V, López-Enríquez S. Dendritic cells: the yin and yang in disease progression. Front Immunol 2024; 14:1321051. [PMID: 38239364 PMCID: PMC10794555 DOI: 10.3389/fimmu.2023.1321051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/12/2023] [Indexed: 01/22/2024] Open
Abstract
Dendritic cells (DCs) are antigen presenting cells that link innate and adaptive immunity. DCs have been historically considered as the most effective and potent cell population to capture, process and present antigens to activate naïve T cells and originate favorable immune responses in many diseases, such as cancer. However, in the last decades, it has been observed that DCs not only promote beneficial responses, but also drive the initiation and progression of some pathologies, including inflammatory bowel disease (IBD). In line with those notions, different therapeutic approaches have been tested to enhance or impair the concentration and role of the different DC subsets. The blockade of inhibitory pathways to promote DCs or DC-based vaccines have been successfully assessed in cancer, whereas the targeting of DCs to inhibit their functionality has proved to be favorable in IBD. In this review, we (a) described the general role of DCs, (b) explained the DC subsets and their role in immunogenicity, (c) analyzed the role of DCs in cancer and therapeutic approaches to promote immunogenic DCs and (d) analyzed the role of DCs in IBD and therapeutic approaches to reduced DC-induced inflammation. Therefore, we aimed to highlight the "yin-yang" role of DCs to improve the understand of this type of cells in disease progression.
Collapse
Affiliation(s)
- Carlos Jiménez-Cortegana
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Francisca Palomares
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Gonzalo Alba
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Consuelo Santa-María
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, Seville, Spain
| | - Luis de la Cruz-Merino
- Clinical Oncology Dept. Medicine Department, University of Seville, Virgen Macarena University Hospital, Seville, Spain
| | - Victor Sánchez-Margalet
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Soledad López-Enríquez
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| |
Collapse
|
6
|
Liu Q, Chen C, He Y, Mai W, Ruan S, Ning Y, Li Y. Notch Signaling Regulates the Function and Phenotype of Dendritic Cells in Helicobacter pylori Infection. Microorganisms 2023; 11:2818. [PMID: 38004829 PMCID: PMC10673485 DOI: 10.3390/microorganisms11112818] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Notch signaling manipulates the function and phenotype of dendritic cells (DCs), as well as the interaction between DCs and CD4+ T cells. However, the role of Notch signaling in Helicobacter pylori (H. pylori) infection remains elusive. Murine bone marrow-derived dendritic cells (BMDCs) were pretreated in the absence or presence of Notch signaling inhibitor DAPT prior to H. pylori stimulation and the levels of Notch components, cytokines and surface markers as well as the differentiation of CD4+ T cells in co-culture were measured using quantitative real-time PCR (qRT-PCR), Western blot, enzyme-linked immunosorbent assay (ELISA) and flow cytometry. Compared with the control, the mRNA expression of all Notch receptors and Notch ligands Dll4 and Jagged1 was up-regulated in H. pylori-stimulated BMDCs. The blockade of Notch signaling by DAPT influenced the production of IL-1β and IL-10 in H. pylori-pulsed BMDCs, and reduced the expression of Notch1, Notch3, Notch4, Dll1, Dll3 and Jagged2. In addition, DAPT pretreatment decreased the expression of maturation markers CD80, CD83, CD86, and major histocompatibility complex class II (MHC-II) of BMDCs, and further skewed Th17/Treg balance toward Treg. Notch signaling regulates the function and phenotype of DCs, thus mediating the differentiation of CD4+ T cells during H. pylori infection.
Collapse
Affiliation(s)
| | | | | | | | | | - Yunshan Ning
- School of Laboratory Medicine and Biotechnology, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China (W.M.)
| | - Yan Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China (W.M.)
| |
Collapse
|
7
|
Hao L, Zhong W, Woo J, Wei X, Ma H, Dong H, Guo W, Sun X, Yue R, Zhao J, Zhang Q, Zhou Z. Conventional type 1 dendritic cells protect against gut barrier disruption via maintaining Akkermansia muciniphila in alcoholic steatohepatitis. Hepatology 2023; 78:896-910. [PMID: 36626632 PMCID: PMC11140646 DOI: 10.1097/hep.0000000000000019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 10/07/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND AIMS Alcohol-perturbed gut immune homeostasis is associated with the development of alcoholic liver disease (ALD). However, the role of intestinal dendritic cells (DCs) in ALD progression is still unknown. This study aimed to investigate the cellular and molecular mechanisms through which intestinal DCs respond to alcohol exposure and contribute to the pathogenesis of ALD. APPROACH AND RESULTS After 8 weeks of alcohol consumption, the number of basic leucine zipper transcription factor ATF-like 3 ( Batf3 )-dependent conventional type 1 DCs (cDC1s) was dramatically decreased in the intestine but not the liver. cDC1 deficient Batf3 knockout mice along with wild-type mice were subjected to chronic-binge ethanol feeding to determine the role of intestinal cDC1s reduction in ALD. cDC1s deficiency exacerbated alcohol-induced gut barrier disruption, bacterial endotoxin translocation into the circulation, and liver injury. Adoptive transfer of cDC1s to alcohol-fed mice ameliorated alcohol-mediated gut barrier dysfunction and liver injury. Further studies revealed that intestinal cDC1s serve as a positive regulator of Akkermansia muciniphila ( A. muciniphila ). Oral administration of A. muciniphila markedly reversed alcoholic steatohepatitis in mice. Mechanistic studies revealed that cDC1s depletion exacerbated alcohol-downregulated intestinal antimicrobial peptides which play a crucial role in maintaining A. muciniphila abundance, by disrupting the IL-12-interferon gamma signaling pathway. Lastly, we identified that intestinal cDC1s were required for the protective role of Lactobacillus reuteri in alcoholic steatohepatitis. CONCLUSIONS This study demonstrated that cDC1s protect alcohol-induced liver injury by maintaining A. muciniphila abundance in mice. Targeting cDC1s may serve as a promising therapeutic approach for treating ALD.
Collapse
Affiliation(s)
- Liuyi Hao
- Center for Translational Biomedical Research, Kannapolis, North Carolina, USA
| | - Wei Zhong
- Center for Translational Biomedical Research, Kannapolis, North Carolina, USA
- Department of Nutrition, Kannapolis, North Carolina, USA
| | - Jongmin Woo
- Center for Translational Biomedical Research, Kannapolis, North Carolina, USA
| | - Xiaoyuan Wei
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, Arkansas, USA
| | - Hao Ma
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Haibo Dong
- Center for Translational Biomedical Research, Kannapolis, North Carolina, USA
| | - Wei Guo
- Center for Translational Biomedical Research, Kannapolis, North Carolina, USA
| | - Xinguo Sun
- Center for Translational Biomedical Research, Kannapolis, North Carolina, USA
| | - Ruichao Yue
- Center for Translational Biomedical Research, Kannapolis, North Carolina, USA
| | - Jiangchao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, Arkansas, USA
| | - Qibin Zhang
- Center for Translational Biomedical Research, Kannapolis, North Carolina, USA
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina, USA
| | - Zhanxiang Zhou
- Center for Translational Biomedical Research, Kannapolis, North Carolina, USA
- Department of Nutrition, Kannapolis, North Carolina, USA
| |
Collapse
|
8
|
Noori M, Fayyaz F, Rezaei N. Impact of Helicobacter pylori infection on the efficacy of immune checkpoint inhibitors for cancer treatment: a meta-analysis. Immunotherapy 2023; 15:657-667. [PMID: 37140002 DOI: 10.2217/imt-2022-0250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023] Open
Abstract
Aim: The present systematic review and meta-analysis was designed to assess the impact of Helicobacter pylori infection on the efficacy of immune checkpoint inhibitors (ICIs). Materials & methods: PubMed, Scopus, Web of Science and EMBASE databases were systematically searched up to 1 February 2023. Results: Three studies comprising 263 patients treated with ICIs were included. The results of pooled analysis showed that H. pylori infection was associated with reduced overall survival and progression-free survival. Furthermore, the rate of progressive disease after administration of ICIs was higher in H. pylori-positive patients relative to H. pylori-negative patients. Conclusion: H. pylori infection status is a novel potential response biomarker for predicting the efficacy of ICIs in different cancers.
Collapse
Affiliation(s)
- Maryam Noori
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Urology Research Center, Tehran University of Medical Science, Tehran, Iran
| | - Farimah Fayyaz
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Helicobacter pylori Chronic-Stage Inflammation Undergoes Fluctuations That Are Altered in tlpA Mutants. Infect Immun 2023; 91:e0032222. [PMID: 36533917 PMCID: PMC9872690 DOI: 10.1128/iai.00322-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Helicobacter pylori colonizes half of the world's population and is responsible for a significant disease burden by causing gastritis, peptic ulcers, and gastric cancer. The development of host inflammation drives these diseases, but there are still open questions in the field about how H. pylori controls this process. We characterized H. pylori inflammation using an 8-month mouse infection time course and comparison of the wild type (WT) and a previously identified mutant lacking the TlpA chemoreceptor that causes elevated inflammation. Our work shows that H. pylori chronic-stage corpus inflammation undergoes surprising fluctuations, with changes in Th17 and eosinophil numbers. The H. pylori tlpA mutant changed the inflammation temporal characteristics, resulting in different inflammation from the wild type at some time points. tlpA mutants have equivalent total and gland colonization in late-stage infections. During early infection, in contrast, they show elevated gland and total colonization compared to those by WT. Our results suggest the chronic inflammation setting is dynamic and may be influenced by colonization properties of early infection.
Collapse
|
10
|
Fuchs S, Gong R, Gerhard M, Mejías-Luque R. Immune Biology and Persistence of Helicobacter pylori in Gastric Diseases. Curr Top Microbiol Immunol 2023; 444:83-115. [PMID: 38231216 DOI: 10.1007/978-3-031-47331-9_4] [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] [Indexed: 01/18/2024]
Abstract
Helicobacter pylori is a prevalent pathogen, which affects more than 40% of the global population. It colonizes the human stomach and persists in its host for several decades or even a lifetime, if left untreated. The persistent infection has been linked to various gastric diseases, including gastritis, peptic ulcers, and an increased risk for gastric cancer. H. pylori infection triggers a strong immune response directed against the bacterium associated with the infiltration of innate phagocytotic immune cells and the induction of a Th1/Th17 response. Even though certain immune cells seem to be capable of controlling the infection, the host is unable to eliminate the bacteria as H. pylori has developed remarkable immune evasion strategies. The bacterium avoids its killing through innate recognition mechanisms and manipulates gastric epithelial cells and immune cells to support its persistence. This chapter focuses on the innate and adaptive immune response induced by H. pylori infection, and immune evasion strategies employed by the bacterium to enable persistent infection.
Collapse
Affiliation(s)
- Sonja Fuchs
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich (TUM), Trogerstraße 30, 81675, Munich, Germany
| | - Ruolan Gong
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich (TUM), Trogerstraße 30, 81675, Munich, Germany
| | - Markus Gerhard
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich (TUM), Trogerstraße 30, 81675, Munich, Germany
| | - Raquel Mejías-Luque
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich (TUM), Trogerstraße 30, 81675, Munich, Germany.
| |
Collapse
|
11
|
Poh AR, O'Brien M, Chisanga D, He H, Baloyan D, Traichel J, Dijkstra C, Chopin M, Nutt S, Whitehead L, Boon L, Parkin A, Lowell C, Pajic M, Shi W, Nikfarjam M, Ernst M. Inhibition of HCK in myeloid cells restricts pancreatic tumor growth and metastasis. Cell Rep 2022; 41:111479. [PMID: 36223746 PMCID: PMC11299506 DOI: 10.1016/j.celrep.2022.111479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 08/18/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with a low 5-year survival rate and is associated with poor response to therapy. Elevated expression of the myeloid-specific hematopoietic cell kinase (HCK) is observed in PDAC and correlates with reduced patient survival. To determine whether aberrant HCK signaling in myeloid cells is involved in PDAC growth and metastasis, we established orthotopic and intrasplenic PDAC tumors in wild-type and HCK knockout mice. Genetic ablation of HCK impaired PDAC growth and metastasis by inducing an immune-stimulatory endotype in myeloid cells, which in turn reduced the desmoplastic microenvironment and enhanced cytotoxic effector cell infiltration. Consequently, genetic ablation or therapeutic inhibition of HCK minimized metastatic spread, enhanced the efficacy of chemotherapy, and overcame resistance to anti-PD1, anti-CTLA4, or stimulatory anti-CD40 immunotherapy. Our results provide strong rationale for HCK to be developed as a therapeutic target to improve the response of PDAC to chemo- and immunotherapy.
Collapse
Affiliation(s)
- Ashleigh R Poh
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Melbourne, VIC 3084, Australia
| | - Megan O'Brien
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Melbourne, VIC 3084, Australia
| | - David Chisanga
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Melbourne, VIC 3084, Australia
| | - Hong He
- Department of Surgery, University of Melbourne and Austin Health, Melbourne, VIC 3084, Australia
| | - David Baloyan
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Melbourne, VIC 3084, Australia
| | - Jasmin Traichel
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg 79104, Germany
| | - Christine Dijkstra
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Melbourne, VIC 3084, Australia
| | - Michaël Chopin
- The Walter and Eliza Hall Institute and University of Melbourne Department of Medical Biology, Melbourne, VIC 3052, Australia
| | - Stephen Nutt
- The Walter and Eliza Hall Institute and University of Melbourne Department of Medical Biology, Melbourne, VIC 3052, Australia
| | - Lachlan Whitehead
- The Walter and Eliza Hall Institute and University of Melbourne Department of Medical Biology, Melbourne, VIC 3052, Australia
| | | | - Ashleigh Parkin
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Clifford Lowell
- University of California San Francisco, San Francisco, CA 94131, USA
| | - Marina Pajic
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Wei Shi
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Melbourne, VIC 3084, Australia; Department of Computing and Information Systems, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Mehrdad Nikfarjam
- Department of Surgery, University of Melbourne and Austin Health, Melbourne, VIC 3084, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Melbourne, VIC 3084, Australia.
| |
Collapse
|
12
|
Ngoi S, Yang Y, Iwanowycz S, Gutierrez J, Li Y, Williams C, Hill M, Chung D, Allen C, Liu B. Migrating Type 2 Dendritic Cells Prime Mucosal Th17 Cells Specific to Small Intestinal Commensal Bacteria. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1200-1211. [PMID: 35995508 PMCID: PMC9492644 DOI: 10.4049/jimmunol.2200204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/12/2022] [Indexed: 01/04/2023]
Abstract
Dendritic cells (DCs) are professional APCs equipped with MHC-restricted Ags, costimulations, and cytokines that effectively prime and differentiate naive T cells into distinct functional subsets. The immune signals that DCs carry reflect the route of Ag uptake and the innate stimuli they received. In the mucosal tissues, owing to the great variety of foreign Ags and inflammatory cues, DCs are predominantly activated and migratory. In the small intestine, CD4 Th17 cells are abundant and have been shown to be regulated by DCs and macrophages. Using a mouse commensal bacteria experimental model, we identified that the early priming step of commensal-driven Th17 cells is controlled by bona fide Zbtb46-expressing DCs. CCR7-dependent migration of type 2 DCs (DC2s) from the small intestine to the mesenteric lymph nodes (MLNs) is essential for the activation of naive CD4 T cells. The migratory DC2 population in the MLNs is almost exclusively Esam+ cells. Single-cell RNA sequencing highlighted the abundance of costimulatory markers (CD40 and OX40) and chemokines (Ccl22 and Cxcl16) on MLN migratory DCs. Further resolution of MLN migratory DC2s revealed that the Th17-polarizing cytokine IL-6 colocalizes with DC2s expressing CD40, Ccl17, and Ccl22. Thus, early Th17 cell differentiation is initiated by a small subset of migratory DC2s in the gut-draining lymph nodes.
Collapse
Affiliation(s)
- Soo Ngoi
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH; and
| | - Yi Yang
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC
| | - Stephen Iwanowycz
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC
| | - Jennifer Gutierrez
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC
| | - Yingqi Li
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC
| | - Christina Williams
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC
| | - Megan Hill
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH; and
| | - Dongjun Chung
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH; and
| | - Carter Allen
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH; and
| | - Bei Liu
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH; and
| |
Collapse
|
13
|
Poh AR, Love CG, Chisanga D, Steer JH, Baloyan D, Chopin M, Nutt S, Rautela J, Huntington ND, Etemadi N, O’Brien M, O’Keefe R, Ellies LG, Macri C, Mintern JD, Whitehead L, Gangadhara G, Boon L, Chand AL, Lowell CA, Shi W, Pixley FJ, Ernst M. Therapeutic inhibition of the SRC-kinase HCK facilitates T cell tumor infiltration and improves response to immunotherapy. SCIENCE ADVANCES 2022; 8:eabl7882. [PMID: 35731867 PMCID: PMC9216510 DOI: 10.1126/sciadv.abl7882] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Although immunotherapy has revolutionized cancer treatment, many immunogenic tumors remain refractory to treatment. This can be largely attributed to an immunologically "cold" tumor microenvironment characterized by an accumulation of immunosuppressive myeloid cells and exclusion of activated T cells. Here, we demonstrate that genetic ablation or therapeutic inhibition of the myeloid-specific hematopoietic cell kinase (HCK) enables activity of antagonistic anti-programmed cell death protein 1 (anti-PD1), anti-CTLA4, or agonistic anti-CD40 immunotherapies in otherwise refractory tumors and augments response in treatment-susceptible tumors. Mechanistically, HCK ablation reprograms tumor-associated macrophages and dendritic cells toward an inflammatory endotype and enhances CD8+ T cell recruitment and activation when combined with immunotherapy in mice. Meanwhile, therapeutic inhibition of HCK in humanized mice engrafted with patient-derived xenografts counteracts tumor immunosuppression, improves T cell recruitment, and impairs tumor growth. Collectively, our results suggest that therapeutic targeting of HCK activity enhances response to immunotherapy by simultaneously stimulating immune cell activation and inhibiting the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Ashleigh R. Poh
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria 3084, Australia
| | - Christopher G. Love
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - David Chisanga
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria 3084, Australia
| | - James H. Steer
- School of Biomedical Sciences, University of Western Australia, Crawley, Western Australia 6009, Australia
| | - David Baloyan
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria 3084, Australia
| | - Michaël Chopin
- Walter and Eliza Hall Institute and Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Stephen Nutt
- Walter and Eliza Hall Institute and Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Jai Rautela
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3186, Australia
- oNKo-Innate Pty Ltd, Moonee Ponds, Victoria 3039, Australia
| | - Nicholas D. Huntington
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3186, Australia
- oNKo-Innate Pty Ltd, Moonee Ponds, Victoria 3039, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3186, Australia
| | - Nima Etemadi
- Walter and Eliza Hall Institute and Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Megan O’Brien
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria 3084, Australia
| | - Ryan O’Keefe
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria 3084, Australia
| | - Lesley G. Ellies
- School of Biomedical Sciences, University of Western Australia, Crawley, Western Australia 6009, Australia
| | - Christophe Macri
- Department of Biochemistry and Pharmacology, University of Melbourne and Bio21 Molecular Science and Biotechnology Institute, Melbourne, Victoria 3010, Australia
| | - Justine D. Mintern
- Department of Biochemistry and Pharmacology, University of Melbourne and Bio21 Molecular Science and Biotechnology Institute, Melbourne, Victoria 3010, Australia
| | - Lachlan Whitehead
- Walter and Eliza Hall Institute and Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Gangadhara Gangadhara
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria 3084, Australia
| | | | - Ashwini L. Chand
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria 3084, Australia
| | | | - Wei Shi
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria 3084, Australia
| | - Fiona J. Pixley
- School of Biomedical Sciences, University of Western Australia, Crawley, Western Australia 6009, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria 3084, Australia
- Corresponding author.
| |
Collapse
|
14
|
Jiang S, Redelman-Sidi G. BCG in Bladder Cancer Immunotherapy. Cancers (Basel) 2022; 14:3073. [PMID: 35804844 PMCID: PMC9264881 DOI: 10.3390/cancers14133073] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/20/2022] [Accepted: 06/20/2022] [Indexed: 01/18/2023] Open
Abstract
BCG is a live attenuated strain of Mycobacterium bovis that is primarily used as a vaccine against tuberculosis. In the past four decades, BCG has also been used for the treatment of non-muscle invasive bladder cancer (NMIBC). In patients with NMIBC, BCG reduces the risk of tumor recurrence and decreases the likelihood of progression to more invasive disease. Despite the long-term clinical experience with BCG, its mechanism of action is still being elucidated. Data from animal models and from human studies suggests that BCG activates both the innate and adaptive arms of the immune system eventually leading to tumor destruction. Herein, we review the current data regarding the mechanism of BCG and summarize the evidence for its clinical efficacy and recommended indications and clinical practice.
Collapse
Affiliation(s)
- Song Jiang
- Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Gil Redelman-Sidi
- Infectious Diseases Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
15
|
Neuper T, Frauenlob T, Posselt G, Horejs-Hoeck J. Beyond the gastric epithelium - the paradox of Helicobacter pylori-induced immune responses. Curr Opin Immunol 2022; 76:102208. [PMID: 35569416 DOI: 10.1016/j.coi.2022.102208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/08/2022] [Accepted: 04/14/2022] [Indexed: 11/03/2022]
Abstract
Chronic infections are typically characterized by an ineffective immune response to the inducing pathogen. While failing to clear the infectious microbe, the provoked inflammatory processes may cause severe tissue damage culminating in functional impairment of the affected organ. The human pathogen Helicobacter pylori is a uniquely successful Gram-negative microorganism inhabiting the gastric mucosa in approximately 50% of the world's population. This bacterial species has evolved spectacular means of evading immune surveillance and influencing host immunity, leading to a fragile equilibrium between proinflammatory and anti-inflammatory signals, the breakdown of which can have serious consequences for the host, including gastric ulceration and cancer. This review highlights novel insights into this delicate interaction between host and pathogen from an immunological perspective.
Collapse
Affiliation(s)
- Theresa Neuper
- Department of Biosciences and Medical Biology, University of Salzburg, Austria
| | - Tobias Frauenlob
- Department of Biosciences and Medical Biology, University of Salzburg, Austria; Cancer Cluster Salzburg (CCS), Austria
| | - Gernot Posselt
- Department of Biosciences and Medical Biology, University of Salzburg, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences and Medical Biology, University of Salzburg, Austria; Cancer Cluster Salzburg (CCS), Austria.
| |
Collapse
|
16
|
Sasaki I, Kato T, Hemmi H, Fukuda-Ohta Y, Wakaki-Nishiyama N, Yamamoto A, Kaisho T. Conventional Type 1 Dendritic Cells in Intestinal Immune Homeostasis. Front Immunol 2022; 13:857954. [PMID: 35693801 PMCID: PMC9184449 DOI: 10.3389/fimmu.2022.857954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/04/2022] [Indexed: 11/15/2022] Open
Abstract
Dendritic cells (DC) play critical roles in linking innate and adaptive immunity. DC are heterogenous and there are subsets with various distinct functions. One DC subset, conventional type 1 DC (cDC1), can be defined by expression of CD8α/CD103 in mice and CD141 in humans, or by expression of a chemokine receptor, XCR1, which is a conserved marker in both mice and human. cDC1 are characterized by high ability to ingest dying cells and to cross-present antigens for generating cytotoxic CD8 T cell responses. Through these activities, cDC1 play crucial roles in immune responses against infectious pathogens or tumors. Meanwhile, cDC1 involvement in homeostatic situations is not fully understood. Analyses by using mutant mice, in which cDC1 are ablated in vivo, revealed that cDC1 are critical for maintaining intestinal immune homeostasis. Here, we review the homeostatic roles of cDC1, focusing upon intestinal immunity.
Collapse
Affiliation(s)
- Izumi Sasaki
- Department of Immunology, Institute for Advanced Medicine, Wakayama Medical University, Wakayama, Japan
- *Correspondence: Izumi Sasaki, ; Tsuneyasu Kaisho,
| | - Takashi Kato
- Department of Immunology, Institute for Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Hiroaki Hemmi
- Department of Immunology, Institute for Advanced Medicine, Wakayama Medical University, Wakayama, Japan
- Laboratory of Immunology, Faculty of Veterinary Medicine, Okayama University of Science, Ehime, Japan
| | - Yuri Fukuda-Ohta
- Department of Immunology, Institute for Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Naoko Wakaki-Nishiyama
- Department of Immunology, Institute for Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Asumi Yamamoto
- Department of Immunology, Institute for Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute for Advanced Medicine, Wakayama Medical University, Wakayama, Japan
- *Correspondence: Izumi Sasaki, ; Tsuneyasu Kaisho,
| |
Collapse
|
17
|
Oster P, Vaillant L, Riva E, McMillan B, Begka C, Truntzer C, Richard C, Leblond MM, Messaoudene M, Machremi E, Limagne E, Ghiringhelli F, Routy B, Verdeil G, Velin D. Helicobacter pylori infection has a detrimental impact on the efficacy of cancer immunotherapies. Gut 2022; 71:457-466. [PMID: 34253574 PMCID: PMC8862014 DOI: 10.1136/gutjnl-2020-323392] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 06/24/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE In this study, we determined whether Helicobacter pylori (H. pylori) infection dampens the efficacy of cancer immunotherapies. DESIGN Using mouse models, we evaluated whether immune checkpoint inhibitors or vaccine-based immunotherapies are effective in reducing tumour volumes of H. pylori-infected mice. In humans, we evaluated the correlation between H. pylori seropositivity and the efficacy of the programmed cell death protein 1 (PD-1) blockade therapy in patients with non-small-cell lung cancer (NSCLC). RESULTS In mice engrafted with MC38 colon adenocarcinoma or B16-OVA melanoma cells, the tumour volumes of non-infected mice undergoing anticytotoxic T-lymphocyte-associated protein 4 and/or programmed death ligand 1 or anti-cancer vaccine treatments were significantly smaller than those of infected mice. We observed a decreased number and activation status of tumour-specific CD8+ T cells in the tumours of infected mice treated with cancer immunotherapies independent of the gut microbiome composition. Additionally, by performing an in vitro co-culture assay, we observed that dendritic cells of infected mice promote lower tumour-specific CD8+ T cell proliferation. We performed retrospective human clinical studies in two independent cohorts. In the Dijon cohort, H. pylori seropositivity was found to be associated with a decreased NSCLC patient survival on anti-PD-1 therapy. The survival median for H. pylori seropositive patients was 6.7 months compared with 15.4 months for seronegative patients (p=0.001). Additionally, in the Montreal cohort, H. pylori seropositivity was found to be associated with an apparent decrease of NSCLC patient progression-free survival on anti-PD-1 therapy. CONCLUSION Our study unveils for the first time that the stomach microbiota affects the response to cancer immunotherapies and that H. pylori serology would be a powerful tool to personalize cancer immunotherapy treatment.
Collapse
Affiliation(s)
- Paul Oster
- Service of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Laurie Vaillant
- Service of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Erika Riva
- Service of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Brynn McMillan
- Service of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Christina Begka
- Service of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Caroline Truntzer
- Department of Medical Oncology, Centre Georges François Leclerc, Dijon, France
| | - Corentin Richard
- Research Centre for the University of Montréal (CRCHUM), Hematology-Oncology Division, Department of Medicine, University of Montreal Healthcare Centre (CHUM), Montreal, Quebec, Canada
| | - Marine M Leblond
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Meriem Messaoudene
- Research Centre for the University of Montréal (CRCHUM), Hematology-Oncology Division, Department of Medicine, University of Montreal Healthcare Centre (CHUM), Montreal, Quebec, Canada
| | - Elisavet Machremi
- Service of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Emeric Limagne
- Department of Medical Oncology, Centre Georges François Leclerc, Dijon, France
| | | | - Bertrand Routy
- Research Centre for the University of Montréal (CRCHUM), Hematology-Oncology Division, Department of Medicine, University of Montreal Healthcare Centre (CHUM), Montreal, Quebec, Canada
| | - Gregory Verdeil
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Dominique Velin
- Service of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
18
|
Artola-Borán M, Fallegger A, Priola M, Jeske R, Waterboer T, Dohlman AB, Shen X, Wild S, He J, Levesque MP, Yousefi S, Simon HU, Cheng PF, Müller A. Mycobacterial infection aggravates Helicobacter pylori-induced gastric preneoplastic pathology by redirection of de novo induced Treg cells. Cell Rep 2022; 38:110359. [PMID: 35139377 DOI: 10.1016/j.celrep.2022.110359] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/12/2021] [Accepted: 01/19/2022] [Indexed: 12/12/2022] Open
Abstract
The two human pathogens Helicobacter pylori and Mycobacterium tuberculosis (Mtb) co-exist in many geographical areas of the world. Here, using a co-infection model of H. pylori and the Mtb relative M. bovis bacillus Calmette-Guérin (BCG), we show that both bacteria affect the colonization and immune control of the respective other pathogen. Co-occurring M. bovis boosts gastric Th1 responses and H. pylori control and aggravates gastric immunopathology. H. pylori in the stomach compromises immune control of M. bovis in the liver and spleen. Prior antibiotic H. pylori eradication or M. bovis-specific immunization reverses the effects of H. pylori. Mechanistically, the mutual effects can be attributed to the redirection of regulatory T cells (Treg cells) to sites of M. bovis infection. Reversal of Treg cell redirection by CXCR3 blockade restores M. bovis control. In conclusion, the simultaneous presence of both pathogens exacerbates the problems associated with each individual infection alone and should possibly be factored into treatment decisions.
Collapse
Affiliation(s)
- Mariela Artola-Borán
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Angela Fallegger
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Martina Priola
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Rima Jeske
- Infection and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Tim Waterboer
- Infection and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Anders B Dohlman
- Department of Biomedical Engineering, Center for Genomics and Computational Biology, Duke Microbiome Center, Duke University, Durham, NC, USA
| | - Xiling Shen
- Department of Biomedical Engineering, Center for Genomics and Computational Biology, Duke Microbiome Center, Duke University, Durham, NC, USA
| | - Sebastian Wild
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Jiazhuo He
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | | | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland; Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia; Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia; Institute of Biochemistry, Medical School Brandenburg, Neuruppin, Germany
| | - Phil F Cheng
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
19
|
Conventional type 1 dendritic cells protect against age-related adipose tissue dysfunction and obesity. Cell Mol Immunol 2022; 19:260-275. [PMID: 34983945 PMCID: PMC8803960 DOI: 10.1038/s41423-021-00812-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 02/08/2023] Open
Abstract
Conventional dendritic cells (cDCs) scan and integrate environmental cues in almost every tissue, including exogenous metabolic signals. While cDCs are critical in maintaining immune balance, their role in preserving energy homeostasis is unclear. Here, we showed that Batf3-deficient mice lacking conventional type 1 DCs (cDC1s) had increased body weight and adiposity during aging. This led to impaired energy expenditure and glucose tolerance, insulin resistance, dyslipidemia, and liver steatosis. cDC1 deficiency caused adipose tissue inflammation that was preceded by a paucity of NK1.1+ invariant NKT (iNKT) cells. Accordingly, among antigen-presenting cells, cDC1s exhibited notable induction of IFN-γ production by iNKT cells, which plays a metabolically protective role in lean adipose tissue. Flt3L treatment, which expands the dendritic cell (DC) compartment, mitigated diet-induced obesity and hyperlipidemia in a Batf3-dependent manner. This effect was partially mediated by NK1.1+ cells. These results reveal a new critical role for the cDC1-iNKT cell axis in the regulation of adipose tissue homeostasis.
Collapse
|
20
|
TGF-β production by eosinophils drives the expansion of peripherally induced neuropilin - RORγt + regulatory T-cells during bacterial and allergen challenge. Mucosal Immunol 2022; 15:504-514. [PMID: 35169233 PMCID: PMC9038533 DOI: 10.1038/s41385-022-00484-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 01/04/2022] [Accepted: 01/13/2022] [Indexed: 02/04/2023]
|
21
|
Carneiro MB, Peters NC. The Paradox of a Phagosomal Lifestyle: How Innate Host Cell- Leishmania amazonensis Interactions Lead to a Progressive Chronic Disease. Front Immunol 2021; 12:728848. [PMID: 34557194 PMCID: PMC8452962 DOI: 10.3389/fimmu.2021.728848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Intracellular phagosomal pathogens represent a formidable challenge for innate immune cells, as, paradoxically, these phagocytic cells can act as both host cells that support pathogen replication and, when properly activated, are the critical cells that mediate pathogen elimination. Infection by parasites of the Leishmania genus provides an excellent model organism to investigate this complex host-pathogen interaction. In this review we focus on the dynamics of Leishmania amazonensis infection and the host innate immune response, including the impact of the adaptive immune response on phagocytic host cell recruitment and activation. L. amazonensis infection represents an important public health problem in South America where, distinct from other Leishmania parasites, it has been associated with all three clinical forms of leishmaniasis in humans: cutaneous, muco-cutaneous and visceral. Experimental observations demonstrate that most experimental mouse strains are susceptible to L. amazonensis infection, including the C57BL/6 mouse, which is resistant to other species such as Leishmania major, Leishmania braziliensis and Leishmania infantum. In general, the CD4+ T helper (Th)1/Th2 paradigm does not sufficiently explain the progressive chronic disease established by L. amazonensis, as strong cell-mediated Th1 immunity, or a lack of Th2 immunity, does not provide protection as would be predicted. Recent findings in which the balance between Th1/Th2 immunity was found to influence permissive host cell availability via recruitment of inflammatory monocytes has also added to the complexity of the Th1/Th2 paradigm. In this review we discuss the roles played by innate cells starting from parasite recognition through to priming of the adaptive immune response. We highlight the relative importance of neutrophils, monocytes, dendritic cells and resident macrophages for the establishment and progressive nature of disease following L. amazonensis infection.
Collapse
Affiliation(s)
- Matheus B Carneiro
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine and Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Nathan C Peters
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine and Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
22
|
Hong J, Gao R, Yang Y. CrepHAN: Cross-species prediction of enhancers by using hierarchical attention networks. Bioinformatics 2021; 37:3436-3443. [PMID: 33978703 DOI: 10.1093/bioinformatics/btab349] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/21/2021] [Accepted: 05/06/2021] [Indexed: 01/17/2023] Open
Abstract
MOTIVATION Enhancers are important functional elements in genome sequences. The identification of enhancers is a very challenging task due to the great diversity of enhancer sequences and the flexible localization on genomes. Till now, the interactions between enhancers and genes have not been fully understood yet. To speed up the studies of the regulatory roles of enhancers, computational tools for the prediction of enhancers have emerged in recent years. Especially, thanks to the ENCODE project and the advances of high-throughput experimental techniques, a large amount of experimentally verified enhancers have been annotated on the human genome, which allows large-scale predictions of unknown enhancers using data-driven methods. However, except for human and some model organisms, the validated enhancer annotations are scarce for most species, leading to more difficulties in the computational identification of enhancers for their genomes. RESULTS In this study, we propose a deep learning-based predictor for enhancers, named CrepHAN, which is featured by a hierarchical attention neural network and word embedding-based representations for DNA sequences. We use the experimentally-supported data of the human genome to train the model, and perform experiments on human and other mammals, including mouse, cow, and dog. The experimental results show that CrepHAN has more advantages on cross-species predictions, and outperforms the existing models by a large margin. Especially, for human-mouse cross-predictions, the AUC score of ROC curve is increased by 0.033∼0.145 on the combined tissue dataset and 0.032∼0.109 on tissue-specific datasets. AVAILABILITY bcmi.sjtu.edu.cn/~yangyang/CrepHAN.html. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Jianwei Hong
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, 800 Dong Chuan Rd., Shanghai 200240, China.,School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ruitian Gao
- Department of Bioinformatics and Biostatistics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yang Yang
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, 800 Dong Chuan Rd., Shanghai 200240, China.,Key Laboratory of Shanghai Education Commission for Intelligent Interaction and Cognitive Engineering, Shanghai, 200240, China
| |
Collapse
|
23
|
Ahmed A, Rakshit S, Adiga V, Dias M, Dwarkanath P, D'Souza G, Vyakarnam A. A century of BCG: Impact on tuberculosis control and beyond. Immunol Rev 2021; 301:98-121. [PMID: 33955564 DOI: 10.1111/imr.12968] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/13/2021] [Accepted: 03/13/2021] [Indexed: 12/14/2022]
Abstract
BCG turns 100 this year and while it might not be the perfect vaccine, it has certainly contributed significantly towards eradication and prevention of spread of tuberculosis (TB). The search for newer and better vaccines for TB is an ongoing endeavor and latest results from trials of candidate TB vaccines such as M72AS01 look promising. However, recent encouraging data from BCG revaccination trials in adults combined with studies on mucosal and intravenous routes of BCG vaccination in non-human primate models have renewed interest in BCG for TB prevention. In addition, several well-demonstrated non-specific effects of BCG, for example, prevention of viral and respiratory infections, give BCG an added advantage. Also, BCG vaccination is currently being widely tested in human clinical trials to determine whether it protects against SARS-CoV-2 infection and/or death with detailed analyses and outcomes from several ongoing trials across the world awaited. Through this review, we attempt to bring together information on various aspects of the BCG-induced immune response, its efficacy in TB control, comparison with other candidate TB vaccines and strategies to improve its efficiency including revaccination and alternate routes of administration. Finally, we discuss the future relevance of BCG use especially in light of its several heterologous benefits.
Collapse
Affiliation(s)
- Asma Ahmed
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Srabanti Rakshit
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Vasista Adiga
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Mary Dias
- Division of Infectious Diseases, St John's Research Institute, Bangalore, India
| | | | - George D'Souza
- Division of Infectious Diseases, St John's Research Institute, Bangalore, India.,Department of Pulmonary Medicine, St John's Medical College, Bangalore, India
| | - Annapurna Vyakarnam
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India.,Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, Guy's Hospital, King's College London, London, UK
| |
Collapse
|
24
|
Abstract
For over 35 years since Mosmann and Coffman proposed the seminal “type 1 T helper (Th1)/type 2 T helper (Th2)” hypothesis in 1986, the immunological community has appreciated that naïve CD4 T cells need to make important decisions upon their activation, namely to differentiate towards a Th1, Th2, Th17 (interleukin-17-producing T helper), follicular T helper (Tfh), or regulatory T cell (Treg) fate to orchestrate a variety of adaptive immune responses. The major molecular underpinnings of the Th1/Th2 effector fate choice had been initially characterized using excellent reductionist in vitro culture systems, through which the transcription factors T-bet and GATA3 were identified as the master regulators for the differentiation of Th1 and Th2 cells, respectively. However, Th1/Th2 cell differentiation and their cellular heterogeneity are usually determined by a combinatorial expression of multiple transcription factors, particularly in vivo, where dendritic cell (DC) and innate lymphoid cell (ILC) subsets can also influence T helper lineage choices. In addition, inflammatory cytokines that are capable of inducing Th17 cell differentiation are also found to be induced during typical Th1- or Th2-related immune responses, resulting in an alternative differentiation pathway, transiting from a Th17 cell phenotype towards Th1 or Th2 cells. In this review, we will discuss the recent advances in the field, focusing on some new players in the transcriptional network, contributions of DCs and ILCs, and alternative differentiation pathways towards understanding the Th1/Th2 effector choice in vivo.
Collapse
Affiliation(s)
- Matthew J Butcher
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
25
|
Abstract
As the professional antigen-presenting cells of the immune system, dendritic cells (DCs) sense the microenvironment and shape the ensuing adaptive immune response. DCs can induce both immune activation and immune tolerance according to the peripheral cues. Recent work has established that DCs comprise several phenotypically and functionally heterogeneous subsets that differentially regulate T lymphocyte differentiation. This review summarizes both mouse and human DC subset phenotypes, development, diversification, and function. We focus on advances in our understanding of how different DC subsets regulate distinct CD4+ T helper (Th) cell differentiation outcomes, including Th1, Th2, Th17, T follicular helper, and T regulatory cells. We review DC subset intrinsic properties, local tissue microenvironments, and other immune cells that together determine Th cell differentiation during homeostasis and inflammation.
Collapse
Affiliation(s)
- Xiangyun Yin
- Department of Laboratory Medicine and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA;
| | - Shuting Chen
- Department of Laboratory Medicine and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA;
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA;
| |
Collapse
|
26
|
An Antibiotic-Impacted Microbiota Compromises the Development of Colonic Regulatory T Cells and Predisposes to Dysregulated Immune Responses. mBio 2021; 12:mBio.03335-20. [PMID: 33531385 PMCID: PMC7858066 DOI: 10.1128/mbio.03335-20] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The assembly of microbial communities that populate all mucosal surfaces of the human body begins right after birth. This process is prone to disruption as newborns and young infants are increasingly exposed to antibiotics, both deliberately for therapeutic purposes, and as a consequence of transmaternal exposure. Antibiotic exposure early in life and other practices impacting the vertical transmission and ordered assembly of a diverse and balanced gut microbiota are associated with a higher risk of immunological and metabolic disorders such as asthma and allergy, autoimmunity, obesity, and susceptibility to opportunistic infections. In this study, we used a model of perinatal exposure to the broad-spectrum antibiotic ampicillin to examine how the acquisition of a dysbiotic microbiota affects neonatal immune system development. We found that the resultant dysbiosis imprints in a manner that is irreversible after weaning, leading to specific and selective alteration of the colonic CD4+ T-cell compartment. In contrast, colonic granulocyte and myeloid lineages and other mucosal T-cell compartments are unaffected. Among colonic CD4+ T cells, we observed the most pronounced effects on neuropilin-negative, RORγt- and Foxp3-positive regulatory T cells, which are largely absent in antibiotic-exposed mice even as they reach adulthood. Immunomagnetically isolated dendritic cells from antibiotic-exposed mice fail to support the generation of Foxp3+ regulatory T cells (Tregs) from naive T cells ex vivo. The perinatally acquired dysbiotic microbiota predisposes to dysregulated effector T-cell responses to Citrobacter rodentium or ovalbumin challenge. The transfer of the antibiotic-impacted, but not healthy, fecal microbiota into germfree recipients recapitulates the selective loss of colonic neuropilin-negative, RORγt- and Foxp3-positive Tregs. The combined data indicate that the early-life acquisition of a dysbiotic microbiota has detrimental effects on the diversity and microbial community composition of offspring that persist into adulthood and predisposes to inappropriate T-cell responses that are linked to compromised immune tolerance.
Collapse
|
27
|
Lung CD103 + Dendritic cells of mice infected with Paracoccidioides brasiliensis contribute to Treg differentiation. Microb Pathog 2020; 150:104696. [PMID: 33359357 DOI: 10.1016/j.micpath.2020.104696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 12/05/2020] [Accepted: 12/09/2020] [Indexed: 01/17/2023]
Abstract
The DC subsets that express αE integrin (CD103) have been described to exert antagonistic functions, driving T cells towards either an inflammatory (Th1/Th17) or immunosuppressive phenotype (regulatory T cells - Treg). These functions depend on the tissue they reside and microenvironment factors or stimuli that this Antigen-presenting cell (APC) subpopulation receive. In this regard, immunoregulatory phenotype has been described in small subsets of CD103+ DCs from lung and intestinal mucosa. The function of this APC subpopulation in pulmonary Paracoccidioides brasiliensis infection is poorly described. Here, we showed that lung CD103+ DCs contribute to Treg differentiation in a pulmonary P. brasiliensis infection model, which was attributed to downregulation of costimulatory molecules analyzed in these APC subtypes 21 days post-infection. Overall, this data suggests that P. brasiliensis infection caused an immunosuppression that has also been observed in patients with the most severe form of Paracoccidioidomycosis (PCM) - a sickness caused by this fungus genus. Furthermore, these results open new perspectives for knowledge of the mechanisms that underlie the higher percentage of Treg cells found in peripheral blood of PCM patients.
Collapse
|
28
|
Soto M, Ramírez L, Solana JC, Cook ECL, Hernández-García E, Charro-Zanca S, Redondo-Urzainqui A, Reguera RM, Balaña-Fouce R, Iborra S. Resistance to Experimental Visceral Leishmaniasis in Mice Infected With Leishmania infantum Requires Batf3. Front Immunol 2020; 11:590934. [PMID: 33362772 PMCID: PMC7758202 DOI: 10.3389/fimmu.2020.590934] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/09/2020] [Indexed: 12/20/2022] Open
Abstract
Unveiling the protective immune response to visceral leishmaniasis is critical for a rational design of vaccines aimed at reducing the impact caused by this fatal, if left untreated, vector-borne disease. In this study we sought to determine the role of the basic leucine zipper transcription factor ATF-like 3 (Batf3) in the evolution of infection with Leishmania infantum, the causative agent of human visceral leishmaniasis in the Mediterranean Basin and Latin America. For that, Batf3-deficient mice in C57BL/6 background were infected with an L. infantum strain expressing the luciferase gene. Bioluminescent imaging, as well as in vitro parasite titration, demonstrated that Batf3-deficient mice were unable to control hepatic parasitosis as opposed to wild-type C57BL/6 mice. The impaired microbicide capacities of L. infantum-infected macrophages from Batf3-deficient mice mainly correlated with a reduction of parasite-specific IFN-γ production. Our results reinforce the implication of Batf3 in the generation of type 1 immunity against infectious diseases.
Collapse
Affiliation(s)
- Manuel Soto
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular, Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Ramírez
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular, Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - José Carlos Solana
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular, Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Emma C L Cook
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Elena Hernández-García
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Sara Charro-Zanca
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Ana Redondo-Urzainqui
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Rosa M Reguera
- Departamento de Ciencias Biomédicas, Universidad de León, León, Spain
| | | | - Salvador Iborra
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
29
|
Gallizioli M, Miró-Mur F, Otxoa-de-Amezaga A, Cugota R, Salas-Perdomo A, Justicia C, Brait VH, Ruiz-Jaén F, Arbaizar-Rovirosa M, Pedragosa J, Bonfill-Teixidor E, Gelderblom M, Magnus T, Cano E, Del Fresno C, Sancho D, Planas AM. Dendritic Cells and Microglia Have Non-redundant Functions in the Inflamed Brain with Protective Effects of Type 1 cDCs. Cell Rep 2020; 33:108291. [PMID: 33086061 PMCID: PMC7578563 DOI: 10.1016/j.celrep.2020.108291] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/02/2020] [Accepted: 09/29/2020] [Indexed: 01/14/2023] Open
Abstract
Brain CD11c+ cells share features with microglia and dendritic cells (DCs). Sterile inflammation increases brain CD11c+ cells, but their phenotype, origin, and functions remain largely unknown. We report that, after cerebral ischemia, microglia attract DCs to the inflamed brain, and astroglia produce Flt3 ligand, supporting development and expansion of CD11c+ cells. CD11c+ cells in the inflamed brain are a complex population derived from proliferating microglia and infiltrating DCs, including a major subset of OX40L+ conventional cDC2, and also cDC1, plasmacytoid, and monocyte-derived DCs. Despite sharing certain morphological features and markers, CD11c+ microglia and DCs display differential expression of pattern recognition receptors and chemokine receptors. DCs excel CD11c- and CD11c+ microglia in the capacity to present antigen through MHCI and MHCII. Of note, cDC1s protect from brain injury after ischemia. We thus reveal aspects of the dynamics and functions of brain DCs in the regulation of inflammation and immunity.
Collapse
Affiliation(s)
- Mattia Gallizioli
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Francesc Miró-Mur
- Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain; Fundació Clínic, Barcelona 08036, Spain
| | - Amaia Otxoa-de-Amezaga
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Roger Cugota
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain
| | - Angélica Salas-Perdomo
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Fundació Clínic, Barcelona 08036, Spain
| | - Carles Justicia
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Vanessa H Brait
- Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Francisca Ruiz-Jaén
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Maria Arbaizar-Rovirosa
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Jordi Pedragosa
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Ester Bonfill-Teixidor
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Eva Cano
- Neuroinflammation Unit, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III, Majadahonda, Madrid 28222, Spain
| | - Carlos Del Fresno
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - David Sancho
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Anna M Planas
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain.
| |
Collapse
|
30
|
Abstract
Understanding the mechanisms involved in induction and regulation of the immune and inflammatory response to Helicobacter pylori is extremely important in determining disease outcomes. H pylori expresses a plethora of factors that influence the host response. Vaccines against H pylori are desperately needed for the prevention of gastric carcinogenesis, especially with the increasing trends in antimicrobial resistance. This review summarizes some important findings, published between 1 April 2019 and 31 March 2020, in the areas of H pylori-mediated inflammation, immunity and vaccines.
Collapse
Affiliation(s)
- Karen Robinson
- School of Medicine, Nottingham Digestive Diseases Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Philippe Lehours
- UMR1053 Bordeaux Research In Translational Oncology, INSERM, Univ. Bordeaux, BaRITOn, Bordeaux, France.,French National Reference Centre for Campylobacters & Helicobacters, Hôpital Pellegrin, Bordeaux, France
| |
Collapse
|
31
|
Zhang X, Artola-Boran M, Fallegger A, Arnold IC, Weber A, Reuter S, Taube C, Müller A. IRF4 Expression Is Required for the Immunoregulatory Activity of Conventional Type 2 Dendritic Cells in Settings of Chronic Bacterial Infection and Cancer. THE JOURNAL OF IMMUNOLOGY 2020; 205:1933-1943. [PMID: 32848032 DOI: 10.4049/jimmunol.2000405] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/24/2020] [Indexed: 01/22/2023]
Abstract
The lamina propria of the gastrointestinal tract and other mucosal surfaces of humans and mice host a network of mononuclear phagocytes that differ in their ontogeny, surface marker and transcription factor expression, and functional specialization. Conventional dendritic cells (DCs) in particular exist as two major subpopulations in both lymphoid and nonlymphoid organs that can be distinguished based on their surface marker and transcription factor expression. In this study, we show in various Th1- and/or Th17-polarized settings of acute and chronic bacterial infection and of tumor growth that the conditional ablation of Irf4 in CD11c+ DCs results in more efficient immune control of Helicobacter pylori, Mycobacterium bovis bacillus Calmette-Guérin, and Citrobacter rodentium and of tumor growth in a syngeneic tumor model. We attribute the phenotype of IRF4ΔDC mice to unrestricted Th1 responses and in particular to IFN-γ- and TNF-α-expressing CD4+ T cells. This activity of IRF4-expressing DCs is linked to a DC-specific immunoregulatory transcriptional program. In contrast, in Th2-polarized settings such as house dust mite-induced allergic airway inflammation, the lack of IRF4 expression in the DC compartment alleviates inflammation and goblet cell metaplasia. The combined data provide evidence for immunoregulatory properties of this versatile DC population in Th1-polarized infection settings.
Collapse
Affiliation(s)
- Xiaozhou Zhang
- Institute of Molecular Cancer Research, University of Zurich, 8057 Zurich, Switzerland
| | - Mariela Artola-Boran
- Institute of Molecular Cancer Research, University of Zurich, 8057 Zurich, Switzerland
| | - Angela Fallegger
- Institute of Molecular Cancer Research, University of Zurich, 8057 Zurich, Switzerland
| | - Isabelle C Arnold
- Institute of Molecular Cancer Research, University of Zurich, 8057 Zurich, Switzerland
| | - Achim Weber
- Institute of Molecular Cancer Research, University of Zurich, 8057 Zurich, Switzerland.,Institute of Pathology and Molecular Pathology, University of Zurich, 8091 Zurich, Switzerland; and
| | - Sebastian Reuter
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, 45239 Essen, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, 45239 Essen, Germany
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zurich, 8057 Zurich, Switzerland;
| |
Collapse
|
32
|
The role of the changing human microbiome in the asthma pandemic. J Allergy Clin Immunol 2020; 144:1457-1466. [PMID: 31812180 DOI: 10.1016/j.jaci.2019.10.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 02/06/2023]
Abstract
Asthma and allergy incidence continue to increase globally. We have made significant strides in treating disease, but it is becoming more apparent that we need to advance our knowledge into the origins of asthmatic disease. Much recent work has indicated that microbiome composition influences immune regulation and that multiple health care factors have driven a loss in microbiome diversity in modern human populations. Evidence is growing of microbiota-driven influences on immune development, asthma susceptibility, and asthma pathogenesis. The focus of this review is to highlight the strides the field has made in characterizing the constituents of the human gastrointestinal microbiota, such as Helicobacter pylori, other members of the neonatal intestinal microbiota, and microbial peptides and metabolites that influence host immunity and immune response to allergens. As we delve further into this field of research, the goal will be to find actionable and clinical interventions to identify at-risk populations earlier to prevent disease onset. Manipulation of the host microbial community during infancy might be an especially promising approach.
Collapse
|
33
|
Neuper T, Frauenlob T, Sarajlic M, Posselt G, Wessler S, Horejs-Hoeck J. TLR2, TLR4 and TLR10 Shape the Cytokine and Chemokine Release of H. pylori-Infected Human DCs. Int J Mol Sci 2020; 21:ijms21113897. [PMID: 32486097 PMCID: PMC7311968 DOI: 10.3390/ijms21113897] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/22/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022] Open
Abstract
Helicobacter pylori (H. pylori) is a stomach pathogen that persistently colonizes the gastric mucosa, often leading to chronic inflammation and gastric pathologies. Although infection with H. pylori is the primary risk factor for gastric cancer, the underlying mechanisms of pathogen persistence and consequential chronic inflammation are still not well understood. Conventional dendritic cells (cDCs), which are among the first immune cells to encounter H. pylori in the gastric lining, and the cytokines and chemokines they secrete, contribute to both acute and chronic inflammation. Therefore, this study aimed to unravel the contributions of specific signaling pathways within human CD1c+ cDCs (cDC2s) to the composition of secreted cytokines and chemokines in H. pylori infection. Here, we show that the type IV secretion system (T4SS) plays only a minor role in H. pylori-induced activation of cDC2s. In contrast, Toll-like receptor 4 (TLR4) signaling drives the secretion of inflammatory mediators, including IL-12 and IL-18, while signaling via TLR10 attenuates the release of IL-1β and other inflammatory cytokines upon H. pylori infection. The TLR2 pathway significantly blocks the release of CXCL1 and CXCL8, while it promotes the secretion of TNFα and GM-CSF. Taken together, these results highlight how specific TLR-signaling pathways in human cDC2s shape the H. pylori-induced cytokine and chemokine milieu, which plays a pivotal role in the onset of an effective immune response.
Collapse
Affiliation(s)
- Theresa Neuper
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria; (T.N.); (T.F.); (M.S.); (G.P.); (S.W.)
| | - Tobias Frauenlob
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria; (T.N.); (T.F.); (M.S.); (G.P.); (S.W.)
| | - Muamera Sarajlic
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria; (T.N.); (T.F.); (M.S.); (G.P.); (S.W.)
| | - Gernot Posselt
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria; (T.N.); (T.F.); (M.S.); (G.P.); (S.W.)
| | - Silja Wessler
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria; (T.N.); (T.F.); (M.S.); (G.P.); (S.W.)
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria; (T.N.); (T.F.); (M.S.); (G.P.); (S.W.)
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
- Correspondence:
| |
Collapse
|
34
|
Hilligan KL, Ronchese F. Antigen presentation by dendritic cells and their instruction of CD4+ T helper cell responses. Cell Mol Immunol 2020; 17:587-599. [PMID: 32433540 DOI: 10.1038/s41423-020-0465-0] [Citation(s) in RCA: 200] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/10/2020] [Indexed: 12/20/2022] Open
Abstract
Dendritic cells are powerful antigen-presenting cells that are essential for the priming of T cell responses. In addition to providing T-cell-receptor ligands and co-stimulatory molecules for naive T cell activation and expansion, dendritic cells are thought to also provide signals for the differentiation of CD4+ T cells into effector T cell populations. The mechanisms by which dendritic cells are able to adapt and respond to the great variety of infectious stimuli they are confronted with, and prime an appropriate CD4+ T cell response, are only partly understood. It is known that in the steady-state dendritic cells are highly heterogenous both in phenotype and transcriptional profile, and that this variability is dependent on developmental lineage, maturation stage, and the tissue environment in which dendritic cells are located. Exposure to infectious agents interfaces with this pre-existing heterogeneity by providing ligands for pattern-recognition and toll-like receptors that are variably expressed on different dendritic cell subsets, and elicit production of cytokines and chemokines to support innate cell activation and drive T cell differentiation. Here we review current information on dendritic cell biology, their heterogeneity, and the properties of different dendritic cell subsets. We then consider the signals required for the development of different types of Th immune responses, and the cellular and molecular evidence implicating different subsets of dendritic cells in providing such signals. We outline how dendritic cell subsets tailor their response according to the infectious agent, and how such transcriptional plasticity enables them to drive different types of immune responses.
Collapse
Affiliation(s)
- Kerry L Hilligan
- Malaghan Institute of Medical Research, Wellington, 6012, New Zealand.,Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Franca Ronchese
- Malaghan Institute of Medical Research, Wellington, 6012, New Zealand.
| |
Collapse
|
35
|
Kim HC, Oh DS, Park JH, Kim HJ, Seo YB, Yoo HJ, Jang HS, Shin J, Kim CW, Kwon MS, Jin HT, Lee SK, Oh JE, Lee HK. Multivalent DNA vaccine protects against genital herpes by T-cell immune induction in vaginal mucosa. Antiviral Res 2020; 177:104755. [PMID: 32112797 DOI: 10.1016/j.antiviral.2020.104755] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 02/21/2020] [Accepted: 02/24/2020] [Indexed: 12/11/2022]
Abstract
Genital herpes is one of the most common sexually transmitted infections (STIs), and it is mainly caused by the neurotropic herpes simplex virus (HSV-2). Not only does this infection cause ulcers, but HSV-2 can also stay in a latent state in the nervous system of the host throughout their lifespan. As a result, many people do not know that they harbor this infection. Moreover, HSV-2 serves as a major risk factor for human immunodeficiency virus (HIV) infection and can be transmitted to the fetus. Despite the high risk of infection and adverse effects, attempts at development of an effective vaccine for HSV-2 have not yet been successful. In this study, we developed a DNA vaccine for HSV-2 (SL-V20). This multivalent DNA vaccine effectively reduced the pathological symptoms of infection and induced efficient elimination of the virus in a mouse model. Intramuscular injection of SL-V20 led to induction of an HSV-2-specific T-cell response in the vagina, the major infection site, and in draining lymph organs. Dendritic cells (DCs), especially basic leucine zipper ATF-like transcription factor 3 (Baft3)+ DCs and partially interferon regulatory factor 4 (Irf4)+ DCs, were involved in this T-cell-mediated protective response, while B cells were dispensable for these prophylactic effects. This study demonstrates that SL-V20 offers a novel and effective vaccine against vaginal HSV-2 infection and may be applicable to patients, pending validation in clinical studies.
Collapse
Affiliation(s)
- Hyeon Cheol Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Dong Sun Oh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jang Hyun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hyun-Jin Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Yong Bok Seo
- SL-VAXiGEN Inc., 700 Daewangpanyo-Ro, Bundang-Gu, Seongnam, Gyeonggi, 13488, Republic of Korea
| | - Hye Jee Yoo
- Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon, 34141, Republic of Korea
| | - Hye Seon Jang
- Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon, 34141, Republic of Korea
| | - Jua Shin
- SL-VAXiGEN Inc., 700 Daewangpanyo-Ro, Bundang-Gu, Seongnam, Gyeonggi, 13488, Republic of Korea
| | - Chae Won Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Myeong Seung Kwon
- Department of Obstetrics and Gynecology, College of Medicine, Myunggok Medical Research Center, Konyang University, Daejeon, Republic of Korea
| | - Hyun-Tak Jin
- ProGen Co., Ltd, 1201, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Sung Ki Lee
- Department of Obstetrics and Gynecology, College of Medicine, Myunggok Medical Research Center, Konyang University, Daejeon, Republic of Korea
| | - Ji Eun Oh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea; Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon, 34141, Republic of Korea; KAIST Institute for Health Science and Technology, KAIST, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
36
|
Zhang X, Arnold IC, Müller A. Mechanisms of persistence, innate immune activation and immunomodulation by the gastric pathogen Helicobacter pylori. Curr Opin Microbiol 2020; 54:1-10. [PMID: 32007716 DOI: 10.1016/j.mib.2020.01.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/03/2020] [Indexed: 12/20/2022]
Abstract
The gastric bacterium Helicobacter pylori efficiently evades innate immune detection and persistently colonizes its human host. Understanding the genetic determinants that H. pylori uses to establish and maintain persistence, along with their cellular targets, is key to our understanding of the pathogenesis of this extraordinarily successful bacterial colonizer of the human stomach. This review highlights recent advances in elucidating innate immune recognition of H. pylori, its interactions with myeloid cells and the consequences that this very local infection has for immune responses at extragastric sites in models of allergy, autoimmunity and parasitic infection. The human-specific, gram-negative gastric colonizer and carcinogen H. pylori represents the prototype of a persistent bacterial pathogen. It is transmitted during early childhood, typically from mother to infant, and is believed to persist in its human host from the cradle to the grave. The tremendous success of H. pylori in infecting and colonizing half of the world's population, and in continuously accompanying humans since they migrated out of Africa over 60000 years ago, can largely be attributed to its ability to manipulate the host immune system to its own advantage, and to thereby ensure its own persistence and chronicity. In his final years as an active PI, Stanley Falkow increasingly recognized the need to understand bacterial persistence strategies as a prerequisite of understanding the pathogenesis of chronic bacterial infections, and, inspired in large part by Denise Monack's work on Salmonella persistence, many of our discussions at the time revolved around this topic. Multiple labs have since made important contributions to our understanding of innate immune detection of H. pylori, the types and polarization of adaptive immune responses that ensue, the ability of H. pylori to skew such immune responses to its advantage, and its ability to manipulate the host immune system with far-reaching, even systemic consequences. This review attempts to cover some of these topics, with a particular focus on the most recent contributions by researchers in the field.
Collapse
Affiliation(s)
- Xiaozhou Zhang
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Isabelle C Arnold
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland.
| |
Collapse
|