1
|
Qin C, Xu C, Zhu Z, Song X, Wang X, Xu W, Zhu M. A study of the association between Helicobacter pylori infection type and pancreatic cancer risk: A systematic review and meta‑analysis. Oncol Lett 2025; 29:174. [PMID: 39975953 PMCID: PMC11837465 DOI: 10.3892/ol.2025.14920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025] Open
Abstract
Pancreatic cancer is a highly invasive malignant tumor with a complex pathogenesis that makes early diagnosis challenging. The potential association between Helicobacter pylori infection and pancreatic cancer risk has been noted; however, the available results are still highly divergent. The aim of the present study was to systematically evaluate the association between different types of H. pylori infection and pancreatic cancer risk as well as to explore the possible causes. A systematic search was conducted using the PubMed, Embase and Cochrane Library databases up to August 2023. The literature quality was evaluated using the Newcastle-Ottawa Scale. All studies that met the criteria were included in the overall meta-analysis to calculate the odds ratios (ORs) and corresponding 95% confidence intervals (CIs). In addition, subgroup analyses were performed based on factors such as diagnostic criteria for H. pylori infection, study region, type of study design and CagA status. The effect of publication bias on the quantitative synthesis results was assessed using the trim-and-fill analysis, and sensitivity analyses were used to verify the robustness of the quantitative synthesis results. A total of 17 studies involving 67,910 participants, including 64,372 controls and 3,538 patients with pancreatic cancer, were included in the present study. The overall analysis showed that no significant association was observed between H. pylori infection and pancreatic cancer risk (OR, 1.15; 95% CI, 0.93-1.41). Further subgroup analyses, which did not consider the effects of study quality, diagnostic criteria, geographical distribution and the type of study design, did not produce new findings that contradicted the results of the overall analysis. CagA+ H. pylori infection did not significantly affect the risk of pancreatic cancer (OR, 0.95; 95% CI, 0.78-1.16), whereas CagA- H. pylori infection may be a possible risk factor for pancreatic cancer (OR, 1.24; 95% CI, 1.004-1.541). The H. pylori infection did not significantly increase the risk of pancreatic cancer. However, it is noteworthy that CagA- H. pylori infection could be a potential factor that elevated the risk of pancreatic cancer.
Collapse
Affiliation(s)
- Chao Qin
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Chonghe Xu
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, P.R. China
| | - Zhongqi Zhu
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Xixi Song
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Xin Wang
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Wei Xu
- Department of Blood Transfusion, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Mei Zhu
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| |
Collapse
|
2
|
Al Omari SM, Khalifeh AH, Moman R, Sawan HM. Knowledge, Attitudes, and Practices Related to Helicobacter pylori and Gastric Disease in Jordan: Implications for Early Detection and Eradication. Infect Drug Resist 2025; 18:1503-1514. [PMID: 40123709 PMCID: PMC11930260 DOI: 10.2147/idr.s508330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
Background Gastric cancer and ulcers are responsible for almost 1 million deaths globally each year, disproportionately affecting low- and middle-income populations. Helicobacter pylori (H. pylori) infection is a major risk factor for both gastric cancer and peptic ulcers, with infection rates surpassing 70% in developing countries and reaching 88% in Jordan. Despite strong evidence linking H. pylori infection to gastric cancer, particularly with CagA-positive strains, public awareness of H. pylori infection, its transmission routes, and associated health risks remains insufficient. Methods This study aimed to assess the knowledge, attitudes, and practices (KAP) related to H. pylori-induced stomach ulcers and cancer in a Jordanian population, focusing on early detection and eradication efforts. A survey was administered to 398 participants to evaluate their understanding of H. pylori and its role in gastric disease. Results The findings revealed that 64.3% of respondents were aware of H. pylori, with 75.9% recognizing its association with gastric ulcers. However, awareness of the transmission routes and potential complications is limited. The frequent use of antacids for symptom relief also highlights the need for better awareness of appropriate treatments. Conclusion Public health education targeting these knowledge gaps could help reduce the incidence of H. pylori-related complications, including gastric cancer, especially in high-prevalence areas such as Jordan. Addressing these deficits and promoting preventive strategies, such as improved hygiene and regular medical check-ups, could facilitate early detection and improve health outcomes for individuals at risk of H. pylori-induced infection.
Collapse
Affiliation(s)
| | | | - Raja Moman
- Faculty of Pharmacy, University of Tripoli, Tripoli, Libya
| | - Hana M Sawan
- Faculty of Pharmacy, Zarqa University, Zarqa, Jordan
| |
Collapse
|
3
|
Muñoz AB, Stepanian J, Solano‐Gutierrez JS, Vale FF, Trespalacios‐Rangel AA. Helicobacter pylori Infection in Colombia: Phylogeny, Resistome, and Virulome. APMIS 2025; 133:e70003. [PMID: 39930978 PMCID: PMC11811748 DOI: 10.1111/apm.70003] [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: 04/17/2024] [Revised: 11/06/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025]
Abstract
Helicobacter pylori is a successful etiologic gastric agent that reaches a prevalence around 80% in Colombia. This bacterium is extremely diverse and has shown a phylogeographic pattern. The objective of this study was to perform an analysis of genomic epidemiology of H. pylori in Colombia. We enriched our set of 29 newly sequenced Colombian H. pylori genomes with additional data from public databases, reaching a total of 221 genomes in our dataset. Phylogenetic characterization was carried out using MLST and whole genome SNP analysis. We also performed a characterized the diversity of virulence factors and mutations associated with antimicrobial resistance. Phylogenetic analyzes showed two new Colombian H. pylori clades. Furthermore, many virulence genotype combinations were found, mutations associated with resistance were found for all the studied antibiotics, highlighting 14.4% of the genomes presented profiles associated with resistance to more than one family of antibiotics. Our analyzes described the genomics of Colombian H. pylori and verify the presence of a population group formed exclusively by Colombian isolates. We demonstrated the great diversity among the isolates and that the analysis by comparative genomics of H. pylori are valuable tools to assess the diversity, virulence, and resistance of H. pylori.
Collapse
Affiliation(s)
- Angela B. Muñoz
- Infectious Diseases Research Group, Microbiology Department, Sciences FacultyPontificia Universidad JaverianaBogotáColombia
- Health Sciences FacultyUniversidad Colegio Mayor de CundinamarcaBogotáColombia
| | - Johanna Stepanian
- Infectious Diseases Research Group, Microbiology Department, Sciences FacultyPontificia Universidad JaverianaBogotáColombia
| | - Juan S. Solano‐Gutierrez
- Texas Tech University—School of Veterinary MedicineAmarilloTexasUSA
- Texas Center for Comparative Cancer Research (TC3R)AmarilloTexasUSA
| | - Filipa F. Vale
- BioISI—Instituto de Biosistemas e Ciências Integrativas, Faculdade de CiênciasUniversidade de LisboaLisboaPortugal
- Research Institute for Medicines (iMed.ULisboa), Faculty of PharmacyUniversidade de LisboaLisboaPortugal
| | - Alba A. Trespalacios‐Rangel
- Infectious Diseases Research Group, Microbiology Department, Sciences FacultyPontificia Universidad JaverianaBogotáColombia
| |
Collapse
|
4
|
Shahi A, Kidane D. Decoding mitochondrial DNA damage and repair associated with H. pylori infection. Front Cell Infect Microbiol 2025; 14:1529441. [PMID: 39906209 PMCID: PMC11790445 DOI: 10.3389/fcimb.2024.1529441] [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: 11/16/2024] [Accepted: 12/19/2024] [Indexed: 02/06/2025] Open
Abstract
Mitochondrial genomic stability is critical to prevent various human inflammatory diseases. Bacterial infection significantly increases oxidative stress, driving mitochondrial genomic instability and initiating inflammatory human disease. Oxidative DNA base damage is predominantly repaired by base excision repair (BER) in the nucleus (nBER) as well as in the mitochondria (mtBER). In this review, we summarize the molecular mechanisms of spontaneous and H. pylori infection-associated oxidative mtDNA damage, mtDNA replication stress, and its impact on innate immune signaling. Additionally, we discuss how mutations located on mitochondria targeting sequence (MTS) of BER genes may contribute to mtDNA genome instability and innate immune signaling activation. Overall, the review summarizes evidence to understand the dynamics of mitochondria genome and the impact of mtBER in innate immune response during H. pylori-associated pathological outcomes.
Collapse
Affiliation(s)
| | - Dawit Kidane
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC, United States
| |
Collapse
|
5
|
Dong Y, Zhu J, Pan N. Recent advances in rapid detection of Helicobacter pylori by lateral flow assay. Arch Microbiol 2025; 207:35. [PMID: 39820420 DOI: 10.1007/s00203-025-04239-w] [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: 12/01/2024] [Revised: 12/28/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025]
Abstract
Infection with H. pylori (Helicobacter pylori) is the most prevalent human infection worldwide and is strongly associated with many gastrointestinal disorders, including gastric cancer. Endoscopy is mainly used to diagnose H. pylori infection in gastric biopsies. However, this approach is invasive, time-consuming and expensive. On the other hand, serology-based methods can be considered as a non-invasive approach to detecting H. pylori infection. The LFA (lateral flow assay) serves as a rapid point-of-care diagnostic tool. This paper-based platform facilitates the detection and quantification of analytes within human fluids such as blood, serum and urine. Due to ease of production, rapid results, and low costs, LFAs have a wide application in clinical laboratories and hospitals. In this comprehensive review, we examined LFA-based approaches for detection of H. pylori infection from human fluids and compare them with other high-sensitivity methods like ELISA (Enzyme-linked immunosorbent assay). Furthermore, we reviewed methods to elevate LFA sensitivity during H. pylori infection including, CRISPR/Cas system and isothermal amplification approaches. The development and optimization of novel labeling agents such as nanozyme to enhance the performance of LFA devices in detecting H. pylori were reviewed. These innovations aim to improve signal amplification and stability, thereby increasing the diagnostic accuracy of LFA devices. A combination of advances in LFA technology and molecular insight could significantly improve diagnostic accuracy, resulting in a significant improvement in clinical and remote diagnostic accuracy.
Collapse
Affiliation(s)
- Yanjin Dong
- Department of Laboratory Medicine, Jinan Second People's Hospital of Shandong Province (Jinan Eye Hospital), No. 148, Jingyi Road, Jinan, 250022, Shandong, China
| | - Jie Zhu
- Department of Laboratory Medicine, Shandong First Medical University Affiliated Provincial Hospital (East Hospital), No. 9677, Jingshi Road, Lixia District, Jinan, 250098, Shandong, China
| | - Ning Pan
- Department of Laboratory Medicine, Jinan Second People's Hospital of Shandong Province (Jinan Eye Hospital), No. 148, Jingyi Road, Jinan, 250022, Shandong, China.
| |
Collapse
|
6
|
Whitmire JM, Windham IH, Makobongo MO, Westland MD, Tran SC, Piñol J, Hui Y, Raheem Alkarkoushi R, Pich OQ, McGee DJ, Piazuelo MB, Melton-Celsa A, Testerman TL, Cover TL, Merrell DS. A unique Helicobacter pylori strain to study gastric cancer development. Microbiol Spectr 2025; 13:e0216324. [PMID: 39641575 PMCID: PMC11705839 DOI: 10.1128/spectrum.02163-24] [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: 08/28/2024] [Accepted: 10/20/2024] [Indexed: 12/07/2024] Open
Abstract
Helicobacter pylori colonizes a majority of the human population worldwide and can trigger development of a variety of gastric diseases. Since the bacterium is classified as a carcinogen, elucidation of the characteristics of H. pylori that influence gastric carcinogenesis is a high priority. To this end, the Mongolian gerbil infection model has proven to be an important tool to study gastric cancer progression. However, only a small number of H. pylori strains have been evaluated in the gerbil model. Thus, to identify additional strains able to colonize and induce disease in this model, several H. pylori strains were used to infect Mongolian gerbils, and stomachs were harvested at multiple timepoints to assess colonization and gastric pathology. The USU101 strain reproducibly colonized Mongolian gerbils and induced gastric inflammation in the majority of the animals 1 month after infection. Adenocarcinoma or dysplasia was observed in the majority of gerbils by 2 months post-infection. To define the contribution of key virulence factors to this process, isogenic strains lacking cagA or vacA, along with restorant strains containing a wild-type (WT) copy of the genes, were studied. The ΔcagA USU101 strain colonized gerbils at levels similar to WT, but did not induce comparable levels of inflammation or disease. In contrast, the ΔvacA USU101 strain did not colonize gerbils, and the stomach pathology resembled that of the mock-infected animals. The restorant USU101 strains expressed the CagA and VacA proteins in vitro, and in vivo experiments with Mongolian gerbils showed a restoration of colonization levels and inflammation scores comparable to those observed in WT USU101. Our studies indicate that the USU101 strain is a valuable tool to study H. pylori-induced disease.IMPORTANCEGastric cancer is the fifth leading cause of cancer-related death globally; the majority of gastric cancers are associated with Helicobacter pylori infection. Infection of Mongolian gerbils with H. pylori has been shown to result in induction of gastric cancer, but few H. pylori strains have been studied in this model; this limits our ability to fully understand gastric cancer pathogenesis in humans because H. pylori strains are notoriously heterogenous. Our studies reveal that USU101 represents a unique H. pylori strain that can be added to our repertoire of strains to study gastric cancer development in the Mongolian gerbil model.
Collapse
Affiliation(s)
| | - Ian H. Windham
- Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Morris O. Makobongo
- Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | | | | | - Jaume Piñol
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Yvonne Hui
- University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | | | - Oscar Q. Pich
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Laboratori de Recerca en Microbiologia i Malalties Infeccioses, Hospital Universitari Parc Taulí, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - David J. McGee
- Department of Microbiology and Immunology, LSU Health Sciences Center-Shreveport, Shreveport, Louisiana, USA
| | | | - Angela Melton-Celsa
- Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Traci L. Testerman
- University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Timothy L. Cover
- Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - D. Scott Merrell
- Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
7
|
Song J, Liu L, Wang F, Bao D. Targeting Ubiquitin-specific Protease 5 Overcomes Chemoresistance via Negatively Regulating p53 in Gastric Cancer. Curr Mol Med 2025; 25:211-221. [PMID: 38347776 DOI: 10.2174/0115665240278762240202103722] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 04/11/2025]
Abstract
BACKGROUND Resistance to chemotherapy is a major obstacle in the clinical management of gastric cancer, and the mechanisms underlying chemoresistance remain largely unknown. AIMS This study aimed to investigate the involvement of ubiquitin-specific protease 5 (USP5), a deubiquitinating enzyme, in gastric cancer chemoresistance. METHODS USP5 expression was analyzed in fifty paired gastric cancer and adjacent normal tissues, chemo-sensitive and chemo-resistant gastric cancer lines using quantitative ELISA. The role of USP5 was determined using loss-of-function and gainof- function methods. USP5-mediated downstream effectors were analyzed using biochemical methods focusing on p53. RESULTS USP5 expression was comparable in tumors and normal in the majority of the cohort. Following chemotherapy treatment, USP5 expression significantly increased in gastric cancer cells, while p53 levels remained unaltered. Overexpression of USP5 amplified growth and migration while decreasing apoptosis induced by serum withdrawal across multiple gastric cancer cell lines. Conversely, USP5 knockdown effectively heightened gastric cancer sensitivity to paclitaxel and 5-FU treatments, particularly targeting chemo-resistant gastric cancer cells by inhibiting proliferation and migration and inducing apoptosis. Additionally, USP5 knockdown increased levels of p53 but not MDM2, increased p53 activity and increased transcription of p53 target genes. In contrast, USP5 overexpression decreased the level and activity of p53 and inhibited transcription of p53 target genes. The anti-proliferative, anti-migratory, and pro-apoptotic effects of USP5 were significantly diminished upon p53 depletion, highlighting the interplay between p53 and USP5 in regulating gastric cancer cell activities. Additionally, USP5 inhibition suppressed chemo-resistant gastric cancer cell migration via suppressing epithelial-mesenchymal transition (EMT) and RhoA activity. CONCLUSION Targeting USP5 inhibition has emerged as a promising alternative therapeutic approach to overcoming chemoresistance in gastric cancer. Additionally, our study sheds light on the novel role of USP5 as a regulator of p53 in gastric cancer.
Collapse
Affiliation(s)
- Jing Song
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, 441000, People's Republic of China
| | - Lei Liu
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, 441000, People's Republic of China
| | - Fang Wang
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, 441000, People's Republic of China
| | - Di Bao
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, 441000, People's Republic of China
| |
Collapse
|
8
|
Liu Y, Yu X, Shen H, Hong Y, Hu G, Niu W, Ge J, Xuan J, Qin JJ, Li Q. Mechanisms of traditional Chinese medicine in the treatment and prevention of gastric cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156003. [PMID: 39305742 DOI: 10.1016/j.phymed.2024.156003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/21/2024] [Accepted: 06/07/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Gastric cancer (GC) ranks as the fifth most prevalent malignancy worldwide. Conventional treatments, including radiotherapy and chemotherapy, often induce severe side effects and significant adverse reactions, and they may also result in drug resistance. Consequently, there is a critical need for the development of new therapeutic agents. Traditional Chinese Medicine (TCM) and natural products are being extensively researched due to their low toxicity, multi-targeted approaches, and diverse pathways. Scholars are increasingly focusing on identifying active anticancer components within TCM. PURPOSE This review aims to summarise research conducted over the past 14 years on the treatment of GC using TCM. The focus is on therapeutic targets, mechanisms, and efficacy of Chinese medicine and natural products, including monomer compounds, extracts or analogues, and active ingredients. METHODS Relevant articles on TCM and GC were retrieved from PubMed using appropriate keywords. The collected articles were screened and classified according to the types of TCM, with an emphasis on the molecular mechanisms underlying the treatment of GC. RESULTS The research on TCM indicates that TCM and natural products can effectively inhibit the metastasis, proliferation, and invasion of tumour cells. They can also induce apoptosis, autophagy and improve the chemosensitivity of drug-resistant cells. Additionally, injections derived from Chinese herbal medicine, when used as an adjunct to conventional chemotherapy, can significantly improve the prognosis of GC patients by reducing chemotherapy toxicity. CONCLUSION This review summarises the progress of TCM treatment of GC over the past 14 years, and discusses its therapeutic application of GC, which proves that TCM is a promising treatment strategy for GC in the future.
Collapse
Affiliation(s)
- Yanyang Liu
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China; Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuefei Yu
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China
| | - Huize Shen
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yangjian Hong
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China; Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gaofeng Hu
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China; Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenyuan Niu
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jiaming Ge
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China; Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jie Xuan
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China; Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiang-Jiang Qin
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| | - Qinglin Li
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China.
| |
Collapse
|
9
|
Lin Z, Assaraf YG, Kwok HF. Peptides for microbe-induced cancers: latest therapeutic strategies and their advanced technologies. Cancer Metastasis Rev 2024; 43:1315-1336. [PMID: 39008152 DOI: 10.1007/s10555-024-10197-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 06/14/2024] [Indexed: 07/16/2024]
Abstract
Cancer is a significant global health concern associated with multiple distinct factors, including microbial and viral infections. Numerous studies have elucidated the role of microorganisms, such as Helicobacter pylori (H. pylori), as well as viruses for example human papillomavirus (HPV), hepatitis B virus (HBV), and hepatitis C virus (HCV), in the development of human malignancies. Substantial attention has been focused on the treatment of these microorganism- and virus-associated cancers, with promising outcomes observed in studies employing peptide-based therapies. The current paper provides an overview of microbe- and virus-induced cancers and their underlying molecular mechanisms. We discuss an assortment of peptide-based therapies which are currently being developed, including tumor-targeting peptides and microbial/viral peptide-based vaccines. We describe the major technological advancements that have been made in the design, screening, and delivery of peptides as anticancer agents. The primary focus of the current review is to provide insight into the latest research and development in this field and to provide a realistic glimpse into the future of peptide-based therapies for microbe- and virus-induced neoplasms.
Collapse
Affiliation(s)
- Ziqi Lin
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Lab, Faculty of Biology, Technion-Israel Instituteof Technology, Haifa, 3200003, Israel
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR.
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR.
| |
Collapse
|
10
|
Koshibu Y, Ubagai T, Yoshino Y, Ono Y. Immunobiological effects of lipopolysaccharide derived from Helicobacter pylori and influence of a proton pump inhibitor lansoprazole on human polymorphonuclear leukocytes. Folia Microbiol (Praha) 2024; 69:1369-1378. [PMID: 39153156 PMCID: PMC11485072 DOI: 10.1007/s12223-024-01188-7] [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: 09/28/2023] [Accepted: 08/01/2024] [Indexed: 08/19/2024]
Abstract
Helicobacter pylori colonizes the human gastric mucosa of more than half of the human population and has a unique lipopolysaccharide (LPS) structure. LPS is the most dominant and suitable pathogen-associated molecular pattern that is detected via pattern recognition receptors. Although the priming effect of H. pylori LPS on reactive oxygen species (ROS) production of PMNs is lower than that of Escherichia coli O111:B4 LPS, LPS released from H. pylori associated with antibiotics eradication therapy may activate PMNs and increase ROS production. In addition, we describe the effects of H. pylori and E. coli O111:B4 LPSs on gene expression and the anti-inflammatory effect of lansoprazole (LPZ) in human polymorphonuclear leukocytes. LPS isolated from H. pylori and E. coli O111:B4 alters toll-like receptor 2 (TLR) and TLR4 expressions similarly. However, LPS from E. coli O111:B4 and H. pylori caused a 1.8-fold and 1.5-fold increase, respectively, in CD14 expression. All LPS subtypes upregulated TNFα and IL6 expression in a concentration-dependent manner. Although E. coli O111:B4 LPS upregulated IL8R mRNA levels, H. pylori LPS did not (≦ 100 ng/mL). Gene expression levels of ITGAM demonstrated no significant change on using both LPSs. These different effects on the gene expression in PMNs may depend on variations in LPS structural modifications related to the acquired immunomodulatory properties of H. pylori LPS. Proton pump inhibitors, i.e., LPZ, are used in combination with antibiotics for the eradication therapy of H. pylori. LPZ and its acid-activated sulphenamide form AG-2000 suppress ROS production of PMNs in a dose-dependent manner. These results suggest that LPZ combination with antibiotics for H. pylori eradication reduces gastric inflammation by suppressing ROS release from PMNs.
Collapse
Affiliation(s)
- Yoji Koshibu
- Department of Microbiology & Immunology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Tsuneyuki Ubagai
- Department of Microbiology & Immunology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605, Japan.
| | - Yusuke Yoshino
- Department of Microbiology & Immunology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Yasuo Ono
- Department of Microbiology & Immunology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605, Japan
- Faculty of Health and Medical Science, Teikyo Heisei University, Tokyo, 170-8445, Japan
| |
Collapse
|
11
|
Zou Q, Wu Y, Zhang S, Li S, Li S, Su Y, Zhang L, Li Q, Zou H, Zhang X, Wang T, Liang S, Yang J, Li C. Escherichia coli and HPV16 coinfection may contribute to the development of cervical cancer. Virulence 2024; 15:2319962. [PMID: 38380669 PMCID: PMC10883084 DOI: 10.1080/21505594.2024.2319962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 02/09/2024] [Indexed: 02/22/2024] Open
Abstract
Persistent human papillomavirus HPV infection is a necessary but insufficient condition for cervical cancer. Microorganisms are crucial environmental factors in cancers susceptibility and progression, recently attracting considerable attention. This study aimed to determine the infection status and relationship between high-risk HPV (HR-HPV) and lower genital tract infectious pathogens in cervical cancer and its precursors. From a retrospective and a prospective cohort analysis, Escherichia coli (E. coli) dominated the pathogens isolated from cervical discharges, and an isolation rate uptrend has been shown recently. HPV16 and E. coli's coinfection rate gradually increased with the severity of cervical intraepithelial neoplasia. The adhesion and invasion abilities of the isolated E. coli to HPV16-positive SiHa cells were evaluated in vitro. The TCGA database and cervical tissues samples analysis showed that IL-10 was upregulated in cervical cancer. IL-10 expression levels increased in tissue samples with the severity of cervical cancer and its precursors with HPV16 and E. coli coinfection. Although no significant changes in IL-10 production were observed in the co-culture supernatant, we hypothesized that Treg immune cells in the tumour microenvironment might be responsible for the local IL-10 upregulation, according to our data showing Foxp3 upregulation and an upward trend with the cervical intraepithelial neoplasia grading to cancer and tumours with E. coli and HPV16 coinfection. Our data provide insights into the possible role of E. coli in cervical cancer progression and suggest that the application of HPV and E. coli screening programs may be an effective strategy to relieve the burden of cervical cancer and its precursor lesions.
Collapse
Affiliation(s)
- Qin Zou
- Department of Clinical Laboratory, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - Yingying Wu
- Department of Clinical Laboratory, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - ShuaiShuai Zhang
- Department of Clinical Laboratory, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Shu Li
- Department of Clinical Laboratory, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - Siyue Li
- Department of Clinical Laboratory, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - Yan Su
- Department of Clinical Laboratory, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - Lei Zhang
- Department of Clinical Laboratory, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - Qian Li
- Department of Clinical Laboratory, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - Hua Zou
- Department of Clinical Laboratory, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - Xinyuan Zhang
- Department of Clinical Laboratory, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - Teng Wang
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shuang Liang
- Department of Pathology, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - Jun Yang
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - Chunli Li
- Department of Clinical Laboratory, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| |
Collapse
|
12
|
Kuhn EMA, Sominsky LA, Chittò M, Schwarz EM, Moriarty TF. Antibacterial Mechanisms and Clinical Impact of Sitafloxacin. Pharmaceuticals (Basel) 2024; 17:1537. [PMID: 39598446 PMCID: PMC11597390 DOI: 10.3390/ph17111537] [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: 10/14/2024] [Revised: 11/09/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Sitafloxacin is a 4th generation fluoroquinolone antibiotic with broad activity against a wide range of Gram-negative and Gram-positive bacteria. It is approved in Japan and used to treat pneumonia and urinary tract infections (UTIs) as well as other upper and lower respiratory infections, genitourinary infections, oral infections and otitis media. Compared to other fluoroquinolones, sitafloxacin displays a low minimal inhibitory concentration (MIC) for many bacterial species but also activity against anaerobes, intracellular bacteria, and persisters. Furthermore, it has also shown strong activity against biofilms of P. aeruginosa and S. aureus in vitro, which was recently validated in vivo with murine models of S. aureus implant-associated bone infection. Although limited in scale at present, the published literature supports the further evaluation of sitafloxacin in implant-related infections and other biofilm-related infections. The aim of this review is to summarize the chemical-positioning-based mechanisms, activity, resistance profile, and future clinical potential of sitafloxacin.
Collapse
Affiliation(s)
- Elian M. A. Kuhn
- AO Research Institute Davos, 7270 Davos, Switzerland; (E.M.A.K.); (M.C.)
- Infection Biology, Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Levy A. Sominsky
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA (E.M.S.)
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Marco Chittò
- AO Research Institute Davos, 7270 Davos, Switzerland; (E.M.A.K.); (M.C.)
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA (E.M.S.)
| | - T. Fintan Moriarty
- AO Research Institute Davos, 7270 Davos, Switzerland; (E.M.A.K.); (M.C.)
| |
Collapse
|
13
|
Fang Z, Zhang W, Wang H, Zhang C, Li J, Chen W, Xu X, Wang L, Ma M, Zhang S, Li Y. Helicobacter pylori promotes gastric cancer progression by activating the TGF-β/Smad2/EMT pathway through HKDC1. Cell Mol Life Sci 2024; 81:453. [PMID: 39545942 PMCID: PMC11568101 DOI: 10.1007/s00018-024-05491-x] [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: 03/27/2024] [Revised: 09/21/2024] [Accepted: 10/24/2024] [Indexed: 11/17/2024]
Abstract
Helicobacter pylori (H. pylori) infection is widely acknowledged as the primary risk factor for gastric cancer, facilitating its progression via the Correa cascade. Concurrently, Hexokinase Domain Containing 1 (HKDC1) has been implicated in the mediation of aerobic glycolysis, contributing to tumorigenesis across various cancers. However, the precise role of HKDC1 in the inflammatory transformation associated with H. pylori-induced gastric cancer remains elusive. In this study, transcriptome sequencing revealed a significant correlation between HKDC1 and H. pylori-induced gastric cancer. Subsequent validation using qRT-PCR, immunohistochemistry, and Western blot analysis confirmed elevated HKDC1 expression in both human and murine gastritis and gastric tumors. Moreover, in vitro and in vivo experiments demonstrated that H. pylori infection up-regulates TGF-β1 and p-Smad2, thereby activating the epithelial-mesenchymal transition (EMT) pathway, with HKDC1 playing a pivotal role. Suppression of HKDC1 expression or pharmacological inhibition of TGF-β1 reversed EMT activation, consequently reducing gastric cancer cell proliferation and metastasis. These results underscore HKDC1's essential contribution to H. pylori-induced gastric cancer progression via EMT activation.
Collapse
Affiliation(s)
- Ziqing Fang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China
| | - Weitong Zhang
- Department of General Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, 241001, China
| | - Huizhen Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China
| | - Chaoyang Zhang
- Department of General Surgery, The Second Affiliated Hospital Zhejiang University, Hangzhou, 310000, China
| | - Jing Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China
| | - Wanjing Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China
| | - Xin Xu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China
| | - Luyang Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China
| | - Mengdi Ma
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China
| | - Shangxin Zhang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China
| | - Yongxiang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China.
| |
Collapse
|
14
|
Bawali P, Brahma A, Rana SR, Pal A, Bhattacharyya A. Helicobacter pylori infection and inflammatory events: the extracellular vesicle-connect in driving gastrointestinal tract cancers. Front Med (Lausanne) 2024; 11:1444242. [PMID: 39610678 PMCID: PMC11602329 DOI: 10.3389/fmed.2024.1444242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/28/2024] [Indexed: 11/30/2024] Open
Affiliation(s)
| | | | | | | | - Asima Bhattacharyya
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, An OCC of Homi Bhabha National Institute, Khurda, Odisha, India
| |
Collapse
|
15
|
Wu H, Yuan H, Zhang J, He T, Deng Y, Chen Y, Zhang Y, Chen W, Wu C. Helicobacter pylori upregulates PAD4 expression via stabilising HIF-1α to exacerbate rheumatoid arthritis. Ann Rheum Dis 2024; 83:1666-1676. [PMID: 39107082 PMCID: PMC11671999 DOI: 10.1136/ard-2023-225306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 07/21/2024] [Indexed: 08/09/2024]
Abstract
OBJECTIVE Helicobacter pylori infection has been reported to aggravate rheumatoid arthritis (RA), but the relevant mechanism remains unclear. This study aimed to investigate the underlying pathogenic mechanism of H. pylori infection in the progression of RA. METHODS The Disease Activity Score (DAS-28) and serum anticitrullinated protein antibody (ACPA) levels were compared between H. pylori-negative and H. pylori-positive patients with RA. MH7A cells were stimulated with polyclonal ACPA purified from the peripheral blood of patients with RA. The citrullination levels were assessed by western blot in GES-1 cells and sera. ChIP, luciferase reporter assays, mass spectrometry and ELISA were applied to explore the molecular mechanism of H. pylori infection in RA progression. RESULTS The DAS-28 and ACPA levels of patients with RA in the H. pylori-positive group were significantly higher than those in the H. pylori-negative group. Polyclonal ACPA derived from H. pylori-positive patients promoted cell proliferation and induced secretion of IL-6 and IL-8. For the first time, we found that H. pylori infection induces cellular protein citrullination by upregulating protein arginine deiminase type 4 (PAD4). Furthermore, we confirmed a direct functional binding of hypoxia-inducible factor 1α on the PADI4 gene promoter. We demonstrated that PAD4 interacts with and citrullinates keratin 1 (K1), and serum and synovial fluid levels of anti-Cit-K1 antibody were markedly increased in H. pylori-infected patients with RA. CONCLUSION Our findings reveal a novel mechanism by which H. pylori infection contributes to RA progression. Therapeutic interventions targeting H. pylori may be a viable strategy for the management of RA.
Collapse
Affiliation(s)
- Hui Wu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Hanmei Yuan
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Jin Zhang
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Taojun He
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yilin Deng
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Ying Chen
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yunqi Zhang
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, Guangdong, China
| | - Weisan Chen
- Biochemistry and Genetics, La Trobe University, Melbourne, Victoria, Australia
| | - Chao Wu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| |
Collapse
|
16
|
Akcelik-Deveci S, Kılıç E, Mansur-Ozen N, Timucin E, Buyukcolak Y, Oktem-Okullu S. Identification of interaction partners of outer inflammatory protein A: Computational and experimental insights into how Helicobacter pylori infects host cells. PLoS One 2024; 19:e0300557. [PMID: 39471168 PMCID: PMC11521304 DOI: 10.1371/journal.pone.0300557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 09/26/2024] [Indexed: 11/01/2024] Open
Abstract
Outer membrane proteins (OMPs) play a key role in facilitating the survival of Helicobacter pylori within the gastric tissue by mediating adherence. Among these proteins, Outer inflammatory protein A (OipA) is a critical factor in H. pylori colonization of the host gastric epithelial cell surface. While the role of OipA in H. pylori attachment and its association with clinical outcomes have been established, the structural mechanisms underlying OipA's action in adherence to gastric epithelial cells remain limited. Our study employed experimental and computational approaches to investigate the interaction partners of OipA on the gastric epithelial cell surface. Initially, we conducted a proteomic analysis using a pull-down assay with recombinant OipA and gastric epithelial cell membrane proteins to identify the OipA interactome. This analysis revealed 704 unique proteins that interacted with OipA. We subsequently analyzed 16 of these OipA partners using molecular modeling tools. Among these 16 partners, we highlight three human proteins, namely Hepatocyte growth factor (HGF), Mesenchymal epithelial transition factor receptor (Met), and Adhesion G Protein-Coupled Receptor B1 (AGRB1) that could play a role in H. pylori adherence to the gastric epithelial cell surface with OipA. Collectively, these findings reveal novel host interactions mediated by OipA, suggesting their potential as therapeutic targets for combating H. pylori infection.
Collapse
Affiliation(s)
- Sümeyye Akcelik-Deveci
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem University, Atasehir, Istanbul, Turkey
| | - Elif Kılıç
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem University, Atasehir, Istanbul, Turkey
| | - Nesteren Mansur-Ozen
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem University, Atasehir, Istanbul, Turkey
| | - Emel Timucin
- Department of Biostatistics and Medical Informatics, School of Medicine, Acibadem University, Atasehir, Istanbul, Turkey
- Department of Biostatistics and Bioinformatics, Institute of Health Sciences, Acibadem University, Atasehir, Istanbul, Turkey
| | - Yaren Buyukcolak
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem University, Atasehir, Istanbul, Turkey
| | - Sinem Oktem-Okullu
- Department of Medical Microbiology, School of Medicine, Acibadem, Atasehir, Istanbul, Turkey
| |
Collapse
|
17
|
Li Y, Farhan MHR, Yang X, Guo Y, Sui Y, Chu J, Huang L, Cheng G. A review on the development of bacterial multi-epitope recombinant protein vaccines via reverse vaccinology. Int J Biol Macromol 2024; 282:136827. [PMID: 39476887 DOI: 10.1016/j.ijbiomac.2024.136827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/04/2024] [Accepted: 10/21/2024] [Indexed: 11/10/2024]
Abstract
Bacterial vaccines play a crucial role in combating bacterial infectious diseases. Apart from the prevention of disease, bacterial vaccines also help to reduce the mortality rates in infected populations. Advancements in vaccine development technologies have addressed the constraints of traditional vaccine design, providing novel approaches for the development of next-generation vaccines. Advancements in reverse vaccinology, bioinformatics, and comparative proteomics have opened horizons in vaccine development. Specifically, the use of protein structural data in crafting multi-epitope vaccines (MEVs) to target pathogens has become an important research focus in vaccinology. In this review, we focused on describing the methodologies and tools for epitope vaccine development, along with recent progress in this field. Moreover, this article also discusses the challenges in epitope vaccine development, providing insights for the future development of bacterial multi-epitope genetically engineered vaccines.
Collapse
Affiliation(s)
- Yuxin Li
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Muhammad Haris Raza Farhan
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Xiaohan Yang
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Ying Guo
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Yuxin Sui
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Jinhua Chu
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Lingli Huang
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China; MOA Laboratory of Risk Assessment for Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Guyue Cheng
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China; MOA Laboratory of Risk Assessment for Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China.
| |
Collapse
|
18
|
Halawa M, Newman PM, Aderibigbe T, Carabetta VJ. Conjugated therapeutic proteins as a treatment for bacteria which trigger cancer development. iScience 2024; 27:111029. [PMID: 39635133 PMCID: PMC11615139 DOI: 10.1016/j.isci.2024.111029] [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: 12/07/2024] Open
Abstract
In recent years, an increasing amount of research has focused on the intricate and complex correlation between bacterial infections and the development of cancer. Some studies even identified specific bacterial species as potential culprits in the initiation of carcinogenesis, which generated a great deal of interest in the creation of innovative therapeutic strategies aimed at addressing both the infection and the subsequent risk of cancer. Among these strategies, there has been a recent emergence of the use of conjugated therapeutic proteins, which represent a highly promising avenue in the field of cancer therapeutics. These proteins offer a dual-targeting approach that seeks to effectively combat both the bacterial infection and the resulting malignancies that may arise because of such infections. This review delves into the landscape of conjugated therapeutic proteins that have been intricately designed with the purpose of specifically targeting bacteria that have been implicated in the induction of cancer.
Collapse
Affiliation(s)
- Mohamed Halawa
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Precious M. Newman
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Tope Aderibigbe
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Valerie J. Carabetta
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| |
Collapse
|
19
|
Dai W, Li YQ, Zhou Y. Clinical implications of the latest advances in gastrointestinal tumor research. World J Gastrointest Oncol 2024; 16:4055-4059. [DOI: 10.4251/wjgo.v16.i10.4055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/25/2024] [Accepted: 06/14/2024] [Indexed: 09/26/2024] Open
Abstract
In this editorial, we provide commentary on six articles recently published in the World Journal of Gastrointestinal Oncology. These articles collectively present the latest findings in the field of gastric and colorectal cancer (CRC) research. The global incidence of gastric cancer varies based on geographical location, age, and sex. The disease predominantly affects middle-aged and elderly individuals, with a slightly higher prevalence in men than in women. CRC is characterized by a low 5-year survival rate and high mortality rate. It primarily affects individuals over the age of 50, and the risk of disease increases with age. Both gastric and CRC pose significant health threats, thus requiring more effective diagnostic, therapeutic, and supportive care strategies to improve patient outcomes. The articles discussed in this editorial encompass topics such as screening, diagnosis, mechanisms of progression, and postoperative recovery in gastric and CRC, and the findings offer valuable insights for clinical decision-making in the diagnosis, treatment, and prognosis of gastrointestinal cancers.
Collapse
Affiliation(s)
- Wei Dai
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu Province, China
| | - Yuan-Qi Li
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu Province, China
| | - You Zhou
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu Province, China
| |
Collapse
|
20
|
Van de Weyer Y, Bentley C, Orlowska A, Barbon AR, Chantrey J. Gastric adenocarcinoma with carcinomatosis and degenerative joint disease of the xiphisternum in a Bactrian camel (Camelus bactrianus). J Comp Pathol 2024; 214:7-11. [PMID: 39163717 DOI: 10.1016/j.jcpa.2024.07.006] [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: 04/04/2024] [Revised: 06/01/2024] [Accepted: 07/19/2024] [Indexed: 08/22/2024]
Abstract
Gastric adenocarcinomas have been sporadically reported in camelids. This report describes a primary gastric adenocarcinoma and subsequent peritoneal carcinomatosis in a 20-year-old female Bactrian camel (Camelus bactrianus). Numerous metastases were present throughout the omentum, liver, abdominal lymph nodes, intestinal serosa, kidneys and lungs. The primary tumour macroscopically resembled an ulcerated crater and originated from the distal four-fifths of the C3 compartment, an anatomical region with naturally prominent gastric rugae and true glands. Moderate numbers of Helicobacter spp colonies were present within gastric pits and necrotic areas of C3. Ménetrier's disease has previously been implicated as a predisposing condition for the development of gastric adenocarcinoma in another camel, but no evidence of this premalignant disorder was found in this case. This camel also suffered from a chronic skin wound of the hump and severe degenerative joint disease of the xiphisternum, the latter of which was presumably associated with excessive pressure on the sternum.
Collapse
Affiliation(s)
- Yannick Van de Weyer
- Institute of Infection, Veterinary and Ecological Sciences, Department of Veterinary Anatomy, Physiology and Pathology, University of Liverpool, Leahurst Campus, Neston CH64 7TE, UK.
| | - Charlotte Bentley
- North of England Zoological Society (Chester Zoo), Caughall Road, Upton By Chester, Chester CH2 1LH, UK
| | - Anna Orlowska
- Institute of Infection, Veterinary and Ecological Sciences, Department of Veterinary Anatomy, Physiology and Pathology, University of Liverpool, Leahurst Campus, Neston CH64 7TE, UK
| | - Alberto R Barbon
- Institute of Infection, Veterinary and Ecological Sciences, Department of Veterinary Anatomy, Physiology and Pathology, University of Liverpool, Leahurst Campus, Neston CH64 7TE, UK; North of England Zoological Society (Chester Zoo), Caughall Road, Upton By Chester, Chester CH2 1LH, UK
| | - Julian Chantrey
- Institute of Infection, Veterinary and Ecological Sciences, Department of Veterinary Anatomy, Physiology and Pathology, University of Liverpool, Leahurst Campus, Neston CH64 7TE, UK
| |
Collapse
|
21
|
Zhang J, Ji X, Liu S, Sun Z, Cao X, Liu B, Li Y, Zhao H. Helicobacter pylori infection promotes liver injury through an exosome-mediated mechanism. Microb Pathog 2024; 195:106898. [PMID: 39208956 DOI: 10.1016/j.micpath.2024.106898] [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: 05/02/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Helicobacter pylori infection has been thought to be associated with liver diseases, although the exact mechanisms remain elusive. This study identified H. pylori-induced liver inflammation and tissue damage in infected mice and examined the exosome-mediated mechanism underlying H. pylori infection's impact on liver injury. Exosomes were isolated from H. pylori-infected gastric epithelial GES-1 cells (Hp-GES-EVs), and the crucial virulence factor CagA was identified within these exosomes. Fluorescent labeling demonstrated that Hp-GES-EVs can be absorbed by liver cells. Treatment with Hp-GES-EVs enhanced the proliferation, migration, and invasion of Hep G2 and Hep 3B cells. Additionally, exposure to Hp-GES-EVs activated NF-κB and PI3K/AKT signaling pathways, which provides a reasonable explanation for the liver inflammation and neoplastic traits. Using a mouse model established via tail vein injection of Hp-GES-EVs, exosome-driven liver injury was evidenced by slight hepatocellular erosion around the central hepatic vein and elevated serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), and IL-6. Administering the exosome inhibitor GW4869 via intraperitoneal injection in mice resulted in a reduction of liver damage caused by H. pylori infection. These findings illuminate the exosome-mediated pathogenesis of H. pylori-induced liver injury and offer valuable insights into the extra-gastrointestinal manifestations of H. pylori infection.
Collapse
Affiliation(s)
| | - Xiaofei Ji
- Binzhou Medical University, Yantai, China
| | | | - Zekun Sun
- Binzhou Medical University, Yantai, China
| | | | | | - Yizheng Li
- Binzhou Medical University, Yantai, China
| | | |
Collapse
|
22
|
Bonny SQ, Zhou X, Khan MF, Rahman MM, Xin Y, Vankadari N, Tikhomirova A, Homman-Ludiye J, Roujeinikova A. Functional and biochemical characterisation of remote homologues of type IV pili proteins PilN and PilO in Helicobacter pylori. IUBMB Life 2024; 76:780-787. [PMID: 38748402 DOI: 10.1002/iub.2828] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/03/2024] [Indexed: 10/19/2024]
Abstract
Helicobacter pylori encodes homologues of PilM, PilN and PilO from bacteria with Type IV pili, where these proteins form a pilus alignment complex. Inactivation of pilO changes H. pylori motility in semi-solid media, suggesting a link to the chemosensory pathways or flagellar motor. Here, we showed that mutation of the pilO or pilN gene in H. pylori strain SS1 reduced the mean linear swimming speed in liquid media, implicating PilO and PilN in the function, or regulation of, the flagellar motor. We also demonstrated that the soluble variants of H. pylori PilN and PilO share common biochemical properties with their Type IV pili counterparts which suggests their adapted function in the bacterial flagellar motor may be similar to that in the Type IV pili.
Collapse
Affiliation(s)
- Sharmin Q Bonny
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Xiaotian Zhou
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Mohammad F Khan
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Mohammad M Rahman
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Yue Xin
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Naveen Vankadari
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Alexandra Tikhomirova
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Jihane Homman-Ludiye
- Monash Micro Imaging, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Anna Roujeinikova
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
23
|
Ghazanfar H, Javed N, Reina R, Thartori O, Ghazanfar A, Patel H. Advances in Diagnostic Modalities for Helicobacter pylori Infection. Life (Basel) 2024; 14:1170. [PMID: 39337953 PMCID: PMC11432972 DOI: 10.3390/life14091170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Helicobacter pylori (H. pylori) infection is a widespread global health issue with a varying prevalence influenced by geography, socioeconomic status, and demographics. In the U.S., the prevalence is lower, though certain groups, such as older adults and immigrants from high-prevalence regions, show higher rates. The decrease in infection rates in developed countries is due to improved sanitation, antibiotics, and healthcare, whereas developing countries continue to experience high rates due to poor living conditions. H. pylori infection can be asymptomatic or cause symptoms like dyspepsia, abdominal pain, bloating, nausea, and loss of appetite. Pathophysiologically, H. pylori contribute to conditions such as gastritis, peptic ulcers, and gastric cancer through mechanisms including urease production and the release of virulence factors, leading to chronic inflammation and an increased cancer risk. Diagnostic methods for H. pylori have progressed significantly. Non-invasive techniques, such as serological assays, stool antigen tests, and urea breath tests, are practical and sensitive. Invasive methods, including endoscopic biopsy and molecular diagnostics, are more definitive but resource intensive. Recent advancements in diagnostic technology, including matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS), biosensor technology, and next-generation sequencing (NGS), promise improved speed, accuracy, and accessibility. These innovations are expected to enhance the detection and management of H. pylori, potentially reducing the global disease burden. This review aims to discuss these diagnostic modalities with a focus on further advances under investigation.
Collapse
Affiliation(s)
- Haider Ghazanfar
- Division of Gastroenterology, BronxCare Health System, Bronx, NY 10457, USA
| | | | - Raul Reina
- BronxCare Health System, Bronx, NY 10457, USA
| | | | - Ali Ghazanfar
- Fauji Foundation Hospital, Rawalpindi 45000, Pakistan
| | - Harish Patel
- Division of Gastroenterology, BronxCare Health System, Bronx, NY 10457, USA
| |
Collapse
|
24
|
Li Q, Shi G, Li Y, Lu R, Liu Z. Integrated analysis of disulfidoptosis-related genes identifies NRP1 as a novel biomarker promoting proliferation of gastric cancer via glutamine mediated energy metabolism. Discov Oncol 2024; 15:337. [PMID: 39110136 PMCID: PMC11306494 DOI: 10.1007/s12672-024-01217-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024] Open
Abstract
The incidence and mortality of gastric cancer rank fifth and fourth worldwide among all malignancies, respectively. Additionally, disulfidoptosis, a recently identified form of cellular demise, is closely linked to the initiation and advancement of malignancies. This study aims to create a novel signature of disulfidptosis-related genes (DRGs) and to further explore its association with the tumor immune microenvironment. Based on our comprehensive study, a prognostic signature consisting of 31 DRGs in stomach adenocarcinoma (STAD) was identified and characterized. Through the integrative analyses involving gene expression profiling, machine learning algorithms, and Cox regression models, the prognostic significance of these DRGs was demonstrated. Our findings highlight their strong predictive power in assessing overall survival across diverse patient datasets, and their better performance than traditional clinicopathological factors. Moreover, the DRGs signature showed association with the characteristics of the tumor microenvironment, which has implications for the immune modulation and therapeutic strategies in STAD. Specifically, NRP1 emerged as a key DRG with elevated expression in STAD, showing correlation with the advanced stages of diseases and poorer outcomes. Functional studies further revealed the role of NRP1 in promoting STAD cell proliferation through the modulation of glutamine metabolism. Overall, our study underscores the clinical relevance of DRGs as biomarker and potential therapeutic targets in STAD management, providing insights into disease biology and personalized treatments.
Collapse
Affiliation(s)
- Qiuhua Li
- Liaoning University of Traditional Chinese Medicine, No. 79 Chongshan East Road, Shenyang, 110033, Liaoning, People's Republic of China
- Department of Oncology, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, 518000, Guangdong, People's Republic of China
| | - Guofeng Shi
- Department of Oncology, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, 518000, Guangdong, People's Republic of China
| | - Yuebo Li
- Liaoning University of Traditional Chinese Medicine, No. 79 Chongshan East Road, Shenyang, 110033, Liaoning, People's Republic of China
| | - Ren Lu
- Liaoning University of Traditional Chinese Medicine, No. 79 Chongshan East Road, Shenyang, 110033, Liaoning, People's Republic of China.
| | - Zhaozhe Liu
- Department of Oncology, General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China.
| |
Collapse
|
25
|
Bum Lee J, Huang Y, Oya Y, Nutzinger J, LE Ang Y, Sooi K, Chul Cho B, Soo RA. Modulating the gut microbiome in non-small cell lung cancer: Challenges and opportunities. Lung Cancer 2024; 194:107862. [PMID: 38959670 DOI: 10.1016/j.lungcan.2024.107862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/05/2024]
Abstract
Despite the efficacy of immunotherapy in non-small cell lung cancer (NSCLC), the majority of the patients experience relapse with limited subsequent treatment options. Preclinical studies of various epithelial tumors, such as melanoma and NSCLC, have shown that harnessing the gut microbiome resulted in improvement of therapeutic responses to immunotherapy. Is this review, we summarize the role of microbiome, including lung and gut microbiome in the context of NSCLC, provide overview of the mechanisms of microbiome in efficacy and toxicity of chemotherapies and immunotherapies, and address current ongoing clinical trials for NSCLC including fecal microbiota transplantation (FMT) and live biotherapeutic products (LBPs).
Collapse
Affiliation(s)
- Jii Bum Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Yiqing Huang
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Yuko Oya
- Department of Respiratory Medicine, Fujita Health University, Toyoake, Japan
| | - Jorn Nutzinger
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Yvonne LE Ang
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Kenneth Sooi
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Ross A Soo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore.
| |
Collapse
|
26
|
Meidaninikjeh S, Mohammadi P, Elikaei A. Bacteriophages and bacterial extracellular vesicles, threat or opportunity? Life Sci 2024; 350:122749. [PMID: 38821215 DOI: 10.1016/j.lfs.2024.122749] [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: 12/04/2023] [Revised: 03/25/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024]
Abstract
Emergence of antimicrobial-resistant bacteria (AMR) is one of the health major problems worldwide. The scientists are looking for a novel method to treat infectious diseases. Phage therapy is considered a suitable approach for treating infectious diseases. However, there are different challenges in this way. Some biological aspects can probably influence on therapeutic results and further investigations are necessary to reach a successful phage therapy. Bacteriophage activity can influence by bacterial defense system. Bacterial extracellular vesicles (BEVs) are one of the bacterial defense mechanisms which can modify the results of bacteriophage activity. BEVs have the significant roles in the gene transferring, invasion, escape, and spreading of bacteriophages. In this review, the defense mechanisms of bacteria against bacteriophages, especially BEVs secretion, the hidden linkage of BEVs and bacteriophages, and its possible consequences on the bacteriophage activity as well phage therapy will be discussed.
Collapse
Affiliation(s)
- Sepideh Meidaninikjeh
- Department of Microbiology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran.
| | - Parisa Mohammadi
- Department of Microbiology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran; Research Center for Applied Microbiology and Microbial Biotechnology, Alzahra University, Tehran, Iran.
| | - Ameneh Elikaei
- Department of Microbiology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran; Research Center for Applied Microbiology and Microbial Biotechnology, Alzahra University, Tehran, Iran.
| |
Collapse
|
27
|
Yan K, Wang X, Han Y, Tian Y, Niu M, Dong X, Li X, Li H, Sun Y. Simultaneous Detection of Helicobacter pylori and Clarithromycin Resistance Mutations Using RAA-CRISPR/Cas13a Assay. Infect Drug Resist 2024; 17:3001-3010. [PMID: 39045109 PMCID: PMC11265216 DOI: 10.2147/idr.s462963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024] Open
Abstract
Background Infection caused by Helicobacter pylori (H. pylori) affects approximately 50% of the global population. It is a major pathogenic factor for chronic gastritis and gastric cancer. Besides, the resistance to antibiotics such as clarithromycin could reduce the eradication rate. Currently, there is an urgent need for a swift, easy to perform, and highly sensitive detection method for H. pylori and clarithromycin resistance. Methods We used FAM/Digoxin labeled primers to amplify specific H. pylori 23S rRNA fragments by Recombinase Aided Amplification (RAA), and resistance mutations were distinguished using CRISPR/Cas13a system combined with lateral flow strip. Twenty-eight saliva samples were analyzed using qPCR, gene sequencing and this method to evaluate the detection efficiency. Results We developed a simultaneous detection method for H. pylori and clarithromycin resistance mutations named sensitive H. pylori easy-read dual detection (SHIELD). The results showed both A2142G and A2143G mutant DNAs causing clarithromycin resistance could be distinguished from the wild type with a concentration of 50 copies/μL, and no cross-reaction with other 5 common gastrointestinal bacteria was observed. For the detection of H. pylori in 28 saliva samples, the positive predictive value of this method was 100% (19/19) in comparison with qPCR. For detecting clarithromycin resistance, the positive predictive value of this method was 84.6% (11/13) compared with gene sequencing. Conclusion SHIELD assay showed high sensitivity and specificity in detecting H. pylori and clarithromycin resistance mutations. It could be a potential measure in the rapid detection of H. pylori, large-scale screening and guiding clinical medication.
Collapse
Affiliation(s)
- Kuocheng Yan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People’s Republic of China
| | - Xiaohui Wang
- Department of Gastroenterology, the Sixth Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, 100080, People’s Republic of China
| | - Yao Han
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People’s Republic of China
| | - Yuan Tian
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People’s Republic of China
| | - Mengwei Niu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People’s Republic of China
| | - Xue Dong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People’s Republic of China
| | - Xiaowei Li
- Department of Gastroenterology, the Sixth Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, 100080, People’s Republic of China
| | - Hao Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People’s Republic of China
| | - Yansong Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People’s Republic of China
| |
Collapse
|
28
|
Sikaroudi MK, Ebrahimi Z, Darzi M, Shateri Z, Nouri M, Masoodi M, Hejazi M, Shidfar F. Does a High Ratio of Dietary Omega-6/Omega-3 Fatty Acids Increase the Risk of Helicobacter pylori Infection? A Case-Control Study. Clin Nutr Res 2024; 13:176-185. [PMID: 39165292 PMCID: PMC11333148 DOI: 10.7762/cnr.2024.13.3.176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 08/22/2024] Open
Abstract
Helicobacter pylori infection is the cause of 90% of non-cardia gastric cancer. Several dietary elements have been identified as possible contributors to H. pylori infection and its advancement through various pathways. Based on the anti-inflammatory and anti-microbial effects of a diet low in omega-6 and high in omega-3 polyunsaturated fatty acids (PUFAs), this study aimed to assess the ratio of dietary omega-6 to omega-3 PUFAs and the risk of developing H. pylori. The present case-control study was conducted on 150 cases with H. pylori infection and 302 controls. The omega-6 to omega-3 ratio was calculated using food intake information sourced from a validated food frequency questionnaire. Physical activity and demographic data were collected through a related questionnaire. The association between the odds of H. pylori infection and the omega-6 to omega-3 ratio was evaluated using logistic regression models. A p value < 0.05 was considered statistically significant. The findings revealed that individuals in the third tertile had significantly higher odds of H. pylori (odds ratio [OR], 2.10; 95% confidence interval [CI], 1.30-3.40) in the crude model. Furthermore, even after adjusting the potential confounders including sex, age, body mass index, physical activity, energy intake, alcohol, and smoking status, this association remained significant (fully adjusted model: OR, 2.00; 95% CI, 1.17-3.34). Our study revealed a higher ratio of omega-6 to omega-3 was related to a higher likelihood of H. pylori infection. Therefore, it is advisable to maintain a balanced intake of PUFAs in the diet.
Collapse
Affiliation(s)
- Masoumeh Khalighi Sikaroudi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran 141556117, Iran
- Colorectal Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Zohreh Ebrahimi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Melika Darzi
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | - Zainab Shateri
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6135715794, Iran
| | - Mehran Nouri
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol 4717647745, Iran
| | - Mohsen Masoodi
- Colorectal Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Mahdi Hejazi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Farzad Shidfar
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Nutritional Sciences Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
| |
Collapse
|
29
|
Gomaa A, Maacha S, Peng D, Soutto M, Genoula M, Bhat N, Cao L, Zhu S, Castells A, Chen Z, Zaika A, McDonald OG, El-Rifai W. SOX9 is regulated by AURKA in response to Helicobacter pylori infection via EIF4E-mediated cap-dependent translation. Cancer Lett 2024; 593:216939. [PMID: 38729556 PMCID: PMC11175609 DOI: 10.1016/j.canlet.2024.216939] [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: 01/09/2024] [Revised: 04/18/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024]
Abstract
Helicobacter pylori (H. pylori) infection is the main risk factor for gastric cancer. The SRY-Box Transcription Factor 9 (SOX9) serves as a marker of stomach stem cells. We detected strong associations between AURKA and SOX9 expression levels in gastric cancers. Utilizing in vitro and in vivo mouse models, we demonstrated that H. pylori infection induced elevated levels of both AURKA and SOX9 proteins. Notably, the SOX9 protein and transcription activity levels were dependent on AURKA expression. AURKA knockdown led to a reduction in the number and size of gastric gland organoids. Conditional knockout of AURKA in mice resulted in a decrease in SOX9 baseline level in AURKA-knockout gastric glands, accompanied by diminished SOX9 induction following H. pylori infection. We found an AURKA-dependent increase in EIF4E and cap-dependent translation with an AURKA-EIF4E-dependent increase in SOX9 polysomal RNA levels. Immunoprecipitation assays demonstrated binding of AURKA to EIF4E with a decrease in EIF4E ubiquitination. Immunohistochemistry analysis on tissue arrays revealed moderate to strong immunostaining of AURKA and SOX9 with a significant correlation in gastric cancer tissues. These findings elucidate the mechanistic role of AURKA in regulating SOX9 levels via cap-dependent translation in response to H. pylori infection in gastric tumorigenesis.
Collapse
Affiliation(s)
- Ahmed Gomaa
- Department of Surgery, University of Miami Miler School of Medicine, Miami, FL, USA; Department of Veterans Affairs, Miami Healthcare System, Miami, FL, USA
| | - Selma Maacha
- Department of Surgery, University of Miami Miler School of Medicine, Miami, FL, USA
| | - Dunfa Peng
- Department of Surgery, University of Miami Miler School of Medicine, Miami, FL, USA
| | - Mohammed Soutto
- Department of Surgery, University of Miami Miler School of Medicine, Miami, FL, USA; Department of Veterans Affairs, Miami Healthcare System, Miami, FL, USA
| | - Melanie Genoula
- Department of Surgery, University of Miami Miler School of Medicine, Miami, FL, USA
| | - Nadeem Bhat
- Department of Surgery, University of Miami Miler School of Medicine, Miami, FL, USA
| | - Longlong Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shoumin Zhu
- Department of Surgery, University of Miami Miler School of Medicine, Miami, FL, USA
| | - Antoni Castells
- Gastroenterology Department, Hospital Clinic, CIBERehd, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Zhibin Chen
- Department of Microbiology and Immunology, University of Miami Miler School of Medicine, Miami, FL, USA
| | - Alexander Zaika
- Department of Surgery, University of Miami Miler School of Medicine, Miami, FL, USA; Department of Veterans Affairs, Miami Healthcare System, Miami, FL, USA
| | - Oliver G McDonald
- Department of Pathology, University of Miami Miler School of Medicine, Miami, FL, USA
| | - Wael El-Rifai
- Department of Surgery, University of Miami Miler School of Medicine, Miami, FL, USA; Department of Veterans Affairs, Miami Healthcare System, Miami, FL, USA.
| |
Collapse
|
30
|
Sedarat Z, Taylor-Robinson AW. Helicobacter pylori Outer Membrane Proteins and Virulence Factors: Potential Targets for Novel Therapies and Vaccines. Pathogens 2024; 13:392. [PMID: 38787244 PMCID: PMC11124246 DOI: 10.3390/pathogens13050392] [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: 02/26/2024] [Revised: 04/12/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
Helicobacter pylori is a gastric oncopathogen that infects over half of the world's human population. It is a Gram-negative, microaerophilic, helix-shaped bacterium that is equipped with flagella, which provide high motility. Colonization of the stomach is asymptomatic in up to 90% of people but is a recognized risk factor for developing various gastric disorders such as gastric ulcers, gastric cancer and gastritis. Invasion of the human stomach occurs via numerous virulence factors such as CagA and VacA. Similarly, outer membrane proteins (OMPs) play an important role in H. pylori pathogenicity as a means to adapt to the epithelial environment and thereby facilitate infection. While some OMPs are porins, others are adhesins. The epithelial cell receptors SabA, BabA, AlpA, OipA, HopQ and HopZ have been extensively researched to evaluate their epidemiology, structure, role and genes. Moreover, numerous studies have been performed to seek to understand the complex relationship between these factors and gastric diseases. Associations exist between different H. pylori virulence factors, the co-expression of which appears to boost the pathogenicity of the bacterium. Improved knowledge of OMPs is a major step towards combatting this global disease. Here, we provide a current overview of different H. pylori OMPs and discuss their pathogenicity, epidemiology and correlation with various gastric diseases.
Collapse
Affiliation(s)
- Zahra Sedarat
- Cellular & Molecular Research Centre, Shahrekord University of Medical Sciences, Shahrekord 8813833435, Iran;
| | - Andrew W. Taylor-Robinson
- College of Health Sciences, VinUniversity, Gia Lam District, Hanoi 67000, Vietnam
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 1904, USA
| |
Collapse
|
31
|
Jenner A, Garcia-Saez AJ. The regulation of the apoptotic pore-An immunological tightrope walk. Adv Immunol 2024; 162:59-108. [PMID: 38866439 DOI: 10.1016/bs.ai.2024.02.004] [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: 06/14/2024]
Abstract
Apoptotic pore formation in mitochondria is the pivotal point for cell death during mitochondrial apoptosis. It is regulated by BCL-2 family proteins in response to various cellular stress triggers and mediates mitochondrial outer membrane permeabilization (MOMP). This allows the release of mitochondrial contents into the cytosol, which triggers rapid cell death and clearance through the activation of caspases. However, under conditions of low caspase activity, the mitochondrial contents released into the cytosol through apoptotic pores serve as inflammatory signals and activate various inflammatory responses. In this chapter, we discuss how the formation of the apoptotic pore is regulated by BCL-2 proteins as well as other cellular or mitochondrial proteins and membrane lipids. Moreover, we highlight the importance of sublethal MOMP in the regulation of mitochondrial-activated inflammation and discuss its physiological consequences in the context of pathogen infection and disease and how it can potentially be exploited therapeutically, for example to improve cancer treatment.
Collapse
Affiliation(s)
- Andreas Jenner
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Ana J Garcia-Saez
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
| |
Collapse
|
32
|
Bali P, Lozano-Pope I, Hernandez J, Estrada MV, Corr M, Turner MA, Bouvet M, Benner C, Obonyo M. TRIF-IFN-I pathway in Helicobacter-induced gastric cancer in an accelerated murine disease model and patient biopsies. iScience 2024; 27:109457. [PMID: 38558931 PMCID: PMC10981133 DOI: 10.1016/j.isci.2024.109457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/20/2023] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
Helicobacter pylori (H. pylori) infection is a known cause of many digestive diseases, including gastritis, peptic ulcers, and gastric cancer. However, the underlying mechanisms by which H. pylori infection triggers these disorders are still not clearly understood. Gastric cancer is a slow progressing disease, which makes it difficult to study. We have developed an accelerated disease progression mouse model, which leverages mice deficient in the myeloid differentiation primary response 88 gene (Myd88-/-) infected with Helicobacter felis (H. felis). Using this model and gastric biopsy samples from patients, we report that activation of the Toll/interleukin-1 receptor (TIR)-domain-containing adaptor inducing interferon-β (TRIF)-type I interferon (IFN-I) signaling pathway promotes Helicobacter-induced disease progression toward severe gastric pathology and gastric cancer development. Further, results implicated downstream targets of this pathway in disease pathogenesis. These findings may facilitate stratification of Helicobacter-infected patients and thus enable treatment prioritization of patients.
Collapse
Affiliation(s)
- Prerna Bali
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Ivonne Lozano-Pope
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jonathan Hernandez
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Monica V. Estrada
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Maripat Corr
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Michael A. Turner
- Department of Surgery, University of California, San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Michael Bouvet
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
- Department of Surgery, University of California, San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Christopher Benner
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Marygorret Obonyo
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
33
|
Ma T, Xu F, Hou Y, Shu Y, Zhao Z, Zhang Y, Bai L, Feng L, Zhong L. SETDB1: Progress and prospects in cancer treatment potential and inhibitor research. Bioorg Chem 2024; 145:107219. [PMID: 38377821 DOI: 10.1016/j.bioorg.2024.107219] [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: 12/27/2023] [Revised: 02/03/2024] [Accepted: 02/14/2024] [Indexed: 02/22/2024]
Abstract
SET domain bifurcated methyltransferase 1 (SETDB1) serves as a histone lysine methyltransferase, catalyzing the di- and tri-methylation of histone H3K9. Mounting evidence indicates that the abnormal expression or activity of SETDB1, either through amplification or mutation, plays a crucial role in tumorigenesis and progression. This is particularly evident in the context of tumor immune evasion and resistance to immune checkpoint blockade therapy. Furthermore, there is a robust association between SETDB1 dysregulation and an unfavorable prognosis across various types of tumors. The oncogenic role of SETDB1 primarily arises from its methyltransferase function, which contributes to the establishment of a condensed and transcriptionally inactive heterochromatin state. This results in the inactivation of genes that typically hinder cancer development and silencing of retrotransposons that could potentially trigger an immune response. These findings underscore the substantial potential for SETDB1 as an anti-tumor therapeutic target. Nevertheless, despite significant strides in recent years in tumor biology research, challenges persist in SETDB1-targeted therapy. To better facilitate the development of anti-tumor therapy targeting SETDB1, we have conducted a comprehensive review of SETDB1 in this account. We present the structure and function of SETDB1, its role in various tumors and immune regulation, as well as the advancements made in SETDB1 antagonists. Furthermore, we discuss the challenges encountered and provide perspectives for the development of SETDB1-targeted anti-tumor therapy.
Collapse
Affiliation(s)
- Tingnan Ma
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Feifei Xu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China; State Key Laboratory of Southwestern Chinese Medicine Resources; Key Laboratory of Standardization of Chinese Herbal Medicines of Ministry of Education, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yingying Hou
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Yongquan Shu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Zhipeng Zhao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Yaru Zhang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Lan Bai
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China.
| | - Lu Feng
- Department of Emergency, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610041, China.
| | - Lei Zhong
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China.
| |
Collapse
|
34
|
Malfertheiner P, Schulz C, Hunt RH. Helicobacter pylori Infection: A 40-Year Journey through Shifting the Paradigm to Transforming the Management. Dig Dis 2024; 42:299-308. [PMID: 38447558 DOI: 10.1159/000538079] [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: 11/05/2023] [Accepted: 02/26/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Helicobacter pylori (H. pylori) was discovered 40 years ago and has set a milestone in human medicine. The discovery led to rejection of the dogma of the acidic stomach as a sterile organ and requested to rewrite the chapters on gastric pathophysiology and gastroduodenal diseases. SUMMARY Over a period of 40 years following the discovery, more than 50,000 articles can be retrieved in PubMed as of today and illustrate the amount and the intensity of research around the role of this bacterium. H. pylori emerged as cause of chronic gastritis and principal cause of peptic ulcer disease (PUD). Eradication of H. pylori became standard of care in management in PUD. The importance of this was highlighted in 2005 with the Nobel Prize in Medicine awarded to Barry Marshall and Robin Warren. H. pylori became eventually recognized for its oncogenic potential in the stomach and as the main risk factor for gastric cancer development. KEY MESSAGES H. pylori gastritis is defined as infectious disease and requires therapy in all infected individuals. Strategies of gastric cancer prevention and development of therapies to overcome the increasing antibiotic resistance are main targets in clinical research of today.
Collapse
Affiliation(s)
- Peter Malfertheiner
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Medical Department 2, LMU, Munich, Germany
| | - Christian Schulz
- Medical Department 2, LMU, Munich, Germany,
- Partner Site Munich, DZIF, Braunschweig, Germany,
| | - Richard H Hunt
- Farncombe Family Digestive Health Research Institute and Division of Gastroenterology, Department of Medicine, McMaster University Faculty of Health Sciences, Hamilton, Ontario, Canada
| |
Collapse
|
35
|
Apoorva E, Jacob R, Rao DN, Kumar S. Helicobacter pylori enhances HLA-C expression in the human gastric adenocarcinoma cells AGS and can protect them from the cytotoxicity of natural killer cells. Helicobacter 2024; 29:e13069. [PMID: 38516860 DOI: 10.1111/hel.13069] [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: 12/14/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 03/23/2024]
Abstract
Helicobacter pylori (H. pylori) seems to play causative roles in gastric cancers. H. pylori has also been detected in established gastric cancers. How the presence of H. pylori modulates immune response to the cancer is unclear. The cytotoxicity of natural killer (NK) cells, toward infected or malignant cells, is controlled by the repertoire of activating and inhibitory receptors expressed on their surface. Here, we studied H. pylori-induced changes in the expression of ligands, of activating and inhibitory receptors of NK cells, in the gastric adenocarcinoma AGS cells, and their impacts on NK cell responses. AGS cells lacked or had low surface expression of the class I major histocompatibility complex (MHC-I) molecules HLA-E and HLA-C-ligands of the major NK cell inhibitory receptors NKG2A and killer-cell Ig-like receptor (KIR), respectively. However, AGS cells had high surface expression of ligands of activating receptors DNAM-1 and CD2, and of the adhesion molecules LFA-1. Consistently, AGS cells were sensitive to killing by NK cells despite the expression of inhibitory KIR on NK cells. Furthermore, H. pylori enhanced HLA-C surface expression on AGS cells. H. pylori infection enhanced HLA-C protein synthesis, which could explain H. pylori-induced HLA-C surface expression. H. pylori infection enhanced HLA-C surface expression also in the hepatoma Huh7 and HepG2 cells. Furthermore, H. pylori-induced HLA-C surface expression on AGS cells promoted inhibition of NK cells by KIR, and thereby protected AGS cells from NK cell cytotoxicity. These results suggest that H. pylori enhances HLA-C expression in host cells and protects them from the cytotoxic attack of NK cells expressing HLA-C-specific inhibitory receptors.
Collapse
Affiliation(s)
- Etikala Apoorva
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rini Jacob
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, Telangana, India
| | - Desirazu N Rao
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Santosh Kumar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
36
|
Zhou X, Rahman MM, Bonny SQ, Xin Y, Liddelow N, Khan MF, Tikhomirova A, Homman-Ludiye J, Roujeinikova A. Pal power: Demonstration of the functional association of the Helicobacter pylori flagellar motor with peptidoglycan-associated lipoprotein (Pal) and its preliminary crystallographic analysis. Biosci Trends 2024; 17:491-498. [PMID: 38072447 DOI: 10.5582/bst.2023.01278] [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: 02/02/2024]
Abstract
The bacterial flagellar motor is a molecular nanomachine, the assembly and regulation of which requires many accessory proteins. Their identity, structure and function are often discovered through characterisation of mutants with impaired motility. Here, we demonstrate the functional association of the Helicobacter pylori peptidoglycan-associated lipoprotein (HpPal) with the flagellar motor by analysing the motility phenotype of the ∆pal mutant, and present the results of the preliminary X-ray crystallographic analysis of its globular C-terminal domain HpPal-C. Purified HpPal-C behaved as a dimer in solution. Crystals of HpPal-C were grown by the hanging drop vapour diffusion method using medium molecular weight polyethylene glycol (PEG) Smear as the precipitating agent. The crystals belong to the primitive orthorhombic space group P1 with unit cell parameters a = 50.7, b = 63.0, c = 75.1 Å. X-ray diffraction data were collected to 1.8 Å resolution on the Australian Synchrotron beamline MX2. Calculation of the Matthews coefficient (VM=2.24 Å3/Da) and molecular replacement showed that the asymmetric unit contains two protein subunits. This study is an important step towards elucidation of the non-canonical role of H. pylori Pal in the regulation, or function of, the flagellar motor.
Collapse
Affiliation(s)
- Xiaotian Zhou
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Mohammad M Rahman
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Sharmin Q Bonny
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Yue Xin
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Nikki Liddelow
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - Mohammad F Khan
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Alexandra Tikhomirova
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Jihane Homman-Ludiye
- Monash Micro Imaging, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Anna Roujeinikova
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
37
|
Zimmerman EH, Ramsey EL, Hunter KE, Villadelgado SM, Phillips CM, Shipman RT, Forsyth MH. The Helicobacter pylori methylome is acid-responsive due to regulation by the two-component system ArsRS and the type I DNA methyltransferase HsdM1 (HP0463). J Bacteriol 2024; 206:e0030923. [PMID: 38179929 PMCID: PMC10810217 DOI: 10.1128/jb.00309-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 12/14/2023] [Indexed: 01/06/2024] Open
Abstract
In addition to its role in genome protection, DNA methylation can regulate gene expression. In this study, we characterized the impact of acidity, phase variation, and the ArsRS TCS on the expression of the Type I m6A DNA methyltransferase HsdM1 (HP0463) of Helicobacter pylori 26695 and their subsequent effects on the methylome. Transcription of hsdM1 increases at least fourfold in the absence of the sensory histidine kinase ArsS, the major acid-sensing protein of H. pylori. hsdM1 exists in the phase-variable operon hsdR1-hsdM1. Phase-locking hsdR1 (HP0464), the restriction endonuclease gene, has significant impacts on the transcription of hsdM1. To determine the impacts of methyltransferase transcription patterns on the methylome, we conducted methylome sequencing on samples cultured at pH 7 or pH 5. We found differentially methylated motifs between these growth conditions and that deletions of arsS and/or hsdM1 interfere with the epigenetic acid response. Deletion of arsS leads to altered activity of HsdM1 and multiple other methyltransferases under both pH conditions indicating that the ArsRS TCS, in addition to direct effects on regulon transcription during acid acclimation, may also indirectly impact gene expression via regulation of the methylome. We determined the target motif of HsdM1 (HP0463) to be the complementary bipartite sequence pair 5'-TCAm6AVN6TGY-3' and 3'-AGTN6GAm6ACA-5'. This complex regulation of DNA methyltransferases, and thus differential methylation patterns, may have implications for the decades-long persistent infection by H. pylori. IMPORTANCE This study expands the possibilities for complex, epigenomic regulation in Helicobacter pylori. We demonstrate that the H. pylori methylome is plastic and acid sensitive via the two-component system ArsRS and the DNA methyltransferase HsdM1. The control of a methyltransferase by ArsRS may allow for a layered response to changing acidity. Likely, an early response whereby ArsR~P affects regulon expression, including the methyltransferase hsdM1. Then, a somewhat later effect as the altered methylome, due to altered HsdM1 expression, subsequently alters the expression of other genes involved in acclimation. The intermediate methylation of certain motifs supports the hypothesis that methyltransferases play a regulatory role. Untangling this additional web of regulation could play a key role in understanding H. pylori colonization and persistence.
Collapse
Affiliation(s)
| | - Erin L. Ramsey
- Department of Biology, William & Mary, Williamsburg, Virginia, USA
| | | | | | | | - Ryan T. Shipman
- Department of Biology, William & Mary, Williamsburg, Virginia, USA
| | - Mark H. Forsyth
- Department of Biology, William & Mary, Williamsburg, Virginia, USA
| |
Collapse
|
38
|
Shuman JHB, Lin AS, Westland MD, Bryant KN, Piazuelo MB, Reyzer ML, Judd AM, McDonald WH, McClain MS, Schey KL, Algood HMS, Cover TL. Remodeling of the gastric environment in Helicobacter pylori-induced atrophic gastritis. mSystems 2024; 9:e0109823. [PMID: 38059647 PMCID: PMC10805037 DOI: 10.1128/msystems.01098-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 12/08/2023] Open
Abstract
Helicobacter pylori colonization of the human stomach is a strong risk factor for gastric cancer. To investigate H. pylori-induced gastric molecular alterations, we used a Mongolian gerbil model of gastric carcinogenesis. Histologic evaluation revealed varying levels of atrophic gastritis (a premalignant condition characterized by parietal and chief cell loss) in H. pylori-infected animals, and transcriptional profiling revealed a loss of markers for these cell types. We then assessed the spatial distribution and relative abundance of proteins in the gastric tissues using imaging mass spectrometry and liquid chromatography with tandem mass spectrometry. We detected striking differences in the protein content of corpus and antrum tissues. Four hundred ninety-two proteins were preferentially localized to the corpus in uninfected animals. The abundance of 91 of these proteins was reduced in H. pylori-infected corpus tissues exhibiting atrophic gastritis compared with infected corpus tissues exhibiting non-atrophic gastritis or uninfected corpus tissues; these included numerous proteins with metabolic functions. Fifty proteins localized to the corpus in uninfected animals were diffusely delocalized throughout the stomach in infected tissues with atrophic gastritis; these included numerous proteins with roles in protein processing. The corresponding alterations were not detected in animals infected with a H. pylori ∆cagT mutant (lacking Cag type IV secretion system activity). These results indicate that H. pylori can cause loss of proteins normally localized to the gastric corpus as well as diffuse delocalization of corpus-specific proteins, resulting in marked changes in the normal gastric molecular partitioning into distinct corpus and antrum regions.IMPORTANCEA normal stomach is organized into distinct regions known as the corpus and antrum, which have different functions, cell types, and gland architectures. Previous studies have primarily used histologic methods to differentiate these regions and detect H. pylori-induced alterations leading to stomach cancer. In this study, we investigated H. pylori-induced gastric molecular alterations in a Mongolian gerbil model of carcinogenesis. We report the detection of numerous proteins that are preferentially localized to the gastric corpus but not the antrum in a normal stomach. We show that stomachs with H. pylori-induced atrophic gastritis (a precancerous condition characterized by the loss of specialized cell types) exhibit marked changes in the abundance and localization of proteins normally localized to the gastric corpus. These results provide new insights into H. pylori-induced gastric molecular alterations that are associated with the development of stomach cancer.
Collapse
Affiliation(s)
- Jennifer H. B. Shuman
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Aung Soe Lin
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mandy D. Westland
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kaeli N. Bryant
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - M. Blanca Piazuelo
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Michelle L. Reyzer
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Audra M. Judd
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - W. Hayes McDonald
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Mark S. McClain
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kevin L. Schey
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Holly M. S. Algood
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Timothy L. Cover
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
39
|
Wu H, Xie X, Tang Q, Huang T, Tang X, Jiao B, Wang R, Zhu X, Ye X, Ma H, Li X. Epiberberine inhibits Helicobacter pylori and reduces host apoptosis and inflammatory damage by down-regulating urease expression. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117046. [PMID: 37586440 DOI: 10.1016/j.jep.2023.117046] [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: 07/24/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE With dramatically increasing antibiotic resistance in Helicobacter pylori (H. pylori), it is urgent to find alternative therapeutic agents. Rhizoma Coptidis is a traditional Chinese medicine for gastrointestinal diseases and shows excellent anti-H. pylori effect. Epiberberine (EPI), as one of the major alkaloids of Rhizoma Coptidis, has been reported to have urease-inhibiting activity, but its scavenging effect on H. pylori and the potential mechanism remain unclear. AIM OF THE STUDY To investigate the inhibitory effect of EPI on H. pylori and explore its multi-action on Helicobacter pylori urease (HPU). MATERIALS AND METHODS Using minimum inhibitory concentration (MIC) assay, minimum bactericidal concentration (MBC) assay, growth inhibition kinetics assay, bacterial resistance development, transmission electron microscope (TEM) assay, and animal experiments to investigate the inhibitory effect of EPI on H. pylori in vitro and in vivo. Using the Berthelot method, molecular docking and thermal displacement experiments to verify that EPI inhibits urease activity by interacting with HPU. Using transcriptome data, Real-Time PCR (RT-PCR) experiments to investigate the alterations in the expression of urease subunit ureB gene after EPI treatment. Using MTT cell viability assay, Hoechst 33342 staining method, JC-1 reagent detection method, western blot experiments, and Griess method to investigate the anti-apoptosis and anti-inflammation actions of EPI on gastric epithelial cells (GES-1) induced by HPU. RESULTS In vitro experiments proved that EPI has significant anti-H. pylori activity without drug resistance, induces H. pylori fragmentation and apoptosis. In vivo experiments showed that EPI has a certain clearance effect of H. pylori, and can reduce gastric inflammation caused by H. pylori infection. Transcriptome data, RT-PCR experiments, and other experiments demonstrate that EPI has a triple effect: (1) inhibiting the expression of HPU subunits ureB, (2) directly inhibiting urease activity by interacting with HPU, and (3) inhibiting HPU-induced apoptosis and inflammation in GES-1. CONCLUSIONS EPI is an excellent anti-H. pylori agent and reduces host apoptosis and inflammation by inhibiting the activity of urease and down-regulating the expression of ureB.
Collapse
Affiliation(s)
- Huimin Wu
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China; Chongqing Haiwang Biological Engineering Co., Chongqing, 409900, China.
| | - Xinrui Xie
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Qin Tang
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China.
| | - Ting Huang
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China.
| | - Xiang Tang
- School of Life Sciences, Southwest University, Chongqing, 400715, China.
| | - Baihua Jiao
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China.
| | - Rui Wang
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China.
| | - Xinhu Zhu
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China.
| | - Xiaoli Ye
- School of Life Sciences, Southwest University, Chongqing, 400715, China.
| | - Hang Ma
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China.
| | - Xuegang Li
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
40
|
Ko KP. Risk Factors of Gastric Cancer and Lifestyle Modification for Prevention. J Gastric Cancer 2024; 24:99-107. [PMID: 38225769 PMCID: PMC10774756 DOI: 10.5230/jgc.2024.24.e10] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/17/2024] Open
Abstract
Gastric cancer has been consistently decreasing worldwide, whereas cardia gastric cancer is on the rise. This indicates that the exposure rates to epidemiological causes are changing. In this study, we aim to review the risk factors for gastric cancer with respect to cardia and non-cardia types. One of the most significant risk factors for gastric cancer is Helicobacter pylori infection. H. pylori infection is known as a risk factor for non-cardia gastric cancer, and there have been results indicating that H. pylori infection is not associated with cardia gastric cancer. However, in the East Asian region, there is epidemiological evidence suggesting that H. pylori infection might be a risk factor for cardia gastric cancer. Smoking and alcohol consumption are known risk factors for gastric cancer, regardless of anatomical location. Obesity is considered a factor in the development of cardia gastric cancer. However, further research is needed to understand the specific relationship with non-cardia gastric cancer. The consumption of high-salt and processed meat is more distinctly associated with non-cardia gastric cancer than in cardia gastric cancer. In addition to these factors, exposure to chemicals and radiation are considered risk factors for gastric cancer. Primary prevention of gastric cancer involves eliminating or avoiding risk factors such as H. pylori eradication and adopting a healthy lifestyle, including quitting smoking, reducing alcohol consumption, maintaining a healthy weight, and having a low-salt diet.
Collapse
Affiliation(s)
- Kwang-Pil Ko
- Clinical Preventive Medicine Center, Seoul National University Bundang Hospital, Seongnam, Korea.
| |
Collapse
|
41
|
Ali F, Alom S, Ali SR, Kondoli B, Sadhu P, Borah C, Kakoti BB, Ghosh SK, Shakya A, Ahmed AB, Singh UP, Bhat HR. Ebselen: A Review on its Synthesis, Derivatives, Anticancer Efficacy and Utility in Combating SARS-COV-2. Mini Rev Med Chem 2024; 24:1203-1225. [PMID: 37711004 DOI: 10.2174/1389557523666230914103339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/26/2023] [Accepted: 06/16/2023] [Indexed: 09/16/2023]
Abstract
Ebselen is a selenoorganic chiral compound with antioxidant properties comparable to glutathione peroxidase. It is also known as 2-phenyl-1,2-benzisoselenazol-3(2H)-one. In studies examining its numerous pharmacological activities, including antioxidant, anticancer, antiviral, and anti- Alzheimer's, ebselen has demonstrated promising results. This review's primary objective was to emphasize the numerous synthesis pathways of ebselen and their efficacy in fighting cancer. The data were collected from multiple sources, including Scopus, PubMed, Google Scholar, Web of Science, and Publons. The starting reagents for the synthesis of ebselen are 2-aminobenzoic acid and N-phenyl benzamide. It was discovered that ebselen has the ability to initiate apoptosis in malignant cells and prevent the formation of new cancer cells by scavenging free radicals. In addition, ebselen increases tumor cell susceptibility to apoptosis by inhibiting TNF-α mediated NF-kB activation. Ebselen can inhibit both doxorubicin and daunorubicin-induced cardiotoxicity. Allopurinol and ebselen administered orally can be used to suppress renal ototoxicity and nephrotoxicity. Due to excessive administration, diclofenac can induce malignancy of the gastrointestinal tract, which ebselen can effectively suppress. Recent research has demonstrated ebselen to inhibit viral function by binding to cysteinecontaining catalytic domains of various viral proteases. It was discovered that ebselen could inhibit the catalytic dyad function of Mpro by forming an irreversible covalent bond between Se and Cys145, thereby altering protease function and inhibiting SARS-CoV-2. Ebselen may also inhibit the activation of endosomal NADPH oxidase of vascular endothelial cells, which is believed to be required for thrombotic complications in COVID-19. In this review, we have included various studies conducted on the anticancer effect of ebselen as well as its inhibition of SARS-CoV-2.
Collapse
Affiliation(s)
- Farak Ali
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, 786004, India
- Girijananda Chowdhury Institute of Pharmaceutical Science, Tezpur Medical College and Hospital, Tezpur, Sonitpur Assam, 784501,India
| | - Shahnaz Alom
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, 786004, India
- Girijananda Chowdhury Institute of Pharmaceutical Science, Tezpur Medical College and Hospital, Tezpur, Sonitpur Assam, 784501,India
| | - Sheikh Rezzak Ali
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, 786004, India
| | - Biswanarayan Kondoli
- Department of Pharmacy, Tripura University, Suryamani Nagar, Agartala, Tripura 799022, India
| | - Prativa Sadhu
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, 786004, India
| | - Chinmoyee Borah
- Girijananda Chowdhury Institute of Pharmaceutical Science, Guwahati, Kamrup, Assam, 781017, India
| | - Bibhuti Bushan Kakoti
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, 786004, India
| | - Surajit Kumar Ghosh
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, 786004, India
| | - Anshul Shakya
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, 786004, India
| | - Abdul Baquee Ahmed
- Girijananda Chowdhury Institute of Pharmaceutical Science,Tezpur Medical College and Hospital, Tezpur, Sonitpur-784501, Assam, India
| | - Udaya Pratap Singh
- Drug Design & Discovery Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, Uttar Pradesh, 211007, India
| | - Hans Raj Bhat
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, 786004, India
| |
Collapse
|
42
|
Abstract
Our perception of microbes has considerably changed since the recognition of their pathogenic potential in the 19th century. The discovery of antibiotics and their subsequent widespread adoption have substantially altered the landscape of medicine, providing us with treatment options for many infectious diseases and enabling the deployment of previously risky interventions (eg, surgical procedures and chemotherapy), while also leading to the rise of AMR. The latter is commonly viewed as the predominant downside of antibiotic use. However, with the increasing recognition that all metazoan organisms rely on a community of microbes (the microbiota) for normal development and for most physiologic processes, the negative impacts of antibiotic use now extend well beyond AMR. Using the iceberg as a metaphor, we argue that the effects of antibiotics on AMR represent the tip of the iceberg, with much greater repercussions stemming from their role in the rise of so-called noncommunicable diseases (including obesity, diabetes, allergic and autoimmune diseases, neurodevelopmental disorders, and certain cancers). We highlight some of the emerging science around the intersection of the microbiome, antibiotic use, and health (including biological costs and future therapeutic avenues), and we advocate a more nuanced approach in evaluating the impacts of proposed antibiotic use, especially in the setting of preexposure and postexposure prophylaxis.
Collapse
Affiliation(s)
- Louis-Patrick Haraoui
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Centre de Recherche Charles-Le Moyne, Greenfield Park, Quebec, Canada
- Humans & the Microbiome Program, Canadian Institute for Advanced Research, Toronto, Ontario, Canada
| | - Martin J Blaser
- Humans & the Microbiome Program, Canadian Institute for Advanced Research, Toronto, Ontario, Canada
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey, USA
- Robert Wood Johnson School of Medicine, Departments of Medicine and Pathology & Laboratory Medicine, New Brunswick, New Jersey, USA
| |
Collapse
|
43
|
Ji J, Jin W, Liu S, Jiao Z, Li X. Probiotics, prebiotics, and postbiotics in health and disease. MedComm (Beijing) 2023; 4:e420. [PMID: 37929014 PMCID: PMC10625129 DOI: 10.1002/mco2.420] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/30/2023] [Accepted: 10/13/2023] [Indexed: 11/07/2023] Open
Abstract
The gut microbiota and its homeostasis play a crucial role in human health. However, for some diseases related to the gut microbiota, current traditional medicines can only relieve symptoms, and it is difficult to solve the root causes or even cause side effects like disturbances in the gut microbiota. Increasing clinical studies and evidences have demonstrated that probiotics, prebiotics, and postbiotics can prevent and treat various diseases, but currently they can only be used as dietary supplements rather than medicines, which restricts the application of probiotics in the field of medicine. Here, this review analyzes the importance of gut microbiota in human health and the current problems of traditional medicines, and systematically summarizes the effectiveness and mechanisms of probiotics, prebiotics, and postbiotics in maintaining health and treating diseases based on animal models and clinical trials. And based on current research outcomes and development trends in this field, the challenges and prospects of their clinical application in maintaining health, alleviating and treating diseases are analyzed. It is hoped to promote the application of probiotics, prebiotics, and postbiotics in disease treatment and open up new frontiers in probiotic research.
Collapse
Affiliation(s)
- Jing Ji
- MOE Key Laboratory of Cell Activities and Stress AdaptationsSchool of Life SciencesLanzhou UniversityLanzhouGansuChina
| | - Weilin Jin
- Institute of Cancer NeuroscienceMedical Frontier Innovation Research CenterThe First Hospital of Lanzhou UniversityThe First Clinical Medical College of Lanzhou UniversityLanzhouGansuChina
| | - Shuang‐Jiang Liu
- State Key Laboratory of Microbial ResourcesInstitute of MicrobiologyChinese Academy of SciencesBeijingChina
| | - Zuoyi Jiao
- Cuiying Biomedical Research CenterThe Second Hospital of Lanzhou UniversityLanzhouGansuChina
| | - Xiangkai Li
- MOE Key Laboratory of Cell Activities and Stress AdaptationsSchool of Life SciencesLanzhou UniversityLanzhouGansuChina
| |
Collapse
|
44
|
Iqbal W, Wang Y, Sun P, Zhou X. Modeling Liver Development and Disease in a Dish. Int J Mol Sci 2023; 24:15921. [PMID: 37958904 PMCID: PMC10650907 DOI: 10.3390/ijms242115921] [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: 09/12/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Historically, biological research has relied primarily on animal models. While this led to the understanding of numerous human biological processes, inherent species-specific differences make it difficult to answer certain liver-related developmental and disease-specific questions. The advent of 3D organoid models that are either derived from pluripotent stem cells or generated from healthy or diseased tissue-derived stem cells have made it possible to recapitulate the biological aspects of human organs. Organoid technology has been instrumental in understanding the disease mechanism and complements animal models. This review underscores the advances in organoid technology and specifically how liver organoids are used to better understand human-specific biological processes in development and disease. We also discuss advances made in the application of organoid models in drug screening and personalized medicine.
Collapse
Affiliation(s)
- Waqas Iqbal
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China; (W.I.); (Y.W.); (P.S.)
- Research Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Yaru Wang
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China; (W.I.); (Y.W.); (P.S.)
- Research Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Pingnan Sun
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China; (W.I.); (Y.W.); (P.S.)
- Research Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Xiaoling Zhou
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China; (W.I.); (Y.W.); (P.S.)
- Research Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
45
|
Burkett WC, Clontz AD, Keku TO, Bae-Jump V. The interplay of obesity, microbiome dynamics, and innovative anti-obesity strategies in the context of endometrial cancer progression and therapeutic approaches. Biochim Biophys Acta Rev Cancer 2023; 1878:189000. [PMID: 37844671 DOI: 10.1016/j.bbcan.2023.189000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 10/09/2023] [Accepted: 10/09/2023] [Indexed: 10/18/2023]
Abstract
Endometrial cancer (EC) is the most common gynecologic malignancy in the United States, and its incidence and mortality are rising. Obesity is more tightly associated with EC than any other cancer. Thus, the rising prevalence of obesity and associated risk factors, including diabetes and insulin resistance, cause alarm. The metabolic derangements of obesity increase the bioavailability of estrogen, hyperinsulinemia, and inflammation in a complex system with direct and indirect effects on the endometrium, resulting in proliferation and, ultimately, carcinogenesis. In addition, the gut dysbiosis associated with obesity helps contribute to these metabolic derangements, priming an individual for developing EC and perhaps affecting treatment efficacy. More recent studies are beginning to explore obesity's effect on the local tumor microbiome of EC and its role in carcinogenesis. Significant and sustained weight loss in individuals can considerably decrease the risk of EC, likely through reversal of the altered metabolism and dysbiosis resulting obesity. Bariatric surgery is the gold standard for successful weight loss and highlights how reversing of the systemic effects of obesity can reduce EC risk. However, the current limited availability, knowledge, and imposed stigma of bariatric surgery prohibits population-level reductions in EC. Therefore, effective and maintainable non-surgical dietary and pharmacologic interventions are needed.
Collapse
Affiliation(s)
- Wesley C Burkett
- University of North Carolina at Chapel Hill, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, United States of America.
| | - Angela D Clontz
- University of North Carolina at Chapel Hill, Nutrition Research Institute, United States of America.
| | - Temitope O Keku
- University of North Carolina at Chapel Hill, Department of Medicine, Center for Gastrointestinal Biology and Disease, United States of America.
| | - Victoria Bae-Jump
- University of North Carolina at Chapel Hill, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, United States of America; University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, United States of America.
| |
Collapse
|
46
|
Wanibuchi K, Hosoda K, Amgalanbaatar A, Ihara M, Takezawa M, Sakai Y, Masui H, Shoji M, Hayashi S, Shimomura H. Aspects for development of novel antibacterial medicines using a vitamin D 3 decomposition product in Helicobacter pylori infection. J Antibiot (Tokyo) 2023; 76:665-672. [PMID: 37658133 DOI: 10.1038/s41429-023-00651-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/05/2023] [Accepted: 08/10/2023] [Indexed: 09/03/2023]
Abstract
A previous study by our group demonstrated that a vitamin D3 decomposition product (VDP1) acts as the selective bactericidal substance on Helicobacter pylori. VDP1 is an indene compound modified with a carbonyl and an alkyl. The alkyl of VDP1 turned out to be a mandatory structure to exert effective bactericidal action on H. pylori. Meanwhile, it still remains to be clarified as to how influence the alteration of the carbonyl in VDP1 has on the anti-H. pylori activity. In this study, we synthesized novel VDP1 derivatives that replaced the carbonyl of VDP1 by various functional groups and investigated the antibacterial action of the VDP1 derivatives on H. pylori. VDP1 derivatives retaining either a hydroxy (VD3-1) or an acetic ester (VD3-3) exhibited more effective bactericidal action to H. pylori than VDP1. The replacement of the carbonyl of VDP1 by either an allyl acetate (VD3-2) or an acrylic acid (VD3-5) provided almost no change to the anti-H. pylori activity. Apart from this, an isomer of VDP1 (VD3-4) slightly improved anti-H. pylori activity of VDP1. Meanwhile, the replacement of the carbonyl of VDP1 by a methyl acrylate (VD3-6) attenuated the anti-H. pylori activity. As with VDP1, its derivatives also were suggested to exert the anti-H. pylori action through the interaction with myristic acid side chains of dimyristoyl-phosphatidylethanolamine, a characteristic membrane lipid constituent of this pathogen. These results indicate that it is capable of developing specific antibacterial medicines for H. pylori targeting the biomembranal dimyristoyl-phosphatidylethanolamine using VDP1 as the fundamental structure.
Collapse
Affiliation(s)
- Kiyofumi Wanibuchi
- Faculty of Pharmaceutical Sciences, Yokohama University of Pharmacy, 601, Matano-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 245-0066, Japan
| | - Kouichi Hosoda
- Nikon Cell Innovation Co., Ltd., 2-4-10, Shinsuna, Koto-ku, Tokyo, 136-0075, Japan
| | - Avarzed Amgalanbaatar
- Department of Graduate Education, Graduate School, Mongolian National University of Medical Sciences, 14210, Zoing street, Sukhbaatar District, Ulaanbaatar, 14210, Mongolia
| | - Masato Ihara
- Faculty of Pharmaceutical Sciences, Yokohama University of Pharmacy, 601, Matano-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 245-0066, Japan
| | - Motoki Takezawa
- Faculty of Pharmaceutical Sciences, Yokohama University of Pharmacy, 601, Matano-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 245-0066, Japan
| | - Yuki Sakai
- Faculty of Pharmaceutical Sciences, Yokohama University of Pharmacy, 601, Matano-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 245-0066, Japan
| | - Hisashi Masui
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601, Japan
| | - Mitsuru Shoji
- Faculty of Pharmaceutical Sciences, Yokohama University of Pharmacy, 601, Matano-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 245-0066, Japan
| | - Shunji Hayashi
- Department of Microbiology, School of Medicine, Kitasato University, 1-15-1, Kitasato, Minami-ku, Sagamihara-shi, Kanagawa, 252-0374, Japan
| | - Hirofumi Shimomura
- Public Health Center of Uki, Kumamoto Prefecture Office, 400-1, Kugu, Matsubase-machi, Uki-shi, Kumamoto, 869-0532, Japan.
| |
Collapse
|
47
|
Hong Q, Wang J, Zhang H, Liu X, Liu Z. Study of the effect of Lactobacillus crispatus FSCDJY67L3 on Helicobacter Pylori eradication: a double-blind randomized controlled clinical trial. Front Immunol 2023; 14:1265995. [PMID: 38022520 PMCID: PMC10645133 DOI: 10.3389/fimmu.2023.1265995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/29/2023] [Indexed: 12/01/2023] Open
Abstract
Helicobacter pylori (H. pylori) is a gram-negative bacterium exhibiting high pathogenicity. Traditional antibiotic treatments are considered ineffective as the H. pylori resistance has increased. Recently, a quadruple therapy strategy of probiotics and antibiotics to eliminate H. pylori was proposed. Probiotics play a therapeutic role as supplements in this process. The present research screened a probiotic strain (Lactobacillus crispatus FSCDJY67L3) that co-aggregates strongly with H. pylori. L. crispatus FSCDJY67L3 was demonstrated to significantly reduce H. pylori load (14C breath test) in clinical trials with H. pylori-positive patients. The Gastrointestinal Symptom Rating Scale (GSRS) score decreased, indicating improvement in the gastrointestinal discomfort of patients. Furthermore, L. crispatus FSCDJY67L3 showed no change in the structure of the intestinal flora of patients. Routine blood indices and blood biochemical indices related to liver and kidney function were also not affected in the patients. Therefore, L. crispatus FSCDJY67L3 may be used clinically as a supplement for the treatment of H. pylori. Clinical Trial Registration https://www.chictr.org.cn/, Chinese Clinical Trial Registry (ChiCTR2100053710).
Collapse
Affiliation(s)
- Qing Hong
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China
| | - Jidong Wang
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China
| | - Huayue Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xiaoming Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Zhenmin Liu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China
| |
Collapse
|
48
|
Huang J, Gong C, Zhou A. Modulation of gut microbiota: a novel approach to enhancing the effects of immune checkpoint inhibitors. Ther Adv Med Oncol 2023; 15:17588359231204854. [PMID: 37841750 PMCID: PMC10571694 DOI: 10.1177/17588359231204854] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023] Open
Abstract
Although immune checkpoint inhibitors (ICIs) have greatly improved the prognosis of some cancer patients, the majority still fail to respond adequately, and the available biomarkers cannot reliably predict drug efficacy. The gut microbiota has received widespread attention among the various intrinsic and extrinsic factors contributing to drug resistance. As an essential regulator of physiological function, the impact of gut microbiota on host immunity and response to cancer therapy is increasingly recognized. Several studies have demonstrated significant differences in gut microbiota between responders and nonresponders. The gut microbiota associated with better clinical outcomes is called 'favorable gut microbiota'. Significantly, interventions can alter the gut microbiota. By shifting the gut microbiota to the 'favorable' one through various modifications, preclinical and clinical studies have yielded more pronounced responses and better clinical outcomes when combined with ICIs treatment, providing novel approaches to improve the efficacy of cancer immunotherapy. These findings may be attributed to the effects of gut microbiota and its metabolites on the immune microenvironment and the systemic immune system, but the underlying mechanisms remain to be discovered. In this review, we summarize the clinical evidence that the gut microbiota is strongly associated with the outcomes of ICI treatment and describe the gut microbiota characteristics associated with better clinical outcomes. We then expand on the current prevalent modalities of gut microbiota regulation, provide a comprehensive overview of preclinical and clinical research advances in improving the therapeutic efficacy and prognosis of ICIs by modulating gut microbiota, and suggest fundamental questions we need to address and potential directions for future research expansion.
Collapse
Affiliation(s)
- Jinglong Huang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Caifeng Gong
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Aiping Zhou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100020, China
| |
Collapse
|
49
|
Modi N, Chen Y, Dong X, Hu X, Lau GW, Wilson KT, Peek RM, Chen LF. BRD4 Regulates Glycolysis-Dependent Nos2 Expression in Macrophages Upon H pylori Infection. Cell Mol Gastroenterol Hepatol 2023; 17:292-308.e1. [PMID: 37820788 PMCID: PMC10829522 DOI: 10.1016/j.jcmgh.2023.10.001] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND & AIMS Metabolic reprogramming is essential for the activation and functions of macrophages, including bacterial killing and cytokine production. Bromodomain-containing protein 4 (BRD4) has emerged as a critical regulator of innate immune response. However, the potential role of BRD4 in the metabolic reprogramming of macrophage activation upon Helicobacter pylori infection remains unclear. METHODS Bone marrow-derived macrophages (BMDMs) from wild-type (WT) and Brd4-myeloid deletion conditional knockout (Brd4-CKO) mice were infected with H pylori. RNA sequencing was performed to evaluate the differential gene expression between WT and Brd4-deficient BMDMs upon infection. An in vivo model of H pylori infection using WT and Brd4-CKO mice was used to confirm the role of BRD4 in innate immune response to infection. RESULTS Depletion of Brd4 in BMDMs showed impaired H pylori-induced glycolysis. In addition, H pylori-induced expression of glycolytic genes, including Slc2a1 and Hk2, was decreased in Brd4-deficient BMDMs. BRD4 was recruited to the promoters of Slc2a1 and Hk2 via hypoxia-inducible factor-1α, facilitating their expression. BRD4-mediated glycolysis stabilized H pylori-induced nitric oxide synthase (Nos2) messenger RNA to produce nitric oxide. The NO-mediated killing of H pylori decreased in Brd4-deficient BMDMs, which was rescued by pyruvate. Furthermore, Brd4-CKO mice infected with H pylori showed reduced gastric inflammation and increased H pylori colonization with reduced inducible NO synthase expression in gastric macrophages. CONCLUSIONS Our study identified BRD4 as a key regulator of hypoxia-inducible factor-1α-dependent glycolysis and macrophage activation. Furthermore, we show a novel regulatory role of BRD4 in innate immunity through glycolysis to stabilize Nos2 messenger RNA for NO production to eliminate H pylori infection.
Collapse
Affiliation(s)
- Nikita Modi
- Department of Biochemistry, College of Liberal Arts & Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Yanheng Chen
- Department of Biochemistry, College of Liberal Arts & Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Xingchen Dong
- Department of Biochemistry, College of Liberal Arts & Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Xiangming Hu
- Department of Biochemistry, College of Liberal Arts & Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Gee W Lau
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Keith T Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee; Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Richard M Peek
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lin-Feng Chen
- Department of Biochemistry, College of Liberal Arts & Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois.
| |
Collapse
|
50
|
Gurunathan S, Kim JH. Bacterial extracellular vesicles: Emerging nanoplatforms for biomedical applications. Microb Pathog 2023; 183:106308. [PMID: 37595812 DOI: 10.1016/j.micpath.2023.106308] [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: 05/12/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
Bacterial extracellular vesicles (BEVs) are nanosized lipid bilayers generated from membranes that are filled with components derived from bacteria. BEVs are important for the physiology, pathogenicity, and interactions between bacteria and their hosts as well. BEVs represent an important mechanism of transport and interaction between cells. Recent advances in biomolecular nanotechnology have enabled the desired properties to be engineered on the surface of BEVs and decoration with desired and diverse biomolecules and nanoparticles, which have potential biomedical applications. BEVs have been the focus of various fields, including nanovaccines, therapeutic agents, and drug delivery vehicles. In this review, we delineate the fundamental aspects of BEVs, including their biogenesis, cargo composition, function, and interactions with host cells. We comprehensively summarize the factors influencing the biogenesis of BEVs. We further highlight the importance of the isolation, purification, and characterization of BEVs because they are essential processes for potential benefits related to host-microbe interactions. In addition, we address recent advancements in BEVs in biomedical applications. Finally, we provide conclusions and future perspectives as well as highlight the remaining challenges of BEVs for different biomedical applications.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Biotechnology, Rathinam College of Arts and Science, Rathinam Techzone Campus, Eachanari, Coimbatore, 641 021, Tamil Nadu, India.
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea.
| |
Collapse
|