1
|
Zong L, Cheng G, Zhao J, Zhuang X, Zheng Z, Liu Z, Song F. Inhibitory Effect of Ursolic Acid on the Migration and Invasion of Doxorubicin-Resistant Breast Cancer. Molecules 2022; 27:1282. [PMID: 35209071 PMCID: PMC8879026 DOI: 10.3390/molecules27041282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/29/2022] [Accepted: 02/11/2022] [Indexed: 12/11/2022] Open
Abstract
The cause of death in most breast cancer patients is disease metastasis and the occurrence of multidrug resistance (MDR). Ornithine decarboxylase (ODC), which is involved into multiple pathways, is closely related to carcinogenesis and development. Ursolic acid (UA), a natural triterpenoid compound, has been shown to reverse the MDR characteristics of tumor cells. However, the effect of UA on the invasion and metastasis of tumor cells with MDR is not known. Therefore, we investigated the effects of UA on invasion and metastasis, ODC-related polyamine metabolism, and MAPK-Erk-VEGF/MMP-9 signaling pathways in a doxorubicin-resistant breast cancer cell (MCF-7/ADR) model. The obtained results showed that UA significantly inhibited the adhesion and migration of MCF-7/ADR cells, and had higher affinities with key active cavity residues of ODC compared to the known inhibitor di-fluoro-methyl-ornithine (DFMO). UA could downregulate ODC, phosphorylated Erk (P-Erk), VEGF, and matrix metalloproteinase-9 (MMP-9) activity. Meanwhile, UA significantly reduced the content of metabolites of the polyamine metabolism. Furthermore, UA increased the intracellular accumulation of Dox in MCF-7/ADR cells. Taken together, UA can inhibit against tumor progression during the treatment of breast cancer with Dox, and possibly modulate the Erk-VEGF/MMP-9 signaling pathways and polyamine metabolism by targeting ODC to exert these effects.
Collapse
Affiliation(s)
- Li Zong
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; (L.Z.); (G.C.); (J.Z.); (Z.Z.); (Z.L.)
- Jilin Province Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Guorong Cheng
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; (L.Z.); (G.C.); (J.Z.); (Z.Z.); (Z.L.)
- Jilin Province Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Jingwu Zhao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; (L.Z.); (G.C.); (J.Z.); (Z.Z.); (Z.L.)
- Jilin Province Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Xiaoyu Zhuang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhong Zheng
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; (L.Z.); (G.C.); (J.Z.); (Z.Z.); (Z.L.)
- Jilin Province Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Zhiqiang Liu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; (L.Z.); (G.C.); (J.Z.); (Z.Z.); (Z.L.)
- Jilin Province Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Fengrui Song
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; (L.Z.); (G.C.); (J.Z.); (Z.Z.); (Z.L.)
- Jilin Province Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| |
Collapse
|
2
|
Abstract
The intimate involvement of pathogens with the heightened risk for developing certain cancers is an area of research that has captured a great deal of attention over the last 10 years. One firmly established paradigm that highlights this aspect of disease progression is in the instance of Helicobacter pylori infection and the contribution it makes in elevating the risk for developing gastric cancer. Whilst the molecular mechanisms that pinpoint the contribution that this microorganism inflicts towards host cells during gastric cancer initiation have come into greater focus, another picture that has also emerged is one that implicates the host's immune system, and the chronic inflammation that can arise therefrom, as being a central contributory factor in disease progression. Consequently, when taken with the underlying role that the extracellular matrix plays in the development of most cancers, and how this dynamic can be modulated by proteases expressed from the tumor or inflammatory cells, a complex and detailed relationship shared between the individual cellular components and their surroundings is coming into focus. In this review article, we draw attention to the emerging role played by the cathepsin proteases in modulating the stage-specific progression of Helicobacter pylori-initiated gastric cancer and the underlying immune response, while highlighting the therapeutic significance of this dynamic and how it may be amenable for novel intervention strategies within a basic research or clinical setting.
Collapse
|
3
|
López-Gómez M, García de Santiago B, Delgado-López PD, Malmierca E, González-Olmedo J, Gómez-Raposo C, Sandoval C, Ruiz-Seco P, Escribano N, Gómez-Cerezo JF, Casado E. Gastrointestinal tumors and infectious agents: A wide field to explore. World J Meta-Anal 2021; 9:505-521. [DOI: 10.13105/wjma.v9.i6.505] [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: 05/01/2021] [Revised: 08/26/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
Infection is currently one of the main contributors to carcinogenesis. In fact, the International Agency for Research on Cancer has categorized eleven biological agents as group I carcinogens. It is estimated that around 16% of the 12.7 million new cancers diagnosed in 2008 were attributable to infectious agents. Although underdeveloped regions carry the highest incidence rates, about 7.4% of infection-related cancer cases occur in developed areas. Physicians are increasingly aware of the potential carcinogenic role of common virus like the Human Papilloma virus in cervical cancer, or the hepatitis B and C viruses in hepatocarcinoma. However, the carcinogenic role of several other infectious agents is less recognized. Given that gastrointestinal malignancies carry an overall poor prognosis, a better understanding of the carcinogenic mechanisms triggered by infectious agents is key to decrease the rate of cancer related deaths. Preventive measures directed to such infections would ideally impact survival. In this paper we review the main pathogenic mechanisms related to the development of gastrointestinal malignancies induced by infectious microorganisms and other pathogens which are currently under investigation.
Collapse
Affiliation(s)
- Miriam López-Gómez
- Medical Oncology Department. Precision Oncology Laboratory, Infanta Sofía University Hospital, San Sebastián de los Reyes 28231, Madrid, Spain
| | - Belén García de Santiago
- Pharmacy Department, Infanta Sofia University Hospital, San Sebastián de los Reyes 28703, Madrid, Spain
| | | | - Eduardo Malmierca
- Internal Medicine Department, Infanta Sofía University Hospital, San Sebastián de los Reyes 28703, Madrid, Spain
| | - Jesús González-Olmedo
- Medical Oncology Department, Infanta Sofia University Hospital, San Sebastián de los Reyes 28703, Madrid, Spain
| | - César Gómez-Raposo
- Medical Oncology Department, Infanta Sofia University Hospital, San Sebastián de los Reyes 28703, Madrid, Spain
| | - Carmen Sandoval
- Medical Oncology Department, Infanta Sofia University Hospital, San Sebastián de los Reyes 28703, Madrid, Spain
| | - Pilar Ruiz-Seco
- Internal Medicine Department, Infanta Sofía University Hospital, San Sebastián de los Reyes 28703, Madrid, Spain
| | - Nora Escribano
- Intensive Care Unit, Jiménez Díaz Foundation, Madrid 28040, Madrid, Spain
| | - Jorge Francisco Gómez-Cerezo
- Internal Medicine Department, Infanta Sofía University Hospital, San Sebastián de los Reyes 28703, Madrid, Spain
| | - Enrique Casado
- Medical Oncology Department, Infanta Sofia University Hospital, San Sebastián de los Reyes 28703, Madrid, Spain
| |
Collapse
|
4
|
Li QZ, Zuo ZW, Zhou ZR, Ji Y. Polyamine homeostasis-based strategies for cancer: The role of combination regimens. Eur J Pharmacol 2021; 910:174456. [PMID: 34464603 DOI: 10.1016/j.ejphar.2021.174456] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/14/2021] [Accepted: 08/26/2021] [Indexed: 01/07/2023]
Abstract
Spermine, spermidine and putrescine polyamines are naturally occurring ubiquitous positively charged amines and are essential metabolites for biological functions in our life. These compounds play a crucial role in many cell processes, including cellular proliferation, growth, and differentiation. Intracellular levels of polyamines depend on their biosynthesis, transport and degradation. Polyamine levels are high in cancer cells, which leads to the promotion of tumor growth, invasion and metastasis. Targeting polyamine metabolism as an anticancer strategy is considerably rational. Due to compensatory mechanisms, a single strategy does not achieve satisfactory clinical effects when using a single agent. Combination regimens are more clinically promising for cancer chemoprevention because they work synergistically with causing little or no adverse effects due to each individual agent being used at lower doses. Moreover, bioactive substances have advantages over single chemical agents because they can affect multiple targets. In this review, we discuss anticancer strategies targeting polyamine metabolism and describe how combination treatments and effective natural active ingredients are promising therapies. The existing research suggests that polyamine metabolic enzymes are important therapeutic targets and that combination therapies can be more effective than monotherapies based on polyamine depletion.
Collapse
Affiliation(s)
- Qi-Zhang Li
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Industrial Fermentation (Ministry of Education), Institute of Biomedical and Pharmaceutical Sciences, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, Hubei, 430068, PR China.
| | - Zan-Wen Zuo
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Industrial Fermentation (Ministry of Education), Institute of Biomedical and Pharmaceutical Sciences, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, Hubei, 430068, PR China
| | - Ze-Rong Zhou
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Industrial Fermentation (Ministry of Education), Institute of Biomedical and Pharmaceutical Sciences, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, Hubei, 430068, PR China
| | - Yan Ji
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Industrial Fermentation (Ministry of Education), Institute of Biomedical and Pharmaceutical Sciences, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, Hubei, 430068, PR China
| |
Collapse
|
5
|
McNamara KM, Gobert AP, Wilson KT. The role of polyamines in gastric cancer. Oncogene 2021; 40:4399-4412. [PMID: 34108618 PMCID: PMC8262120 DOI: 10.1038/s41388-021-01862-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/07/2021] [Accepted: 05/24/2021] [Indexed: 02/05/2023]
Abstract
Advancements in our understanding of polyamine molecular and cellular functions have led to increased interest in targeting polyamine metabolism for anticancer therapeutic benefits. The polyamines putrescine, spermidine, and spermine are polycationic alkylamines commonly found in all living cells and are essential for cellular growth and survival. This review summarizes the existing research on polyamine metabolism and function, specifically the role of polyamines in gastric immune cell and epithelial cell function. Polyamines have been implicated in a multitude of cancers, but in this review, we focus on the role of polyamine dysregulation in the context of Helicobacter pylori-induced gastritis and subsequent progression to gastric cancer. Due to the emerging implication of polyamines in cancer development, there is an increasing number of promising clinical trials using agents to target the polyamine metabolic pathway for potential chemoprevention and anticancer therapy.
Collapse
Affiliation(s)
- Kara M. McNamara
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Program in Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alain P. Gobert
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Keith T. Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Program in Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, USA,Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA,Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
6
|
Suharsono H, Wibawa DN, Muttaqin Z, Agustina KK. Structure of cytotoxic associated antigen A protein of Helicobacter pylori from Bali and Lombok isolates of Indonesia. Vet World 2020; 13:1319-1326. [PMID: 32848306 PMCID: PMC7429398 DOI: 10.14202/vetworld.2020.1319-1326] [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: 12/31/2019] [Accepted: 05/21/2020] [Indexed: 11/16/2022] Open
Abstract
Background and Aim: Helicobacterpylori is a well-known zoonotic agent with worldwide distribution. In Indonesia, only one report regarding the variation within the cytotoxic associated antigen A (CagA) protein of H. pylori has been described in the literature, which was conducted in Manado, South Sulawesi. There remains no report concerning the structure of this protein, particularly for the Bali and Lombok isolates. The objective of this study was to investigate the diversity of H. pylori CagA amino acid sequences of Bali and Lombok isolates, to predict their molecular structures and conduct toxicity examination of CagA on gastric cells. Materials and Methods: A total of 36 samples were used in equal proportions for each pathologic condition. DNA extraction was performed to subculture H. pylori Bali isolates. The amplification of the CagA 3′ variable region was carried out using the primers P1 (5′-GATAACAGGCAAGCTTTTTGAGG-3′) and P2 (5′-CTGCAAAAGATTGTTTGGCAG-3′). The W2, W9, and W35 fragments were selected as a representation of H. pylori Bali isolates, which were modeled through the threading modeling approach using I-TASSER. Results: According to the 12 CagA sequences obtained and phylogenetic analyses, the H. pylori strain originating from Bali can be grouped within the East Asian genotypes and is identical to the Lombok strain. In addition, the Bali isolates are phylogenetically more closely related to Southeast Asian strains, particularly the Filipino strain. The relationship between degree of inflammation induced and CagA-positive infection was not statistically significant. Conclusion: The structure of the H. pylori Bali isolate is identical to that of Lombok isolate, which belongs to the same group of East Asian genotypes, and bacterial virulence is not related to structure.
Collapse
Affiliation(s)
- Hamong Suharsono
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Udayana University, Denpasar, Indonesia
| | - Dewa Nyoman Wibawa
- Internal Medicine Laboratory, Faculty of Medicine Udayana University, Denpasar, Indonesia
| | - Zainul Muttaqin
- Biomedical Research Unit, West Nusa Tenggara General Hospital, Lombok, Indonesia
| | - Kadek Karang Agustina
- Department of Veterinary Public Health, Faculty of Veterinary Medicine Udayana University, Denpasar, Indonesia
| |
Collapse
|
7
|
Guo Y, Zhang T, Shi Y, Zhang J, Li M, Lu F, Zhang J, Chen X, Ding S. Helicobacter pylori inhibits GKN1 expression via the CagA/p-ERK/AUF1 pathway. Helicobacter 2020; 25:e12665. [PMID: 31657090 DOI: 10.1111/hel.12665] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Recent studies have shown that gastrokine 1 (GKN1), an important tumor suppressor gene, is downregulated in Helicobacter pylori (H. pylori) infected gastric mucosa and gastric cancer. However, the underlying mechanism is poorly understood. Herein, we investigated the potential mechanism of H. pylori-induced GKN1 downregulation. MATERIALS AND METHODS GKN1 and AU-rich element RNA-binding factor 1 (AUF1) expressions were assessed by quantitative real-time PCR, Western blot, or immunohistochemistry in H. pylori-infected tissues and H. pylori co-cultured cell lines. The regulation of AUF1 on GKN1 was determined by RNA pulldown assay, RNA immunoprecipitation, mRNA turnover, and luciferase activity assays. The involvement of phosphorylated extra-cellular signal-regulated kinase (p-ERK) or CagA in H. pylori-induced AUF1 expression was verified using p-ERK inhibitor or CagA knockout H. pylori. In addition, the cell proliferation and migration capacities of AUF1-knockdown cells were investigated. RESULTS GKN1 expression progressively decreased from H. pylori-infected gastritis to gastric cancer tissues. H. pylori co-culture also induced significant GKN1 reduction in GES-1 and BGC-823 cells. Besides, the mRNA level of GKN1 and AUF1 in human gastric mucosa showed negative correlation significantly. AUF1 knockdown resulted in upregulation of GKN1 expression and promoted GKN1 mRNA decay by binding the 3' untranslated region of GKN1 mRNA H. pylori-induced AUF1 expression was associated with p-ERK activation and CagA. Furthermore, knockdown of AUF1 significantly inhibited cell viability, migration ability, and arrested fewer cells in S-phase. CONCLUSION Our data demonstrated that H. pylori infection downregulated GKN1 expression via the CagA/p-ERK/AUF1 pathway. AUF1 promoted gastric cancer at least partly through downregulating GKN1, which presented a novel potential target for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Yanlei Guo
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Ting Zhang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Science, Peking University Health Science Center, Beijing, China
| | - Yanyan Shi
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, China
| | - Jing Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Mingyu Li
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Science, Peking University Health Science Center, Beijing, China
| | - Fengmin Lu
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Science, Peking University Health Science Center, Beijing, China
| | - Jing Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Xiangmei Chen
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Science, Peking University Health Science Center, Beijing, China
| | - Shigang Ding
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
8
|
Xu X, Zhang X, Xing H, Liu Z, Jia J, Jin C, Zhang Y. Importin-4 functions as a driving force in human primary gastric cancer. J Cell Biochem 2019; 120:12638-12646. [PMID: 30861176 DOI: 10.1002/jcb.28530] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/02/2019] [Accepted: 01/14/2019] [Indexed: 11/08/2022]
Abstract
OBJECTIVES Importin-4 (IPO4) is responsible for transporting histones H3 and H4 into the nucleus for chromatin assembly. But, the role of IPO4 in cancer, especially in gastric cancer (GC), has not been fully understood. We aim to determine the expression and function of IPO4 in GC. MATERIALS AND METHODS Bioinformatics analysis was used to study the association of IPO4 and GC using GEO data and the Kaplan-Meier plotter. The quantitative real-time polymerase chain reaction and Western blot analysis were used to determine the IPO4 level in GC cells and tissues. Small interfering RNAs (siRNAs) were used to knockdown endogenous IPO4 expression in GC cells. Cell counting kit-8 (CCK-8), colony formation and transwell assays were used to examine the effect of IPO4 on cell proliferation and migration. RESULTS IPO4 mRNA is overexpressed in GC tissues using bioinformatics analysis of three groups' transcriptome data, and high level of IPO4 is negatively correlated with poor long-term survival using the Kaplan-Meier plotter analysis. Western blot analysis further shows that IPO4 protein levels are also overexpressed in GC tissues and a number of GC cell lines. Endogenous IPO4 level can be inhibited by specific siRNA effectively. Importantly, CCK-8, colony formation, and transwell assays demonstrate that IPO4 knockdown by siRNA impairs GC cell proliferation and migration. CONCLUSIONS Our data suggest that IPO4 contributes to GC progression and poor prognosis, and may function as a driving force in GC progression.
Collapse
Affiliation(s)
- Xia Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Xinchao Zhang
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huaixin Xing
- Department of Anesthesiology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China.,Department of Anesthesiology, Shandong Institue of Cancer Prevention and Control Affiliated to Shandong Academy of Medical Sciences, Jinan, China
| | - Zhifang Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Jihui Jia
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, Department of Microbiology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Chunyuan Jin
- Department of Environmental Medicine & Biochemistry and Molecular Pharmacology, New York University School of Medicine, Tuxedo, New York
| | - Yingjie Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China.,Department of Radiation Oncology, Shandong Institute of Cancer Prevention and Control Affiliated to Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
9
|
Maleki Kakelar H, Barzegari A, Dehghani J, Hanifian S, Saeedi N, Barar J, Omidi Y. Pathogenicity of Helicobacter pylori in cancer development and impacts of vaccination. Gastric Cancer 2019; 22:23-36. [PMID: 30145749 DOI: 10.1007/s10120-018-0867-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/14/2018] [Indexed: 02/07/2023]
Abstract
Helicobacter pylori affect around 50% of the population worldwide. More importantly, the gastric infection induced by this bacterium is deemed to be associated with the progression of distal gastric carcinoma and gastric mucosal lymphoma in the human. H. pylori infection and its prevalent genotype significantly differ across various geographical regions. Based on numerous virulence factors, H. pylori can target different cellular proteins to modulate the variety of inflammatory responses and initiate numerous "hits" on the gastric mucosa. Such reactions lead to serious complications, including gastritis and peptic ulceration, gastric cancer and gastric mucosa-associated lymphoid structure lymphoma. Therefore, H. pylori have been considered as the type I carcinogen by the Global Firm for Research on Cancer. During the two past decades, different reports revealed that H. pylori possess oncogenic potentials in the gastric mucosa through a complicated interplay between the bacterial factors, various facets, and the environmental factors. Accordingly, numerous signaling pathways could be triggered in the development of gastrointestinal diseases (e.g., gastric cancer). Therefore, the main strategy for the treatment of gastric cancer is controlling the disease far before its onset using preventive/curative vaccination. Increasing the efficiency of vaccines may be achieved by new trials of vaccine modalities, which is used to optimize the cellular immunity. Taken all, H. pylori infection may impose severe complications, for resolving of which extensive researches are essential in terms of immune responses to H. pylori. We envision that H. pylori-mediated diseases can be controlled by advanced vaccines and immunotherapies.
Collapse
Affiliation(s)
- Hadi Maleki Kakelar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaber Dehghani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahram Hanifian
- Department of Food Science and Technology, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Nazli Saeedi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, 5165665811, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, 5165665811, Iran.
| |
Collapse
|
10
|
van Elsland D, Neefjes J. Bacterial infections and cancer. EMBO Rep 2018; 19:embr.201846632. [PMID: 30348892 PMCID: PMC6216254 DOI: 10.15252/embr.201846632] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/10/2018] [Accepted: 09/24/2018] [Indexed: 12/30/2022] Open
Abstract
Infections are estimated to contribute to 20% of all human tumours. These are mainly caused by viruses, which explains why a direct bacterial contribution to cancer formation has been largely ignored. While epidemiological data link bacterial infections to particular cancers, tumour formation is generally assumed to be solely caused by the ensuing inflammation responses. Yet, many bacteria directly manipulate their host cell in various phases of their infection cycle. Such manipulations can affect host cell integrity and can contribute to cancer formation. We here describe how bacterial surface moieties, bacterial protein toxins and bacterial effector proteins can induce host cell DNA damage, and thereby can interfere with essential host cell signalling pathways involved in cell proliferation, apoptosis, differentiation and immune signalling.
Collapse
Affiliation(s)
- Daphne van Elsland
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center LUMC, Leiden, The Netherlands
| | - Jacques Neefjes
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center LUMC, Leiden, The Netherlands
| |
Collapse
|
11
|
Zhao SJ, Shen YF, Li Q, He YJ, Zhang YK, Hu LP, Jiang YQ, Xu NW, Wang YJ, Li J, Wang YH, Liu F, Zhang R, Yin GY, Tang JH, Zhou D, Zhang ZG. SLIT2/ROBO1 axis contributes to the Warburg effect in osteosarcoma through activation of SRC/ERK/c-MYC/PFKFB2 pathway. Cell Death Dis 2018; 9:390. [PMID: 29523788 PMCID: PMC5844886 DOI: 10.1038/s41419-018-0419-y] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/15/2018] [Accepted: 02/14/2018] [Indexed: 12/20/2022]
Abstract
Cellular metabolic reprogramming is the main characteristic of cancer cells and identification of targets using this metabolic pattern is extremely important to treat cancers, such as osteosarcoma (OS). In this study, SLIT2 and ROBO1 were upregulated in OS, and higher expression of ROBO1 was associated with worse overall survival rate. Furthermore, in vitro and in vivo experiments demonstrated that the SLIT2/ROBO1 axis promotes proliferation, inhibits apoptosis, and contributes to the Warburg effect in OS cells. Mechanistically, the SLIT2/ROBO1 axis exerted cancer-promoting effects on OS via activation of the SRC/ERK/c-MYC/PFKFB2 pathway. Taken together, the findings reveal a previously unappreciated function of SLIT2/ROBO1 signaling in OS, which is intertwined with metabolic alterations that promote cancer progression. Targeting the SLIT2/ROBO1 axis may be a potential therapeutic approach for patients with OS.
Collapse
Affiliation(s)
- Shu-Jie Zhao
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
- Department of Orthopedics, The Affiliated Hospital of Nanjing Medical University, Changzhou No.2 People's Hospital, Changzhou, 213003, Jiangsu, China
| | - Yi-Fei Shen
- Department of Orthopedics, The Affiliated Hospital of Nanjing Medical University, Changzhou No.2 People's Hospital, Changzhou, 213003, Jiangsu, China
| | - Qing Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yun-Jie He
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yun-Kun Zhang
- Department of Orthopedics, The Affiliated Hospital of Nanjing Medical University, Changzhou No.2 People's Hospital, Changzhou, 213003, Jiangsu, China
| | - Li-Peng Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yu-Qing Jiang
- Department of Orthopedics, The Affiliated Hospital of Nanjing Medical University, Changzhou No.2 People's Hospital, Changzhou, 213003, Jiangsu, China
| | - Nan-Wei Xu
- Department of Orthopedics, The Affiliated Hospital of Nanjing Medical University, Changzhou No.2 People's Hospital, Changzhou, 213003, Jiangsu, China
| | - Yu-Ji Wang
- Department of Orthopedics, The Affiliated Hospital of Nanjing Medical University, Changzhou No.2 People's Hospital, Changzhou, 213003, Jiangsu, China
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ya-Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Fei Liu
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Rong Zhang
- Department of Obstetrics and Gynecology, Fengxian Hospital, Southern Medical University, Shanghai, 201499, China
| | - Guo-Yong Yin
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Jin-Hai Tang
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Dong Zhou
- Department of Orthopedics, The Affiliated Hospital of Nanjing Medical University, Changzhou No.2 People's Hospital, Changzhou, 213003, Jiangsu, China.
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
12
|
Gobert AP, Wilson KT. Polyamine- and NADPH-dependent generation of ROS during Helicobacter pylori infection: A blessing in disguise. Free Radic Biol Med 2017; 105:16-27. [PMID: 27682363 PMCID: PMC5366100 DOI: 10.1016/j.freeradbiomed.2016.09.024] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 09/16/2016] [Accepted: 09/23/2016] [Indexed: 12/20/2022]
Abstract
Helicobacter pylori is a Gram-negative bacterium that specifically colonizes the gastric ecological niche. During the infectious process, which results in diseases ranging from chronic gastritis to gastric cancer, the host response is characterized by the activation of the innate immunity of gastric epithelial cells and macrophages. These cells thus produce effector molecules such as reactive oxygen species (ROS) to counteract the infection. The generation of ROS in response to H. pylori involves two canonical pathways: 1) the NADPH-dependent reduction of molecular oxygen to generate O2•-, which can dismute to generate ROS; and 2) the back-conversion of the polyamine spermine into spermidine through the enzyme spermine oxidase, leading to H2O2 production. Although these products have the potential to affect the survival of bacteria, H. pylori has acquired numerous strategies to counteract their deleterious effects. Nonetheless, ROS-mediated oxidative DNA damage and mutations may participate in the adaptation of H. pylori to its ecological niche. Lastly, ROS have been shown to play a major role in the development of the inflammation and carcinogenesis. It is the purpose of this review to summarize the literature about the production of ROS during H. pylori infection and their role in this infectious gastric disease.
Collapse
Affiliation(s)
- Alain P Gobert
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, United States; Center for Mucosal Inflammation and Cancer, United States
| | - Keith T Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, United States; Department of Pathology, Microbiology, and Immunology, United States; Department of Cancer Biology, United States; Center for Mucosal Inflammation and Cancer, United States; Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, United States.
| |
Collapse
|
13
|
Wei Y, Zhao L, He W, Yang J, Geng C, Chen Y, Liu T, Chen H, Li Y. Benzo[a]pyrene promotes gastric cancer cell proliferation and metastasis likely through the Aryl hydrocarbon receptor and ERK-dependent induction of MMP9 and c-myc. Int J Oncol 2016; 49:2055-2063. [PMID: 27601158 DOI: 10.3892/ijo.2016.3674] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 08/01/2016] [Indexed: 11/05/2022] Open
Abstract
Gastric cancer (GC) is the fifth most common cancer worldwide and the third leading cause of global cancer-related death. Benzo[a]pyrene (BaP), a Group Ⅰ carcinogen categorized by the IARC, is a cumulative foodborne carcinogen and ubiquitous environmental pollutant with potent carcinogenic properties. However, the function and mechanism of BaP exposure on GC progression remains unclear. We investigated the role of BaP in human GC progression to identify potential mechanism underlining its carcinogenic activity. After exposure to various concentrations of BaP, human GC cells SGC-7901 and MNK-45 showed an increased capability of proliferation, migration and invasion. Further study indicated that BaP promotes the expression of matrix metalloproteinase-9 (MMP9) and c-myc at mRNA and protein level, and activates Aryl hydrocarbon receptor (AhR) and ERK pathway. Moreover, BaP-induced overexpression of MMP9 and c-myc were attenuated by the ERK inhibitor U0126 and AhR inhibitor resveratrol, respectively. These data suggest that BaP promotes proliferation and metastasis of GC cells through upregulation of MMP9 and c-myc expression, and this was likely mediated via the AhR and ERK signaling pathway.
Collapse
Affiliation(s)
- Yucai Wei
- The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Lei Zhao
- The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Wenting He
- The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Jingwei Yang
- The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Chunyu Geng
- The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Yusheng Chen
- The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Tao Liu
- The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Hao Chen
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Yumin Li
- The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| |
Collapse
|
14
|
Ahn HJ, Lee DS. Helicobacter pylori in gastric carcinogenesis. World J Gastrointest Oncol 2015; 7:455-65. [PMID: 26690981 PMCID: PMC4678392 DOI: 10.4251/wjgo.v7.i12.455] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 10/10/2015] [Accepted: 11/03/2015] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer still is a major concern as the third most common cancer worldwide, despite declining rates of incidence in many Western countries. Helicobacter pylori (H. pylori) is the major cause of gastric carcinogenesis, and its infection insults gastric mucosa leading to the occurrence of atrophic gastritis which progress to intestinal metaplasia, dysplasia, early gastric cancer, and advanced gastric cancer consequently. This review focuses on multiple factors including microbial virulence factors, host genetic factors, and environmental factors, which can heighten the chance of occurrence of gastric adenocarcinoma due to H. pylori infection. Bacterial virulence factors are key components in controlling the immune response associated with the induction of carcinogenesis, and cagA and vacA are the most well-known pathogenic factors. Host genetic polymorphisms contribute to regulating the inflammatory response to H. pylori and will become increasingly important with advancing techniques. Environmental factors such as high salt and smoking may also play a role in gastric carcinogenesis. It is important to understand the virulence factors, host genetic factors, and environmental factors interacting in the multistep process of gastric carcinogenesis. To conclude, prevention via H. pylori eradication and controlling environmental factors such as diet, smoking, and alcohol is an important strategy to avoid H. pylori-associated gastric carcinogenesis.
Collapse
|
15
|
De Falco M, Lucariello A, Iaquinto S, Esposito V, Guerra G, De Luca A. Molecular Mechanisms of Helicobacter pylori Pathogenesis. J Cell Physiol 2015; 230:1702-7. [PMID: 25639461 DOI: 10.1002/jcp.24933] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 01/16/2015] [Indexed: 12/12/2022]
Abstract
Helicobacter pylori infects 50% of mankind. The vast majority of H. pylori infection occurs in the developing countries where up to 80% of the middle-aged adults may be infected. Bacterial infection causes an inflammatory response that proceeds through a series of intermediated stages of precancerous lesions (gastritis, atrophy, intestinal metaplasia, and dysplasia). Among infected individuals, approximately 10% develops severe gastric lesions such as peptic ulcer disease, 1-3% progresses to gastric cancer (GC) with a low 5-year survival rate, and 0.1% develops mucosa-associated lymphoid tissue (MALT). GC is one of the most common cancer and the third leading cause of cancer-related deaths worldwide. In this review, we have summarized the most recent papers about molecular mechanisms of H. pylori pathogenesis. The main important steps of H. pylori infection such as adhesion, entry in epithelial gastric cells, activation of intracellular pathways until epigenetic modifications have been described.
Collapse
Affiliation(s)
- Maria De Falco
- Department of Biology, University Federico II of Naples, Naples, Italy; National Institute of Biostructures and Biosystems (INBB), Rome, Italy
| | | | | | | | | | | |
Collapse
|
16
|
Yong X, Tang B, Li BS, Xie R, Hu CJ, Luo G, Qin Y, Dong H, Yang SM. Helicobacter pylori virulence factor CagA promotes tumorigenesis of gastric cancer via multiple signaling pathways. Cell Commun Signal 2015; 13:30. [PMID: 26160167 PMCID: PMC4702319 DOI: 10.1186/s12964-015-0111-0] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 07/03/2015] [Indexed: 12/12/2022] Open
Abstract
Helicobacter pylori (H. pylori) infection is strongly associated with the development of gastric diseases but also with several extragastric diseases. The clinical outcomes caused by H. pylori infection are considered to be associated with a complex combination of host susceptibility, environmental factors and bacterial isolates. Infections involving H. pylori strains that possess the virulence factor CagA have a worse clinical outcome than those involving CagA-negative strains. It is remarkable that CagA-positive H. pylori increase the risk for gastric cancer over the risk associated with H. pylori infection alone. CagA behaves as a bacterial oncoprotein playing a key role in H. pylori-induced gastric cancer. Activation of oncogenic signaling pathways and inactivation of tumor suppressor pathways are two crucial events in the development of gastric cancer. CagA shows the ability to affect the expression or function of vital protein in oncogenic or tumor suppressor signaling pathways via several molecular mechanisms, such as direct binding or interaction, phosphorylation of vital signaling proteins and methylation of tumor suppressor genes. As a result, CagA continuously dysregulates of these signaling pathways and promotes tumorigenesis. Recent research has enriched our understanding of how CagA effects on these signaling pathways. This review summarizes the results of the most relevant studies, discusses the complex molecular mechanism involved and attempts to delineate the entire signaling pathway.
Collapse
Affiliation(s)
- Xin Yong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| | - Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| | - Bo-Sheng Li
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| | - Rui Xie
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| | - Chang-Jiang Hu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| | - Gang Luo
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| | - Yong Qin
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| | - Hui Dong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| | - Shi-Ming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| |
Collapse
|
17
|
Xu Y, Yan Y, Hou MX, Liu YE. NaCl pretreatment attenuates H.pylori-induced DNA damage and exacerbates proliferation of gastric epithelial cells (GES-1). Infect Agent Cancer 2015; 10:8. [PMID: 25859277 PMCID: PMC4391598 DOI: 10.1186/s13027-015-0003-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 02/06/2015] [Indexed: 12/11/2022] Open
Abstract
Background Both H. pylori infection and high salt (NaCl) diet are risks of gastric cancer, however, the interaction pattern of the two is not very clear. Our objective was to investigate the effects of NaCl-pretreated H. pylori on DNA damage and proliferation of gastric epithelial cell (GES-1). Methods GES-1 cells were co-cultured with H.pylori or NaCl-pretreated H. pylori (with 30% NaCl) for 24 h. The morphological changes of all cells were observed by inverted phase contrast microscopy and transmission electron microscopy. Oxidative DNA damage was examined by immunofluorescence. Alterations in mitochondrial membrane potential and apoptosis rate were detected by flow cytometry and western blot, and expression of Ki-67, PCNA and P21 were evaluated using the immunocytochemical staining. Results GES-1 cells co-cultured with NaCl-pretreated H.pylori exhibited morphological changes and oxidative DNA damage. Although no significant disruption of the mitochondrial membrane potential (ΔΨm) and apoptotic rate were observed compared with control groups, there were significant decreased in Bax and Caspase3 proteins and increased in Bcl-2 protein in GES-1 cells infected with H. pylori30 when compared with GES-1 cells cultured with H. pylori. In addition, we found a proliferative effect on GES-1 cells with an increased expression of Ki-67 and PCNA as well as a decreased p21 expression, through which the cells may acquire the potential for malignant transformation. Conclusion NaCl-pretreated H. pylori possessed the ability to cause cell injury and promote proliferation in gastric epithelial cells.
Collapse
Affiliation(s)
- Ying Xu
- Radiation oncology Department of General Hospital of Shenyang Military Command, Shenyang, l10016 China
| | - Ying Yan
- Radiation oncology Department of General Hospital of Shenyang Military Command, Shenyang, l10016 China
| | - Ming-Xiao Hou
- Emergency Medicine Department of General Hospital of Shenyang Military Command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, 83 Wenhua Road, Shenhe District, Shenyang, l10016 China
| | - Yun-En Liu
- Emergency Medicine Department of General Hospital of Shenyang Military Command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, 83 Wenhua Road, Shenhe District, Shenyang, l10016 China
| |
Collapse
|
18
|
Wang XQ, Terry PD, Cheng L, Yan H, Wang JS, Wu WA, Hu SK. Interactions between pork consumption, CagA status and IL-1B-31 genotypes in gastric cancer. World J Gastroenterol 2014; 20:8151-8157. [PMID: 25009387 PMCID: PMC4081686 DOI: 10.3748/wjg.v20.i25.8151] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Revised: 02/09/2014] [Accepted: 04/03/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore potential interactions among Helicobacter pylori (H. pylori), CagA status, interleukin (IL)-1B-31 genotypes, and non-cardiac gastric cancer (GC) risk.
METHODS: A case-control study of non-cardia GC was performed at 3 hospitals located in Xi’an, China, between September 2008 and July 2010. We included 171 patients with histologically diagnosed primary non-cardia GC and 367 population based controls (matched by sex, age and city of residence). A standardized questionnaire was used to obtain information regarding potential risk factors, including pork consumption. H. pylori CagA status was assessed by enzyme-linked immunosorbent assay, and IL-1B-31 genotypes were determined by polymerase chain reaction-restriction fragment length polymorphism. Multivariate unconditional logistic regression was used to explore potential interactions among the factors.
RESULTS: The CagA appeared to confer an increased risk of GC (OR = 1.81, 95%CI: 1.25-2.61). The main associations with IL-1B-31C allele here were 0.98 (95%CI: 0.59-1.63) for CC vs TT and 0.99 (95%CI: 0.64-1.51) for C Carriers vs TT. However, no associations were observed for CagA or IL-1B-31 genotype status among subjects who reported low pork consumption (P for interaction = 0.11). In contrast, high pork consumption and IL-1B-31C genotypes appeared to synergistically increase GC risk (P for interaction = 0.048) after adjusting for confounding factors, particularly among subjects with CagA (OR = 3.07, 95%CI: 1.17-10.79). We did not observe effect modification of pork consumption by H. pylori CagA status, or between H. pylori CagA status and IL-1B-31 genotypes after adjustment for pork consumption and other factors.
CONCLUSION: These interaction relationships among CagA, IL-1B-31 and pork consumption may have implications for development of the preventive strategies for the early detection of non-cardiac GC.
Collapse
|
19
|
Helicobacter pylori-induced gastric inflammation and gastric cancer. Cancer Lett 2014; 345:196-202. [PMID: 23981572 DOI: 10.1016/j.canlet.2013.08.016] [Citation(s) in RCA: 545] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Revised: 08/07/2013] [Accepted: 08/13/2013] [Indexed: 02/06/2023]
|
20
|
Pastrello C, Pasini E, Kotlyar M, Otasek D, Wong S, Sangrar W, Rahmati S, Jurisica I. Integration, visualization and analysis of human interactome. Biochem Biophys Res Commun 2014; 445:757-73. [DOI: 10.1016/j.bbrc.2014.01.151] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 01/24/2014] [Indexed: 02/06/2023]
|
21
|
Fonseca MV, Swanson MS. Nutrient salvaging and metabolism by the intracellular pathogen Legionella pneumophila. Front Cell Infect Microbiol 2014; 4:12. [PMID: 24575391 PMCID: PMC3920079 DOI: 10.3389/fcimb.2014.00012] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 01/23/2014] [Indexed: 11/13/2022] Open
Abstract
The Gram-negative bacterium Legionella pneumophila is ubiquitous in freshwater environments as a free-swimming organism, resident of biofilms, or parasite of protozoa. If the bacterium is aerosolized and inhaled by a susceptible human host, it can infect alveolar macrophages and cause a severe pneumonia known as Legionnaires' disease. A sophisticated cell differentiation program equips L. pneumophila to persist in both extracellular and intracellular niches. During its life cycle, L. pneumophila alternates between at least two distinct forms: a transmissive form equipped to infect host cells and evade lysosomal degradation, and a replicative form that multiplies within a phagosomal compartment that it has retooled to its advantage. The efficient changeover between transmissive and replicative states is fundamental to L. pneumophila's fitness as an intracellular pathogen. The transmission and replication programs of L. pneumophila are governed by a number of metabolic cues that signal whether conditions are favorable for replication or instead trigger escape from a spent host. Several lines of experimental evidence gathered over the past decade establish strong links between metabolism, cellular differentiation, and virulence of L. pneumophila. Herein, we focus on current knowledge of the metabolic components employed by intracellular L. pneumophila for cell differentiation, nutrient salvaging and utilization of host factors. Specifically, we highlight the metabolic cues that are coupled to bacterial differentiation, nutrient acquisition systems, and the strategies utilized by L. pneumophila to exploit host metabolites for intracellular replication.
Collapse
Affiliation(s)
- Maris V Fonseca
- Science and Mathematics Division, Monroe County Community College Monroe, MI, USA
| | - Michele S Swanson
- Department of Microbiology and Immunology, University of Michigan Medical School Ann Arbor, MI, USA
| |
Collapse
|
22
|
Farinati F, Cardin R, Piciocchi M, Rodríguez-Castro K, Maddalo G, Rugge M. Helicobacter pylori Infection – The Link Between Oxidative Damage, Cell Proliferation, Apoptosis, and Gastric Cancer. SYSTEMS BIOLOGY OF FREE RADICALS AND ANTIOXIDANTS 2014:1871-1891. [DOI: 10.1007/978-3-642-30018-9_211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
23
|
Ornithine decarboxylase and glutamate decarboxylase 65 as prognostic markers of gallbladder malignancy: a clinicopathological study in benign and malignant lesions of the gallbladder. Mol Med Rep 2012; 7:413-8. [PMID: 23152127 DOI: 10.3892/mmr.2012.1178] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 10/31/2012] [Indexed: 12/16/2022] Open
Abstract
Ornithine decarboxylase (ODC) plays a critical role in cell proliferation and is overexpressed in a variety of cancers. Furthermore, γ-aminobutyric acid (GABA) content and glutamate decarboxylase (GAD) activity are increased in neoplastic tissues in colon and breast cancer. However, few studies have examined these molecules in gallbladder cancer specimens. We observed the expression levels of ODC and GAD65 in benign and malignant lesions of the gallbladder and investigated their clinicopathological significance for the first time. The expression levels of ODC and GAD65 in specimens from gallbladder adenocarcinoma (n=108), peritumoral tissues (n=46), adenomatous polyps (n=15) and chronic cholecystitis (n=35) were detected using immunohistochemical methods. Kaplan-Meier survival and Cox regression analyses were carried out to explore the clinical and pathological correlations. The levels of positive staining of ODC and GAD65 were significantly higher in gallbladder adenocarcinoma than in peritumoral tissues, adenomatous polyps and chronic cholecystitis. The Kaplan‑Meier survival analysis and Cox regression analysis showed that the expression of ODC and GAD65 correlated significantly with the one-year survival rate and the mean survival time of the patients postoperatively. We conclude that the overexpression of ODC and GAD65 are significant in the carcinogenesis and progression of gallbladder adenocarcinoma. They may be important biological markers for the evaluation of biological behaviors and the prognosis of gallbladder adenocarcinoma.
Collapse
|
24
|
Abstract
Although Helicobacter pylori infection is highly prevalent in the global human population, the majority of infected individuals remain asymptomatic. A complex combination of host, environmental, and bacterial factors are considered to determine susceptibility and severity of outcome in the subset of individuals that develop clinical disease. These factors collectively determine the ability of H. pylori to colonize the gastric mucosa and profoundly influence the nature of the interaction that ensues. Many studies over the last year provide new insight into H. pylori virulence strategies and the activities of critical bacterial determinants that modulate the host environment. These latter include the secreted proteins CagA and VacA and adhesins BabA and OipA, which directly interact with host tissues. Observations from several studies extend the functional repertoire of CagA and the cag type IV secretion system in particular, providing further mechanistic understanding of how these important determinants engage and activate host signalling pathways important in the development of disease.
Collapse
Affiliation(s)
- Robin M Delahay
- Centre for Biomolecular Sciences and Nottingham Digestive Diseases Centre, University of Nottingham, Nottingham, UK.
| | | |
Collapse
|
25
|
Zheng RN, Li SR, Masahiro A. Helicobacter pylori CagA and gastric carcinogenesis. Asian Pac J Cancer Prev 2012; 13:6305-10. [PMID: 23464450 DOI: 10.7314/apjcp.2012.13.12.6305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVES This study aimed to demonstrate the tyrosine phosphorylation motif (TPM) and 3' region structure of the Helicobacter pylori CagA gene as well as its SHP-2 binding activity in AGS cells and relation to gastric carcinogenesis. METHODS Sixteen clinical isolate H. pylori strains from eight duodenal ulcer and eight gastric adenocarcinoma patients were studied for CagA repeat sequence EPIYA motifs, C-terminal structure, and western blot analysis of CagA protein expression, translocation, and SHP-2 binding in AGS cells. RESULTS Except for strain 547, all strains from the gastric adenocarcinoma patients were positive for CagA by PCR and had three EPIYA copy motifs. Western blotting showed that all strains were positive for CagA protein expression (100%), CagA protein translocation (100%), and SHP-2 binding (100%). CagA protein expression was significantly higher in the gastric adenocarcinoma patients than in the duodenal ulcer patients (P=0.0023). CagA protein translocation and SHP-2 binding in the gastric adenocarcinoma patients were higher than those in the duodenal ulcer patients, but no significant differences were found between the two groups (P=0.59, P=0.21, respectively). CONCLUSIONS The TPMs and 3' region structures of the H. pylori CagA gene in the duodenal ulcer and gastric adenocarcinoma patients have no significant differences.
Collapse
Affiliation(s)
- Ri-Nan Zheng
- Department of Gastroenterology, Yanda International Hospital, East Beijing, China.
| | | | | |
Collapse
|