1
|
Wizenty J, Sigal M. Helicobacter pylori, microbiota and gastric cancer - principles of microorganism-driven carcinogenesis. Nat Rev Gastroenterol Hepatol 2025; 22:296-313. [PMID: 40011753 DOI: 10.1038/s41575-025-01042-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2025] [Indexed: 02/28/2025]
Abstract
The demonstration that Helicobacter pylori is a pathogenic bacterium with marked carcinogenic potential has paved the way for new preventive approaches for gastric cancer. Although decades of research have uncovered complex interactions of H. pylori with epithelial cells, current insights have refined our view on H. pylori-associated carcinogenesis. Specifically, the cell-type-specific effects on gastric stem and progenitor cells deep in gastric glands provide a new view on the ability of the bacteria to colonize long-term, manipulate host responses and promote gastric pathology. Furthermore, new, large-scale epidemiological data have shed light on factors that determine why only a subset of carriers progress to gastric cancer. Currently, technological advances have brought yet another revelation: H. pylori is far from the only microorganism able to colonize the stomach. Instead, the stomach is colonized by a diverse gastric microbiota, and there is emerging evidence for the occurrence and pathological effect of dysbiosis resulting from an aberrant interplay between H. pylori and the gastric mucosa. With the weight of this evidence mounting, here we consider how the lessons learned from H. pylori research inform and synergize with this emerging field to bring a more comprehensive understanding of the role of microbes in gastric carcinogenesis.
Collapse
Affiliation(s)
- Jonas Wizenty
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy and BIH Charité Clinician Scientist Program, Berlin, Germany
| | - Michael Sigal
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
| |
Collapse
|
2
|
Du O, Yan YL, Yang HY, Yang YX, Wu AG, Guo YK, Li K, Qiao G, Du JR, Long FY. ALPK1 signaling pathway activation by HMGB1 drives microglial pyroptosis and ferroptosis and brain injury after acute ischemic stroke. Int Immunopharmacol 2025; 149:114229. [PMID: 39933362 DOI: 10.1016/j.intimp.2025.114229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 01/26/2025] [Accepted: 02/02/2025] [Indexed: 02/13/2025]
Abstract
Pyroptosis and ferroptosis emerge as remarkable contributors to neuronal death and inflammation following ischemic stroke. High mobility group box 1 (HMGB1), a principal damage-associated molecular pattern (DAMP), is implicated in pyroptosis and ferroptosis post-stroke. Our previous research has demonstrated that alpha kinase 1 (ALPK1), a novel cytoplasmic pattern recognition receptor (PRR), plays an important role in mediating inflammatory damage following ischemic stroke. However, the interaction between ALPK1 and HMGB1, and their combined impact on pyroptosis and ferroptosis post-ischemic stroke remain unexplored, which is what this study aims to investigate. Initially, we observed that ALPK1 ablation attenuated ischemic brain injury of transient middle cerebral artery occlusion (tMCAO) mice. Moreover, recombinant HMGB1 (rHMGB1) stimulation induced the greatest upregulation of ALPK1 expression in microglia compared to astrocytes and neurons. Further investigation using co-immunofluorescence, co-immunoprecipitation, pull-down assay, and molecular docking revealed an interaction between HMGB1 and ALPK1. Additionally, the exacerbation of ischemic brain injury and the induction of microglial pyroptosis and ferroptosis by rHMGB1 treatment in tMCAO mice were significantly mitigated through ALPK1 deficiency by inhibiting the NLRP3/Caspase-1/GSDMD and JAK2/STAT3 signaling pathways. The inhibitory effects of ALPK1 deficiency on pyroptosis and ferroptosis induced by rHMGB1 in microglial cells were further substantiated. Finally, glycyrrhizic acid (GA), an inhibitor of HMGB1, exhibited significant neuroprotective effects in both tMCAO mice and BV2 cells subjected to oxygen-glucose deprivation/reperfusion (OGD/R) by downregulating ALPK1 expression and inhibiting microglial pyroptosis and ferroptosis. Collectively, these findings suggest that HMGB1 may interact with ALPK1 to drive microglial pyroptosis and ferroptosis via the activation of the ALPK1/NF-κB/NLRP3/GSDMD and JAK2/STAT3 signaling pathways, thereby exacerbating brain injury following acute ischemic stroke.
Collapse
Affiliation(s)
- Ou Du
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Ya-Ling Yan
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Han-Yinan Yang
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Yu-Xin Yang
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drug Ability Evaluation, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yin-Kun Guo
- Department of Radiology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kuan Li
- Department of Radiology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Gan Qiao
- Sichuan Key Medical Laboratory of New Drug Discovery and Drug Ability Evaluation, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jun-Rong Du
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
| | - Fang-Yi Long
- Laboratory Medicine Center, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, Sichuan, China.
| |
Collapse
|
3
|
Liu B, Bukhari I, Li F, Ren F, Xia X, Hu B, Liu H, Meyer TF, Marshall BJ, Tay A, Fu Y, Wu W, Tang Y, Mi Y, Zheng PY. Enhanced LRP8 expression induced by Helicobacter pylori drives gastric cancer progression by facilitating β-Catenin nuclear translocation. J Adv Res 2025; 69:299-312. [PMID: 38609049 PMCID: PMC11954824 DOI: 10.1016/j.jare.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024] Open
Abstract
INTRODUCTION Helicobacter pylori (H. pylori) infection has been associated with gastric carcinogenesis. However, the precise involvement of LRP8, the low-density lipoprotein receptor-related protein 8, in H. pylori pathogenesis and gastric cancer (GC) remains poorly understood. OBJECTIVES To investigate the potential role of LRP8 in H. pylori infection and gastric carcinogenesis. METHODS Three-dimensional human-derived gastric organoids (hGO) and gastric cancer organoids (hGCO) were synthesized from the tissues obtained from human donors. In this work, multi-omics combined with in vivo and in vitro studies were conducted to investigate the potential involvement of LRP8 in H. pylori-induced GC. RESULTS We found that H. pylori infection significantly upregulated the expression of LRP8 in human GC tissues, cells, organoids, and mouse gastric mucous. In particular, LRP8 exhibited a distinct enrichment in cancer stem cells (CSC). Functionally, silencing of LRP8 affected the formation and proliferation of tumor spheroids, while increased expression of LRP8 was associated with increased proliferation and stemness of GC cells and organoids. Mechanistically, LRP8 promotes the binding of E-cadherin to β-catenin, thereby promoting nuclear translocation and transcriptional activity of β-catenin. Furthermore, LRP8 interacts with the cytotoxin-associated gene A (CagA) to form the CagA/LRP8/β-catenin complex. This complex further amplifies H. pylori-induced β-catenin nuclear translocation, leading to increased transcription of inflammatory factors and CSC markers. Clinical analysis demonstrated that abnormal overexpression of LRP8 is correlated with a poor prognosis and resistance to 5-Fluorouracil in patients with GC. CONCLUSION Our findings provide valuable information on the molecular intricacies of H. pylori-induced gastric carcinogenesis, offering potential therapeutic targets and prognostic markers for GC.
Collapse
Affiliation(s)
- Bin Liu
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Ihtisham Bukhari
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Fazhan Li
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Feifei Ren
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xue Xia
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Baitong Hu
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Haipeng Liu
- Clinical and Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Thomas F Meyer
- Max Planck Institute for Infection Biology, Department of Molecular Biology, 10117 Berlin, Germany; Laboratory of Infection Oncology, Institute of Clinical Molecular Biology (IKMB), Christian-Albrechts University of Kiel, Kiel, Germany
| | - Barry J Marshall
- Helicobacter Pylori Research Laboratory, School of Biomedical Sciences, Marshall Centre for Infectious Disease Research and Training, University of Western Australia, Nedlands 6009, Australia
| | - Alfred Tay
- Helicobacter Pylori Research Laboratory, School of Biomedical Sciences, Marshall Centre for Infectious Disease Research and Training, University of Western Australia, Nedlands 6009, Australia
| | - Yuming Fu
- Gastrointestinal Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Wanqing Wu
- Gastrointestinal Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Youcai Tang
- Department of Pediatrics, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yang Mi
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Peng-Yuan Zheng
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| |
Collapse
|
4
|
Sibony-Benyamini H, Jbara R, Shubash Napso T, Abu-Rahmoun L, Vizenblit D, Easton-Mor M, Perez S, Brandis A, Leshem T, Peretz A, Maman Y. The landcape of Helicobacter pylori-mediated DNA breaks links bacterial genotoxicity to its oncogenic potential. Genome Med 2025; 17:14. [PMID: 39994739 PMCID: PMC11853333 DOI: 10.1186/s13073-025-01439-3] [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: 04/18/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) infection is a significant risk factor for gastric cancer (GC) development. A growing body of evidence suggests a causal link between infection with H. pylori and increased DNA breakage in the host cells. While several mechanisms have been proposed for this damage, their relative impact on the overall bacterial genotoxicity is unknown. Moreover, the link between the formation of DNA damage following infection and the emergence of cancerous structural variants (SV) in the genome of infected cells remained unexplored. METHODS We constructed a high-resolution map of genomic H. pylori-induced recurrent break sites using the END-seq method on AGS human gastric cells before and after infection. We next applied END-seq to cycling and arrested cells to identify the role of DNA replication on break formation. Recurrent H. pylori-mediated break sites were further characterized by analyzing published RNA-seq, DRIP-seq, and GRO-seq data at these sites. γH2AX staining and comet assay were used for DNA breakage quantification. Liquid chromatography-mass spectrometry (LC-MS) assay was used to quantify cellular concentrations of dNTPs. RESULTS Our data indicated that sites of recurrent H. pylori-mediated DNA breaks are ubiquitous across cell types, localized at replication-related fragile sites, and their breakage is dependent on replication. Consistent with that, we found that H. pylori inflicts nucleotide depletion, and that rescuing the cellular nucleotide pool largely reduced H. pylori-induced DNA breaks. Intriguingly, we found that this genotoxic mechanism operates independently of H. pylori cag pathogenicity island (CagPAI) that encodes for the bacterial type 4 secretion system (T4SS), and its virulence factor, CagA, which was previously implicated in increasing DNA damage by downregulating the DNA damage response. Finally, we show that sites of recurrent H. pylori-mediated breaks coincide with chromosomal deletions observed in patients with intestinal-type GC and that this link potentially elucidates the persistent transcriptional alterations observed in cancer driver genes. CONCLUSIONS Our findings indicate that dNTP depletion by H. pylori is a key component of its genotoxicity and suggest a link between H. pylori genotoxicity and its oncogenic potential.
Collapse
Affiliation(s)
| | - Rose Jbara
- Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Zefat, Israel
| | - Tania Shubash Napso
- Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Zefat, Israel
| | - Layan Abu-Rahmoun
- Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Zefat, Israel
| | - Daniel Vizenblit
- Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Zefat, Israel
- Baruch Padeh Medical Center, Poriya, Israel
| | - Michal Easton-Mor
- Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Zefat, Israel
| | - Shira Perez
- Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Zefat, Israel
| | | | | | - Avi Peretz
- Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Zefat, Israel
- Baruch Padeh Medical Center, Poriya, Israel
| | - Yaakov Maman
- Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Zefat, Israel.
| |
Collapse
|
5
|
Artola-Borán M, Kirsche L, Fallegger A, Leary P, Tanriover M, Goodwin T, Geiger G, Hapfelmeier S, Yousefi S, Simon HU, Arnold IC, Müller A. IgA facilitates the persistence of the mucosal pathogen Helicobacter pylori. Mucosal Immunol 2025; 18:232-247. [PMID: 39581230 DOI: 10.1016/j.mucimm.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/29/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024]
Abstract
IgA antibodies have an important role in clearing mucosal pathogens. In this study, we have examined the contribution of IgA to the immune control of the gastrointestinal bacterial pathogens Helicobacter pylori and Citrobacter rodentium. Both bacteria trigger a strong local IgA response that results in bacterial IgA coating in mice and in gastritis patients. Class switching to IgA depends on Peyer's patches, T-cells, eosinophils, and eosinophil-derived TGF-β in both models. In the case of H. pylori, IgA secretion and bacterial coating also depend on a functional bacterial type IV secretion system, which drives the generation of Th17 cells and the IL-17-dependent expression of the polymeric immunoglobulin receptor PIGR. IgA-/- mice are hypercolonized with C. rodentium in all examined tissues, suffer from more severe weight loss and develop more colitis. In contrast, H. pylori is controlled more efficiently in IgA-/- mice than their WT counterparts. The effects of IgA deficiency of the offspring can be compensated by maternal IgA delivered by WT foster mothers. We attribute the improved immune control observed in IgA-/- mice to IgA-mediated protection from complement killing, as H. pylori colonization is restored to wild type levels in a composite strain lacking both IgA and the central complement component C3. IgA antibodies can thus have protective or detrimental activities depending on the infectious agent.
Collapse
Affiliation(s)
- Mariela Artola-Borán
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Lydia Kirsche
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Angela Fallegger
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Peter Leary
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland; Functional Genomics Center Zürich, University of Zürich/ETHZ, Zürich, Switzerland
| | - Mine Tanriover
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Tanja Goodwin
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Gavin Geiger
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | | | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland; Institute of Biochemistry, Brandenburg Medical School, 16816 Neuruppin, Germany
| | - Isabelle C Arnold
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland; Comprehensive Cancer Center Zürich, Zürich, Switzerland.
| |
Collapse
|
6
|
Sirit IS, Peek RM. Decoding the Ability of Helicobacter pylori to Evade Immune Recognition and Cause Disease. Cell Mol Gastroenterol Hepatol 2025; 19:101470. [PMID: 39889829 PMCID: PMC11946503 DOI: 10.1016/j.jcmgh.2025.101470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 02/03/2025]
Abstract
Helicobacter pylori (H pylori) successfully and chronically colonizes the gastric mucosa of approximately 43% of the world's population. Infection with this organism is the strongest known risk factor for the development of gastric cancer, and disease development is dependent on several interactive components. One H pylori determinant that augments cancer risk is the strain-specific cag type IV secretion system, which not only translocates a pro-inflammatory and oncogenic protein, CagA, into host cells but also DNA, peptidoglycan, and a lipopolysaccharide intermediate, heptose-1,7-bisphosphate. However, cognate interactions between certain microbial and host constituents can also attenuate pro-inflammatory responses, and H pylori harbors multiple effectors that function differently than the respective counterparts in other mucosal pathogens. In this review, we discuss current data related to mechanisms utilized by H pylori to evade the immune response, sustain its longevity in the host, and further disease progression, as well as implications for developing targeted, immune-based eradication strategies.
Collapse
Affiliation(s)
- Isabella S Sirit
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Molecular Pathology and Immunology Training Program, Vanderbilt University, Nashville, Tennessee
| | - Richard M Peek
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
7
|
Qiu L, Lu X, Xue W, Fu H, Deng S, Li L, Chen M, Wang Y. Ischemic stroke susceptibility associated with ALPK1 single nucleotide polymorphisms by inhibiting URAT1 in uric acid hemostasis. Gene 2025; 934:149017. [PMID: 39437898 DOI: 10.1016/j.gene.2024.149017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVES Ischemic stroke (IS) prevalence rising annually, the necessity of discovering non-interventional genetic influences is progressing. Single nucleotide polymorphism (SNP) plays a pivotal role in stable inheritance of disease susceptibility. Based on the relationship between Alpha- Kinase 1 (ALPK1) and traditional IS risk factors especially hyperuricemia, our study investigated the association and function of ALPK1 SNPs with IS susceptibility. METHODS A case-control study of 1539 patients and 933 controls from northeast China was conducted. Genotyping information of ALPK1 rs2074379 and rs2074388 was collected. Four types of plasmids including rs2074379/rs2074388 G/G, A/G, G/A, and A/A were transfected into 293T cells to observe ALPK1 and SLC22A12 expression. Possible ALPK1 structures of different SNPs were predicted online. RESULTS Genotype GG (OR = 1.371, CI = 1.029-1.828, P = 0.031) and GA (OR = 1.326, CI = 1.110-1.584, P = 0.002) of rs2074379 and GA of rs2074388 (OR = 1.359, CI = 1.137-1.624, P = 0.001) were found significantly susceptible to IS, with G allele on sites to be a risk allele. Rs2074379 had a multiplicative interaction with hyperuricemia (OR = 1.637, CI = 1.157-2.315, P = 0.005). Uric acid levels differed in genotypes (P < 0.001). The expression of ALPK1 (P < 0.01) and SLC22A12 in membrane urate transporter 1 (URAT1) protein (P < 0.05) functionally changed with G allele on either site. With glycine changing into aspartic acid at rs2074388, the protein secondary structure changed, but the ALPK1 protein subtype remained still. CONCLUSIONS ALPK1 rs2074379 and rs2074388 SNPs were functionally associated with IS susceptibility. The wild allele progressed IS risk probably by reducing ALPK1 expression and inhibiting URAT1 raising the uric acid level, contributing to further exploration of pathogenetic mechanisms of stroke. Chinese Clinical Trial Registration number: ChiCTR-COC-17013559.
Collapse
Affiliation(s)
- Luying Qiu
- Department of Neurology, Key Laboratory for Neurological Big Data of Liaoning Province, Shenyang Clinical Medical Research Center for Difficult and Serious Diseases of the Nervous System, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning Province 110001, China
| | - Xiaoqin Lu
- Department of Neurology, Key Laboratory for Neurological Big Data of Liaoning Province, Shenyang Clinical Medical Research Center for Difficult and Serious Diseases of the Nervous System, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning Province 110001, China
| | - Weishuang Xue
- Department of Neurology, Key Laboratory for Neurological Big Data of Liaoning Province, Shenyang Clinical Medical Research Center for Difficult and Serious Diseases of the Nervous System, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning Province 110001, China
| | - Hefei Fu
- Department of Neurology, Key Laboratory for Neurological Big Data of Liaoning Province, Shenyang Clinical Medical Research Center for Difficult and Serious Diseases of the Nervous System, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning Province 110001, China
| | - Shumin Deng
- Department of Neurology, Key Laboratory for Neurological Big Data of Liaoning Province, Shenyang Clinical Medical Research Center for Difficult and Serious Diseases of the Nervous System, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning Province 110001, China
| | - Long Li
- Department of Neurosurgery, The First Hospital of China Medical University, China Medical University, Shenyang 110001, China
| | - Meilin Chen
- Department of Neurology, Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100069, China
| | - Yanzhe Wang
- Department of Neurology, Key Laboratory for Neurological Big Data of Liaoning Province, Shenyang Clinical Medical Research Center for Difficult and Serious Diseases of the Nervous System, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning Province 110001, China.
| |
Collapse
|
8
|
Martin A, Caron S, Marcotte M, Bronnec P, Garneret E, Martel N, Maalouf G, Sève P, Saadoun D, Jamilloux Y, Henry T. IFN-γ licenses normal and pathogenic ALPK1/TIFA pathway in human monocytes. iScience 2025; 28:111563. [PMID: 39868044 PMCID: PMC11758396 DOI: 10.1016/j.isci.2024.111563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 10/22/2024] [Accepted: 12/06/2024] [Indexed: 01/28/2025] Open
Abstract
Alpha-kinase 1 (ALPK1) is an immune receptor sensing the bacterial nucleotide sugar ADP-heptose. ALPK1 phosphorylates TIFA leading to its oligomerization and downstream NF-κB activation. Specific mutations in ALPK1 are associated with an autoinflammatory syndrome termed ROSAH and with spiradenoma (skin cancers with sweat gland differentiation). This study investigated ALPK1 responses in human mononuclear cells and demonstrates that human mononuclear cells have distinct abilities to respond to ADP-heptose. Notably, IFN-γ is required to license the ALPK1/TIFA pathway in monocytes, while it was dispensable for the responsiveness of B cells. IFN-γ induced TIFA upregulation in monocytes, and TIFA induction was sufficient to recapitulate the licensing effect of IFN-γ. IFN-γ treatment promoted the phenotypic expression of pathogenic ALPK1 mutations. The licensing effect of IFN-γ in monocytes was blocked by JAK inhibitors. These findings underscore the critical role of IFN-γ in ALPK1 function and suggest JAK inhibitors as potential therapies for ALPK1-related inflammatory conditions.
Collapse
Affiliation(s)
- Amandine Martin
- CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, University Lyon, F-69007 Lyon, France
| | - Solène Caron
- CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, University Lyon, F-69007 Lyon, France
| | - Mélissa Marcotte
- CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, University Lyon, F-69007 Lyon, France
| | - Pauline Bronnec
- CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, University Lyon, F-69007 Lyon, France
| | - Etienne Garneret
- CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, University Lyon, F-69007 Lyon, France
| | - Nora Martel
- CeRéMAIA: Centre National de Références Maladies Autoinflammatoires et Amylose Inflammatoire, 69000 Lyon, France
- Internal Medicine, University Hospital Croix-Rousse, Hospices Civils de Lyon, 69000 Lyon, France
| | - Georgina Maalouf
- CeRéMAIA: Centre National de Références Maladies Autoinflammatoires et Amylose Inflammatoire, 69000 Lyon, France
- Department of Internal Medicine and Clinical Immunology, Sorbonne Universités, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Centre National de Références Maladies Auto-immunes et Systémiques Rares, INSERM, UMR S959, Immunology-Immunopathology-Immunotherapy (I3), 83 Boulevard de L’hôpital, 75013 Paris, France
| | - Pascal Sève
- CeRéMAIA: Centre National de Références Maladies Autoinflammatoires et Amylose Inflammatoire, 69000 Lyon, France
- Internal Medicine, University Hospital Croix-Rousse, Hospices Civils de Lyon, 69000 Lyon, France
| | - David Saadoun
- CeRéMAIA: Centre National de Références Maladies Autoinflammatoires et Amylose Inflammatoire, 69000 Lyon, France
- Department of Internal Medicine and Clinical Immunology, Sorbonne Universités, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Centre National de Références Maladies Auto-immunes et Systémiques Rares, INSERM, UMR S959, Immunology-Immunopathology-Immunotherapy (I3), 83 Boulevard de L’hôpital, 75013 Paris, France
| | - Yvan Jamilloux
- CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, University Lyon, F-69007 Lyon, France
- CeRéMAIA: Centre National de Références Maladies Autoinflammatoires et Amylose Inflammatoire, 69000 Lyon, France
- Internal Medicine, University Hospital Croix-Rousse, Hospices Civils de Lyon, 69000 Lyon, France
| | - Thomas Henry
- CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, University Lyon, F-69007 Lyon, France
- CeRéMAIA: Centre National de Références Maladies Autoinflammatoires et Amylose Inflammatoire, 69000 Lyon, France
| |
Collapse
|
9
|
Liu YT, Cao LY, Sun ZJ. The emerging roles of liquid-liquid phase separation in tumor immunity. Int Immunopharmacol 2024; 143:113212. [PMID: 39353387 DOI: 10.1016/j.intimp.2024.113212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 09/02/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024]
Abstract
Recent advancements in tumor immunotherapy, particularly PD-1 targeted therapy, have shown significant promise, marking major progress in tumor treatment approaches. Despite this, the development of resistance to therapy and mechanisms of immune evasion by tumors pose considerable obstacles to the broad application of immunotherapy. This necessitates a deeper exploration of complex immune signaling pathways integral to tumor immunity. This review aims to critically analyze the role of liquid-liquid phase separation (LLPS) within tumor immunity, specifically its impact on immune signaling pathways and its potential to foster the development of novel cancer therapies. LLPS, a biophysical process newly recognized for its ability to spontaneously segregate and organize biomacromolecules into liquid-like condensates through weak multivalent interactions, offers a novel perspective on the formation of signaling clusters and the functionality of immune molecules. The review delves into the micromolecular mechanisms behind the creation of signaling condensates via LLPS and reviews recent progress in adjusting signaling pathways pertinent to tumor immunity, including the T cell receptor (TCR), B cell receptor (BCR), immune checkpoints, and innate immune pathways such as the cGAS-STING pathway, stress granules, and the ADP-heptose-ALPK1 signaling axis. Furthermore, it considers the prospects of utilizing LLPS to generate groundbreaking cancer therapies capable of navigating past current treatment barriers. Through an extensive examination of LLPS's impact on tumor immunity, the review seeks to highlight novel therapeutic strategies and address the challenges and future directions in this rapidly evolving field.
Collapse
Affiliation(s)
- Yuan-Tong Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China; Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lin-Yu Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
10
|
Kuang W, Xu J, Xu F, Huang W, Majid M, Shi H, Yuan X, Ruan Y, Hu X. Current study of pathogenetic mechanisms and therapeutics of chronic atrophic gastritis: a comprehensive review. Front Cell Dev Biol 2024; 12:1513426. [PMID: 39720008 PMCID: PMC11666564 DOI: 10.3389/fcell.2024.1513426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024] Open
Abstract
Chronic atrophic gastritis (CAG) is a prevalent digestive system disease characterized by atrophy of the gastric mucosa and the disappearance of inherent gastric glands. According to the theory of Correa's cascade, CAG is an important pathological stage in the transformation from normal condition to gastric carcinoma. In recent years, the global incidence of CAG has been increasing due to pathogenic factors, including Helicobacter pylori infection, bile reflux, and the consumption of processed meats. In this review, we comprehensively described the etiology and clinical diagnosis of CAG. We focused on elucidating the regulatory mechanisms and promising therapeutic targets in CAG, with the expectation of providing insights and theoretical support for future research on CAG.
Collapse
Affiliation(s)
- Weihong Kuang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Traditional Chinese Medicines for Prevention and Treatment of Digestive Diseases, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Jialin Xu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Traditional Chinese Medicines for Prevention and Treatment of Digestive Diseases, Guangdong Medical University, Dongguan, China
| | - Fenting Xu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Traditional Chinese Medicines for Prevention and Treatment of Digestive Diseases, Guangdong Medical University, Dongguan, China
| | - Weizhen Huang
- Cancer Center, The First Huizhou Affiliated Hospital, Guangdong Medical University, Huizhou, China
| | - Muhammad Majid
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Traditional Chinese Medicines for Prevention and Treatment of Digestive Diseases, Guangdong Medical University, Dongguan, China
| | - Hui Shi
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Traditional Chinese Medicines for Prevention and Treatment of Digestive Diseases, Guangdong Medical University, Dongguan, China
| | - Xia Yuan
- Cancer Center, The First Huizhou Affiliated Hospital, Guangdong Medical University, Huizhou, China
| | - Yongdui Ruan
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Traditional Chinese Medicines for Prevention and Treatment of Digestive Diseases, Guangdong Medical University, Dongguan, China
- Cancer Center, The First Huizhou Affiliated Hospital, Guangdong Medical University, Huizhou, China
| | - Xianjing Hu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Traditional Chinese Medicines for Prevention and Treatment of Digestive Diseases, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Department of Acupuncture, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| |
Collapse
|
11
|
Nascakova Z, He J, Papa G, Francas B, Azizi F, Müller A. Helicobacter pylori induces the expression of Lgr5 and stem cell properties in gastric target cells. Life Sci Alliance 2024; 7:e202402783. [PMID: 39191487 PMCID: PMC11350067 DOI: 10.26508/lsa.202402783] [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: 04/22/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024] Open
Abstract
Helicobacter pylori infection predisposes carriers to a high risk of developing gastric cancer. The cell-of-origin of antral gastric cancer is the Lgr5+ stem cell. Here, we show that infection of antrum-derived gastric organoid cells with H. pylori increases the expression of the stem cell marker Lgr5 as determined by immunofluorescence microscopy, qRT-PCR, and Western blotting, both when cells are grown and infected as monolayers and when cells are exposed to H. pylori in 3D structures. H. pylori exposure increases stemness properties as determined by spheroid formation assay. Lgr5 expression and the acquisition of stemness depend on a functional type IV secretion system (T4SS) and at least partly on the T4SS effector CagA. The pharmacological inhibition or genetic ablation of NF-κB reverses the increase in Lgr5 and spheroid formation. Constitutively active Wnt/β-catenin signaling because of Apc inactivation exacerbates H. pylori-induced Lgr5 expression and stemness, both of which persist even after eradication of the infection. The combined data indicate that H. pylori has stemness-inducing properties that depend on its ability to activate NF-κB signaling.
Collapse
Affiliation(s)
- Zuzana Nascakova
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Jiazhuo He
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Giovanni Papa
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Biel Francas
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Flora Azizi
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
- Comprehensive Cancer Center Zürich, Zürich, Switzerland
| |
Collapse
|
12
|
Linz B, Sticht H, Tegtmeyer N, Backert S. Cancer-associated SNPs in bacteria: lessons from Helicobacter pylori. Trends Microbiol 2024; 32:847-857. [PMID: 38485609 DOI: 10.1016/j.tim.2024.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 09/06/2024]
Abstract
Several single-nucleotide polymorphisms (SNPs) in human chromosomes are known to predispose to cancer. However, cancer-associated SNPs in bacterial pathogens were unknown until discovered in the stomach pathogen Helicobacter pylori. Those include an alanine-threonine polymorphism in the EPIYA-B phosphorylation motif of the injected effector protein CagA that affects cancer risk by modifying inflammatory responses and loss of host cell polarity. A serine-to-leucine change in serine protease HtrA is associated with boosted proteolytic cleavage of epithelial junction proteins and introduction of DNA double-strand breaks (DSBs) in host chromosomes, which co-operatively elicit malignant alterations. In addition, H. pylori genome-wide association studies (GWAS) identified several other SNPs potentially associated with increased gastric cancer (GC) risk. Here we discuss the clinical importance, evolutionary origin, and functional advantage of the H. pylori SNPs. These exciting new data highlight cancer-associated SNPs in bacteria, which should be explored in more detail in future studies.
Collapse
Affiliation(s)
- Bodo Linz
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudtstr. 5, 91058 Erlangen, Germany
| | - Heinrich Sticht
- Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg; 91054 Erlangen, Germany
| | - Nicole Tegtmeyer
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudtstr. 5, 91058 Erlangen, Germany
| | - Steffen Backert
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudtstr. 5, 91058 Erlangen, Germany.
| |
Collapse
|
13
|
Gong T, Zhang X, Liu X, Ye Y, Tian Z, Yin S, Zhang M, Tang J, Liu Y. Exosomal Tenascin-C primes macrophage pyroptosis amplifying aberrant inflammation during sepsis-induced acute lung injury. Transl Res 2024; 270:66-80. [PMID: 38604333 DOI: 10.1016/j.trsl.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 03/15/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
Sepsis-induced acute lung injury (ALI) is a serious complication of sepsis and the predominant cause of death. Exosomes released by lung tissue cells critically influence the progression of ALI during sepsis by modulating the inflammatory microenvironment. However, the molecular mechanisms by which exosome-mediated intercellular signaling exacerbates ALI in septic infection remain undefined. Our study found increased levels of exosomal Tenascin-C (TNC) in the plasma of both patients and mice with ALI, showing a strong association with disease progression. By integrating exosomal proteomics with transcriptome sequencing and experimental validation, we elucidated that LPS induce unresolved endoplasmic reticulum stress (ERs) in alveolar epithelial cells (AECs), ultimately leading to the release of exosomal TNC through the activation of PERK-eIF2α and the transcription factor CHOP. In the sepsis mouse model with TNC knockout, we noted a marked reduction in macrophage pyroptosis. Our detailed investigations found that exosomal TNC binds to TLR4 on macrophages, resulting in an augmented production of ROS, subsequent mitochondrial damage, activation of the NF-κB signaling pathway, and induction of DNA damage response. These interconnected events culminate in macrophage pyroptosis, thereby amplifying the release of inflammatory cytokines. Our findings demonstrate that exosomal Tenascin-C, released from AECs under unresolved ER stress, exacerbates acute lung injury by intensifying sepsis-associated inflammatory responses. This research provides new insights into the complex cellular interactions underlying sepsis-induced ALI.
Collapse
Affiliation(s)
- Ting Gong
- Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, No.1333, Xinhu Road, Baoan District, Shenzhen 518110, Guangdong, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| | - Xuedi Zhang
- Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, No.1333, Xinhu Road, Baoan District, Shenzhen 518110, Guangdong, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xiaolei Liu
- Department of Anaesthetics, Affiliated Hospital of Guangdong Medical University, No.57 People Avenue South, Zhanjiang, 524001, Guangdong, China
| | - Yinfeng Ye
- Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, No.1333, Xinhu Road, Baoan District, Shenzhen 518110, Guangdong, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhiyuan Tian
- Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, No.1333, Xinhu Road, Baoan District, Shenzhen 518110, Guangdong, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Shuang Yin
- Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, No.1333, Xinhu Road, Baoan District, Shenzhen 518110, Guangdong, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Min Zhang
- Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, No.1333, Xinhu Road, Baoan District, Shenzhen 518110, Guangdong, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jing Tang
- Department of Anaesthetics, Affiliated Hospital of Guangdong Medical University, No.57 People Avenue South, Zhanjiang, 524001, Guangdong, China.
| | - Youtan Liu
- Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, No.1333, Xinhu Road, Baoan District, Shenzhen 518110, Guangdong, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
14
|
Zhang Y, Liu T, He W. The application of organoids in cancers associated with pathogenic infections. Clin Exp Med 2024; 24:168. [PMID: 39052148 PMCID: PMC11272814 DOI: 10.1007/s10238-024-01435-8] [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: 04/15/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024]
Abstract
Cancers associated with pathogen infections are gradually becoming important threats to human health globally, and it is of great significance to study the mechanisms of pathogen carcinogenesis. Current mechanistic studies rely on animal and two-dimensional (2D) cell culture models, but traditional methods have been proven insufficient for the rapid modeling of diseases caused by new pathogens. Therefore, research focus has shifted to organoid models, which can replicate the structural and genetic characteristics of the target tissues or organs in vitro, providing new platforms for the study of pathogen-induced oncogenic mechanisms. This review summarizes the application of organoid technology in the studies of four pathogen-associated cancers: gastric cancer linked to Helicobacter pylori, liver cancer associated with hepatitis B virus or hepatitis C virus, colorectal cancer caused by Escherichia coli, and cervical cancer related to human papillomavirus. This review also proposes several limitations of organoid technology to optimize organoid models and advance the treatment of cancer associated with pathogen infections in the future.
Collapse
Affiliation(s)
- Yuyu Zhang
- Department of the Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou, 730030, China
- Digestive System Tumor Prevention and Treatment and Translational Medicine Engineering Innovation Center of Lanzhou University, Lanzhou, 730030, China
- Digestive System Tumor Translational Medicine Engineering Research Center of Gansu Province, Lanzhou, 730030, China
| | - Tao Liu
- Department of the Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China.
- Second Hospital of Lanzhou University, Lanzhou University, Lanzhou, 730000, China.
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou, 730030, China.
- Digestive System Tumor Prevention and Treatment and Translational Medicine Engineering Innovation Center of Lanzhou University, Lanzhou, 730030, China.
- Digestive System Tumor Translational Medicine Engineering Research Center of Gansu Province, Lanzhou, 730030, China.
| | - Wenting He
- Department of the Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China.
- Second Hospital of Lanzhou University, Lanzhou University, Lanzhou, 730000, China.
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou, 730030, China.
- Digestive System Tumor Prevention and Treatment and Translational Medicine Engineering Innovation Center of Lanzhou University, Lanzhou, 730030, China.
- Digestive System Tumor Translational Medicine Engineering Research Center of Gansu Province, Lanzhou, 730030, China.
| |
Collapse
|
15
|
Hochvaldova L, Posselt G, Wessler S, Kvítek L, Panáček A. Implications of silver nanoparticles for H. pylori infection: modulation of CagA function and signaling. Front Cell Infect Microbiol 2024; 14:1419568. [PMID: 38983115 PMCID: PMC11231068 DOI: 10.3389/fcimb.2024.1419568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/10/2024] [Indexed: 07/11/2024] Open
Abstract
Background Helicobacter pylori infection poses a significant health burden worldwide, and its virulence factor CagA plays a pivotal role in its pathogenesis. Methods In this study, the interaction between H. pylori-infected AGS cells and silver nanoparticles (AgNPs) was investigated, with a focus on the modulation of CagA-mediated responses, investigated by western blotting. Both, the dose-dependent efficacy against H. pylori (growth curves, CFU assay) and the impact of the nanoparticles on AGS cells (MTT assay) were elucidated. Results AGS cells infected with H. pylori displayed dramatic morphological changes, characterized by elongation and a migratory phenotype, attributed to CagA activity. Preincubation of H. pylori with AgNPs affected these morphological changes in a concentration-dependent manner, suggesting a correlation between AgNPs concentration and CagA function. Conclusion Our study highlights the nuanced interplay between host-pathogen interactions and the therapeutic potential of AgNPs in combating H. pylori infection and offers valuable insights into the multifaceted dynamics of CagA mediated responses.
Collapse
Affiliation(s)
- Lucie Hochvaldova
- Department of Physical Chemistry, Palacky University Olomouc, Olomouc, Czechia
| | - Gernot Posselt
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, Austria
- Center for Tumor Biology and Immunology (CTBI), Paris Lodron University of Salzburg, Salzburg, Austria
| | - Silja Wessler
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, Austria
- Center for Tumor Biology and Immunology (CTBI), Paris Lodron University of Salzburg, Salzburg, Austria
| | - Libor Kvítek
- Department of Physical Chemistry, Palacky University Olomouc, Olomouc, Czechia
| | - Aleš Panáček
- Department of Physical Chemistry, Palacky University Olomouc, Olomouc, Czechia
| |
Collapse
|
16
|
Zhang X, He Y, Zhang X, Fu B, Song Z, Wang L, Fu R, Lu X, Xing J, Lv J, Guo M, Huo X, Liu X, Lu J, Du X, Ge Z, Chen Z, Li C. Sustained exposure to Helicobacter pylori induces immune tolerance by desensitizing TLR6. Gastric Cancer 2024; 27:324-342. [PMID: 38310631 PMCID: PMC10896808 DOI: 10.1007/s10120-023-01461-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 12/19/2023] [Indexed: 02/06/2024]
Abstract
Helicobacter pylori (H. pylori, Hp) has been designated a class I carcinogen and is closely associated with severe gastric diseases. During colonization in the gastric mucosa, H. pylori develops immune escape by inducing host immune tolerance. The gastric epithelium acts as the first line of defense against H. pylori, with Toll-like receptors (TLRs) in gastric epithelial cells being sensitive to H. pylori components and subsequently activating the innate immune system. However, the mechanism of immune tolerance induced by H. pylori through the TLR signalling pathway has not been fully elucidated. In this research, we detected the expression of TLRs and inflammatory cytokines in GES-1 cells upon sustained exposure to H. pylori or H. pylori lysate from 1 to 30 generations and in Mongolian gerbils infected with H. pylori for 5 to 90 weeks. We found that the levels of TLR6 and inflammatory cytokines first increased and then dropped during the course of H. pylori treatment in vitro and in vivo. The restoration of TLR6 potentiated the expression of IL-1β and IL-8 in GES-1 cells, which recruited neutrophils and reduced the colonization of H. pylori in the gastric mucosa of gerbils. Mechanistically, we found that persistent infection with H. pylori reduces the sensitivity of TLR6 to bacterial components and regulates the expression of inflammatory cytokines in GES-1 cells through TLR6/JNK signaling. The TLR6 agonist obviously alleviated inflammation in vitro and in vivo. Promising results suggest that TLR6 may be a potential candidate immunotherapy drug for H. pylori infection.
Collapse
Affiliation(s)
- Xiulin Zhang
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, Beijing, People's Republic of China
| | - Yang He
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
- School of Nursing, Dalian Medical University, Dalian, People's Republic of China
| | - Xiaolu Zhang
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Bo Fu
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Zidai Song
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Liang Wang
- Peking University Ninth School of Clinical Medicine, Beijing, People's Republic of China
| | - Rui Fu
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Xuancheng Lu
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Jin Xing
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Jianyi Lv
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Meng Guo
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Xueyun Huo
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Xin Liu
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Jing Lu
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Xiaoyan Du
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Zhongming Ge
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, USA
| | - Zhenwen Chen
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Changlong Li
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China.
| |
Collapse
|
17
|
Isik E, Shukla K, Pospisilova M, König C, Andrs M, Rao S, Rosano V, Dobrovolna J, Krejci L, Janscak P. MutSβ-MutLβ-FANCJ axis mediates the restart of DNA replication after fork stalling at cotranscriptional G4/R-loops. SCIENCE ADVANCES 2024; 10:eadk2685. [PMID: 38324687 PMCID: PMC10849593 DOI: 10.1126/sciadv.adk2685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 01/08/2024] [Indexed: 02/09/2024]
Abstract
Transcription-replication conflicts (TRCs) induce formation of cotranscriptional RNA:DNA hybrids (R-loops) stabilized by G-quadruplexes (G4s) on the displaced DNA strand, which can cause fork stalling. Although it is known that these stalled forks can resume DNA synthesis in a process initiated by MUS81 endonuclease, how TRC-associated G4/R-loops are removed to allow fork passage remains unclear. Here, we identify the mismatch repair protein MutSβ, an MLH1-PMS1 heterodimer termed MutLβ, and the G4-resolving helicase FANCJ as factors that are required for MUS81-initiated restart of DNA replication at TRC sites in human cells. This DNA repair process depends on the G4-binding activity of MutSβ, the helicase activity of FANCJ, and the binding of FANCJ to MLH1. Furthermore, we show that MutSβ, MutLβ, and MLH1-FANCJ interaction mediate FANCJ recruitment to G4s. These data suggest that MutSβ, MutLβ, and FANCJ act in conjunction to eliminate G4/R-loops at TRC sites, allowing replication restart.
Collapse
Affiliation(s)
- Esin Isik
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Kaustubh Shukla
- Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 143 00 Prague, Czech Republic
| | - Michaela Pospisilova
- Department of Biology and National Centre for Biomolecular Research, Masaryk University, Kamenice 5/A7, Brno 62500, Czech Republic
- International Clinical Research Center, St Anne's University Hospital, Pekarska 53, Brno 656 91, Czech Republic
| | - Christiane König
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Martin Andrs
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Satyajeet Rao
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Vinicio Rosano
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Jana Dobrovolna
- Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 143 00 Prague, Czech Republic
| | - Lumir Krejci
- Department of Biology and National Centre for Biomolecular Research, Masaryk University, Kamenice 5/A7, Brno 62500, Czech Republic
- International Clinical Research Center, St Anne's University Hospital, Pekarska 53, Brno 656 91, Czech Republic
| | - Pavel Janscak
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 143 00 Prague, Czech Republic
| |
Collapse
|
18
|
Guo X, Qiao G, Wang J, Yang C, Zhao M, Zhang Q, Wan Y. TIFA contributes to periodontitis in diabetic mice via activating the NF‑κB signaling pathway. Mol Med Rep 2024; 29:23. [PMID: 38099344 PMCID: PMC10784739 DOI: 10.3892/mmr.2023.13146] [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: 05/31/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
Diabetic periodontitis (DP) refers to destruction of periodontal tissue and absorption of bone tissue in diabetic patients. Tumor necrosis factor receptor‑associated factor (TRAF)‑interacting protein with forkhead‑associated domain (TIFA) as a crucial regulator of inflammation activates the NF‑κB signaling pathway to regulate cell biological behavior. However, the function and mechanism of TIFA on DP suffer from a lack of research. In the present study, TIFA was upregulated in the periodontal tissue of a DP mouse model. In addition, the expression of TIFA in RAW264.7 cells was induced by high glucose (HG) culture and increased by lipopolysaccharide (LPS) from Porphyromonas gingivalis treatment in a time‑dependent manner. Knockdown of TIFA significantly reduced the levels of inflammatory cytokines, including TNF‑α, IL‑6, IL‑1β and monocyte chemoattractant protein‑1, in HG and LPS‑induced RAW264.7 cells. The nuclear translocation of NF‑κB p65 was induced by HG and LPS and was clearly suppressed by absence of TIFA. The expression of downstream factors Nod‑like receptor family pyrin domain‑containing 3 and apoptosis‑associated speck‑like protein was inhibited by silencing TIFA. Moreover, TIFA was increased by receptor activator of NF‑κB (RANK) ligand (RANKL) in a concentration dependent manner. The expression of cathepsin K, MMP9 and nuclear factor of activated T cells cytoplasmic 1 was downregulated by depletion of TIFA. RANKL‑induced osteoclast differentiation was inhibited by silencing of TIFA. Meanwhile, the decrease of TIFA blocked activation of the NF‑κB pathway in RANKL‑treated RAW264.7 cells. In conclusion, TIFA as a promoter regulates the inflammation and osteoclast differentiation via activating the NF‑κB signaling pathway.
Collapse
Affiliation(s)
- Xiaoqian Guo
- Department of Periodontology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
- Ningxia Key Laboratory of Oral Disease Research, School of Stomatology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Guangwei Qiao
- Department of Oral and Maxillofacial Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Jingjiao Wang
- Department of Periodontology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Changyi Yang
- Department of Periodontology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Min Zhao
- Department of Periodontology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Qian Zhang
- Department of Periodontology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yingbiao Wan
- Department of Prosthodontics and Oral Implantology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
19
|
Liu S, Zhuang Z, Liu F, Yuan X, Zhang Z, Liang X, Li X, Chen Y. Identification of potential biomarkers and infiltrating immune cells from scalp psoriasis. Gene 2024; 893:147918. [PMID: 37871808 DOI: 10.1016/j.gene.2023.147918] [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: 09/21/2023] [Revised: 10/16/2023] [Accepted: 10/20/2023] [Indexed: 10/25/2023]
Abstract
BACKGROUND Scalp psoriasis seriously affects the appearance and psychological status of patients. The aim of this study was to investigate the effect and potential mechanism of RPL9 and TIFA in scalp psoriasis, so as to provide a precise and effective way for the clinical treatment of scalp psoriasis. METHODS The Gene Expression Omnibus (GEO) database was employed to download the GSE75343 dataset to search for differentially expressed genes (DEGs) in scalp psoriasis through Sangerbox. Gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) enrichment analysis, functional enrichment analysis, immune cell infiltration analysis, immune responses and correlation analysis with 12 hub genes were performed. Then, STRING was used to develop a protein-protein interaction (PPI) network, used Cytoscape to locate hub genes, and SVM-RFE and random forest were utilized to identified RPL9 as the targeted gene. TIFA-RPL9 interaction predictions were made viathe Open Targets Platform and Uniprot. Further, the RPL9 and TIFA expression, molecular mechanism, and function were assessed in scalp psoriasis. RESULTS Immunohistochemistry, qPCR, and western blotting verified that RPL9 and TIFA were highly expressed in lesional tissues of scalp psoriasis and IL17A-stimulated HaCaT cells. RPL9 knockdown effectively suppressed the proliferative capacity of IL17A-stimulated HaCaT cells in the CCK8 assay. The co-immunoprecipitation results revealed that RPL9 could interact with TIFA in IL17A-stimulated HaCaT cells. In qPCR and western blotting, RPL9 knockdown significantly inhibited TIFA at the mRNA and protein levels in IL17A-stimulated HaCaT cells. In ELISA, the secretion of TNF-α was markedly inhibited after downregulating RPL9 in IL17A-stimulated HaCaT cells. CONCLUSION To our knowledge, we have elucidated the expression and role of RPL9 and TIFA in scalp psoriatic skin and keratinocytes, and our findings confirm that RPL9 might act as a candidate therapeutic target for scalp psoriasis.
Collapse
Affiliation(s)
- Shougang Liu
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Zhe Zhuang
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Dermatology, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Fanghua Liu
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Dermatology, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xiuqing Yuan
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Zeqiao Zhang
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoqian Liang
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Xinhui Li
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Yongfeng Chen
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
20
|
Lu J, Liu X, Li X, Li H, Shi L, Xia X, He BL, Meyer TF, Li X, Sun H, Yang X. Copper regulates the host innate immune response against bacterial infection via activation of ALPK1 kinase. Proc Natl Acad Sci U S A 2024; 121:e2311630121. [PMID: 38232278 PMCID: PMC10823219 DOI: 10.1073/pnas.2311630121] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 11/22/2023] [Indexed: 01/19/2024] Open
Abstract
Copper is an essential trace element for the human body, and its requirement for optimistic immune functions has been recognized for decades. How copper is involved in the innate immune pathway, however, remains to be clarified. Here, we report that copper serves as a signal molecule to regulate the kinase activity of alpha-kinase 1 (ALPK1), a cytosolic pattern-recognition receptor (PRR), and therefore promotes host cell defense against bacterial infection. We show that in response to infection, host cells actively accumulate copper in the cytosol, and the accumulated cytosolic copper enhances host cell defense against evading pathogens, including intracellular and, unexpectedly, extracellular bacteria. Subsequently, we demonstrate that copper activates the innate immune pathway of host cells in an ALPK1-dependent manner. Further mechanistic studies reveal that copper binds to ALPK1 directly and is essential for the kinase activity of this cytosolic PRR. Moreover, the binding of copper to ALPK1 enhances the sensitivity of ALPK1 to the bacterial metabolite ADP-heptose and eventually prompts host cells to elicit an enhanced immune response during bacterial infection. Finally, using a zebrafish in vivo model, we show that a copper-treated host shows an increased production of proinflammatory cytokines, enhanced recruitment of phagosome cells, and promoted bacterial clearance. Our findings uncover a previously unrecognized role of copper in the modulation of host innate immune response against bacterial pathogens and advance our knowledge on the cross talk between cytosolic copper homeostasis and immune system.
Collapse
Affiliation(s)
- Jing Lu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, China
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, China
| | - Xue Liu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, China
- Department of Chemistry, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, China
| | - Xinghua Li
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, China
| | - Hongyan Li
- Department of Chemistry, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Chinese Academy of Sciences-The University of Hong Kong Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Liwa Shi
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, China
| | - Xin Xia
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, China
| | - Bai-liang He
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, China
| | - Thomas F. Meyer
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin10117, Germany
- Laboratory of Infection Oncology, Institute of Clinical Molecular Biology, Christian Albrecht’s University of Kiel, University Hospital Schleswig Holstein, Kiel24105, Germany
| | - Xiaofeng Li
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, China
| | - Hongzhe Sun
- Department of Chemistry, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Chinese Academy of Sciences-The University of Hong Kong Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xinming Yang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, China
| |
Collapse
|
21
|
Tran SC, Bryant KN, Cover TL. The Helicobacter pylori cag pathogenicity island as a determinant of gastric cancer risk. Gut Microbes 2024; 16:2314201. [PMID: 38391242 PMCID: PMC10896142 DOI: 10.1080/19490976.2024.2314201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 01/31/2024] [Indexed: 02/24/2024] Open
Abstract
Helicobacter pylori strains can be broadly classified into two groups based on whether they contain or lack a chromosomal region known as the cag pathogenicity island (cag PAI). Colonization of the human stomach with cag PAI-positive strains is associated with an increased risk of gastric cancer and peptic ulcer disease, compared to colonization with cag PAI-negative strains. The cag PAI encodes a secreted effector protein (CagA) and components of a type IV secretion system (Cag T4SS) that delivers CagA and non-protein substrates into host cells. Animal model experiments indicate that CagA and the Cag T4SS stimulate a gastric mucosal inflammatory response and contribute to the development of gastric cancer. In this review, we discuss recent studies defining structural and functional features of CagA and the Cag T4SS and mechanisms by which H. pylori strains containing the cag PAI promote the development of gastric cancer and peptic ulcer disease.
Collapse
Affiliation(s)
- Sirena C. Tran
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kaeli N. Bryant
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Timothy L. Cover
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
22
|
Neuper T, Frauenlob T, Dang HH, Krenn PW, Posselt G, Regl C, Fortelny N, Schäpertöns V, Unger MS, Üblagger G, Neureiter D, Mühlbacher I, Weitzendorfer M, Singhartinger F, Emmanuel K, Huber CG, Wessler S, Aberger F, Horejs-Hoeck J. ADP-heptose attenuates Helicobacter pylori-induced dendritic cell activation. Gut Microbes 2024; 16:2402543. [PMID: 39288239 PMCID: PMC11409497 DOI: 10.1080/19490976.2024.2402543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 09/19/2024] Open
Abstract
Sophisticated immune evasion strategies enable Helicobacter pylori (H. pylori) to colonize the gastric mucosa of approximately half of the world's population. Persistent infection and the resulting chronic inflammation are a major cause of gastric cancer. To understand the intricate interplay between H. pylori and host immunity, spatial profiling was used to monitor immune cells in H. pylori infected gastric tissue. Dendritic cell (DC) and T cell phenotypes were further investigated in gastric organoid/immune cell co-cultures and mechanistic insights were acquired by proteomics of human DCs. Here, we show that ADP-heptose, a bacterial metabolite originally reported to act as a bona fide PAMP, reduces H. pylori-induced DC maturation and subsequent T cell responses. Mechanistically, we report that H. pylori uptake and subsequent DC activation by an ADP-heptose deficient H. pylori strain depends on TLR2. Moreover, ADP-heptose attenuates full-fledged activation of primary human DCs in the context of H. pylori infection by impairing type I IFN signaling. This study reveals that ADP-heptose mitigates host immunity during H. pylori infection.
Collapse
Affiliation(s)
- Theresa Neuper
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Tobias Frauenlob
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Hieu-Hoa Dang
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Peter W Krenn
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Gernot Posselt
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Christof Regl
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Nikolaus Fortelny
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Veronika Schäpertöns
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
| | - Michael S Unger
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
| | - Gunda Üblagger
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
| | - Daniel Neureiter
- Cancer Cluster Salzburg, Salzburg, Austria
- Institute of Pathology, Uniklinikum Salzburg, Salzburg, Austria
| | - Iris Mühlbacher
- Department of General, Visceral and Thoracic Surgery, Paracelsus Medical University/Salzburger Landeskliniken (SALK), Salzburg, Austria
| | - Michael Weitzendorfer
- Department of General, Visceral and Thoracic Surgery, Paracelsus Medical University/Salzburger Landeskliniken (SALK), Salzburg, Austria
| | - Franz Singhartinger
- Department of General, Visceral and Thoracic Surgery, Paracelsus Medical University/Salzburger Landeskliniken (SALK), Salzburg, Austria
| | - Klaus Emmanuel
- Cancer Cluster Salzburg, Salzburg, Austria
- Department of General, Visceral and Thoracic Surgery, Paracelsus Medical University/Salzburger Landeskliniken (SALK), Salzburg, Austria
| | - Christian G Huber
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Silja Wessler
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Fritz Aberger
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| |
Collapse
|
23
|
Martin-Gallausiaux C, Salesse L, Garcia-Weber D, Marinelli L, Beguet-Crespel F, Brochard V, Le Gléau C, Jamet A, Doré J, Blottière HM, Arrieumerlou C, Lapaque N. Fusobacterium nucleatum promotes inflammatory and anti-apoptotic responses in colorectal cancer cells via ADP-heptose release and ALPK1/TIFA axis activation. Gut Microbes 2024; 16:2295384. [PMID: 38126163 PMCID: PMC10761154 DOI: 10.1080/19490976.2023.2295384] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
The anaerobic bacterium Fusobacterium nucleatum is significantly associated with human colorectal cancer (CRC) and is considered a significant contributor to the disease. The mechanisms underlying the promotion of intestinal tumor formation by F. nucleatum have only been partially uncovered. Here, we showed that F. nucleatum releases a metabolite into the microenvironment that strongly activates NF-κB in intestinal epithelial cells via the ALPK1/TIFA/TRAF6 pathway. Furthermore, we showed that the released molecule had the biological characteristics of ADP-heptose. We observed that F. nucleatum induction of this pathway increased the expression of the inflammatory cytokine IL-8 and two anti-apoptotic genes known to be implicated in CRC, BIRC3 and TNFAIP3. Finally, it promoted the survival of CRC cells and reduced 5-fluorouracil chemosensitivity in vitro. Taken together, our results emphasize the importance of the ALPK1/TIFA pathway in Fusobacterium induced-CRC pathogenesis, and identify the role of ADP-H in this process.
Collapse
Affiliation(s)
| | - Laurène Salesse
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | | | - Ludovica Marinelli
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | | | - Vincent Brochard
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Camille Le Gléau
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Alexandre Jamet
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Joël Doré
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Université Paris-Saclay, INRAE, Metagenopolis, Jouy-en-Josas, France
| | - Hervé M. Blottière
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Université Paris-Saclay, INRAE, Metagenopolis, Jouy-en-Josas, France
| | | | - Nicolas Lapaque
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| |
Collapse
|
24
|
Zhang X, Zhang K, Yan L, Wang P, Zhao F, Hu S. The role of toll-like receptors in immune tolerance induced by Helicobacter pylori infection. Helicobacter 2023; 28:e13020. [PMID: 37691007 DOI: 10.1111/hel.13020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/11/2023] [Accepted: 08/29/2023] [Indexed: 09/12/2023]
Abstract
Helicobacter pylori (H. pylori) is a gram-negative, microaerobic bacterium that colonizes the gastric mucosa in about half of the world's population. H. pylori infection can lead to various diseases. Chronic infection by H. pylori exposes the gastric mucosa to bacterial components such as lipopolysaccharide (LPS), outer membrane vesicles (OMVs), and several toxic proteins. Infected with H. pylori activates the release of pro-inflammatory factors and triggers inflammatory responses that damage the gastric mucosa. As the only microorganism that permanently colonizes the human stomach, H. pylori can suppress host immunity to achieve long-term colonization. Toll-like receptors (TLRs) play a crucial role in T-cell activation, promoting innate immune responses and immune tolerance during H. pylori infection. Among the 10 TLRs found in humans, TLR2, TLR4, TLR5, and TLR9 have been thoroughly investigated in relation to H. pylori-linked immune regulation. In the present review, we provide a comprehensive analysis of the various mechanisms employed by different TLRs in the induction of immune tolerance upon H. pylori infection, which will contribute to the research of pathogenic mechanism of H. pylori.
Collapse
Affiliation(s)
- Xiulin Zhang
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, China
| | - Ke Zhang
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, China
| | - Linlin Yan
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, China
| | - Pengfei Wang
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, China
| | - Fan Zhao
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, China
| | - Shoukui Hu
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, China
| |
Collapse
|
25
|
Ma TS, Worth KR, Maher C, Ng N, Beghè C, Gromak N, Rose AM, Hammond EM. Hypoxia-induced transcriptional stress is mediated by ROS-induced R-loops. Nucleic Acids Res 2023; 51:11584-11599. [PMID: 37843099 PMCID: PMC10681727 DOI: 10.1093/nar/gkad858] [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: 05/02/2023] [Revised: 08/21/2023] [Accepted: 09/27/2023] [Indexed: 10/17/2023] Open
Abstract
Hypoxia is a common feature of solid tumors and is associated with poor patient prognosis, therapy resistance and metastasis. Radiobiological hypoxia (<0.1% O2) is one of the few physiologically relevant stresses that activates both the replication stress/DNA damage response and the unfolded protein response. Recently, we found that hypoxia also leads to the robust accumulation of R-loops, which led us to question here both the mechanism and consequence of hypoxia-induced R-loops. Interestingly, we found that the mechanism of R-loop accumulation in hypoxia is dependent on non-DNA damaging levels of reactive oxygen species. We show that hypoxia-induced R-loops play a critical role in the transcriptional stress response, evidenced by the repression of ribosomal RNA synthesis and the translocation of nucleolin from the nucleolus into the nucleoplasm. Upon depletion of R-loops, we observed a rescue of both rRNA transcription and nucleolin translocation in hypoxia. Mechanistically, R-loops accumulate on the rDNA in hypoxia and promote the deposition of heterochromatic H3K9me2 which leads to the inhibition of Pol I-mediated transcription of rRNA. These data highlight a novel mechanistic insight into the hypoxia-induced transcriptional stress response through the ROS-R-loop-H3K9me2 axis. Overall, this study highlights the contribution of transcriptional stress to hypoxia-mediated tumorigenesis.
Collapse
Affiliation(s)
- Tiffany S Ma
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Katja R Worth
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Conor Maher
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Natalie Ng
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Chiara Beghè
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Natalia Gromak
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Anna M Rose
- Department of Pediatrics, University of Oxford, Oxford OX3 9DU, UK
| | - Ester M Hammond
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| |
Collapse
|
26
|
Zhang S, Wang Q, Ye J, Fan Q, Lin X, Gou Z, Azzam MM, Wang Y, Jiang S. Transcriptome and proteome profile of jejunum in chickens challenged with Salmonella Typhimurium revealed the effects of dietary bilberry anthocyanin on immune function. Front Microbiol 2023; 14:1266977. [PMID: 38053560 PMCID: PMC10694457 DOI: 10.3389/fmicb.2023.1266977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/13/2023] [Indexed: 12/07/2023] Open
Abstract
Introduction The present study investigated the effects of bilberry anthocyanin (BA) on immune function when alleviating Salmonella Typhimurium (S. Typhimurium) infection in chickens. Methods A total of 180 newly hatched yellow-feathered male chicks were assigned to three groups (CON, SI, and SI + BA). Birds in CON and SI were fed a basal diet, and those in SI + BA were supplemented with 100 mg/kg BA for 18 days. Birds in SI and SI + BA received 0.5 ml suspension of S. Typhimurium (2 × 109 CFU/ml) by oral gavage at 14 and 16 days of age, and those in CON received equal volumes of sterile PBS. Results At day 18, (1) dietary BA alleviated weight loss of chickens caused by S. Typhimurium infection (P < 0.01). (2) Supplementation with BA reduced the relative weight of the bursa of Fabricius (P < 0.01) and jejunal villus height (P < 0.05) and increased the number of goblet cells (P < 0.01) and the expression of MUC2 (P < 0.05) in jejunal mucosa, compared with birds in SI. (3) Supplementation with BA decreased (P < 0.05) the concentration of immunoglobulins and cytokines in plasma (IgA, IL-1β, IL-8, and IFN-β) and jejunal mucosa (IgG, IgM, sIgA, IL-1β, IL-6, IL-8, TNF-α, IFN-β, and IFN-γ) of S. Typhimurium-infected chickens. (4) BA regulated a variety of biological processes, especially the defense response to bacteria and humoral immune response, and suppressed cytokine-cytokine receptor interaction and intestinal immune network for IgA production pathways by downregulating 6 immune-related proteins. Conclusion In summary, the impaired growth performance and disruption of jejunal morphology caused by S. Typhimurium were alleviated by dietary BA by affecting the expression of immune-related genes and proteins, and signaling pathways are related to immune response associated with immune cytokine receptors and production in jejunum.
Collapse
Affiliation(s)
- Sheng Zhang
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Qin Wang
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Jinling Ye
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Qiuli Fan
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Xiajing Lin
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Zhongyong Gou
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Mahmoud M. Azzam
- Department of Animal Production College of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Yibing Wang
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Shouqun Jiang
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| |
Collapse
|
27
|
He J, Nascakova Z, Leary P, Papa G, Valenta T, Basler K, Müller A. Inactivation of the tumor suppressor gene Apc synergizes with H. pylori to induce DNA damage in murine gastric stem and progenitor cells. SCIENCE ADVANCES 2023; 9:eadh0322. [PMID: 37967175 PMCID: PMC10651120 DOI: 10.1126/sciadv.adh0322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 10/16/2023] [Indexed: 11/17/2023]
Abstract
Helicobacter pylori infection is a major risk factor for the development of gastric cancer. The bacteria reside in close proximity to gastric surface mucous as well as stem and progenitor cells. Here, we take advantage of wild-type and genetically engineered murine gastric organoids and organoid-derived monolayers to study the cellular targets of H. pylori-induced DNA damage and replication stress and to explore possible interactions with preexisting gastric cancer driver mutations. We find using alkaline comet assay, single-molecule DNA fiber assays, and immunofluorescence microscopy of DNA repair foci that H. pylori induces transcription-dependent DNA damage in actively replicating, Leucine-rich-repeat containing G-Protein-Coupled Receptor 5 (Lgr5)-positive antral stem and progenitor cells and their Troy-positive corpus counterparts, but not in other gastric epithelial lineages. Infection-dependent DNA damage is aggravated by Apc inactivation, but not by Trp53 or Smad4 loss, or Erbb2 overexpression. Our data suggest that H. pylori induces DNA damage in stem and progenitor cells, especially in settings of hyperproliferation due to constitutively active Wnt signaling.
Collapse
Affiliation(s)
- Jiazhuo He
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Zuzana Nascakova
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Peter Leary
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
- Functional Genomics Center Zürich, University of Zürich/ETHZ, Zürich, Switzerland
| | - Giovanni Papa
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Tomas Valenta
- Department of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Konrad Basler
- Department of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- Comprehensive Cancer Center Zürich, Zürich, Switzerland
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
- Comprehensive Cancer Center Zürich, Zürich, Switzerland
| |
Collapse
|
28
|
Lou S, Wang J, Chen J, Xie H, Chen H, Zhou B, Zhang B, Hou J, Jiang DK. The Role of ALPK1 in Inhibiting Hepatitis B Virus Replication Facilitates the Identification of ALPK1 P660L Variant for Predicting Response to Pegylated Interferon α Therapy. J Infect Dis 2023; 228:694-703. [PMID: 36932045 DOI: 10.1093/infdis/jiad065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/06/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Alpha kinase 1 (ALPK1) agonist has recently been reported to demonstrate anti-hepatitis B virus (HBV) efficacy via activating NF-κB signaling, which is crucial for maximizing interferon (IFN) responses. Here, we investigated the impact of ALPK1 on HBV replication and explored ALPK1 variants for predicting the response to pegylated IFN-α (PegIFN-α) treatment. METHODS The potential anti-HBV effect of ALPK1 was evaluated in HBV-integrated and HBV-infected hepatoma cells. The potentially functional genetic variants of ALPK1 were screened out, and their correlations with PegIFN-α treatment response were assessed in 945 hepatitis B e antigen (HBeAg)-positive patients with chronic hepatitis B (CHB). RESULTS We revealed that ALPK1 inhibited HBV replication in hepatocytes via activating the JAK-STAT pathway. ALPK1 overexpression improved the anti-HBV effect of IFN-α in cell models. A missense variant, rs35389530 (P660L), of ALPK1 was strongly associated with combined response (CR; namely, HBeAg seroconversion and HBV DNA level <3.3log10 IU/mL) to PegIFN-α treatment in patients with CHB (P = 2.12 × 10-6). Moreover, a polygenic score integrating ALPK1_rs35389530 and 2 additional genetic variants was further significantly associated with CR (Ptrend = 9.28 × 10-7), hepatitis B surface antigen (HBsAg) level (Ptrend = .0002), and HBsAg loss (Ptrend = .025). CONCLUSIONS The anti-HBV effects of ALPK1 through activating JAK-STAT pathway provides a new perspective for CHB therapy. ALPK1_rs35389530 and polygenic score are potential biomarkers to predict PegIFN-α treatment response and may be used for optimizing CHB treatment.
Collapse
Affiliation(s)
- Shuang Lou
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Institute of Liver Diseases, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The Key Laboratory of Molecular Pathology (Hepatic Diseases) of Guangxi, Department of Pathology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Jialin Wang
- Food Safety and Health Research Center, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jiaxuan Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Institute of Liver Diseases, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haisheng Xie
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Institute of Liver Diseases, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haitao Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Institute of Liver Diseases, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Bin Zhou
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Institute of Liver Diseases, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bo Zhang
- Food Safety and Health Research Center, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jinlin Hou
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Institute of Liver Diseases, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - De-Ke Jiang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Institute of Liver Diseases, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The Key Laboratory of Molecular Pathology (Hepatic Diseases) of Guangxi, Department of Pathology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| |
Collapse
|
29
|
Sharafutdinov I, Tegtmeyer N, Linz B, Rohde M, Vieth M, Tay ACY, Lamichhane B, Tuan VP, Fauzia KA, Sticht H, Yamaoka Y, Marshall BJ, Backert S. A single-nucleotide polymorphism in Helicobacter pylori promotes gastric cancer development. Cell Host Microbe 2023; 31:1345-1358.e6. [PMID: 37490912 DOI: 10.1016/j.chom.2023.06.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/23/2023] [Accepted: 06/27/2023] [Indexed: 07/27/2023]
Abstract
Single-nucleotide polymorphisms (SNPs) in various human genes are key factors in carcinogenesis. However, whether SNPs in bacterial pathogens are similarly crucial in cancer development is unknown. Here, we analyzed 1,043 genomes of the stomach pathogen Helicobacter pylori and pinpointed a SNP in the serine protease HtrA (position serine/leucine 171) that significantly correlates with gastric cancer. Our functional studies reveal that the 171S-to-171L mutation triggers HtrA trimer formation and enhances proteolytic activity and cleavage of epithelial junction proteins occludin and tumor-suppressor E-cadherin. 171L-type HtrA, but not 171S-HtrA-possessing H. pylori, inflicts severe epithelial damage, enhances injection of oncoprotein CagA into epithelial cells, increases NF-κB-mediated inflammation and cell proliferation through nuclear accumulation of β-catenin, and promotes host DNA double-strand breaks, collectively triggering malignant changes. These findings highlight the 171S/L HtrA mutation as a unique bacterial cancer-associated SNP and as a potential biomarker for risk predictions in H. pylori infections.
Collapse
Affiliation(s)
- Irshad Sharafutdinov
- Division of Microbiology, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Nicole Tegtmeyer
- Division of Microbiology, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Bodo Linz
- Division of Microbiology, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Michael Vieth
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Klinikum Bayreuth, 95445 Bayreuth, Germany
| | - Alfred Chin-Yen Tay
- Helicobacter Research Laboratory, Marshall Centre for Infectious Diseases Research and Training, School of Pathology and Laboratory Medicine, University of Western Australia, 6009 Perth, Australia
| | - Binit Lamichhane
- Helicobacter Research Laboratory, Marshall Centre for Infectious Diseases Research and Training, School of Pathology and Laboratory Medicine, University of Western Australia, 6009 Perth, Australia
| | - Vo Phuoc Tuan
- Department of Endoscopy, Choray Hospital, Ho Chi Minh, Vietnam; Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Kartika Afrida Fauzia
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu, Oita, Japan; Department of Public Health and Preventive Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Heinrich Sticht
- Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu, Oita, Japan; Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, Houston, TX 77030, USA
| | - Barry J Marshall
- Helicobacter Research Laboratory, Marshall Centre for Infectious Diseases Research and Training, School of Pathology and Laboratory Medicine, University of Western Australia, 6009 Perth, Australia; University of Western Australia, Marshall Centre, M504, Crawley, WA, Australia; Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, China
| | - Steffen Backert
- Division of Microbiology, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany.
| |
Collapse
|
30
|
Abstract
The versatile type IV secretion system (T4SS) nanomachine plays a pivotal role in bacterial pathogenesis and the propagation of antibiotic resistance determinants throughout microbial populations. In addition to paradigmatic DNA conjugation machineries, diverse T4SSs enable the delivery of multifarious effector proteins to target prokaryotic and eukaryotic cells, mediate DNA export and uptake from the extracellular milieu, and in rare examples, facilitate transkingdom DNA translocation. Recent advances have identified new mechanisms underlying unilateral nucleic acid transport through the T4SS apparatus, highlighting both functional plasticity and evolutionary adaptations that enable novel capabilities. In this review, we describe the molecular mechanisms underscoring DNA translocation through diverse T4SS machineries, emphasizing the architectural features that implement DNA exchange across the bacterial membrane and license transverse DNA release across kingdom boundaries. We further detail how recent studies have addressed outstanding questions surrounding the mechanisms by which nanomachine architectures and substrate recruitment strategies contribute to T4SS functional diversity.
Collapse
Affiliation(s)
- Mackenzie E. Ryan
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Prashant P. Damke
- Department of Veterinary Sciences, University of Kentucky College of Agriculture, Lexington, Kentucky, USA
| | - Carrie L. Shaffer
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
- Department of Veterinary Sciences, University of Kentucky College of Agriculture, Lexington, Kentucky, USA
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| |
Collapse
|
31
|
Faass L, Hauke M, Stein SC, Josenhans C. Innate activation of human neutrophils and neutrophil-like cells by the pro-inflammatory bacterial metabolite ADP-heptose and Helicobacter pylori. Int J Med Microbiol 2023; 313:151585. [PMID: 37399704 DOI: 10.1016/j.ijmm.2023.151585] [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: 03/22/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/05/2023] Open
Abstract
Lipopolysaccharide inner core heptose metabolites, including ADP-heptose, play a substantial role in the activation of cell-autonomous innate immune responses in eukaryotic cells, via the ALPK1-TIFA signaling pathway, as demonstrated for various pathogenic bacteria. The important role of LPS heptose metabolites during Helicobacter pylori infection of the human gastric niche has been demonstrated for gastric epithelial cells and macrophages, while the role of heptose metabolites on human neutrophils has not been investigated. In this study, we aimed to gain a better understanding of the activation potential of bacterial heptose metabolites for human neutrophil cells. To do so, we used pure ADP-heptose and, as a bacterial model, H. pylori, which can transport heptose metabolites into the human host cell via the Cag Type 4 Secretion System (CagT4SS). Main questions were how bacterial heptose metabolites impact on the pro-inflammatory activation, alone and in the bacterial context, and how they influence maturation of human neutrophils. Results of the present study demonstrated that neutrophils respond with high sensitivity to pure heptose metabolites, and that global regulation networks and neutrophil maturation are influenced by heptose exposure. Furthermore, activation of human neutrophils by live H. pylori is strongly impacted by the presence of LPS heptose metabolites and the functionality of its CagT4SS. Similar activities were determined in cell culture neutrophils of different maturation states and in human primary neutrophils. In conclusion, we demonstrated that specific heptose metabolites or bacteria producing heptoses exhibit a strong activity on cell-autonomous innate responses of human neutrophils.
Collapse
Affiliation(s)
- Larissa Faass
- Max von Pettenkofer Institute for Medical Microbiology and Hospital Epidemiology, Ludwig Maximilians-University München, Pettenkoferstrasse 9a, 80336 München, Germany
| | - Martina Hauke
- Max von Pettenkofer Institute for Medical Microbiology and Hospital Epidemiology, Ludwig Maximilians-University München, Pettenkoferstrasse 9a, 80336 München, Germany
| | - Saskia C Stein
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Christine Josenhans
- Max von Pettenkofer Institute for Medical Microbiology and Hospital Epidemiology, Ludwig Maximilians-University München, Pettenkoferstrasse 9a, 80336 München, Germany; DZIF Partner Site Munich, Germany.
| |
Collapse
|
32
|
Hauke M, Metz F, Rapp J, Faass L, Bats SH, Radziej S, Link H, Eisenreich W, Josenhans C. Helicobacter pylori Modulates Heptose Metabolite Biosynthesis and Heptose-Dependent Innate Immune Host Cell Activation by Multiple Mechanisms. Microbiol Spectr 2023; 11:e0313222. [PMID: 37129481 PMCID: PMC10269868 DOI: 10.1128/spectrum.03132-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 04/05/2023] [Indexed: 05/03/2023] Open
Abstract
Heptose metabolites including ADP-d-glycero-β-d-manno-heptose (ADP-heptose) are involved in bacterial lipopolysaccharide and cell envelope biosynthesis. Recently, heptoses were also identified to have potent proinflammatory activity on human cells as novel microbe-associated molecular patterns. The gastric pathogenic bacterium Helicobacter pylori produces heptose metabolites, which it transports into human cells through its Cag type 4 secretion system. Using H. pylori as a model, we have addressed the question of how proinflammatory ADP-heptose biosynthesis can be regulated by bacteria. We have characterized the interstrain variability and regulation of heptose biosynthesis genes and the modulation of heptose metabolite production by H. pylori, which impact cell-autonomous proinflammatory human cell activation. HldE, a central enzyme of heptose metabolite biosynthesis, showed strong sequence variability between strains and was also variably expressed between strains. Amounts of gene transcripts in the hldE gene cluster displayed intrastrain and interstrain differences, were modulated by host cell contact and the presence of the cag pathogenicity island, and were affected by carbon starvation regulator A (CsrA). We reconstituted four steps of the H. pylori lipopolysaccharide (LPS) heptose biosynthetic pathway in vitro using recombinant purified GmhA, HldE, and GmhB proteins. On the basis of one- and two-dimensional nuclear magnetic resonance (NMR) spectroscopy and mass spectrometry, the structures of major reaction products were identified as β-d-ADP-heptose and β-heptose-1-monophosphate. A proinflammatory heptose-monophosphate variant was also identified for the first time as a novel cell-active product in H. pylori bacteria. Separate purified HldE subdomains and variant HldE allowed us to uncover additional strain variation in generating heptose metabolites. IMPORTANCE Bacterial heptose metabolites, intermediates of lipopolysaccharide (LPS) biosynthesis, are novel microbe-associated molecular patterns (MAMPs) that activate proinflammatory signaling. In the gastric pathogen Helicobacter pylori, heptoses are transferred into host cells by the Cag type IV secretion system, which is also involved in carcinogenesis. Little is known about how H. pylori, which is highly strain variable, regulates heptose biosynthesis and downstream host cell activation. We report here that the regulation of proinflammatory heptose production by H. pylori is strain specific. Heptose gene cluster activity is modulated by the presence of an active cag pathogenicity island (cagPAI), contact with human cells, and the carbon starvation regulator A. Reconstitution with purified biosynthesis enzymes and purified bacterial lysates allowed us to biochemically characterize heptose pathway products, identifying a heptose-monophosphate variant as a novel proinflammatory metabolite. These findings emphasize that the bacteria use heptose biosynthesis to fine-tune inflammation and also highlight opportunities to mine the heptose biosynthesis pathway as a potential therapeutic target against infection, inflammation, and cancer.
Collapse
Affiliation(s)
- Martina Hauke
- Max von Pettenkofer Institute, Ludwig Maximilians University Munich, München, Germany
| | - Felix Metz
- Max von Pettenkofer Institute, Ludwig Maximilians University Munich, München, Germany
| | - Johanna Rapp
- Bacterial Metabolomics, CMFI, University Tübingen, Tübingen, Germany
| | - Larissa Faass
- Max von Pettenkofer Institute, Ludwig Maximilians University Munich, München, Germany
| | - Simon H. Bats
- Max von Pettenkofer Institute, Ludwig Maximilians University Munich, München, Germany
| | - Sandra Radziej
- Bavarian NMR Center–Structural Membrane Biochemistry, Department of Chemistry, Technical University Munich, Garching, Germany
| | - Hannes Link
- Bacterial Metabolomics, CMFI, University Tübingen, Tübingen, Germany
| | - Wolfgang Eisenreich
- Bavarian NMR Center–Structural Membrane Biochemistry, Department of Chemistry, Technical University Munich, Garching, Germany
| | - Christine Josenhans
- Max von Pettenkofer Institute, Ludwig Maximilians University Munich, München, Germany
| |
Collapse
|
33
|
Sidor K, Skirecki T. A Bittersweet Kiss of Gram-Negative Bacteria: The Role of ADP-Heptose in the Pathogenesis of Infection. Microorganisms 2023; 11:1316. [PMID: 37317291 DOI: 10.3390/microorganisms11051316] [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] [Received: 02/27/2023] [Revised: 05/10/2023] [Accepted: 05/15/2023] [Indexed: 06/16/2023] Open
Abstract
Due to the global crisis caused by the dramatic rise of drug resistance among Gram-negative bacteria, there is an urgent need for a thorough understanding of the pathogenesis of infections of such an etiology. In light of the limited availability of new antibiotics, therapies aimed at host-pathogen interactions emerge as potential treatment modalities. Thus, understanding the mechanism of pathogen recognition by the host and immune evasion appear to be the key scientific issues. Until recently, lipopolysaccharide (LPS) was recognized as a major pathogen-associated molecular pattern (PAMP) of Gram-negative bacteria. However, recently, ADP-L-glycero-β-D-manno-heptose (ADP-heptose), an intermediate carbohydrate metabolite of the LPS biosynthesis pathway, was discovered to activate the hosts' innate immunity. Therefore, ADP-heptose is regarded as a novel PAMP of Gram-negative bacteria that is recognized by the cytosolic alpha kinase-1 (ALPK1) protein. The conservative nature of this molecule makes it an intriguing player in host-pathogen interactions, especially in the context of changes in LPS structure or even in its loss by certain resistant pathogens. Here, we present the ADP-heptose metabolism, outline the mechanisms of its recognition and the activation of its immunity, and summarize the role of ADP-heptose in the pathogenesis of infection. Finally, we hypothesize about the routes of the entry of this sugar into cytosol and point to emerging questions that require further research.
Collapse
Affiliation(s)
- Karolina Sidor
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Tomasz Skirecki
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| |
Collapse
|
34
|
Wu H, Sun Q, Dong H, Qiao J, Lin Y, Yu C, Li Y. Gastroprotective action of the extract of Corydalis yanhusuo in Helicobacter pylori infection and its bioactive component, dehydrocorydaline. JOURNAL OF ETHNOPHARMACOLOGY 2023; 307:116173. [PMID: 36681166 DOI: 10.1016/j.jep.2023.116173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/01/2023] [Accepted: 01/13/2023] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Helicobacter pylori (H. pylori) infection is a frequent chronic infection. Persistent infection is the strongest risk factor for developing gastric complications leading to gastric cancer. The antibiotic resistance of current anti-H. pylori drugs lead to the search for novel candidates from medicinal plants. Traditionally, Corydalis yanhusuo (Y.H. Chou & Chun C.Hsu) W.T. Wang ex Z.Y. Su & C.Y. Wu (Papaveraceae) has been used for the treatment of digestive system diseases in China. So, it's essential to explore and confirm the anti-H. pylori activity of C. yanhusuo and characterize the pharmacologically active compounds. AIM OF THE STUDY This study aims to evaluate the efficacy of C. yanhusuo as complementary or alternative modes of treatment against H. pylori-related diseases and ascertain the active substances of C. yanhusuo to develop non-toxic, natural, and inexpensive products. MATERIALS AND METHODS C. yanhusuo was subjected to solid-liquid extraction with water (WECY), ethanol EECY), and chloroform (CECY). The extracts were screened by agar diffusion assay, the minimum inhibitory concentrations (MIC), the minimum bactericidal (MBC) for their in vitro antimicrobial activity, and by Berthelot reaction for urease inhibition. To assess the in vivo action, H. pylori-induced C57BL/6 mice were used to detect RUT biopsy, perform visual and histopathological analyses and evaluate IgG expression. Furthermore, we compared the anti-H. pylori activities of major alkaloids in CECY to identify the bioactive constituents. RESULTS Among the three C. yanhusuo extracts, CECY showed the maximum in vitro antibacterial activity. Administration of CECY significantly inhibited the survival of H. pylori colonized in the gastric mucosa and alleviated gastric damage along with a reduction in the expression levels of IgG in H. pylori-infected mice. Berberine and dehydrocorydaline exhibited obvious anti-H. pylori activity with MIC of 25 and 12.5 μg/mL, respectively. CONCLUSION C. yanhusuo extracts showed anti-H. pylori activity in different degrees. Among them, CECY showed significant anti-H. pylori, gastroprotective and anti-inflammatory activities in vivo and in vitro. Dehydrocorydalmine, an active alkaloid compound isolated from C. yanhusuo, warranted further investigation for its potential anti-H. pylori activity.
Collapse
Affiliation(s)
- Hao Wu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Qingyue Sun
- No. one Clinical Medicine School of Binzhou Medical University, Binzhou Medical University, Yantai, 264003, China
| | - Huirong Dong
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Jiasen Qiao
- No. one Clinical Medicine School of Binzhou Medical University, Binzhou Medical University, Yantai, 264003, China
| | - Ying Lin
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Chen Yu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| | - Yanni Li
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
35
|
Pani G. Fusobacterium & Co. at the Stem of Cancer: Microbe-Cancer Stem Cell Interactions in Colorectal Carcinogenesis. Cancers (Basel) 2023; 15:cancers15092583. [PMID: 37174049 PMCID: PMC10177588 DOI: 10.3390/cancers15092583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 04/27/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Adult stem cells lie at the crossroads of tissue repair, inflammation, and malignancy. Intestinal microbiota and microbe-host interactions are pivotal to maintaining gut homeostasis and response to injury, and participate in colorectal carcinogenesis. Yet, limited knowledge is available on whether and how bacteria directly crosstalk with intestinal stem cells (ISC), particularly cancerous stem-like cells (CR-CSC), as engines for colorectal cancer initiation, maintenance, and metastatic dissemination. Among several bacterial species alleged to initiate or promote colorectal cancer (CRC), the pathobiont Fusobacterium Nucleatum has recently drawn significant attention for its epidemiologic association and mechanistic linkage with the disease. We will therefore focus on current evidence for an F. nucleatum-CRCSC axis in tumor development, highlighting the commonalities and differences between F. nucleatum-associated colorectal carcinogenesis and gastric cancer driven by Helicobacter Pylori. We will explore the diverse facets of the bacteria-CSC interaction, analyzing the signals and pathways whereby bacteria either confer "stemness" properties to tumor cells or primarily target stem-like elements within the heterogeneous tumor cell populations. We will also discuss the extent to which CR-CSC cells are competent for innate immune responses and participate in establishing a tumor-promoting microenvironment. Finally, by capitalizing on the expanding knowledge of how the microbiota and ISC crosstalk in intestinal homeostasis and response to injury, we will speculate on the possibility that CRC arises as an aberrant repair response promoted by pathogenic bacteria upon direct stimulation of intestinal stem cells.
Collapse
Affiliation(s)
- Giovambattista Pani
- Department of Translational Medicine and Surgery, Section of General Pathology, Faculty of Medicine, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, L. go A. Gemelli 8, 00168 Rome, Italy
| |
Collapse
|
36
|
Duizer C, de Zoete MR. The Role of Microbiota-Derived Metabolites in Colorectal Cancer. Int J Mol Sci 2023; 24:8024. [PMID: 37175726 PMCID: PMC10178193 DOI: 10.3390/ijms24098024] [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: 03/19/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
The impact of bacterial members of the microbiota on the development of colorectal cancer (CRC) has become clear in recent years. However, exactly how bacteria contribute to the development of cancer is often still up for debate. The impact of bacteria-derived metabolites, which can influence the development of CRC either in a promoting or inhibiting manner, is undeniable. Here, we discuss the effects of the most well-studied bacteria-derived metabolites associated with CRC, including secondary bile acids, short-chain fatty acids, trimethylamine-N-oxide and indoles. We show that the effects of individual metabolites on CRC development are often nuanced and dose- and location-dependent. In the coming years, the array of metabolites involved in CRC development will undoubtedly increase further, which will emphasize the need to focus on causation and mechanisms and the clearly defined roles of bacterial species within the microbiota.
Collapse
Affiliation(s)
| | - Marcel R. de Zoete
- Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
37
|
Beccaceci G, Sigal M. Unwelcome guests - the role of gland-associated Helicobacter pylori infection in gastric carcinogenesis. Front Oncol 2023; 13:1171003. [PMID: 37152042 PMCID: PMC10160455 DOI: 10.3389/fonc.2023.1171003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/04/2023] [Indexed: 05/09/2023] Open
Abstract
Helicobacter pylori (H. pylori) are Gram-negative bacteria that cause chronic gastritis and are considered the main risk factor for the development of gastric cancer. H. pylori have evolved to survive the harsh luminal environment of the stomach and are known to cause damage and signaling aberrations in gastric epithelial cells, which can result in premalignant and malignant pathology. As well as colonizing the gastric mucus and surface epithelial cells, a subpopulation of H. pylori can invade deep into the gastric glands and directly interact with progenitor and stem cells. Gland colonization therefore bears the potential to cause direct injury to long-lived cells. Moreover, this bacterial subpopulation triggers a series of host responses that cause an enhanced proliferation of stem cells. Here, we review recent insights into how gastric gland colonization by H. pylori is established, the resulting pro-carcinogenic epithelial signaling alterations, as well as new insights into stem cell responses to infection. Together these point towards a critical role of gland-associated H. pylori in the development of gastric cancer.
Collapse
Affiliation(s)
- Giulia Beccaceci
- Medical Department, Division of Gastroenterology and Hepatology, Charité-Universtitätsmedizin Berlin, Berlin, Germany
- The Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Michael Sigal
- Medical Department, Division of Gastroenterology and Hepatology, Charité-Universtitätsmedizin Berlin, Berlin, Germany
- The Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
38
|
Malfertheiner P, Camargo MC, El-Omar E, Liou JM, Peek R, Schulz C, Smith SI, Suerbaum S. Helicobacter pylori infection. Nat Rev Dis Primers 2023; 9:19. [PMID: 37081005 PMCID: PMC11558793 DOI: 10.1038/s41572-023-00431-8] [Citation(s) in RCA: 314] [Impact Index Per Article: 157.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2023] [Indexed: 04/22/2023]
Abstract
Helicobacter pylori infection causes chronic gastritis, which can progress to severe gastroduodenal pathologies, including peptic ulcer, gastric cancer and gastric mucosa-associated lymphoid tissue lymphoma. H. pylori is usually transmitted in childhood and persists for life if untreated. The infection affects around half of the population in the world but prevalence varies according to location and sanitation standards. H. pylori has unique properties to colonize gastric epithelium in an acidic environment. The pathophysiology of H. pylori infection is dependent on complex bacterial virulence mechanisms and their interaction with the host immune system and environmental factors, resulting in distinct gastritis phenotypes that determine possible progression to different gastroduodenal pathologies. The causative role of H. pylori infection in gastric cancer development presents the opportunity for preventive screen-and-treat strategies. Invasive, endoscopy-based and non-invasive methods, including breath, stool and serological tests, are used in the diagnosis of H. pylori infection. Their use depends on the specific individual patient history and local availability. H. pylori treatment consists of a strong acid suppressant in various combinations with antibiotics and/or bismuth. The dramatic increase in resistance to key antibiotics used in H. pylori eradication demands antibiotic susceptibility testing, surveillance of resistance and antibiotic stewardship.
Collapse
Affiliation(s)
- Peter Malfertheiner
- Medical Department II, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany.
- Medical Department Klinik of Gastroenterology, Hepatology and Infectiology, Otto-von-Guericke Universität, Magdeburg, Germany.
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Emad El-Omar
- Microbiome Research Centre, St George & Sutherland Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Jyh-Ming Liou
- Department of Internal Medicine, National Taiwan University Cancer Center, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Richard Peek
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christian Schulz
- Medical Department II, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
- DZIF Deutsches Zentrum für Infektionsforschung, Partner Site Munich, Munich, Germany
| | - Stella I Smith
- Department of Molecular Biology and Biotechnology, Nigerian Institute of Medical Research, Yaba, Lagos, Nigeria
| | - Sebastian Suerbaum
- DZIF Deutsches Zentrum für Infektionsforschung, Partner Site Munich, Munich, Germany
- Max von Pettenkofer Institute, Faculty of Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- National Reference Center for Helicobacter pylori, Munich, Germany
| |
Collapse
|
39
|
Liu Y, Niu L, Li N, Wang Y, Liu M, Su X, Bao X, Yin B, Shen S. Bacterial-Mediated Tumor Therapy: Old Treatment in a New Context. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205641. [PMID: 36908053 PMCID: PMC10131876 DOI: 10.1002/advs.202205641] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/09/2023] [Indexed: 06/18/2023]
Abstract
Targeted therapy and immunotherapy have brought hopes for precision cancer treatment. However, complex physiological barriers and tumor immunosuppression result in poor efficacy, side effects, and resistance to antitumor therapies. Bacteria-mediated antitumor therapy provides new options to address these challenges. Thanks to their special characteristics, bacteria have excellent ability to destroy tumor cells from the inside and induce innate and adaptive antitumor immune responses. Furthermore, bacterial components, including bacterial vesicles, spores, toxins, metabolites, and other active substances, similarly inherit their unique targeting properties and antitumor capabilities. Bacteria and their accessory products can even be reprogrammed to produce and deliver antitumor agents according to clinical needs. This review first discusses the role of different bacteria in the development of tumorigenesis and the latest advances in bacteria-based delivery platforms and the existing obstacles for application. Moreover, the prospect and challenges of clinical transformation of engineered bacteria are also summarized.
Collapse
Affiliation(s)
- Yao Liu
- Key Laboratory of Spine and Spinal Cord Injury Repairand Regeneration of Ministry of EducationOrthopaedic Department of Tongji Hospital, The Institute for Biomedical Engineering and Nano ScienceTongji University School of MedicineShanghai200092P. R. China
- Pharmacy Department and Center for Medical Research and InnovationShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Lili Niu
- Central LaboratoryFirst Affiliated HospitalInstitute (College) of Integrative MedicineDalian Medical UniversityDalian116021China
| | - Nannan Li
- Central LaboratoryFirst Affiliated HospitalInstitute (College) of Integrative MedicineDalian Medical UniversityDalian116021China
| | - Yang Wang
- Central LaboratoryFirst Affiliated HospitalInstitute (College) of Integrative MedicineDalian Medical UniversityDalian116021China
| | - Mingyang Liu
- Department of Surgical Oncology and General SurgeryThe First Hospital of China Medical University155 North Nanjing Street, Heping DistrictShenyang110001China
| | - Xiaomin Su
- Central LaboratoryFirst Affiliated HospitalInstitute (College) of Integrative MedicineDalian Medical UniversityDalian116021China
| | - Xuhui Bao
- Institute for Therapeutic Cancer VaccinesFudan University Pudong Medical CenterShanghai201399China
| | - Bo Yin
- Institute for Therapeutic Cancer Vaccines and Department of OncologyFudan University Pudong Medical CenterShanghai201399China
| | - Shun Shen
- Pharmacy Department and Center for Medical Research and InnovationShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| |
Collapse
|
40
|
Lopes C, Almeida TC, Pimentel-Nunes P, Dinis-Ribeiro M, Pereira C. Linking dysbiosis to precancerous stomach through inflammation: Deeper than and beyond imaging. Front Immunol 2023; 14:1134785. [PMID: 37063848 PMCID: PMC10102473 DOI: 10.3389/fimmu.2023.1134785] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/17/2023] [Indexed: 04/03/2023] Open
Abstract
Upper gastrointestinal endoscopy is considered the gold standard for gastric lesions detection and surveillance, but it is still associated with a non-negligible rate of missing conditions. In the Era of Personalized Medicine, biomarkers could be the key to overcome missed lesions or to better predict recurrence, pushing the frontier of endoscopy to functional endoscopy. In the last decade, microbiota in gastric cancer has been extensively explored, with gastric carcinogenesis being associated with progressive dysbiosis. Helicobacter pylori infection has been considered the main causative agent of gastritis due to its interference in disrupting the acidic environment of the stomach through inflammatory mediators. Thus, does inflammation bridge the gap between gastric dysbiosis and the gastric carcinogenesis cascade and could the microbiota-inflammation axis-derived biomarkers be the answer to the unmet challenge of functional upper endoscopy? To address this question, in this review, the available evidence on the role of gastric dysbiosis and chronic inflammation in precancerous conditions of the stomach is summarized, particularly targeting the nuclear factor-κB (NF-κB), toll-like receptors (TLRs) and cyclooxygenase-2 (COX-2) pathways. Additionally, the potential of liquid biopsies as a non-invasive source and the clinical utility of studied biomarkers is also explored. Overall, and although most studies offer a mechanistic perspective linking a strong proinflammatory Th1 cell response associated with, but not limited to, chronic infection with Helicobacter pylori, promising data recently published highlights not only the diagnostic value of microbial biomarkers but also the potential of gastric juice as a liquid biopsy pushing forward the concept of functional endoscopy and personalized care in gastric cancer early diagnosis and surveillance.
Collapse
Affiliation(s)
- Catarina Lopes
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI‐IPOP)/Rise@CI‐IPOP (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- CINTESIS – Center for Health Technology and Services Research, University of Porto, Porto, Portugal
- ICBAS-UP – Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Tatiana C. Almeida
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI‐IPOP)/Rise@CI‐IPOP (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Pedro Pimentel-Nunes
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto (FMUP), Porto, Portugal
- Department of Gastroenterology, Unilabs, Porto, Portugal
| | - Mário Dinis-Ribeiro
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI‐IPOP)/Rise@CI‐IPOP (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- Department of Gastroenterology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Carina Pereira
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI‐IPOP)/Rise@CI‐IPOP (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- CINTESIS – Center for Health Technology and Services Research, University of Porto, Porto, Portugal
- *Correspondence: Carina Pereira,
| |
Collapse
|
41
|
Affiliation(s)
- Anne Müller
- From the Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Jiazhuo He
- From the Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| |
Collapse
|
42
|
Usui Y, Taniyama Y, Endo M, Koyanagi YN, Kasugai Y, Oze I, Ito H, Imoto I, Tanaka T, Tajika M, Niwa Y, Iwasaki Y, Aoi T, Hakozaki N, Takata S, Suzuki K, Terao C, Hatakeyama M, Hirata M, Sugano K, Yoshida T, Kamatani Y, Nakagawa H, Matsuda K, Murakami Y, Spurdle AB, Matsuo K, Momozawa Y. Helicobacter pylori, Homologous-Recombination Genes, and Gastric Cancer. N Engl J Med 2023; 388:1181-1190. [PMID: 36988593 DOI: 10.1056/nejmoa2211807] [Citation(s) in RCA: 112] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
BACKGROUND Helicobacter pylori infection is a well-known risk factor for gastric cancer. However, the contribution of germline pathogenic variants in cancer-predisposing genes and their effect, when combined with H. pylori infection, on the risk of gastric cancer has not been widely evaluated. METHODS We evaluated the association between germline pathogenic variants in 27 cancer-predisposing genes and the risk of gastric cancer in a sample of 10,426 patients with gastric cancer and 38,153 controls from BioBank Japan. We also assessed the combined effect of pathogenic variants and H. pylori infection status on the risk of gastric cancer and calculated the cumulative risk in 1433 patients with gastric cancer and 5997 controls from the Hospital-based Epidemiologic Research Program at Aichi Cancer Center (HERPACC). RESULTS Germline pathogenic variants in nine genes (APC, ATM, BRCA1, BRCA2, CDH1, MLH1, MSH2, MSH6, and PALB2) were associated with the risk of gastric cancer. We found an interaction between H. pylori infection and pathogenic variants in homologous-recombination genes with respect to the risk of gastric cancer in the sample from HERPACC (relative excess risk due to the interaction, 16.01; 95% confidence interval [CI], 2.22 to 29.81; P = 0.02). At 85 years of age, persons with H. pylori infection and a pathogenic variant had a higher cumulative risk of gastric cancer than noncarriers infected with H. pylori (45.5% [95% CI, 20.7 to 62.6] vs. 14.4% [95% CI, 12.2 to 16.6]). CONCLUSIONS H. pylori infection modified the risk of gastric cancer associated with germline pathogenic variants in homologous-recombination genes. (Funded by the Japan Agency for Medical Research and Development and others.).
Collapse
Affiliation(s)
- Yoshiaki Usui
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Yukari Taniyama
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Mikiko Endo
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Yuriko N Koyanagi
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Yumiko Kasugai
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Isao Oze
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Hidemi Ito
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Issei Imoto
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Tsutomu Tanaka
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Masahiro Tajika
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Yasumasa Niwa
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Yusuke Iwasaki
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Tomomi Aoi
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Nozomi Hakozaki
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Sadaaki Takata
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Kunihiko Suzuki
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Chikashi Terao
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Masanori Hatakeyama
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Makoto Hirata
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Kokichi Sugano
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Teruhiko Yoshida
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Yoichiro Kamatani
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Hidewaki Nakagawa
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Koichi Matsuda
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Yoshinori Murakami
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Amanda B Spurdle
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Keitaro Matsuo
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Yukihide Momozawa
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| |
Collapse
|
43
|
Kemper L, Hensel A. Campylobacter jejuni: targeting host cells, adhesion, invasion, and survival. Appl Microbiol Biotechnol 2023; 107:2725-2754. [PMID: 36941439 PMCID: PMC10027602 DOI: 10.1007/s00253-023-12456-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/23/2023]
Abstract
Campylobacter jejuni, causing strong enteritis, is an unusual bacterium with numerous peculiarities. Chemotactically controlled motility in viscous milieu allows targeted navigation to intestinal mucus and colonization. By phase variation, quorum sensing, extensive O-and N-glycosylation and use of the flagellum as type-3-secretion system C. jejuni adapts effectively to environmental conditions. C. jejuni utilizes proteases to open cell-cell junctions and subsequently transmigrates paracellularly. Fibronectin at the basolateral side of polarized epithelial cells serves as binding site for adhesins CadF and FlpA, leading to intracellular signaling, which again triggers membrane ruffling and reduced host cell migration by focal adhesion. Cell contacts of C. jejuni results in its secretion of invasion antigens, which induce membrane ruffling by paxillin-independent pathway. In addition to fibronectin-binding proteins, other adhesins with other target structures and lectins and their corresponding sugar structures are involved in host-pathogen interaction. Invasion into the intestinal epithelial cell depends on host cell structures. Fibronectin, clathrin, and dynein influence cytoskeletal restructuring, endocytosis, and vesicular transport, through different mechanisms. C. jejuni can persist over a 72-h period in the cell. Campylobacter-containing vacuoles, avoid fusion with lysosomes and enter the perinuclear space via dynein, inducing signaling pathways. Secretion of cytolethal distending toxin directs the cell into programmed cell death, including the pyroptotic release of proinflammatory substances from the destroyed cell compartments. The immune system reacts with an inflammatory cascade by participation of numerous immune cells. The development of autoantibodies, directed not only against lipooligosaccharides, but also against endogenous gangliosides, triggers autoimmune diseases. Lesions of the epithelium result in loss of electrolytes, water, and blood, leading to diarrhea, which flushes out mucus containing C. jejuni. Together with the response of the immune system, this limits infection time. Based on the structural interactions between host cell and bacterium, the numerous virulence mechanisms, signaling, and effects that characterize the infection process of C. jejuni, a wide variety of targets for attenuation of the pathogen can be characterized. The review summarizes strategies of C. jejuni for host-pathogen interaction and should stimulate innovative research towards improved definition of targets for future drug development. KEY POINTS: • Bacterial adhesion of Campylobacter to host cells and invasion into host cells are strictly coordinated processes, which can serve as targets to prevent infection. • Reaction and signalling of host cell depend on the cell type. • Campylobacter virulence factors can be used as targets for development of antivirulence drug compounds.
Collapse
Affiliation(s)
- Leon Kemper
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany
| | - Andreas Hensel
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany.
| |
Collapse
|
44
|
Sokolova O, Maubach G, Naumann M. Helicobacter pylori regulates TIFA turnover in gastric epithelial cells. Eur J Cell Biol 2023; 102:151307. [PMID: 36965415 DOI: 10.1016/j.ejcb.2023.151307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/01/2023] [Accepted: 03/11/2023] [Indexed: 03/19/2023] Open
Abstract
The human pathogen Helicobacter pylori induces a strong inflammatory response in gastric mucosa manifested by the recruitment of neutrophils and macrophages to the places of infection, and by changes in epithelial integrity and function. At the molecular level, this innate immune response is essentially dependent on the activation of NF-κB transcription factors regulating the expression of chemotactic factors, e.g., IL-8. Recently, it has been demonstrated that the NF-κB signaling pathway is triggered by the bacterial heptose metabolites, which activate the host ALPK1-TIFA axis. TIFA has been suggested to promote oligomerization and activity of the E3 ubiquitin ligase TRAF6, which further stimulates TAK1-IKK signaling. Here, we demonstrate that ALPK1-dependent TIFA activation in H. pylori-infected gastric epithelial cells is followed in time by a decline in TIFA levels, and that this process is impeded by inhibitors of the proteasomal and lysosomal degradation. According to our data, TRAF2, TRAF6, TAK1 or NEMO are not required for TIFA degradation. Additionally, H. pylori promotes the interaction of TIFA with free polyubiquitin as well as with optineurin, TAX1BP1 and LAMP1, which are known protein adaptors involved in intracellular trafficking to lysosomes.
Collapse
Affiliation(s)
- Olga Sokolova
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, 39120 Magdeburg, Germany.
| | - Gunter Maubach
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, 39120 Magdeburg, Germany
| |
Collapse
|
45
|
Innate immune activation and modulatory factors of Helicobacter pylori towards phagocytic and nonphagocytic cells. Curr Opin Immunol 2023; 82:102301. [PMID: 36933362 DOI: 10.1016/j.coi.2023.102301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/17/2023] [Indexed: 03/18/2023]
Abstract
Helicobacter pylori is an intriguing obligate host-associated human pathogen with a specific host interaction biology, which has been shaped by thousands of years of host-pathogen coevolution. Molecular mechanisms of interaction of H. pylori with the local immune cells in the human system are less well defined than epithelial cell interactions, although various myeloid cells, including neutrophils and other phagocytes, are locally present or attracted to the sites of infection and interact with H. pylori. We have recently addressed the question of novel bacterial innate immune stimuli, including bacterial cell envelope metabolites, that can activate and modulate cell responses via the H. pylori Cag type IV secretion system. This review article gives an overview of what is currently known about the interaction modes and mechanisms of H. pylori with diverse human cell types, with a focus on bacterial metabolites and cells of the myeloid lineage including phagocytic and antigen-presenting cells.
Collapse
|
46
|
Stoy H, Zwicky K, Kuster D, Lang KS, Krietsch J, Crossley MP, Schmid JA, Cimprich KA, Merrikh H, Lopes M. Direct visualization of transcription-replication conflicts reveals post-replicative DNA:RNA hybrids. Nat Struct Mol Biol 2023; 30:348-359. [PMID: 36864174 PMCID: PMC10023573 DOI: 10.1038/s41594-023-00928-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 01/23/2023] [Indexed: 03/04/2023]
Abstract
Transcription-replication collisions (TRCs) are crucial determinants of genome instability. R-loops were linked to head-on TRCs and proposed to obstruct replication fork progression. The underlying mechanisms, however, remained elusive due to the lack of direct visualization and of non-ambiguous research tools. Here, we ascertained the stability of estrogen-induced R-loops on the human genome, visualized them directly by electron microscopy (EM), and measured R-loop frequency and size at the single-molecule level. Combining EM and immuno-labeling on locus-specific head-on TRCs in bacteria, we observed the frequent accumulation of DNA:RNA hybrids behind replication forks. These post-replicative structures are linked to fork slowing and reversal across conflict regions and are distinct from physiological DNA:RNA hybrids at Okazaki fragments. Comet assays on nascent DNA revealed a marked delay in nascent DNA maturation in multiple conditions previously linked to R-loop accumulation. Altogether, our findings suggest that TRC-associated replication interference entails transactions that follow initial R-loop bypass by the replication fork.
Collapse
Affiliation(s)
- Henriette Stoy
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Katharina Zwicky
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Danina Kuster
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Kevin S Lang
- Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN, USA
| | - Jana Krietsch
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Magdalena P Crossley
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jonas A Schmid
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Karlene A Cimprich
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Houra Merrikh
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Massimo Lopes
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
47
|
He C, Zhang H, Guo Z, Mo Z. A cuproptosis-related signature for predicting the prognosis of gastric cancer. J Gastrointest Oncol 2023; 14:146-164. [PMID: 36915443 PMCID: PMC10007928 DOI: 10.21037/jgo-23-62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/15/2023] [Indexed: 03/03/2023] Open
Abstract
Background Gastric cancer (GC) is one of the most common malignancies. Cuproptosis is a newly discovered type of cell death caused by protein toxicity stress, with copper having considerable importance in GC development. Methods First, differentially expressed (DE) cuproptosis-related genes (CRGs) were screened in GC. The tumor mutation burden (TMB) of CRGs was analyzed. We then performed enrichment analyses of DE-CRGs. Next, we constructed a GC cuproptosis-related (CR) signature (CRs) using Cox and least absolute shrinkage and selection operator (LASSO) regression analyses. The predictive efficacy was assessed using receiver operating characteristic (ROC) curves. Furthermore, we performed gene set enrichment analysis (GSEA). Different methods were used to assess tumor immunity of the CRs, and the Wilcoxon test was used to examine the expressions of m6A-, m7G-, and ferroptosis-related genes. The "pRRophetic" R package (The R Foundation for Statistical Computing) was used to predict the half maximal inhibitory concentration IC50 of common chemotherapeutic agents. Finally, the expression of CRGs in different clusters was analyzed using single-cell RNA sequencing (scRNA-seq). Results We identified 8 DE-CRGs in GC. There were 9 CRGs with TMB values >1%. We constructed gene expression networks and CRs for GC. The DE-CRGs were involved in important mitochondrial metabolic pathways, and the CRs was a valuable independent prognosis factor. The GSEA revealed that angiogenesis and metabolic-related pathways were enriched in the high-risk group, whereas the low-risk group showed enrichment in DNA replication mismatch and repair pathways. The expressions of immunological checkpoints, ferroptosis-, m6A-, and m7G-related genes, type II interferon (INF) response, major histocompatibility complex (MHC class-I), and the IC50 of the copper-based carrier drug elesclomol were significantly different between the 2 groups of the CRs. Furthermore, the scRNA-seq analysis showed that most CRGs were mainly upregulated in endothelial cells. Conclusions The novel CRs could predict the prognosis of GC.
Collapse
Affiliation(s)
- Chunmei He
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, China
| | - Hao Zhang
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, China
| | - Zehao Guo
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, China
| | - Zhijing Mo
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, China
| |
Collapse
|
48
|
Hashimoto N, Kitai R, Fujita S, Yamauchi T, Isozaki M, Kikuta KI. Single-Cell Analysis of Unidirectional Migration of Glioblastoma Cells Using a Fiber-Based Scaffold. ACS APPLIED BIO MATERIALS 2023; 6:765-773. [PMID: 36758146 PMCID: PMC9945112 DOI: 10.1021/acsabm.2c00958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Glioblastoma (GBM) is a malignant incurable brain tumor in which immature neoplastic cells infiltrate brain tissue by spreading along nerve fibers. The aim of the study was to compare the migration abilities of glioma cells with those of other cancer cells and elucidate the migratory profiles underlying the differential migration of glioma cells using a fiber-based quantitative migration assay. Here, wound healing and transwell assays were used to assess cell mobility in four cell lines: U87-MG glioblastoma cells, MDA-MB-231 breast cancer cells, HCT116 colorectal cancer cells, and MKN45 gastric cancer cells. We also assessed cell mobility using a fiber model that mimics nerve fibers. Time-lapse video microscopy was used to observe cell migration and morphology. The cytoskeleton arrangement was assessed in the fiber model and compared with that in the conventional cell culture model. The conventional evaluation of cell migration ability revealed that the migration ability of breast cancer and glioblastoma cell lines was higher than that of colon cancer and gastric cancer cell lines. The fiber model confirmed that the glioblastoma cell line had a significantly higher migration ability than other cell lines. Tubulin levels were significantly higher in the glioblastoma cells than in other cell lines. In conclusion, the developed fiber-based culture model revealed the specific migratory profile of GBM cells during invasion.
Collapse
Affiliation(s)
- Norichika Hashimoto
- Division of Medicine, Department of Neurosurgery, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan.,Department of Neurosurgery, Fukui General Hospital, 58-16-1 Egami-cho, Fukui-shi, Fukui 910-8561, Japan
| | - Ryuhei Kitai
- Division of Medicine, Department of Neurosurgery, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan.,Department of Neurosurgery, Kaga Medical Center, Kaga, Ri 36, Sakumi-machi, Kaga-shi, Ishikawa 922-8522, Japan
| | - Satoshi Fujita
- Department of Frontier Fiber Technology and Science, Graduate School of Engineering, University of Fukui, 3-9-1, Bunkyo, Fukui-shi, Fukui 910-8507, Japan.,Organization for Life Science Advancement Programs, University of Fukui, 3-9-1, Bunkyo, Fukui-shi, Fukui 910-8507, Japan
| | - Takahiro Yamauchi
- Division of Medicine, Department of Neurosurgery, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan.,Organization for Life Science Advancement Programs, University of Fukui, 3-9-1, Bunkyo, Fukui-shi, Fukui 910-8507, Japan
| | - Makoto Isozaki
- Division of Medicine, Department of Neurosurgery, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan
| | - Ken-Ichiro Kikuta
- Division of Medicine, Department of Neurosurgery, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan.,Organization for Life Science Advancement Programs, University of Fukui, 3-9-1, Bunkyo, Fukui-shi, Fukui 910-8507, Japan
| |
Collapse
|
49
|
Cui X, Li Y, Yuan S, Huang Y, Chen X, Han Y, Liu Z, Li Z, Xiao Y, Wang Y, Sun L, Liu H, Zhu X. Alpha-kinase1 promotes tubular injury and interstitial inflammation in diabetic nephropathy by canonical pyroptosis pathway. Biol Res 2023; 56:5. [PMID: 36732854 PMCID: PMC9893546 DOI: 10.1186/s40659-023-00416-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 01/18/2023] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Alpha-kinase 1 (ALPK1) is a master regulator in inflammation and has been proved to promote renal fibrosis by promoting the production of IL-1β in diabetic nephropathy (DN) mice. Pyroptosis is involved in high glucose (HG)-induced tubular cells injury, characterized by activation of Gasdermin D (GSDMD) and the release of IL-1β and IL-18, resulting in inflammatory injury in DN. It is reasonable to assume that ALPK1 is involved in pyroptosis-related tubular injury in DN. However, the mechanism remains poorly defined. METHODS Immunohistochemistry (IHC) staining was performed to detect the expression of pyroptosis- and fibrosis-related proteins in renal sections of DN patients and DN mice. DN models were induced through injection of streptozotocin combined with a high-fat diet. Protein levels of ALPK1, NF-κB, Caspase-1, GSDMD, IL-1β, IL-18 and α-SMA were detected by Western blot. HK-2 cells treated with high-glucose (HG) served as an in vitro model. ALPK1 small interfering RNA (siRNA) was transfected into HK-2 cells to down-regulate ALPK1. The pyroptosis rates were determined by flow cytometry. The concentrations of IL-1β and IL-18 were evaluated by ELISA kits. Immunofluorescence staining was used to observe translocation of NF-κB and GSDMD. RESULTS The heat map of differentially expressed genes showed that ALPK1, Caspase-1 and GSDMD were upregulated in the DN group. The expression levels of ALPK1, Caspase-1, GSDMD and CD68 were increased in renal biopsy tissues of DN patients by IHC. ALPK1expression and CD68+ macrophages were positively correlated with tubular injury in DN patients. Western blot analysis showed increased expressions of ALPK1, phospho-NF-κB P65, GSDMD-NT, and IL-1β in renal tissues of DN mice and HK-2 cells, accompanied with increased renal fibrosis-related proteins (FN, α-SMA) and macrophages infiltration in interstitial areas. Inhibition of ALPK1 attenuated HG-induced upregulation expressions of NF-κB, pyroptosis-related proteins Caspase-1, GSDMD-NT, IL-1β, IL-18, α-SMA, and pyroptosis level in HK-2 cells. Also, the intensity and nuclear translocation of NF-κB and membranous translocation of GSDMD were ameliorated in HG-treated HK-2 cells after treatment with ALPK1 siRNA. CONCLUSIONS Our data suggest that ALPK1/NF-κB pathway initiated canonical caspase-1-GSDMD pyroptosis pathway, resulting in tubular injury and interstitial inflammation of DN.
Collapse
Affiliation(s)
- Xinyuan Cui
- grid.452708.c0000 0004 1803 0208Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan China
| | - Yifu Li
- grid.452708.c0000 0004 1803 0208Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan China ,grid.452708.c0000 0004 1803 0208Center for Medical Research, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shuguang Yuan
- grid.452708.c0000 0004 1803 0208Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan China
| | - Yao Huang
- grid.452708.c0000 0004 1803 0208Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan China
| | - Xiaojun Chen
- grid.452708.c0000 0004 1803 0208Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan China
| | - Yachun Han
- grid.452708.c0000 0004 1803 0208Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan China
| | - Zhiwen Liu
- grid.452708.c0000 0004 1803 0208Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan China
| | - Zheng Li
- grid.452708.c0000 0004 1803 0208Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan China
| | - Yang Xiao
- grid.452708.c0000 0004 1803 0208Key Laboratory of Diabetes Immunology, Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Youliang Wang
- grid.452708.c0000 0004 1803 0208Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan China
| | - Lin Sun
- grid.452708.c0000 0004 1803 0208Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan China
| | - Hong Liu
- grid.452708.c0000 0004 1803 0208Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan China
| | - Xuejing Zhu
- grid.452708.c0000 0004 1803 0208Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan China
| |
Collapse
|
50
|
Hatakeyama M. Impact of the Helicobacter pylori Oncoprotein CagA in Gastric Carcinogenesis. Curr Top Microbiol Immunol 2023; 444:239-257. [PMID: 38231221 DOI: 10.1007/978-3-031-47331-9_9] [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 CagA is the first and only bacterial oncoprotein etiologically associated with human cancer. Upon delivery into gastric epithelial cells via bacterial type IV secretion, CagA acts as a pathogenic/pro-oncogenic scaffold that interacts with and functionally perturbs multiple host proteins such as pro-oncogenic SHP2 phosphatase and polarity-regulating kinase PAR1b/MARK2. Although H. pylori infection is established during early childhood, gastric cancer generally develops in elderly individuals, indicating that oncogenic CagA activity is effectively counteracted at a younger age. Moreover, the eradication of cagA-positive H. pylori cannot cure established gastric cancer, indicating that H. pylori CagA-triggered gastric carcinogenesis proceeds via a hit-and-run mechanism. In addition to its direct oncogenic action, CagA induces BRCAness, a cellular status characterized by replication fork destabilization and loss of error-free homologous recombination-mediated DNA double-strand breaks (DSBs) by inhibiting cytoplasmic-to-nuclear localization of the BRCA1 tumor suppressor. This causes genomic instability that leads to the accumulation of excess mutations in the host cell genome, which may underlie hit-and-run gastric carcinogenesis. The close connection between CagA and BRCAness was corroborated by a recent large-scale case-control study that revealed that the risk of gastric cancer in individuals carrying pathogenic variants of genes that induce BRCAness (such as BRCA1 and BRCA2) dramatically increases upon infection with cagA-positive H. pylori. Accordingly, CagA-mediated BRCAness plays a crucial role in the development of gastric cancer in conjunction with the direct oncogenic action of CagA.
Collapse
Affiliation(s)
- Masanori Hatakeyama
- Institute of Microbial Chemistry, Laboratory of Microbial Carcinogenesis, Microbial Chemistry Research Foundation, 3-14-23 Kamiosaki, Shinagawa-Ku, Tokyo, 141-0021, Japan.
- Institute for Genetic Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-Ku, Sapporo, 060-0815, Japan.
| |
Collapse
|