1
|
Cruz-Lebrón A, Faiez TS, Hess MM, Sfanos KS. Diet and the microbiome as mediators of prostate cancer risk, progression, and therapy response. Urol Oncol 2025; 43:209-220. [PMID: 39757039 DOI: 10.1016/j.urolonc.2024.12.001] [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: 05/06/2024] [Revised: 11/20/2024] [Accepted: 12/01/2024] [Indexed: 01/07/2025]
Abstract
Complex relationships between the human microbiome and cancer are increasingly recognized for cancer sites that harbor commensal microbial communities such as the gut, genitourinary tract, and skin. For organ sites that likely do not contain commensal microbiota, there is still a substantial capacity for the human-associated microbiota to influence disease etiology across the cancer spectrum. We propose such a relationship for prostate cancer, the most commonly diagnosed cancer in males in the United States. This review explores the current evidence for a role for the urinary and gut microbiota in prostate cancer risk, via both direct interactions (prostate infections) and long-distance interactions such as via the metabolism of procarcinogenic or anticarcinogenic dietary metabolites. We further explore a newly recognized role of the gut microbiota in mediating cancer treatment response or resistance either via production of androgens and/or procarcinogenic metabolites or via direct metabolism of anticancer drugs that are used to treat advanced disease. Overall, we present the current state of knowledge relating to how the human microbiome mediates prostate cancer risk, progression, and therapy response, as well as suggest future research directions for the field.
Collapse
Affiliation(s)
- Angélica Cruz-Lebrón
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Megan M Hess
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Karen S Sfanos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
2
|
Lu J, Tong Q. From pathogenesis to treatment: the impact of bacteria on cancer. Front Microbiol 2024; 15:1462749. [PMID: 39360320 PMCID: PMC11445166 DOI: 10.3389/fmicb.2024.1462749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024] Open
Abstract
The intricate relationship between cancer and bacteria has garnered increasing attention in recent years. While traditional cancer research has primarily focused on tumor cells and genetic mutations, emerging evidence highlights the significant role of microbial communities within the tumor microenvironment in cancer development and progression. This review aims to provide a comprehensive overview of the current understanding of the complex interplay between cancer and bacteria. We explore the diverse ways in which bacteria influence tumorigenesis and tumor behavior, discussing direct interactions between bacteria and tumor cells, their impact on tumor immunity, and the potential modulation of the tumor microenvironment. Additionally, we delve into the mechanisms through which bacterial metabolites and extracellular products May affect cancer pathways. By conducting a thorough analysis of the existing literature, we underscore the multifaceted and intricate relationship between bacteria and cancer. Understanding this complex interplay could pave the way for novel therapeutic approaches and preventive strategies in cancer treatment.
Collapse
Affiliation(s)
| | - Qiang Tong
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
3
|
Wang L, Zheng YB, Yin S, Li KP, Wang JH, Bao EH, Zhu PY. Causal relationship between gut microbiota and prostate cancer contributes to the gut-prostate axis: insights from a Mendelian randomization study. Discov Oncol 2024; 15:58. [PMID: 38431915 PMCID: PMC10909808 DOI: 10.1007/s12672-024-00925-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/01/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Changes in gut microbiota abundance have been linked to prostate cancer development. However, the causality of the gut-prostate axis remains unclear. METHODS The genome-wide association study (GWAS) data for gut microbiota sourced from MiBioGen (n = 14,306), alongside prostate cancer summary data from PRACTICAL (n = 140,254) and FinnGen Consortium (n = 133,164). Inverse-variance-weighted (IVW) was mainly used to compute odds ratios (OR) and 95% confidence intervals (Cl), after diligently scrutinizing potential sources of heterogeneity and horizontal pleiotropy via the rigorous utilization of Cochran's Q test, the MR-PRESSO method, and MR-Egger. We used meta-analysis methods in random effects to combine the Mendelian randomization (MR) estimates from the two sources. RESULTS The pooled analyses of MR results show that genus Eubacterium fissicatena (OR = 1.07, 95% CI 1.01 to 1.13, P = 0.011) and genus Odoribacter (OR = 1.14, 95% CI 1.01 to 1.27, P = 0.025) were positively associated with prostate cancer. However, genus Adlercreutzia (OR = 0.89, 95% CI 0.83 to 0.96, P = 0.002), Roseburia (OR = 0.90, 95% CI 0.83 to 0.99, P = 0.03), Holdemania (OR = 0.92, 95% CI 0.86 to 0.97, P = 0.005), Flavonifractor (OR = 0.85, 95% CI 0.74 to 0.98, P = 0.024) and Allisonella (OR = 0.93, 95% CI 0.89 to 0.98, P = 0.011) seems to be a protective factor for prostate cancer. Sensitivity analysis found no significant heterogeneity, horizontal pleiotropy, or reverse causal links in all causal associations. CONCLUSION This MR study lends support to a causal relationship between genetically predicted gut microbiota and prostate cancer. Research on the gut-prostate axis, along with further multi-omics analyses, holds significant implications for the prevention and treatment of prostate cancer.
Collapse
Affiliation(s)
- Li Wang
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yong-Bo Zheng
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Shan Yin
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Kun-Peng Li
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Jia-Hao Wang
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Er-Hao Bao
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Ping-Yu Zhu
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| |
Collapse
|
4
|
Kandalai S, Li H, Zhang N, Peng H, Zheng Q. The human microbiome and cancer: a diagnostic and therapeutic perspective. Cancer Biol Ther 2023; 24:2240084. [PMID: 37498047 PMCID: PMC10376920 DOI: 10.1080/15384047.2023.2240084] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/09/2023] [Accepted: 07/19/2023] [Indexed: 07/28/2023] Open
Abstract
Recent evidence has shown that the human microbiome is associated with various diseases, including cancer. The salivary microbiome, fecal microbiome, and circulating microbial DNA in blood plasma have all been used experimentally as diagnostic biomarkers for many types of cancer. The microbiomes present within local tissue, other regions, and tumors themselves have been shown to promote and restrict the development and progression of cancer, most often by affecting cancer cells or the host immune system. These microbes have also been shown to impact the efficacy of various cancer therapies, including radiation, chemotherapy, and immunotherapy. Here, we review the research advances focused on how microbes impact these different facets and why they are important to the clinical care of cancer. It is only by better understanding the roles these microbes play in the diagnosis, development, progression, and treatment of cancer, that we will be able to catch and treat cancer early.
Collapse
Affiliation(s)
- Shruthi Kandalai
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, USA
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Huapeng Li
- Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Nan Zhang
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, USA
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Haidong Peng
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, USA
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Qingfei Zheng
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, USA
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, USA
- Department of Biological Chemistry and Pharmacology, College of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
5
|
Jędrusiak A, Fortuna W, Majewska J, Górski A, Jończyk-Matysiak E. Phage Interactions with the Nervous System in Health and Disease. Cells 2023; 12:1720. [PMID: 37443756 PMCID: PMC10341288 DOI: 10.3390/cells12131720] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
The central nervous system manages all of our activities (e.g., direct thinking and decision-making processes). It receives information from the environment and responds to environmental stimuli. Bacterial viruses (bacteriophages, phages) are the most numerous structures occurring in the biosphere and are also found in the human organism. Therefore, understanding how phages may influence this system is of great importance and is the purpose of this review. We have focused on the effect of natural bacteriophages in the central nervous system, linking them to those present in the gut microbiota, creating the gut-brain axis network, as well as their interdependence. Importantly, based on the current knowledge in the field of phage application (e.g., intranasal) in the treatment of bacterial diseases associated with the brain and nervous system, bacteriophages may have significant therapeutic potential. Moreover, it was indicated that bacteriophages may influence cognitive processing. In addition, phages (via phage display technology) appear promising as a targeted therapeutic tool in the treatment of, among other things, brain cancers. The information collected and reviewed in this work indicates that phages and their impact on the nervous system is a fascinating and, so far, underexplored field. Therefore, the aim of this review is not only to summarize currently available information on the association of phages with the nervous system, but also to stimulate future studies that could pave the way for novel therapeutic approaches potentially useful in treating bacterial and non-bacterial neural diseases.
Collapse
Affiliation(s)
- Adam Jędrusiak
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (A.J.); (J.M.); (A.G.)
| | - Wojciech Fortuna
- Department of Neurosurgery, Wroclaw Medical University, Borowska 213, 54-427 Wroclaw, Poland;
- Phage Therapy Unit, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Joanna Majewska
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (A.J.); (J.M.); (A.G.)
| | - Andrzej Górski
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (A.J.); (J.M.); (A.G.)
- Phage Therapy Unit, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
- Infant Jesus Hospital, The Medical University of Warsaw, 02-006 Warsaw, Poland
| | - Ewa Jończyk-Matysiak
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (A.J.); (J.M.); (A.G.)
| |
Collapse
|
6
|
Lin M, Sun X, Lv L. New insights and options into the mechanisms and effects of combined targeted therapy and immunotherapy in prostate cancer. Mol Ther Oncolytics 2023; 29:91-106. [PMID: 37215386 PMCID: PMC10199166 DOI: 10.1016/j.omto.2023.04.007] [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: 05/24/2023] Open
Abstract
Chronic inflammation is believed to drive prostate carcinogenesis by producing reactive oxygen species or reactive nitrogen species to induce DNA damage. This effect might subsequently cause epigenetic and genomic alterations, leading to malignant transformation. Although established therapeutic advances have extended overall survival, tumors in patients with advanced prostate cancer are prone to metastasis, transformation into metastatic castration-resistant prostate cancer, and therapeutic resistance. The tumor microenvironment (TME) of prostate cancer is involved in carcinogenesis, invasion and drug resistance. A plethora of preclinical studies have focused on immune-based therapies. Understanding the intricate TME system in prostate cancer may hold much promise for developing novel therapies, designing combinational therapeutic strategies, and further overcoming resistance to established treatments to improve the lives of prostate cancer patients. In this review, we discuss nonimmune components and various immune cells within the TME and their putative roles during prostate cancer initiation, progression, and metastasis. We also outline the updated fundamental research focusing on therapeutic advances of targeted therapy as well as combinational options for prostate cancer.
Collapse
Affiliation(s)
- Mingen Lin
- Nourse Centre for Pet Nutrition, Wuhu 241200, China
| | - Xue Sun
- Nourse Centre for Pet Nutrition, Wuhu 241200, China
| | - Lei Lv
- Nourse Centre for Pet Nutrition, Wuhu 241200, China
- Shanghai Chowsing Pet Products Co., Ltd, Shanghai 201103, China
| |
Collapse
|
7
|
Oseni SO, Naar C, Pavlović M, Asghar W, Hartmann JX, Fields GB, Esiobu N, Kumi-Diaka J. The Molecular Basis and Clinical Consequences of Chronic Inflammation in Prostatic Diseases: Prostatitis, Benign Prostatic Hyperplasia, and Prostate Cancer. Cancers (Basel) 2023; 15:3110. [PMID: 37370720 DOI: 10.3390/cancers15123110] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/23/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic inflammation is now recognized as one of the major risk factors and molecular hallmarks of chronic prostatitis, benign prostatic hyperplasia (BPH), and prostate tumorigenesis. However, the molecular mechanisms by which chronic inflammation signaling contributes to the pathogenesis of these prostate diseases are poorly understood. Previous efforts to therapeutically target the upstream (e.g., TLRs and IL1-Rs) and downstream (e.g., NF-κB subunits and cytokines) inflammatory signaling molecules in people with these conditions have been clinically ambiguous and unsatisfactory, hence fostering the recent paradigm shift towards unraveling and understanding the functional roles and clinical significance of the novel and relatively underexplored inflammatory molecules and pathways that could become potential therapeutic targets in managing prostatic diseases. In this review article, we exclusively discuss the causal and molecular drivers of prostatitis, BPH, and prostate tumorigenesis, as well as the potential impacts of microbiome dysbiosis and chronic inflammation in promoting prostate pathologies. We specifically focus on the importance of some of the underexplored druggable inflammatory molecules, by discussing how their aberrant signaling could promote prostate cancer (PCa) stemness, neuroendocrine differentiation, castration resistance, metabolic reprogramming, and immunosuppression. The potential contribution of the IL1R-TLR-IRAK-NF-κBs signaling molecules and NLR/inflammasomes in prostate pathologies, as well as the prospective benefits of selectively targeting the midstream molecules in the various inflammatory cascades, are also discussed. Though this review concentrates more on PCa, we envision that the information could be applied to other prostate diseases. In conclusion, we have underlined the molecular mechanisms and signaling pathways that may need to be targeted and/or further investigated to better understand the association between chronic inflammation and prostate diseases.
Collapse
Affiliation(s)
- Saheed Oluwasina Oseni
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Corey Naar
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Mirjana Pavlović
- Department of Computer and Electrical Engineering, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Waseem Asghar
- Department of Computer and Electrical Engineering, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - James X Hartmann
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Gregg B Fields
- Department of Chemistry & Biochemistry, and I-HEALTH, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Nwadiuto Esiobu
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - James Kumi-Diaka
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| |
Collapse
|
8
|
Doulberis M, Papaefthymiou A, Kountouras J. Special Issue “Extragastric Disorders of Helicobacter pylori Infection: From Diagnosis to Treatment”: Editorial. Microorganisms 2023; 11:microorganisms11030677. [PMID: 36985250 PMCID: PMC10059884 DOI: 10.3390/microorganisms11030677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/09/2023] Open
Abstract
Gut microorganisms represent a very attractive field of contemporary biomedical research since they exhibit complex interactions with their host and shape immunity in health and disease [...]
Collapse
Affiliation(s)
- Michael Doulberis
- Gastroklinik, Private Gastroenterological Practice, 8810 Horgen, Switzerland
- Gastroenterology Section, Hirslanden Klinik im Park, 8027 Zurich, Switzerland
- Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, 5001 Aarau, Switzerland
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Apostolis Papaefthymiou
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Pancreaticobiliary Medicine Unit, University College London Hospitals (UCLH), London W1W 6DN, UK
| | - Jannis Kountouras
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Correspondence:
| |
Collapse
|
9
|
Tóth Z, Bezzegh A, Tordé Á, Vásárhelyi B, Gyarmati B. Short term ciprofloxacin and clindamycin combination antibiotic therapy before and after transrectal ultrasound scan and prostate biopsy: Its impact on major components of gut microbiome. Mol Cell Probes 2022; 66:101874. [PMID: 36400114 DOI: 10.1016/j.mcp.2022.101874] [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: 10/11/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 11/17/2022]
Abstract
The perturbation of gut microbiome is a risk factor for a number of adverse conditions. Among other factors antibiotic therapy is a common culprit. We characterized the short-term alteration of gut microbiome after antibiotic therapy. Nine patients (age (median [range]): 67 [57-75 years]) were subjected to prostate biopsy. Ciprofloxacin and clindamycin, 500 mg and 150 mg, respectively, were administered twice a day; this combination therapy was started the day before and continued until 5th and 8th day, respectively, following biopsy. 16s RNA sequencing data from fecal swabs taken before antibiotic therapy and 14 days after biopsy were analysed. At phylum level, the abundance of Actinobacteria and Firmicutes decreased, while that of Bacteroides and Proteobacteria increased after antibiotic therapy. The ratio of Firmicutes:Bacteroides inversed (from 2.81 to 0.74, p = 0.035). At order level, the abundance of Bacteroidales and Veillonellales increased, while that of Clostridiales and Coriobacteriales decreased. At genus level the abundance of Bacteroides increased, while those of Roseburia, Faecalibacterium and Collinsella decreased. These findings indicate that short-term antibiotic exposure skews gut microbiome composition. The current level of knowledge does not allow to decide whether this skewness is detrimental and has any long-term effect on disease including prostate pathology.
Collapse
Affiliation(s)
- Zoltán Tóth
- Uzsoki Hospital, Department of Urology, Budapest, Uzsoki u. 29-41, 1145, Hungary.
| | - Attila Bezzegh
- Dr. Manninger Jenő Center for Traumatology, Department of Laboratory, Budapest, Fiumei út 17, 1044, Hungary.
| | - Ákos Tordé
- Uzsoki Hospital, Department of Urology, Budapest, Uzsoki u. 29-41, 1145, Hungary.
| | - Barna Vásárhelyi
- Semmelweis University, Department of Laboratory Medicine, Budapest, Nagyvárad tér 4, Floor 14, H-1089, Hungary.
| | - Béla Gyarmati
- Uzsoki Hospital, Department of Gynecology and Obstetrics, Budapest, Uzsoki u. 29-41, 1145, Hungary.
| |
Collapse
|
10
|
Liu H, Zeng L, Yang Y, Huang Z, Guo C, Huang L, Niu X, Zhang C, Wang H. Bcl-3 regulates the function of Th17 cells through raptor mediated glycolysis metabolism. Front Immunol 2022; 13:929785. [PMID: 36159779 PMCID: PMC9500237 DOI: 10.3389/fimmu.2022.929785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/19/2022] [Indexed: 11/22/2022] Open
Abstract
Bcl-3 is an atypical IκB family member that regulates transcription in the nucleus by binding to the p50/p52 homologous dimer subunit. Although various studies illustrate the important role of Bcl-3 in physiological function, its role in metabolism is still unclear. We found that Bcl-3 has a metabolic regulatory effect on autoimmunity. Bcl-3-depleted mice are unable to develop experimental autoimmune encephalomyelitis. The disease resistance was linked to an increase in lactate levels in Th17 cells, and lactate could alleviate EAE development in WT mice. Bcl-3 deficient mice had more differentiated Th17 cells and an increased extracellular acidification rate in these cells. Concurrently, their ultimate respiration rate and respiratory reserve capacity were significantly lower than wild-type mice. However, adding GNE-140 (LADH inhibitor) to Bcl-3-deficient Th17 cells could reverse the phenomenon, and lactate supplementation could increase the glycolysis metabolism of Th17 cells in WT mice. Mechanically, Bcl-3 could interact with Raptor through ANK and RNC domains. Therefore, Bcl-3 regulates Th17 pathogenicity by promoting Raptor mediated energy metabolism, revealing a novel regulation of adaptive immunity.
Collapse
Affiliation(s)
- Hui Liu
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Lin Zeng
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Yang Yang
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhen Huang
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Chunlei Guo
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Liwenhui Huang
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Xinqing Niu
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Chenguang Zhang
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Hui Wang
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
11
|
Deng H, Muthupalani S, Erdman S, Liu H, Niu Z, Wang TC, Fox JG. Translocation of Helicobacter hepaticus synergizes with myeloid-derived suppressor cells and contributes to breast carcinogenesis. Oncoimmunology 2022; 11:2057399. [PMID: 35371619 PMCID: PMC8966989 DOI: 10.1080/2162402x.2022.2057399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Microbial dysbiosis plays an important role in the development of intestinal diseases. Recent studies suggest a link between intestinal bacteria and mammary cancer. Here, we report that female ApcMin/+ mice infected with Helicobacter hepaticus exhibited an increased mammary and small/large intestine tumor burden compared with uninfected littermates. H. hepaticus DNA was detected in small/large intestine, mammary tumors, and adjacent lymph nodes, suggesting a migration pathway. CD11b+Gr1+ myeloid-derived suppressor cells (MDSCs) infiltrated and expressed high levels of Wnts, likely enhancing tumorigenesis through activation of Wnt/β-catenin pathway. Our previous studies indicated that histidine decarboxylase (Hdc) marks a population of myeloid-biased hematopoietic stem cells and granulocytic MDSCs. Cytokines/chemokines secreted by IL-17-expressing mast cells and tumor tissues promoted Hdc+ MDSCs expansion and trafficking toward mammary tumors. Adoptive transfer of MDSCs isolated from H. hepaticus-infected mice increased MDSCs frequencies in peripheral blood, mesenteric lymph nodes, mammary gland, and lymph nodes in recipient ApcMin/+ mice. The adoptive transfer of H. hepaticus primed MDSCs also increased the size and number of mammary tumors. Our results demonstrate that H. hepaticus can translocate from the intestine to mammary tissues to promote mammary tumorigenesis with MDSCs. Targeting bacteria and MDSCs may be useful for the prevention and therapy of extraintestinal cancers. Abbreviations: Helicobacter hepaticus, Hh; myeloid-derived suppressor cell, MDSC; histidine decarboxylase, Hdc; Breast cancer, BC; T regulatory, TR; inflammatory bowel disease, IBD; fluorescence in situ hybridization, FISH; myeloid-biased hematopoietic stem cells, MB-HSCs; granulocytic MDSCs, PMN-MDSCs; Lipopolysaccharide, LPS; Toll-like receptors, TLRs; Mast cells, MCs; Granulocyte-macrophage colony-stimulating factor, GM-CSF; epithelial–mesenchymal transition, EMT; Intestinal epithelial cells, IECs.
Collapse
Affiliation(s)
- Huan Deng
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Division of Digestive and Liver Diseases and Herbert Irving Cancer Research Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | | | - Susan Erdman
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Haibo Liu
- Division of Digestive and Liver Diseases and Herbert Irving Cancer Research Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhengchuan Niu
- Division of Digestive and Liver Diseases and Herbert Irving Cancer Research Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Timothy C. Wang
- Division of Digestive and Liver Diseases and Herbert Irving Cancer Research Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - James G. Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
12
|
Yang X, Guo Y, Chen C, Shao B, Zhao L, Zhou Q, Liu J, Wang G, Yuan W, Sun Z. Interaction between intestinal microbiota and tumour immunity in the tumour microenvironment. Immunology 2021; 164:476-493. [PMID: 34322877 PMCID: PMC8517597 DOI: 10.1111/imm.13397] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/18/2021] [Accepted: 07/20/2021] [Indexed: 11/27/2022] Open
Abstract
In recent years, an increasing number of studies have reported that intestinal microbiota have an important effect on tumour immunity by affecting the tumour microenvironment (TME). The intestinal microbiota are closely associated with various immune cells, such as T lymphocytes, natural killer cells (NK cells) and macrophages. Some bacteria, such as Akkermansia muciniphila (A. muciniphila) and Lactobacillus reuteri (L. reuteri), have been shown to improve the effect of tumour immunity. Furthermore, microbial imbalance, such as the increased abundance of Fusobacterium nucleatum (F. nucleatum) and Helicobacter hepaticus (H. hepaticus), generally causes tumour formation and progression. In addition, some microbiota also play important roles in tumour immunotherapy, especially PD-L1-related therapies. Therefore, what is the relationship between these processes and how do they affect each other? In this review, we summarize the interactions and corresponding mechanisms among the intestinal microbiota, immune system and TME to facilitate the research and development of new targeted drugs and provide new approaches to tumour therapy.
Collapse
Affiliation(s)
- Xiuxiu Yang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- School of MedicineZhengzhou UniversityZhengzhouChina
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Yaxin Guo
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
- Department of Basic MedicalAcademy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Academy of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Chen Chen
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
- School of Life SciencesZhengzhou UniversityZhengzhouChina
| | - Bo Shao
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Luyang Zhao
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
- Department of Basic MedicalAcademy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Academy of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Quanbo Zhou
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jinbo Liu
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Guixian Wang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Weitang Yuan
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhenqiang Sun
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| |
Collapse
|
13
|
Hull R, Lolas G, Makrogkikas S, Jensen LD, Syrigos KN, Evangelou G, Padayachy L, Egbor C, Mehrotra R, Makhafola TJ, Oyomno M, Dlamini Z. Microbiomics in Collusion with the Nervous System in Carcinogenesis: Diagnosis, Pathogenesis and Treatment. Microorganisms 2021; 9:2129. [PMID: 34683450 PMCID: PMC8538279 DOI: 10.3390/microorganisms9102129] [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: 08/19/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 11/17/2022] Open
Abstract
The influence of the naturally occurring population of microbes on various human diseases has been a topic of much recent interest. Not surprisingly, continuously growing attention is devoted to the existence of a gut brain axis, where the microbiota present in the gut can affect the nervous system through the release of metabolites, stimulation of the immune system, changing the permeability of the blood-brain barrier or activating the vagus nerves. Many of the methods that stimulate the nervous system can also lead to the development of cancer by manipulating pathways associated with the hallmarks of cancer. Moreover, neurogenesis or the creation of new nervous tissue, is associated with the development and progression of cancer in a similar manner as the blood and lymphatic systems. Finally, microbes can secrete neurotransmitters, which can stimulate cancer growth and development. In this review we discuss the latest evidence that support the importance of microbiota and peripheral nerves in cancer development and dissemination.
Collapse
Affiliation(s)
- Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.H.); (G.L.); (L.P.); (C.E.); (R.M.); (T.J.M.); (M.O.)
| | - Georgios Lolas
- SAMRC Precision Oncology Research Unit (PORU), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.H.); (G.L.); (L.P.); (C.E.); (R.M.); (T.J.M.); (M.O.)
- Department of Medicine, National & Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.S.); (G.E.)
| | | | - Lasse D. Jensen
- Division of Cardiovascular Medicine, Department of Health, Medicine and Caring Sciences, Faculty of Medicine, Linköping University, 581 83 Linköping, Sweden;
| | - Konstantinos N. Syrigos
- Department of Medicine, National & Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.S.); (G.E.)
| | - George Evangelou
- Department of Medicine, National & Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.S.); (G.E.)
| | - Llewellyn Padayachy
- SAMRC Precision Oncology Research Unit (PORU), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.H.); (G.L.); (L.P.); (C.E.); (R.M.); (T.J.M.); (M.O.)
- Department of Neurosurgery, University of Pretoria, Hatfield 0028, South Africa
| | - Cyril Egbor
- SAMRC Precision Oncology Research Unit (PORU), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.H.); (G.L.); (L.P.); (C.E.); (R.M.); (T.J.M.); (M.O.)
- Department of Neurosurgery, University of Pretoria, Hatfield 0028, South Africa
| | - Ravi Mehrotra
- SAMRC Precision Oncology Research Unit (PORU), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.H.); (G.L.); (L.P.); (C.E.); (R.M.); (T.J.M.); (M.O.)
- Centre for Health Innovation and Policy (CHIP) Foundation, Noida 201301, India
- Datar Cancer Genetics, Nashik 422010, India
| | - Tshepiso Jan Makhafola
- SAMRC Precision Oncology Research Unit (PORU), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.H.); (G.L.); (L.P.); (C.E.); (R.M.); (T.J.M.); (M.O.)
- Centre for Quality of Health and Living, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein 9300, South Africa
| | - Meryl Oyomno
- SAMRC Precision Oncology Research Unit (PORU), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.H.); (G.L.); (L.P.); (C.E.); (R.M.); (T.J.M.); (M.O.)
- Department of Surgery, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0007, South Africa
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.H.); (G.L.); (L.P.); (C.E.); (R.M.); (T.J.M.); (M.O.)
| |
Collapse
|
14
|
Leystra AA, Harvey KN, Kaunga E, Hensley H, Vanderveer LA, Devarajan K, Clapper ML. High Variability in Cellular Proliferation, Gene Expression, and Cytokine Production in the Nonneoplastic Colonic Epithelium of Young Apc+/Min-FCCC Mice. Front Oncol 2021; 11:705562. [PMID: 34513688 PMCID: PMC8429936 DOI: 10.3389/fonc.2021.705562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/09/2021] [Indexed: 12/31/2022] Open
Abstract
An urgent need exists to identify efficacious therapeutic preventive interventions for individuals who are at high risk of developing colorectal cancer. To maximize the benefits of preventive intervention, it is vital to identify the time interval during which the initiation of a preventive intervention will lead to an optimal outcome. The goal of the present study was to determine if oncogenic events can be detected in the nonneoplastic colonic mucosa of Apc+/Min-FCCC mice prior to formation of the first adenoma, thus defining an earlier point of intervention along the cancer continuum. Tissues taken at three potential points of intervention were characterized: prior to Apc mutation (wild type Apc+/+-FCCC mice); after initiation but prior to colon adenoma formation (tumor-free Apc+/Min-FCCC mice); and after formation of the first colon adenoma (tumor-bearing Apc+/Min-FCCC mice). Experimentation focused on molecular processes that are dysregulated in early colon lesions: 1) cellular proliferation (proliferative index and size of the proliferative zone); 2) cellular stemness (expression of Ascl2, Grem1, Lgr5 and Muc2); 3) EGFR signaling (expression of Ereg); and 4) inflammation (expression of Mmp9, Ptsg2, and Reg4, as well as secretion of 18 cytokines involved in immune activation and response). Interestingly, the nonneoplastic colonic mucosa of wild type, tumor-free Apc+/Min-FCCC , and tumor-bearing Apc+/Min-FCCC mice did not display significant differences in average epithelial cell proliferation (fold change 0.8-1.3, p≥0.11), mucosal gene expression (fold change 0.8-1.4, p≥0.22), or secretion of specific cytokines from colonic mucosa (fold change 0.2-1.5, p≥0.06). However, the level of cytokine secretion was highly variable, with many (22% of wild type, 31% of tumor-free Apc+/Min-FCCC , and 31% of tumor-bearing Apc+/Min-FCCC ) mice categorized as outliers (> 1.5 x interquartile ranges below the first quartile or above the third quartile) due to elevated expression of at least one cytokine. In summary, no differences were observed in proliferation, stemness, and EGFR signaling in the colonic mucosa of wild type vs Apc+/Min-FCCC mice, with low baseline cytokine expression, prior to the formation of the first colon adenoma. The results of this study provide valuable baseline data to inform the design of future cancer prevention studies.
Collapse
Affiliation(s)
- Alyssa A. Leystra
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Kristen N. Harvey
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Esther Kaunga
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Harvey Hensley
- Biological Imaging Facility, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Lisa A. Vanderveer
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Karthik Devarajan
- Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Margie L. Clapper
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| |
Collapse
|
15
|
Thomas MU, Messex JK, Dang T, Abdulkadir SA, Jorcyk CL, Liou GY. Macrophages expedite cell proliferation of prostate intraepithelial neoplasia through their downstream target ERK. FEBS J 2020; 288:1871-1886. [PMID: 32865335 DOI: 10.1111/febs.15541] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 07/23/2020] [Accepted: 08/24/2020] [Indexed: 12/18/2022]
Abstract
The risk factors for prostate cancer include a high-fat diet and obesity, both of which are associated with an altered cell environment including increased inflammation. It has been shown that chronic inflammation due to a high-fat diet or bacterial infection has the potential to accelerate prostate cancer as well as its precursor, prostatic intraepithelial neoplasia (PIN), development. However, the underlying mechanism of how chronic inflammation promotes prostate cancer development, especially PIN, remains unclear. In this study, we showed that more macrophages were present in PIN areas as compared to the normal areas of human prostate. When co-culturing PIN cells with macrophages in 3D, more PIN cells had nuclear localized cyclin D1, indicating that macrophages enhanced PIN cell proliferation. We identified ICAM-1 and CCL2 as chemoattractants expressed by PIN cells to recruit macrophages. Furthermore, we discovered that macrophage-secreted cytokines including C5a, CXCL1, and CCL2 were responsible for increased PIN cell proliferation. These three cytokines activated ERK and JNK signaling in PIN cells through a ligand-receptor interaction. However, only blockade of ERK abolished macrophage cytokines-induced cell proliferation of PIN. Overall, our results provide a mechanistic view on how macrophages activated through chronic inflammation can expedite PIN progression during prostate cancer development. The information from our work can facilitate a comprehensive understanding of prostate cancer development, which is required for improvement of current strategies for prostate cancer therapy.
Collapse
Affiliation(s)
- Mikalah U Thomas
- Department of Biological Sciences, Clark Atlanta University, GA, USA
| | - Justin K Messex
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, GA, USA
| | - Tu Dang
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, GA, USA
| | - Sarki A Abdulkadir
- Department of Urology, Northwestern University, Chicago, IL, USA.,Department of Pathology, Northwestern University, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Cheryl L Jorcyk
- Department of Biological Science, Boise State University, ID, USA
| | - Geou-Yarh Liou
- Department of Biological Sciences, Clark Atlanta University, GA, USA.,Center for Cancer Research and Therapeutic Development, Clark Atlanta University, GA, USA
| |
Collapse
|
16
|
de Bono JS, Guo C, Gurel B, De Marzo AM, Sfanos KS, Mani RS, Gil J, Drake CG, Alimonti A. Prostate carcinogenesis: inflammatory storms. Nat Rev Cancer 2020; 20:455-469. [PMID: 32546840 DOI: 10.1038/s41568-020-0267-9] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/24/2020] [Indexed: 02/06/2023]
Abstract
Prostate cancer is a major cause of cancer morbidity and mortality. Intra-prostatic inflammation is a risk factor for prostate carcinogenesis, with diet, chemical injury and an altered microbiome being causally implicated. Intra-prostatic inflammatory cell recruitment and expansion can ultimately promote DNA double-strand breaks and androgen receptor activation in prostate epithelial cells. The activation of the senescence-associated secretory phenotype fuels further 'inflammatory storms', with free radicals leading to further DNA damage. This drives the overexpression of DNA repair and tumour suppressor genes, rendering these genes susceptible to mutagenic insults, with carcinogenesis accelerated by germline DNA repair gene defects. We provide updates on recent advances in elucidating prostate carcinogenesis and explore novel therapeutic and prevention strategies harnessing these discoveries.
Collapse
Affiliation(s)
- Johann S de Bono
- The Institute of Cancer Research, London, UK.
- The Royal Marsden NHS Foundation Trust, Sutton, UK.
| | - Christina Guo
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Bora Gurel
- The Institute of Cancer Research, London, UK
| | | | - Karen S Sfanos
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ram S Mani
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jesús Gil
- MRC London Institute of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | | | - Andrea Alimonti
- Institute of Oncology Research, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
- Department of Medicine, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| |
Collapse
|
17
|
Melamed E, Lee MW. Multiple Sclerosis and Cancer: The Ying-Yang Effect of Disease Modifying Therapies. Front Immunol 2020; 10:2954. [PMID: 31998289 PMCID: PMC6965059 DOI: 10.3389/fimmu.2019.02954] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 12/02/2019] [Indexed: 12/17/2022] Open
Abstract
Over the past two decades, the field of multiple sclerosis (MS) has been transformed by the rapidly expanding arsenal of new disease modifying therapies (DMTs). Current DMTs for MS aim to modulate innate and adaptive immune responses toward a less inflammatory phenotype. Since the immune system is also critical for identifying and eliminating malignant cells, immunosuppression from DMTs may predictably increase the risk of cancer development in MS patients. Compared with healthy controls, patients with autoimmune conditions, such as MS, may already have a higher risk of developing certain malignancies and this risk may further be magnified by DMT treatments. For those patients who develop both MS and cancer, these comorbid presentations create a challenge for clinicians on how to therapeutically address management of cancer in the context of MS autoimmunity. As there are currently no accepted guidelines for managing MS patients with prior history of or newly developed malignancy, we undertook this review to evaluate the molecular mechanisms of current DMTs and their potential for instigating and treating cancer in patients living with MS.
Collapse
Affiliation(s)
- Esther Melamed
- Department of Neurology, Dell Medical School, Austin, TX, United States
| | - Michael William Lee
- Department of Oncology, Department of Medical Education, Dell Medical School, Austin, TX, United States
| |
Collapse
|
18
|
Kumar M, Singh P, Murugesan S, Vetizou M, McCulloch J, Badger JH, Trinchieri G, Al Khodor S. Microbiome as an Immunological Modifier. Methods Mol Biol 2020; 2055:595-638. [PMID: 31502171 PMCID: PMC8276114 DOI: 10.1007/978-1-4939-9773-2_27] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Humans are living ecosystems composed of human cells and microbes. The microbiome is the collection of microbes (microbiota) and their genes. Recent breakthroughs in the high-throughput sequencing technologies have made it possible for us to understand the composition of the human microbiome. Launched by the National Institutes of Health in USA, the human microbiome project indicated that our bodies harbor a wide array of microbes, specific to each body site with interpersonal and intrapersonal variabilities. Numerous studies have indicated that several factors influence the development of the microbiome including genetics, diet, use of antibiotics, and lifestyle, among others. The microbiome and its mediators are in a continuous cross talk with the host immune system; hence, any imbalance on one side is reflected on the other. Dysbiosis (microbiota imbalance) was shown in many diseases and pathological conditions such as inflammatory bowel disease, celiac disease, multiple sclerosis, rheumatoid arthritis, asthma, diabetes, and cancer. The microbial composition mirrors inflammation variations in certain disease conditions, within various stages of the same disease; hence, it has the potential to be used as a biomarker.
Collapse
Affiliation(s)
- Manoj Kumar
- Division of Translational Medicine, Research Department, Sidra Medicine, Doha, Qatar
| | - Parul Singh
- Division of Translational Medicine, Research Department, Sidra Medicine, Doha, Qatar
| | - Selvasankar Murugesan
- Division of Translational Medicine, Research Department, Sidra Medicine, Doha, Qatar
| | - Marie Vetizou
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - John McCulloch
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan H Badger
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Souhaila Al Khodor
- Division of Translational Medicine, Research Department, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
19
|
Zhang B, Gu J, Liu J, Huang B, Li J. Fecal Microbiota Taxonomic Shifts in Chinese Multiple Myeloma Patients Analyzed by Quantitative Polimerase Chain Reaction (QPCR) and 16S rRNA High-Throughput Sequencing. Med Sci Monit 2019; 25:8269-8280. [PMID: 31678982 PMCID: PMC6855177 DOI: 10.12659/msm.919988] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Increasing evidence has suggested that gut flora play an important role in tumor progression and prognosis. However, the relationship between fecal microbiota and hematologic malignancy requires further investigation. This study aimed to characterize the relationship of the fecal microbial community in multiple myeloma (MM) patients. Material/Methods A total of 40 MM patients and healthy controls (n=17) were retrospectively collected from the First Affiliated Hospital of Sun Yat-sen University between October 2018 and May 2019. The fecal samples were collected for 16S rRNA high-throughput sequencing for the fecal microbial community, as well as diversity and correlation analysis. Furthermore, 21 MM patients and their family members were selected for the matched pair analysis to confirm the fecal microbiota taxonomic changes by qRT-PCR assay. Results Diversity analysis showed that diversity measured by Shannon index was lower in MM patients compared with healthy controls. At the phylum level, higher abundances of Proteobacteria but lower abundances of Actinobacteria were identified in the MM group in comparison with the healthy control group. At the genus level, the proportion of Bacteroides, Faecalibacterium, and Roseburia was significantly higher in the MM group. The matched pair analysis showed that Pseudomonas aeruginosa and Faecalibacterium were significantly more abundant in the MM group. Further analysis on prognostic risk factors revealed that the Faecalibacterium prausnitzii level was significantly correlated with ISS stage. Conclusions Our study highlights the imbalanced composition and diversity of the gastrointestinal microbiome in MM patients, which could be further used as a potential biomarker for MM risk screening, therapeutic strategies, and prognostic prediction.
Collapse
Affiliation(s)
- Bingqing Zhang
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| | - Jingli Gu
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| | - Junru Liu
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| | - Beihui Huang
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| | - Juan Li
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| |
Collapse
|
20
|
Sha S, Ni L, Stefil M, Dixon M, Mouraviev V. The human gastrointestinal microbiota and prostate cancer development and treatment. Investig Clin Urol 2019; 61:S43-S50. [PMID: 32055753 PMCID: PMC7004837 DOI: 10.4111/icu.2020.61.s1.s43] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/16/2019] [Indexed: 01/14/2023] Open
Abstract
The human gastrointestinal microbiome contains commensal bacteria and other microbiota that have been gaining increasing attention in the context of cancer development and response to treatment. Microbiota play a role in the maintenance of host barrier surfaces that contribute to both local inflammation and other systemic metabolic functions. In the context of prostate cancer, the gastrointestinal microbiome may play a role through metabolism of estrogen, an increase of which has been linked to the induction of prostatic neoplasia. Specific microbiota such as Bacteroides, Streptococcus, Bacteroides massiliensis, Faecalibacterium prausnitzii, Eubacterium rectalie, and Mycoplasma genitalium have been associated with differing risks of prostate cancer development or extensiveness of prostate cancer disease. In this Review, we discuss gastrointestinal microbiota's effects on prostate cancer development, the ability of the microbiome to regulate chemotherapy for prostate cancer treatment, and the importance of using Next Generation Sequencing to further discern the microbiome's systemic influence on prostate cancer.
Collapse
Affiliation(s)
- Sybil Sha
- Dartmouth Medical School, Hanover, NH, USA
| | - Liqiang Ni
- University of Central Florida, Orlando, FL, USA
| | | | | | | |
Collapse
|
21
|
|
22
|
Kay J, Thadhani E, Samson L, Engelward B. Inflammation-induced DNA damage, mutations and cancer. DNA Repair (Amst) 2019; 83:102673. [PMID: 31387777 DOI: 10.1016/j.dnarep.2019.102673] [Citation(s) in RCA: 255] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 06/15/2019] [Accepted: 07/18/2019] [Indexed: 12/22/2022]
Abstract
The relationships between inflammation and cancer are varied and complex. An important connection linking inflammation to cancer development is DNA damage. During inflammation reactive oxygen and nitrogen species (RONS) are created to combat pathogens and to stimulate tissue repair and regeneration, but these chemicals can also damage DNA, which in turn can promote mutations that initiate and promote cancer. DNA repair pathways are essential for preventing DNA damage from causing mutations and cytotoxicity, but RONS can interfere with repair mechanisms, reducing their efficacy. Further, cellular responses to DNA damage, such as damage signaling and cytotoxicity, can promote inflammation, creating a positive feedback loop. Despite coordination of DNA repair and oxidative stress responses, there are nevertheless examples whereby inflammation has been shown to promote mutagenesis, tissue damage, and ultimately carcinogenesis. Here, we discuss the DNA damage-mediated associations between inflammation, mutagenesis and cancer.
Collapse
Affiliation(s)
- Jennifer Kay
- Department of Biological Engineering, United States.
| | | | - Leona Samson
- Department of Biological Engineering, United States; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States
| | | |
Collapse
|
23
|
Abstract
Nickel is an essential cofactor for some pathogen virulence factors. Due to its low availability in hosts, pathogens must efficiently transport the metal and then balance its ready intracellular availability for enzyme maturation with metal toxicity concerns. The most notable virulence-associated components are the Ni-enzymes hydrogenase and urease. Both enzymes, along with their associated nickel transporters, storage reservoirs, and maturation enzymes have been best-studied in the gastric pathogen Helicobacter pylori, a bacterium which depends heavily on nickel. Molecular hydrogen utilization is associated with efficient host colonization by the Helicobacters, which include both gastric and liver pathogens. Translocation of a H. pylori carcinogenic toxin into host epithelial cells is powered by H2 use. The multiple [NiFe] hydrogenases of Salmonella enterica Typhimurium are important in host colonization, while ureases play important roles in both prokaryotic (Proteus mirabilis and Staphylococcus spp.) and eukaryotic (Cryptoccoccus genus) pathogens associated with urinary tract infections. Other Ni-requiring enzymes, such as Ni-acireductone dioxygenase (ARD), Ni-superoxide dismutase (SOD), and Ni-glyoxalase I (GloI) play important metabolic or detoxifying roles in other pathogens. Nickel-requiring enzymes are likely important for virulence of at least 40 prokaryotic and nine eukaryotic pathogenic species, as described herein. The potential for pathogenic roles of many new Ni-binding components exists, based on recent experimental data and on the key roles that Ni enzymes play in a diverse array of pathogens.
Collapse
|
24
|
Cross talk between neutrophils and the microbiota. Blood 2019; 133:2168-2177. [PMID: 30898860 DOI: 10.1182/blood-2018-11-844555] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 12/24/2018] [Indexed: 12/18/2022] Open
Abstract
The microbiota has emerged as an important regulator of the host immunity by the induction, functional modulation, or suppression of local and systemic immune responses. In return, the host immune system restricts translocation and fine tunes the composition and distribution of the microbiota to maintain a beneficial symbiosis. This paradigm applies to neutrophils, a critical component of the innate immunity, allowing their production and function to be influenced by microbial components and metabolites derived from the microbiota, and engaging them in the process of microbiota containment and regulation. The cross talk between neutrophils and the microbiota adjusts the magnitude of neutrophil-mediated inflammation on challenge while preventing neutrophil responses against commensals under steady state. Here, we review the major molecular and cellular mediators of the interactions between neutrophils and the microbiota and discuss their interplay and contribution in chronic inflammatory diseases and cancer.
Collapse
|
25
|
Abstract
Lactobacillus rhamnosus GG (LGG) was the first strain belonging to the genus Lactobacillus to be patented in 1989 thanks to its ability to survive and to proliferate at gastric acid pH and in medium containing bile, and to adhere to enterocytes. Furthermore LGG is able to produces both a biofilm that can mechanically protect the mucosa, and different soluble factors beneficial to the gut by enhancing intestinal crypt survival, diminishing apoptosis of the intestinal epithelium, and preserving cytoskeletal integrity. Moreover LGG thanks to its lectin-like protein 1 and 2 inhibits some pathogens such as Salmonella species. Finally LGG is able to promote type 1 immune-responsiveness by reducing the expression of several activation and inflammation markers on monocytes and by increasing the production of interleukin-10, interleukin-12 and tumor necrosis factor-α in macrophages. A large number of research data on Lactobacillus GG is the basis for the use of this probiotic for human health. In this review we have considered predominantly randomized controlled trials, meta-analysis, Cochrane Review, guide lines of Scientific Societies and anyway studies whose results were evaluated by means of relative risk, odds ratio, weighted mean difference 95% confidence interval. The effectiveness of LGG in gastrointestinal infections and diarrhea, antibiotic and Clostridium difficile associated diarrhea, irritable bowel syndrome, inflammatory bowel disease, respiratory tract infections, allergy, cardiovascular diseases, nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, cystic fibrosis, cancer, elderly end sport were analyzed.
Collapse
|
26
|
Mohammed H, Varoni EM, Cochis A, Cordaro M, Gallenzi P, Patini R, Staderini E, Lajolo C, Rimondini L, Rocchetti V. Oral Dysbiosis in Pancreatic Cancer and Liver Cirrhosis: A Review of the Literature. Biomedicines 2018; 6:biomedicines6040115. [PMID: 30544974 PMCID: PMC6316311 DOI: 10.3390/biomedicines6040115] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/03/2018] [Accepted: 12/07/2018] [Indexed: 02/07/2023] Open
Abstract
The human body is naturally colonized by a huge number of different commensal microbial species, in a relatively stable equilibrium. When this microbial community undergoes dysbiosis at any part of the body, it interacts with the innate immune system and results in a poor health status, locally or systemically. Research studies show that bacteria are capable of significantly influencing specific cells of the immune system, resulting in many diseases, including a neoplastic response. Amongst the multiple different types of diseases, pancreatic cancer and liver cirrhosis were significantly considered in this paper, as they are major fatal diseases. Recently, these two diseases were shown to be associated with increased or decreased numbers of certain oral bacterial species. These findings open the way for a broader perception and more specific investigative studies, to better understand the possible future treatment and prevention. This review aims to describe the correlation between oral dysbiosis and both pancreatic cancer and liver cirrhotic diseases, as well as demonstrating the possible diagnostic and treatment modalities, relying on the oral microbiota, itself, as prospective, simple, applicable non-invasive approaches to patients, by focusing on the state of the art. PubMed was electronically searched, using the following key words: "oral microbiota" and "pancreatic cancer" (PC), "liver cirrhosis", "systemic involvement", and "inflammatory mediators". Oral dysbiosis is a common problem related to poor oral or systemic health conditions. Oral pathogens can disseminate to distant body organs via the local, oral blood circulation, or pass through the gastrointestinal tract and enter into the systemic circulation. Once oral pathogens reach an organ, they modify the immune response and stimulate the release of the inflammatory mediators, this results in a disease. Recent studies have reported a correlation between oral dysbiosis and the increased risk of pancreatic and liver diseases and provided evidence of the presence of oral pathogens in diseased organs. The profound impact that microbial communities have on human health, provides a wide domain towards precisely investigating and clearly understanding the mechanism of many diseases, including cancer. Oral microbiota is an essential contributor to health status and imbalance in this community was correlated to oral and systemic diseases. The presence of elevated numbers of certain oral bacteria, particularly P. gingivalis, as well as elevated levels of blood serum antibodies, against this bacterial species, was associated with a higher risk of pancreatic cancer and liver cirrhosis incidence. Attempts are increasingly directed towards investigating the composition of oral microbiome as a simple diagnostic approach in multiple diseases, including pancreatic and liver pathosis. Moreover, treatment efforts are concerned in the recruitment of microbiota, for remedial purposes of the aforementioned and other different diseases. Further investigation is required to confirm and clarify the role of oral microbiota in enhancing pancreatic and liver diseases. Improving the treatment modalities requires an exertion of more effort, especially, concerning the microbiome engineering and oral microbiota transplantation.
Collapse
Affiliation(s)
- Hiba Mohammed
- Department of Health Sciences, Università del Piemonte Orientale UPO, 28100 Novara, Italy.
- Fondazione Novara Sviluppo, 28100 Novara, Italy.
| | - Elena Maria Varoni
- Department of Biomedical Sciences, Surgery and Dentistry, Università degli Studi di Milano, 20142 Milano, Italy.
| | - Andrea Cochis
- Department of Health Sciences, Università del Piemonte Orientale UPO, 28100 Novara, Italy.
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), 28100 Novara, Italy.
| | - Massimo Cordaro
- Institute of Dentistry and Maxillofacial Surgery, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Patrizia Gallenzi
- Institute of Dentistry and Maxillofacial Surgery, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Romeo Patini
- Institute of Dentistry and Maxillofacial Surgery, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Edoardo Staderini
- Institute of Dentistry and Maxillofacial Surgery, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Carlo Lajolo
- Institute of Dentistry and Maxillofacial Surgery, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Lia Rimondini
- Department of Health Sciences, Università del Piemonte Orientale UPO, 28100 Novara, Italy.
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), 28100 Novara, Italy.
| | - Vincenzo Rocchetti
- Fondazione Novara Sviluppo, 28100 Novara, Italy.
- Department of Clinical and Experimental Medicine, Università del Piemonte Orientale UPO, 28100 Novara, Italy.
| |
Collapse
|
27
|
Zakariah M, Khan S, Chaudhary AA, Rolfo C, Ben Ismail MM, Alotaibi YA. To Decipher the Mycoplasma hominis Proteins Targeting into the Endoplasmic Reticulum and Their Implications in Prostate Cancer Etiology Using Next-Generation Sequencing Data. Molecules 2018; 23:994. [PMID: 29695086 PMCID: PMC6099661 DOI: 10.3390/molecules23050994] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/16/2018] [Accepted: 04/18/2018] [Indexed: 12/21/2022] Open
Abstract
Cancer was initially considered a genetic disease. However, recent studies have revealed the connection between bacterial infections and growth of different types of cancer. The enteroinvasive strain of Mycoplasma hominis alters the normal behavior of host cells that may result in the growth of prostate cancer. The role of M. hominis in the growth and development of prostate cancer still remains unclear. The infection may regulate several factors that influence prostate cancer growth in susceptible individuals. The aim of this study was to predict M. hominis proteins targeted into the endoplasmic reticulum (ER) of the host cell, and their potential role in the induction of prostate cancer. From the whole proteome of M. hominis, 19 proteins were predicted to be targeted into the ER of host cells. The results of our study predict that several proteins of M. hominis may be targeted to the host cell ER, and possibly alter the normal pattern of protein folding. These predicted proteins can modify the normal function of the host cell. Thus, the intercellular infection of M. hominis in host cells may serve as a potential factor in prostate cancer etiology.
Collapse
Affiliation(s)
- Mohammed Zakariah
- Research Center, College of Computer and Information Science, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Shahanavaj Khan
- Nanomedicine & Biotechnology Research Unit, Department of Pharmaceutics, College of Pharmacy, P.O. Box 2457, King Saud University, Riyadh 11451, Saudi Arabia.
- Department of Bioscience, Shri Ram Group of College (SRGC), Muzaffarnagar 251002, UP, India.
| | - Anis Ahmad Chaudhary
- Department of Pharmacology, College of Medicine, Al-Imam Mohammad Ibn Saud Islamic University, Riyadh 11451, Saudi Arabia.
| | - Christian Rolfo
- Phase I-Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital, "Centre for Oncological Research (CORE)", 2650 Edegem, Belgium.
| | - Mohamed Maher Ben Ismail
- Computer Science Department, College of Computer and Information Sciences, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Yousef Ajami Alotaibi
- Computer Science Department, College of Computer and Information Sciences, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
28
|
Doulberis M, Kotronis G, Thomann R, Polyzos SA, Boziki M, Gialamprinou D, Deretzi G, Katsinelos P, Kountouras J. Review: Impact of Helicobacter pylori on Alzheimer's disease: What do we know so far? Helicobacter 2018; 23. [PMID: 29181894 DOI: 10.1111/hel.12454] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Helicobacter pylori has changed radically gastroenterologic world, offering a new concept in patients' management. Over time, more medical data gave rise to diverse distant, extragastric manifestations and interactions of the "new" discovered bacterium. Special interest appeared within the field of neurodegenerative diseases and particularly Alzheimer's disease, as the latter and Helicobacter pylori infection are associated with a large public health burden and Alzheimer's disease ranks as the leading cause of disability. However, the relationship between Helicobacter pylori infection and Alzheimer's disease remains uncertain. METHODS We performed a narrative review regarding a possible connection between Helicobacter pylori and Alzheimer's disease. All accessible relevant (pre)clinical studies written in English were included. Both affected pathologies were briefly analyzed, and relevant studies are discussed, trying to focus on the possible pathogenetic role of this bacterium in Alzheimer's disease. RESULTS Data stemming from both epidemiologic studies and animal experiments seem to be rather encouraging, tending to confirm the hypothesis that Helicobacter pylori infection might influence the course of Alzheimer's disease pleiotropically. Possible main mechanisms may include the bacterium's access to the brain via the oral-nasal-olfactory pathway or by circulating monocytes (infected with Helicobacter pylori due to defective autophagy) through disrupted blood-brain barrier, thereby possibly triggering neurodegeneration. CONCLUSIONS Current data suggest that Helicobacter pylori infection might influence the pathophysiology of Alzheimer's disease. However, further large-scale randomized controlled trials are mandatory to clarify a possible favorable effect of Helicobacter pylori eradication on Alzheimer's disease pathophysiology, before the recommendation of short-term and cost-effective therapeutic regimens against Helicobacter pylori-related Alzheimer's disease.
Collapse
Affiliation(s)
- Michael Doulberis
- Department of Internal Medicine, Bürgerspital Hospital, Solothurn, Switzerland
| | - Georgios Kotronis
- Department of Internal Medicine, Agios Pavlos General Hospital, Thessaloniki, Macedonia, Greece
| | - Robert Thomann
- Department of Internal Medicine, Bürgerspital Hospital, Solothurn, Switzerland
| | - Stergios A Polyzos
- Department of Internal Medicine, Ippokration Hospital, Second Medical Clinic, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Marina Boziki
- Department of Internal Medicine, Ippokration Hospital, Second Medical Clinic, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Dimitra Gialamprinou
- Department of Pediatrics, Papageorgiou General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Georgia Deretzi
- Department of Neurology, Papageorgiou General Hospital, Multiple Sclerosis Unit, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Panagiotis Katsinelos
- Department of Internal Medicine, Ippokration Hospital, Second Medical Clinic, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Jannis Kountouras
- Department of Internal Medicine, Ippokration Hospital, Second Medical Clinic, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| |
Collapse
|
29
|
Wang F, Huang ZA, Chen X, Zhu Z, Wen Z, Zhao J, Yan GY. LRLSHMDA: Laplacian Regularized Least Squares for Human Microbe-Disease Association prediction. Sci Rep 2017; 7:7601. [PMID: 28790448 PMCID: PMC5548838 DOI: 10.1038/s41598-017-08127-2] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 07/04/2017] [Indexed: 02/08/2023] Open
Abstract
An increasing number of evidences indicate microbes are implicated in human physiological mechanisms, including complicated disease pathology. Some microbes have been demonstrated to be associated with diverse important human diseases or disorders. Through investigating these disease-related microbes, we can obtain a better understanding of human disease mechanisms for advancing medical scientific progress in terms of disease diagnosis, treatment, prevention, prognosis and drug discovery. Based on the known microbe-disease association network, we developed a semi-supervised computational model of Laplacian Regularized Least Squares for Human Microbe-Disease Association (LRLSHMDA) by introducing Gaussian interaction profile kernel similarity calculation and Laplacian regularized least squares classifier. LRLSHMDA reached the reliable AUCs of 0.8909 and 0.7657 based on the global and local leave-one-out cross validations, respectively. In the framework of 5-fold cross validation, average AUC value of 0.8794 +/-0.0029 further demonstrated its promising prediction ability. In case studies, 9, 9 and 8 of top-10 predicted microbes have been manually certified to be associated with asthma, colorectal carcinoma and chronic obstructive pulmonary disease by published literature evidence. Our proposed model achieves better prediction performance relative to the previous model. We expect that LRLSHMDA could offer insights into identifying more promising human microbe-disease associations in the future.
Collapse
Affiliation(s)
- Fan Wang
- School of Mechatronic Engineering, China University of Mining and Technology, Xuzhou, 221116, China
- Jiangsu Key Laboratory of Mine Mechanical and Electrical Equipment, China University of Mining and Technology, Xuzhou, 221116, China
| | - Zhi-An Huang
- College of Computer Science and Software Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Xing Chen
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou, 221116, China.
| | - Zexuan Zhu
- College of Computer Science and Software Engineering, Shenzhen University, Shenzhen, 518060, China.
| | - Zhenkun Wen
- College of Computer Science and Software Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Jiyun Zhao
- School of Mechatronic Engineering, China University of Mining and Technology, Xuzhou, 221116, China
| | - Gui-Ying Yan
- Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
30
|
Doulberis M, Saleh C, Beyenburg S. Is there an Association between Migraine and Gastrointestinal Disorders? J Clin Neurol 2017; 13:215-226. [PMID: 28748672 PMCID: PMC5532317 DOI: 10.3988/jcn.2017.13.3.215] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 03/02/2017] [Accepted: 03/07/2017] [Indexed: 02/06/2023] Open
Abstract
Migraine is a primary episodic headache disorder that represents a substantial burden and disability worldwide. Its pathogenesis is multifactorial and remains hitherto poorly elucidated. An interesting but less-well-known association is that between migraine and gastrointestinal disorders. We have reviewed the literature for relevant papers reporting on the clinical association between migraine and gastrointestinal symptoms. Several studies have shown different gastrointestinal diseases to be associated with migraine, but the underlining pathophysiology remains elusive. The data gathered and analyzed have shown great variability across studies, making it impossible to draw definitive conclusions. Further research is required to elucidate this potential relationship. An understanding of the relationship between migraine and gastrointestinal disorders is of great clinical importance for prompt diagnosis and treatment.
Collapse
Affiliation(s)
- Michael Doulberis
- Department of Internal Medicine, Bürgerspital Hospital of Solothurn, Solothurn, Switzerland
| | - Christian Saleh
- Department of Neurology, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg.
| | - Stefan Beyenburg
- Department of Neurology, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| |
Collapse
|
31
|
Vyshenska D, Lam KC, Shulzhenko N, Morgun A. Interplay between viruses and bacterial microbiota in cancer development. Semin Immunol 2017; 32:14-24. [PMID: 28602713 DOI: 10.1016/j.smim.2017.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 05/03/2017] [Accepted: 05/30/2017] [Indexed: 12/29/2022]
Abstract
During the last few decades we have become accustomed to the idea that viruses can cause tumors. It is much less considered and discussed, however, that most people infected with oncoviruses will never develop cancer. Therefore, the genetic and environmental factors that tip the scales from clearance of viral infection to development of cancer are currently an area of active investigation. Microbiota has recently emerged as a potentially critical factor that would affect this balance by increasing or decreasing the ability of viral infection to promote carcinogenesis. In this review, we provide a model of microbiome contribution to the development of oncogenic viral infections and viral associated cancers, give examples of this process in human tumors, and describe the challenges that prevent progress in the field as well as their potential solutions.
Collapse
Affiliation(s)
- Dariia Vyshenska
- College of Pharmacy, Oregon State University, 1601 SW Jefferson Way, Corvallis, OR 97331, USA
| | - Khiem C Lam
- College of Pharmacy, Oregon State University, 1601 SW Jefferson Way, Corvallis, OR 97331, USA
| | - Natalia Shulzhenko
- College of Veterinary Medicine, Oregon State University, 208 Dryden Hall, Corvallis, OR 97331, USA.
| | - Andrey Morgun
- College of Pharmacy, Oregon State University, 1601 SW Jefferson Way, Corvallis, OR 97331, USA.
| |
Collapse
|
32
|
Bracken TC, Cooper CA, Ali Z, Truong H, Moore JM. Helicobacter Infection Significantly Alters Pregnancy Success in Laboratory Mice. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2017; 56:322-329. [PMID: 28535868 PMCID: PMC5438927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 04/22/2016] [Accepted: 12/07/2016] [Indexed: 06/07/2023]
Abstract
Helicobacter spp. are gram-negative, helically shaped bacteria that cause gastric and enterohepatic infections in mammalian species. Although Helicobacter infection frequently is implicated to interfere with reproductive success, few experimental data support these claims. We therefore retrospectively investigated the effect of Helicobacter infection on murine pregnancy outcome after the identification of endemic Helicobacter infection in an animal research facility. Multiplex conventional PCR analysis was used to characterize Helicobacter infection status in one inbred and 2 transgenic strains of mice in 2 self-contained rooms assigned to the same investigator. Outcomes of timed-mating experiments were compared among Helicobacter spp.-infected and uninfected mice of the same strain; Helicobacter infection was eradicated from the colony through fostering with uninfected dams. Although Helicobacter infection affected fecundity in only one strain of transgenic mouse, the total number of embryos per gravid uterus was significantly reduced in C57BL/6J mice that were infected with a single Helicobacter species, H. typhlonius. Helicobacter infection was also associated with a significant increase in the number of resorbing embryos per uterus and significant decreases in pregnancy-associated weight gain relative to uninfected mice in C57BL6/J mice and one transgenic strain. Helicobacter spp.-infected mice of all tested strains exhibited higher frequency of intrauterine hemorrhaging relative to uninfected mice. These results indicate that naturally-acquired Helicobacter infection not only reduces the productivity of a research animal breeding colony, but also negatively impacts embryo health. Despite these deleterious effects, these data suggest that colonies can be rederived to be Helicobacter-free by Cesarean section and fostering with uninfected dams. This paper provides the first evidence that H. typhlonius infection is sufficient to interfere with reproductive success and embryo health of C57BL/6J mice. Animal research facilities should therefore implement Helicobacter spp. surveillance and control practices to avoid confounding experimental results and to improve breeding colony efficiency.
Collapse
Affiliation(s)
- Tara C Bracken
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | - Caitlin A Cooper
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | - Zil Ali
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | - Ha Truong
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | - Julie M Moore
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia;,
| |
Collapse
|
33
|
Abstract
The microbiota is composed of commensal bacteria and other microorganisms that live on the epithelial barriers of the host. The commensal microbiota is important for the health and survival of the organism. Microbiota influences physiological functions from the maintenance of barrier homeostasis locally to the regulation of metabolism, haematopoiesis, inflammation, immunity and other functions systemically. The microbiota is also involved in the initiation, progression and dissemination of cancer both at epithelial barriers and in sterile tissues. Recently, it has become evident that microbiota, and particularly the gut microbiota, modulates the response to cancer therapy and susceptibility to toxic side effects. In this Review, we discuss the evidence for the ability of the microbiota to modulate chemotherapy, radiotherapy and immunotherapy with a focus on the microbial species involved, their mechanism of action and the possibility of targeting the microbiota to improve anticancer efficacy while preventing toxicity.
Collapse
Affiliation(s)
- Soumen Roy
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
34
|
Erdman SE, Poutahidis T. Gut microbiota modulate host immune cells in cancer development and growth. Free Radic Biol Med 2017; 105:28-34. [PMID: 27840315 PMCID: PMC5831246 DOI: 10.1016/j.freeradbiomed.2016.11.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 10/25/2016] [Accepted: 11/06/2016] [Indexed: 12/20/2022]
Abstract
Emerging evidence shows that microbe interactions with the host immune system impact diverse aspects of cancer development and treatment. As a result, exciting new opportunities exist for engineering diets and microbe cocktails to lower cancer risks with fewer adverse clinical effects than traditional strategies. Microbe-based therapies may ultimately be used to reinforce host immune balance and extinguish cancer for generations to come.
Collapse
Affiliation(s)
- Susan E Erdman
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Theofilos Poutahidis
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Pathology, College of Veterinary Medicine, Aristotle University, Thessaloniki, Greece
| |
Collapse
|
35
|
Miyahira AK, Roychowdhury S, Goswami S, Ippolito JE, Priceman SJ, Pritchard CC, Sfanos KS, Subudhi SK, Simons JW, Pienta KJ, Soule HR. Beyond Seed and Soil: Understanding and Targeting Metastatic Prostate Cancer; Report From the 2016 Coffey-Holden Prostate Cancer Academy Meeting. Prostate 2017; 77:123-144. [PMID: 27679977 DOI: 10.1002/pros.23260] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/12/2016] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The 2016 Coffey-Holden Prostate Cancer Academy (CHPCA) Meeting, "Beyond Seed and Soil: Understanding and Targeting Metastatic Prostate Cancer," was held from June 23 to June 26, 2016, in Coronado, California. METHODS For the 4th year in a row, the Prostate Cancer Foundation (PCF) hosted the CHPCA Meeting, a think tank-structured scientific conference, which focuses on a specific topic of critical unmet need on the biology and treatment of advanced prostate cancer. The 2016 CHPCA Meeting was attended by 71 investigators from prostate cancer and other fields, who discussed the biology, study methodologies, treatment strategies, and critical unmet needs concerning metastatic prostate cancer, with the ultimate goal of advancing strategies to treat and eliminate this disease. RESULTS The major topics of discussion included: the molecular landscape and molecular heterogeneity of metastatic prostate cancer, the role of the metastatic microenvironment, optimizing immunotherapy in metastatic prostate cancer, learning from exceptional responders and non-responders, targeting DNA repair deficiency in advanced prostate cancer, developing and applying novel biomarkers and imaging techniques, and potential roles for the microbiome in prostate cancer. DISCUSSION This article reviews the topics presented and discussions held at the CHPCA Meeting, with a focus on the unknowns and next steps needed to advance our understanding of the biology and most effective treatment strategies for metastatic prostate cancer. Prostate 77:123-144, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Sameek Roychowdhury
- Comprehensive Cancer Center, Department of Internal Medicine, Ohio State University, Columbus, Ohio
- Division of Medical Oncology, Ohio State University, Cincinnati, Ohio
| | - Sangeeta Goswami
- Department of Genitourinary Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Joseph E Ippolito
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Saul J Priceman
- Departments of Hematology and Hematopoietic Cell Transplantation, and Immuno-Oncology, Beckman Research Institute at City of Hope National Medical Center, Duarte, California
| | - Colin C Pritchard
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Karen S Sfanos
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Urology, The James Buchanan Brady Urological Institute, Baltimore, Maryland
| | - Sumit K Subudhi
- Department of Genitourinary Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | | | - Kenneth J Pienta
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Urology, The James Buchanan Brady Urological Institute, Baltimore, Maryland
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | |
Collapse
|
36
|
Dzutsev A, Badger JH, Perez-Chanona E, Roy S, Salcedo R, Smith CK, Trinchieri G. Microbes and Cancer. Annu Rev Immunol 2017; 35:199-228. [PMID: 28142322 DOI: 10.1146/annurev-immunol-051116-052133] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Commensal microorganisms (the microbiota) live on all the surface barriers of our body and are particularly abundant and diverse in the distal gut. The microbiota and its larger host represent a metaorganism in which the cross talk between microbes and host cells is necessary for health, survival, and regulation of physiological functions locally, at the barrier level, and systemically. The ancestral molecular and cellular mechanisms stemming from the earliest interactions between prokaryotes and eukaryotes have evolved to mediate microbe-dependent host physiology and tissue homeostasis, including innate and adaptive resistance to infections and tissue repair. Mostly because of its effects on metabolism, cellular proliferation, inflammation, and immunity, the microbiota regulates cancer at the level of predisposing conditions, initiation, genetic instability, susceptibility to host immune response, progression, comorbidity, and response to therapy. Here, we review the mechanisms underlying the interaction of the microbiota with cancer and the evidence suggesting that the microbiota could be targeted to improve therapy while attenuating adverse reactions.
Collapse
Affiliation(s)
- Amiran Dzutsev
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892,
| | - Jonathan H Badger
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892,
| | - Ernesto Perez-Chanona
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892,
| | - Soumen Roy
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892,
| | - Rosalba Salcedo
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892,
| | - Carolyne K Smith
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892,
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892,
| |
Collapse
|
37
|
Gagnaire A, Nadel B, Raoult D, Neefjes J, Gorvel JP. Collateral damage: insights into bacterial mechanisms that predispose host cells to cancer. Nat Rev Microbiol 2017; 15:109-128. [DOI: 10.1038/nrmicro.2016.171] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
38
|
Poutahidis T, Erdman SE. Commensal bacteria modulate the tumor microenvironment. Cancer Lett 2016; 380:356-8. [PMID: 26739062 PMCID: PMC4942371 DOI: 10.1016/j.canlet.2015.12.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/01/2015] [Accepted: 12/18/2015] [Indexed: 12/22/2022]
Abstract
It has been recently shown that gut microbes modulate whole host immune and hormonal factors impacting the fate of distant preneoplastic lesions toward malignancy or regression. This raises the possibility that the tumor microenvironment interacts with broader systemic microbial-immune networks. These accumulated findings suggest novel therapeutic opportunities for holobiont engineering in emerging tumor microenvironments.
Collapse
Affiliation(s)
- Theofilos Poutahidis
- Division of Comparative Medicine, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Laboratory of Pathology, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, Greece 54124
| | - Susan E Erdman
- Division of Comparative Medicine, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.
| |
Collapse
|
39
|
Han JY, Lim YJ, Choi JA, Lee JH, Jo SH, Oh SM, Song CH. The Role of Prostate Apoptosis Response-4 (Par-4) in Mycobacterium tuberculosis Infected Macrophages. Sci Rep 2016; 6:32079. [PMID: 27552917 PMCID: PMC4995434 DOI: 10.1038/srep32079] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 08/02/2016] [Indexed: 01/30/2023] Open
Abstract
Prostate apoptosis response-4 (Par-4) is a tumor suppressor protein that forms a complex with glucose-regulated protein 78 (GRP78) to induce apoptosis. Previously, we reported that ER stress-induced apoptosis is a critical host defense mechanism against Mycobacterium tuberculosis (Mtb). We sought to understand the role of Par-4 during ER stress-induced apoptosis in response to mycobacterial infection. Par-4 and GRP78 protein levels increased in response Mtb (strain: H37Ra) infection. Furthermore, Par-4 and GRP78 translocate to the surface of Mtb H37Ra-infected macrophages and induce apoptosis via caspase activation. NF-κB activation, Mtb-mediated ER stress, and Par-4 production were significantly diminished in macrophages with inhibited ROS production. To test Par-4 function during mycobacterial infection, we analyzed intracellular survival of Mtb H37Ra in macrophages with Par-4 overexpression or knockdown. Mtb H37Ra growth was significantly reduced in Par-4 overexpressing macrophages and increased in knockdown macrophages. We also observed increased Par-4, GRP78, and caspases activation in Bacillus Calmette-Guérin (BCG)-infected prostate cancer cells. Our data demonstrate that Par-4 is associated with ER stress-induced apoptosis resulting in reduced intracellular survival of mycobacteria. BCG treatment increases Par-4-dependent caspase activation in prostate cancer cells. These results suggest ER stress-induced Par-4 acts as an important defense mechanism against mycobacterial infection and regulates cancer.
Collapse
Affiliation(s)
- Ji-Ye Han
- Department of Medical Science, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, Republic of Korea.,Department of Microbiology, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Yun-Ji Lim
- Department of Medical Science, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, Republic of Korea.,Department of Microbiology, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Ji-Ae Choi
- Department of Medical Science, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, Republic of Korea.,Department of Microbiology, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Jung-Hwan Lee
- Department of Medical Science, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, Republic of Korea.,Department of Microbiology, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Sung-Hee Jo
- Department of Medical Science, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, Republic of Korea.,Department of Microbiology, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Sung-Man Oh
- Department of Medical Science, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, Republic of Korea.,Department of Microbiology, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Chang-Hwa Song
- Department of Medical Science, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, Republic of Korea.,Department of Microbiology, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea.,Research Institute for Medical Sciences, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| |
Collapse
|
40
|
Khan S, Zakariah M, Palaniappan S. Computational prediction of Mycoplasma hominis proteins targeting in nucleus of host cell and their implication in prostate cancer etiology. Tumour Biol 2016; 37:10805-10813. [PMID: 26874727 DOI: 10.1007/s13277-016-4970-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/02/2016] [Indexed: 12/16/2022] Open
Abstract
Cancer has long been assumed to be a genetic disease. However, recent evidence supports the enigmatic connection of bacterial infection with the growth and development of various types of cancers. The cause and mechanism of the growth and development of prostate cancer due to Mycoplasma hominis remain unclear. Prostate cancer cells are infected and colonized by enteroinvasive M. hominis, which controls several factors that can affect prostate cancer growth in susceptible persons. We investigated M. hominis proteins targeting the nucleus of host cells and their implications in prostate cancer etiology. Many vital processes are controlled in the nucleus, where the proteins targeting M. hominis may have various potential implications. A total of 29/563 M. hominis proteins were predicted to target the nucleus of host cells. These include numerous proteins with the capability to alter normal growth activities. In conclusion, our results emphasize that various proteins of M. hominis targeted the nucleus of host cells and were involved in prostate cancer etiology through different mechanisms and strategies.
Collapse
Affiliation(s)
- Shahanavaj Khan
- Nanomedicine & Biotechnology Research Unit, Department of Pharmaceutics, College of Pharmacy, King Saud University, PO Box 2457, Riyadh, 11451, Saudi Arabia.
- Department of Bioscience, Shri Ram Group of College (SRGC), Muzaffarnagar, UP, India.
| | - Mohammed Zakariah
- Research Center, College of Computer and Information Science, King Saud University, Riyadh, Saudi Arabia
| | - Sellappan Palaniappan
- School of Science and Engineering, Malaysia University of Science and Technology, Petaling Jaya, Malaysia
| |
Collapse
|
41
|
Benefits of short-chain fatty acids and their receptors in inflammation and carcinogenesis. Pharmacol Ther 2016; 164:144-51. [PMID: 27113407 DOI: 10.1016/j.pharmthera.2016.04.007] [Citation(s) in RCA: 383] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Indexed: 02/08/2023]
Abstract
Epidemiological studies have linked increased incidence of inflammatory diseases and intestinal cancers in the developed parts of the world to the consumption of diets poor in dietary fibers and rich in refined carbohydrates. Gut bacteria residing in the intestinal lumen exclusively metabolize dietary fibers. Butyrate, propionate and acetate, which are collectively called short-chain fatty acids (SCFAs), are generated by fermentation of dietary fibers by gut microbiota. Evidences indicate that SCFAs are key players in regulating beneficial effect of dietary fibers and gut microbiota on our health. SCFAs interact with metabolite-sensing G protein-coupled receptors GPR41, GPR43 and GPR109A expressed in gut epithelium and immune cells. These interactions induce mechanisms that play a key role in maintaining homeostasis in gut and other organs. This review summarizes the protective roles of GPR41, GPR43 and GPR109A in dietary fibers-, gut microbiota- and SCFAs-mediated suppression of inflammation and carcinogenesis in gut and other organs.
Collapse
|
42
|
Rybicki BA, Rundle A, Kryvenko ON, Mitrache N, Do KC, Jankowski M, Chitale DA, Trudeau S, Belinsky SA, Tang D. Methylation in benign prostate and risk of disease progression in men subsequently diagnosed with prostate cancer. Int J Cancer 2016; 138:2884-93. [PMID: 26860439 DOI: 10.1002/ijc.30038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/25/2016] [Indexed: 12/31/2022]
Abstract
In DNA from prostate tumors, methylation patterns in gene promoter regions can be a biomarker for disease progression. It remains unclear whether methylation patterns in benign prostate tissue--prior to malignant transformation--may provide similar prognostic information. To determine whether early methylation events predict prostate cancer outcomes, we evaluated histologically benign prostate specimens from 353 men who eventually developed prostate cancer and received "definitive" treatment [radical prostatectomy (58%) or radiation therapy (42%)]. Cases were drawn from a large hospital-based cohort of men with benign prostate biopsy specimens collected between 1990 and 2002. Risk of disease progression associated with methylation was estimated using time-to-event analyses. Average follow-up was over 5 years; biochemical recurrence (BCR) occurred in 91 cases (26%). In White men, methylation of the APC gene was associated with increased risk of BCR, even after adjusting for standard clinical risk factors for prostate cancer progression (adjusted hazard ratio (aHR) = 2.26; 95%CI 1.23-4.16). APC methylation was most strongly associated with a significant increased risk of BCR in White men with low prostate specific antigen at cohort entry (HR = 3.66; 95%CI 1.51-8.85). In additional stratified analyses, we found that methylation of the RARB gene significantly increased risk of BCR in African American cases who demonstrated methylation of at least one of the other four genes under study (HR = 3.80; 95%CI 1.07-13.53). These findings may have implications in the early identification of aggressive prostate cancer as well as reducing unnecessary medical procedures and emotional distress for men who present with markers of indolent disease.
Collapse
Affiliation(s)
- Benjamin A Rybicki
- Department of Public Health Sciences, Henry Ford Hospital, Detroit, MI.,Josephine Ford Cancer Institute, Henry Ford Hospital, Detroit, MI
| | - Andrew Rundle
- Department of Epidemiology, Columbia University, New York, NY
| | - Oleksandr N Kryvenko
- Department of Pathology and Urology, University of Miami Miller School of Medicine, Miami, FL
| | - Nicoleta Mitrache
- Department of Public Health Sciences, Henry Ford Hospital, Detroit, MI
| | - Kieu C Do
- Lung Cancer Division, Lovelace Respiratory Research Institute, Albuquerque, NM
| | | | - Dhananjay A Chitale
- Josephine Ford Cancer Institute, Henry Ford Hospital, Detroit, MI.,Department of Surgical Pathology, Henry Ford Hospital, Detroit, MI
| | - Sheri Trudeau
- Department of Public Health Sciences, Henry Ford Hospital, Detroit, MI
| | - Steven A Belinsky
- Lung Cancer Division, Lovelace Respiratory Research Institute, Albuquerque, NM
| | - Deliang Tang
- Department of Environmental Health Sciences, Columbia University, New York, NY
| |
Collapse
|
43
|
Lakritz JR, Poutahidis T, Mirabal S, Varian BJ, Levkovich T, Ibrahim YM, Ward JM, Teng EC, Fisher B, Parry N, Lesage S, Alberg N, Gourishetti S, Fox JG, Ge Z, Erdman SE. Gut bacteria require neutrophils to promote mammary tumorigenesis. Oncotarget 2016; 6:9387-96. [PMID: 25831236 PMCID: PMC4496224 DOI: 10.18632/oncotarget.3328] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 02/09/2015] [Indexed: 12/26/2022] Open
Abstract
Recent studies suggest that gastrointestinal tract microbiota modulate cancer development in distant non-intestinal tissues. Here we tested mechanistic hypotheses using a targeted pathogenic gut microbial infection animal model with a predilection to breast cancer. FVB-Tg(C3-1-TAg)cJeg/JegJ female mice were infected by gastric gavage with Helicobacter hepaticus at three-months-of-age putting them at increased risk for mammary tumor development. Tumorigenesis was multifocal and characterized by extensive infiltrates of myeloperoxidase-positive neutrophils otherwise implicated in cancer progression in humans and animal models. To test whether neutrophils were important in etiopathogenesis in this bacteria-triggered model system, we next systemically depleted mice of neutrophils using thrice weekly intraperitoneal injections with anti-Ly-6G antibody. We found that antibody depletion entirely inhibited tumor development in this H. hepaticus-infected model. These data demonstrate that host neutrophil-associated immune responses to intestinal tract microbes significantly impact cancer progression in distal tissues such as mammary glands, and identify gut microbes as novel targets for extra-intestinal cancer therapy.
Collapse
Affiliation(s)
- Jessica R Lakritz
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Theofilos Poutahidis
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Laboratory of Pathology, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, Greece 54124
| | - Sheyla Mirabal
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Bernard J Varian
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tatiana Levkovich
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yassin M Ibrahim
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Ellen C Teng
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Brett Fisher
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Nicola Parry
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Stephanie Lesage
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Natalie Alberg
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sravya Gourishetti
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - James G Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Zhongming Ge
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Susan E Erdman
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
44
|
Jain S, Suklabaidya S, Das B, Raghav SK, Batra SK, Senapati S. TLR4 activation by lipopolysaccharide confers survival advantage to growth factor deprived prostate cancer cells. Prostate 2015; 75:1020-33. [PMID: 25833062 DOI: 10.1002/pros.22983] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 01/23/2015] [Indexed: 02/01/2023]
Abstract
BACKGROUND Prostate cancer (PCa) cells express Toll-like receptor-4 (TLR4), a known pro-tumorigenic molecule for different cancer cells. The cancer cells residing in the avascular region of the tumor confront various metabolic stresses and continuously adapt mechanisms to overcome them. We hypothesized that TLR4 activation might provide direct survival advantage to metabolically stressed PCa cells. METHODS We first investigated the effect of LPS on survival of serum deprived PCa cells. To understand the molecular mechanisms involved in TLR4 mediated PCa survival, we next investigated change in expression of markers for apoptosis, senescence and autophagy. Ultimately, the effect of LPS on established prostate tumors was confirmed in vivo using a syngeneic rat model for PCa. RESULTS Lipopolysaccharide (LPS)-mediated TLR4 activation significantly enhanced survival of serum deprived (SD) PC3, DU145 and MAT-LyLu PCa cells. TLR4 inhibition by a specific inhibitor resulted in rapid death of SD-PC3 cells, which was significantly suppressed by LPS. Interestingly, LPS treatment suppressed macroautophagy in SD-PC3 cells and increased expression of CCL2 (C-C motif ligand-2), a known autophagy inhibitor and pro-survival factor. Intra-tumor LPS injection resulted in increased tumor mass, induced TLR4 activation, suppressed autophagy, and increased the macrophage population in MAT-LyLu-tumors. CONCLUSIONS Our study reveals that bacterial LPS enhance survival of PCa cells under conditions of nutrient stress through TLR4 activation. Moreover, LPS induces overexpression of CCL2 involved in the suppression of starvation-induced macroautophagy in PCa cells, and enhanced macrophage population in prostate tumors in vivo. Taken together, the current study suggests the importance of bacterial infection or TLR4-activation in prostate cancer pathogenesis.
Collapse
Affiliation(s)
- Sumeet Jain
- Institute of Life Sciences, Bhubaneswar, Odisha, India
- Manipal University, Manipal, Karnataka, India
| | - Sujit Suklabaidya
- Institute of Life Sciences, Bhubaneswar, Odisha, India
- Manipal University, Manipal, Karnataka, India
| | - Biswajit Das
- Institute of Life Sciences, Bhubaneswar, Odisha, India
| | | | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, Nebraska
| | | |
Collapse
|
45
|
Molecular Mechanisms and Therapeutic Effects of (-)-Epicatechin and Other Polyphenols in Cancer, Inflammation, Diabetes, and Neurodegeneration. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:181260. [PMID: 26180580 PMCID: PMC4477097 DOI: 10.1155/2015/181260] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 12/23/2014] [Accepted: 12/31/2014] [Indexed: 01/13/2023]
Abstract
With recent insight into the mechanisms involved in diseases, such as cardiovascular disease, cancer, stroke, neurodegenerative diseases, and diabetes, more efficient modes of treatment are now being assessed. Traditional medicine including the use of natural products is widely practiced around the world, assuming that certain natural products contain the healing properties that may in fact have a preventative role in many of the diseases plaguing the human population. This paper reviews the biological effects of a group of natural compounds called polyphenols, including apigenin, epigallocatechin gallate, genistein, and (-)-epicatechin, with a focus on the latter. (-)-Epicatechin has several unique features responsible for a variety of its effects. One of these is its ability to interact with and neutralize reactive oxygen species (ROS) in the cell. (-)-Epicatechin also modulates cell signaling including the MAP kinase pathway, which is involved in cell proliferation. Mutations in this pathway are often associated with malignancies, and the use of (-)-epicatechin holds promise as a preventative agent and as an adjunct for chemotherapy and radiation therapy to improve outcome. This paper discusses the potential of some phenolic compounds to maintain, protect, and possibly reinstate health.
Collapse
|
46
|
Erdman SE, Poutahidis T. Gut bacteria and cancer. Biochim Biophys Acta Rev Cancer 2015; 1856:86-90. [PMID: 26050963 DOI: 10.1016/j.bbcan.2015.05.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 05/24/2015] [Indexed: 02/07/2023]
Abstract
Microbiota on the mucosal surfaces of the gastrointestinal (GI) tract greatly outnumbers the cells in the human body. Effects of antibiotics indicate that GI tract bacteria may be determining the fate of distal cancers. Recent data implicate dysregulated host responses to enteric bacteria leading to cancers in extra-intestinal sites. Together these findings point to novel anti-cancer strategies aimed at promoting GI tract homeostasis.
Collapse
Affiliation(s)
- Susan E Erdman
- Division of Comparative Medicine, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States.
| | - Theofilos Poutahidis
- Division of Comparative Medicine, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States; Laboratory of Pathology, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, 54124, Greece
| |
Collapse
|
47
|
Zhang YJ, Li S, Gan RY, Zhou T, Xu DP, Li HB. Impacts of gut bacteria on human health and diseases. Int J Mol Sci 2015; 16:7493-519. [PMID: 25849657 PMCID: PMC4425030 DOI: 10.3390/ijms16047493] [Citation(s) in RCA: 577] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 03/23/2015] [Accepted: 03/26/2015] [Indexed: 02/07/2023] Open
Abstract
Gut bacteria are an important component of the microbiota ecosystem in the human gut, which is colonized by 1014 microbes, ten times more than the human cells. Gut bacteria play an important role in human health, such as supplying essential nutrients, synthesizing vitamin K, aiding in the digestion of cellulose, and promoting angiogenesis and enteric nerve function. However, they can also be potentially harmful due to the change of their composition when the gut ecosystem undergoes abnormal changes in the light of the use of antibiotics, illness, stress, aging, bad dietary habits, and lifestyle. Dysbiosis of the gut bacteria communities can cause many chronic diseases, such as inflammatory bowel disease, obesity, cancer, and autism. This review summarizes and discusses the roles and potential mechanisms of gut bacteria in human health and diseases.
Collapse
Affiliation(s)
- Yu-Jie Zhang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Sha Li
- School of Chinese Medicine, The University of Hong Kong, Sassoon Road, Hong Kong, China.
| | - Ren-You Gan
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China.
| | - Tong Zhou
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Dong-Ping Xu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
48
|
Dzutsev A, Goldszmid RS, Viaud S, Zitvogel L, Trinchieri G. The role of the microbiota in inflammation, carcinogenesis, and cancer therapy. Eur J Immunol 2014; 45:17-31. [PMID: 25328099 DOI: 10.1002/eji.201444972] [Citation(s) in RCA: 202] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/19/2014] [Accepted: 10/13/2014] [Indexed: 12/11/2022]
Abstract
Commensal microorganisms colonize barrier surfaces of all multicellular organisms, including those of humans. For more than 500 million years, commensal microorganisms and their hosts have coevolved and adapted to each other. As a result, the commensal microbiota affects many immune and nonimmune functions of their hosts, and de facto the two together comprise one metaorganism. The commensal microbiota communicates with the host via biologically active molecules. Recently, it has been reported that microbial imbalance may play a critical role in the development of multiple diseases, such as cancer, autoimmune conditions, and increased susceptibility to infection. In this review, we focus on the role of the commensal microbiota in the development, progression, and immune evasion of cancer, as well as some modulatory effects on the treatment of cancer. In particular, we discuss the mechanisms of microbiota-mediated regulation of innate and adaptive immune responses to tumors, and the consequences on cancer progression and whether tumors subsequently become resistant or susceptible to different anticancer therapeutic regiments.
Collapse
Affiliation(s)
- Amiran Dzutsev
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA; Leidos Biomedical Research, Inc, Frederick, MD, USA
| | | | | | | | | |
Collapse
|
49
|
Ménard A, Péré-Védrenne C, Haesebrouck F, Flahou B. Gastric and enterohepatic helicobacters other than Helicobacter pylori. Helicobacter 2014; 19 Suppl 1:59-67. [PMID: 25167947 DOI: 10.1111/hel.12162] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
During the past year, research on non-Helicobacter pylori species has intensified. H. valdiviensis was isolated from wild birds, and putative novel species have been isolated from Bengal tigers and Australian marsupials. Various genomes have been sequenced: H. bilis, H. canis, H. macacae, H. fennelliae, H. cetorum, and H. suis. Several studies highlighted the virulence of non-H. pylori species including H. cinaedi in humans and hyperlipidemic mice or H. macacae in geriatric rhesus monkeys with intestinal adenocarcinoma. Not surprisingly, increased attention has been paid to the position of Helicobacter species in the microbiota of children and animal species (mice, chickens, penguins, and migrating birds). A large number of experimental studies have been performed in animal models of Helicobacter induced typhlocolitis, showing that the gastrointestinal microbial community is involved in modulation of host pathways leading to chronic inflammation. Animal models of H. suis, H. heilmannii, and H. felis infection have been used to study the development of severe inflammation-related pathologies, including gastric MALT lymphoma and adenocarcinoma.
Collapse
Affiliation(s)
- Armelle Ménard
- Laboratoire de Bactériologie, Centre National de Référence des Campylobacters et Hélicobacters, Université de Bordeaux, F33076, Bordeaux, France; INSERM U853, F33076, Bordeaux, France
| | | | | | | |
Collapse
|
50
|
Poutahidis T, Kleinewietfeld M, Erdman SE. Gut microbiota and the paradox of cancer immunotherapy. Front Immunol 2014; 5:157. [PMID: 24778636 PMCID: PMC3985000 DOI: 10.3389/fimmu.2014.00157] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 03/25/2014] [Indexed: 12/13/2022] Open
Abstract
It is recently shown that beneficial environmental microbes stimulate integrated immune and neuroendocrine factors throughout the body, consequently modulating regulatory T-lymphocyte phenotypes, maintaining systemic immune balance, and determining the fate of preneoplastic lesions toward regression while sustaining whole body good health. Stimulated by a gut microbiota-centric systemic homeostasis hypothesis, we set out to explore the influence of the gut microbiome to explain the paradoxical roles of regulatory T-lymphocytes in cancer development and growth. This paradigm shift places cancer prevention and treatment into a new broader context of holobiont engineering to cultivate a tumor-suppressive macroenvironment.
Collapse
Affiliation(s)
- Theofilos Poutahidis
- Division of Comparative Medicine, Massachusetts Institute of Technology , Cambridge, MA , USA ; Laboratory of Pathology, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki , Thessaloniki , Greece
| | - Markus Kleinewietfeld
- Departments of Neurology and Immunobiology, Yale School of Medicine , New Haven, CT , USA ; Broad Institute, Massachusetts Institute of Technology and Harvard University , Cambridge, MA , USA ; Faculty of Medicine, Dresden University of Technology (TUD) , Dresden , Germany
| | - Susan E Erdman
- Division of Comparative Medicine, Massachusetts Institute of Technology , Cambridge, MA , USA
| |
Collapse
|