551
|
Massironi S, Facciotti F, Cavalcoli F, Amoroso C, Rausa E, Centonze G, Cribiù FM, Invernizzi P, Milione M. Intratumor Microbiome in Neuroendocrine Neoplasms: A New Partner of Tumor Microenvironment? A Pilot Study. Cells 2022; 11:692. [PMID: 35203339 PMCID: PMC8870382 DOI: 10.3390/cells11040692] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 02/05/2023] Open
Abstract
Neuroendocrine neoplasms (NENs) are rare neoplasms with heterogeneous clinical behavior. Alteration in human microbiota was reported in association with carcinogenesis in different solid tumors. However, few studies addressed the role of microbiota in NEN. We here aimed at evaluating the presence of bacterial infiltration in neuroendocrine tumoral tissue. To assess the presence of bacteria, 20 specimens from pancreatic NEN (pan-NEN) and 20 from intestinal NEN (I-NEN) were evaluated through Fluorescent In situ Hybridization and confocal microscopy. Demographic data, pre-operative investigations, operative findings, pathological diagnosis, follow-up, and survival data were evaluated. Among I-NEN, bacteria were detected in 15/20 (75%) specimens, with high variability in microbial distribution. In eight patients, a high infiltration of microorganisms was observed. Among pan-NEN, 18/20 (90%) showed microorganisms' infiltration, with a homogeneous microbial distribution. Bacterial localization in pan-NEN was observed in the proximity of blood vessels. A higher bacterial infiltration in the tumoral specimen as compared with non-tumoral tissue was reported in 10/20 pan-NEN (50%). No significant differences were observed in mean bacterial count according to age, sex, ki67%, site, tumor stage. Mean bacterial count did not result to be a predictor of disease-specific survival. This preliminary study demonstrates the presence of a significant microbiota in the NEN microenvironment. Further research is needed to investigate the potential etiological or clinical role of microbiota in NEN.
Collapse
Affiliation(s)
- Sara Massironi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Ospedale San Gerardo, Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | - Federica Facciotti
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, 20126 Milan, Italy;
| | - Federica Cavalcoli
- Diagnostic and Therapeutic Endoscopy Unit, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy;
| | - Chiara Amoroso
- Department of Experimental Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Emanuele Rausa
- General, Emergency and Trauma Surgery Department, Papa Giovanni XXIII Hospital, 24121 Bergamo, Italy;
| | - Giovanni Centonze
- First Pathology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (G.C.); (M.M.)
| | - Fulvia Milena Cribiù
- Division of Pathology, Fondazione IRCCS Cá Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Ospedale San Gerardo, Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | - Massimo Milione
- First Pathology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (G.C.); (M.M.)
| |
Collapse
|
552
|
Zhang T, Gao G, Sakandar HA, Kwok LY, Sun Z. Gut Dysbiosis in Pancreatic Diseases: A Causative Factor and a Novel Therapeutic Target. Front Nutr 2022; 9:814269. [PMID: 35242797 PMCID: PMC8885515 DOI: 10.3389/fnut.2022.814269] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/21/2022] [Indexed: 12/12/2022] Open
Abstract
Pancreatic-related disorders such as pancreatitis, pancreatic cancer, and type 1 diabetes mellitus (T1DM) impose a substantial challenge to human health and wellbeing. Even though our understanding of the initiation and progression of pancreatic diseases has broadened over time, no effective therapeutics is yet available for these disorders. Mounting evidence suggests that gut dysbiosis is closely related to human health and disease, and pancreatic diseases are no exception. Now much effort is under way to explore the correlation and eventually potential causation between the gut microbiome and the course of pancreatic diseases, as well as to develop novel preventive and/or therapeutic strategies of targeted microbiome modulation by probiotics, prebiotics, synbiotics, postbiotics, and fecal microbiota transplantation (FMT) for these multifactorial disorders. Attempts to dissect the intestinal microbial landscape and its metabolic profile might enable deep insight into a holistic picture of these complex conditions. This article aims to review the subtle yet intimate nexus loop between the gut microbiome and pancreatic diseases, with a particular focus on current evidence supporting the feasibility of preventing and controlling pancreatic diseases via microbiome-based therapeutics and therapies.
Collapse
Affiliation(s)
- Tao Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Guangqi Gao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Hafiz Arbab Sakandar
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Zhihong Sun
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
- *Correspondence: Zhihong Sun
| |
Collapse
|
553
|
Abstract
In this issue of Cancer Cell, Aftab et al. identify a pro-inflammatory cytokine, IL-33, that is released as a chemoattractant for type 2 immune cells in response to the intratumoral mycobiome. Depletion of fungi or deletion of IL-33 in cancer cells significantly decreases pancreatic ductal adenocarcinoma (PDAC) tumor progression and increases survival.
Collapse
Affiliation(s)
- Xin Li
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA; Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Urology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Deepak Saxena
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA; Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Surgery, NYU Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
554
|
Doocey CM, Finn K, Murphy C, Guinane CM. The impact of the human microbiome in tumorigenesis, cancer progression, and biotherapeutic development. BMC Microbiol 2022; 22:53. [PMID: 35151278 PMCID: PMC8840051 DOI: 10.1186/s12866-022-02465-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 02/03/2022] [Indexed: 02/08/2023] Open
Abstract
Abstract
Background
Cancer impacts millions of lives globally each year, with approximately 10 million cancer-related deaths recorded worldwide in 2020. Mounting research has recognised the human microbiome as a key area of interest in the pathophysiology of various human diseases including cancer tumorigenesis, progression and in disease outcome. It is suggested that approximately 20% of human cancers may be linked to microbes. Certain residents of the human microbiome have been identified as potentially playing a role, including: Helicobacter pylori, Fusobacterium nucleatum, Escherichia coli, Bacteroides fragilis and Porphyromonas gingivalis.
Main body
In this review, we explore the current evidence that indicate a link between the human microbiome and cancer. Microbiome compositional changes have been well documented in cancer patients. Furthermore, pathogenic microbes harbouring specific virulence factors have been implicated in driving the carcinogenic activity of various malignancies including colorectal, gastric and pancreatic cancer. The associated genetic mechanisms with possible roles in cancer will be outlined. It will be indicated which microbes have a potential direct link with cancer cell proliferation, tumorigenesis and disease progression. Recent studies have also linked certain microbial cytotoxins and probiotic strains to cancer cell death, suggesting their potential to target the tumour microenvironment given that cancer cells are integral to its composition. Studies pertaining to such cytotoxic activity have suggested the benefit of microbial therapies in oncological treatment regimes. It is also apparent that bacterial pathogenic protein products encoded for by certain loci may have potential as oncogenic therapeutic targets given their possible role in tumorigenesis.
Conclusion
Research investigating the impact of the human microbiome in cancer has recently gathered pace. Vast amounts of evidence indicate the human microbiome as a potential player in tumorigenesis and progression. Promise in the development of cancer biomarkers and in targeted oncological therapies has also been demonstrated, although more studies are needed. Despite extensive in vitro and in vivo research, clinical studies involving large cohorts of human patients are lacking. The current literature suggests that further intensive research is necessary to validate both the role of the human microbiome in cancer, and the use of microbiome modification in cancer therapy.
Collapse
|
555
|
Abstract
The microbiome affects establishment and growth of tumors as well as response to immune-based therapies. In this issue of Immunity, Hezaveh et al. (2022) reveal that metabolites of dietary tryptophan generated by the gut microbiota activate the aryl hydrocarbon receptor in myeloid cells, promoting an immune suppressive tumor microenvironment and facilitating pancreatic ductal adenocarcinoma growth.
Collapse
Affiliation(s)
- Timothy L Frankel
- Department of Surgery, University of Michigan, Ann Arbor, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA.
| |
Collapse
|
556
|
Hezaveh K, Shinde RS, Klötgen A, Halaby MJ, Lamorte S, Ciudad MT, Quevedo R, Neufeld L, Liu ZQ, Jin R, Grünwald BT, Foerster EG, Chaharlangi D, Guo M, Makhijani P, Zhang X, Pugh TJ, Pinto DM, Co IL, McGuigan AP, Jang GH, Khokha R, Ohashi PS, O’Kane GM, Gallinger S, Navarre WW, Maughan H, Philpott DJ, Brooks DG, McGaha TL. Tryptophan-derived microbial metabolites activate the aryl hydrocarbon receptor in tumor-associated macrophages to suppress anti-tumor immunity. Immunity 2022; 55:324-340.e8. [PMID: 35139353 PMCID: PMC8888129 DOI: 10.1016/j.immuni.2022.01.006] [Citation(s) in RCA: 319] [Impact Index Per Article: 106.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 10/19/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a sensor of products of tryptophan metabolism and a potent modulator of immunity. Here, we examined the impact of AhR in tumor-associated macrophage (TAM) function in pancreatic ductal adenocarcinoma (PDAC). TAMs exhibited high AhR activity and Ahr-deficient macrophages developed an inflammatory phenotype. Deletion of Ahr in myeloid cells or pharmacologic inhibition of AhR reduced PDAC growth, improved efficacy of immune checkpoint blockade, and increased intra-tumoral frequencies of IFNγ+CD8+ T cells. Macrophage tryptophan metabolism was not required for this effect. Rather, macrophage AhR activity was dependent on Lactobacillus metabolization of dietary tryptophan to indoles. Removal of dietary tryptophan reduced TAM AhR activity and promoted intra-tumoral accumulation of TNFα+IFNγ+CD8+ T cells; provision of dietary indoles blocked this effect. In patients with PDAC, high AHR expression associated with rapid disease progression and mortality, as well as with an immune-suppressive TAM phenotype, suggesting conservation of this regulatory axis in human disease.
Collapse
Affiliation(s)
- Kebria Hezaveh
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,These authors contributed equally,Present address: Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceutical R&D, Astra Zeneca, Gothenburg, 431 50, Sweden
| | - Rahul S. Shinde
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,These authors contributed equally,Present address: Immunology, Microenvironment, and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Andreas Klötgen
- Department of Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany
| | - Marie Jo Halaby
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Sara Lamorte
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - M. Teresa Ciudad
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Rene Quevedo
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Luke Neufeld
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Zhe Qi Liu
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Robbie Jin
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Barbara T. Grünwald
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | | | - Danica Chaharlangi
- Department of Molecular Genetics, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Mengdi Guo
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Priya Makhijani
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Xin Zhang
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Trevor J. Pugh
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Medical Biophysics, The University of Toronto, Toronto, ON M5G 1L7, Canada,The Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Devanand M. Pinto
- National Research Council, Human Health Therapeutics, Halifax, NS B3H 3Z1, Canada
| | - Ileana L. Co
- Institute of Biomedical Engineering, The University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Alison P. McGuigan
- Institute of Biomedical Engineering, The University of Toronto, Toronto, ON M5S 3G9, Canada,Department of Chemical Engineering and Applied Chemistry, The University of Toronto, Toronto, ON M5S 3E5, Canada
| | - Gun Ho Jang
- The Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Rama Khokha
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada,Department of Medical Biophysics, The University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Pamela S. Ohashi
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Grainne M. O’Kane
- The Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada,Division of Medical Oncology, Department of Medicine, The University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Steven Gallinger
- The Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada,Department of Laboratory Medicine and Pathobiology, The University of Toronto, Toronto, ON M5S 1A8, Canada,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - William W. Navarre
- Department of Molecular Genetics, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | - Dana J. Philpott
- Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - David G. Brooks
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tracy L. McGaha
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada,Lead contact,Correspondence:
| |
Collapse
|
557
|
Bacterial Translocation in Gastrointestinal Cancers and Cancer Treatment. Biomedicines 2022; 10:biomedicines10020380. [PMID: 35203589 PMCID: PMC8962358 DOI: 10.3390/biomedicines10020380] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 11/16/2022] Open
Abstract
In recent years, there has been increasing evidence that gut microbiota is associated with the onset and exacerbation of various diseases, such as gastrointestinal cancer. For instance, it is well known that local inflammation of the intestinal tract in colorectal cancer that is caused by the increased number of Fusobacterium, due to changes in the intestinal bacterial flora, is involved in carcinogenesis. In contrast, gut bacteria or their products, pathogen-associated molecular patterns, not only cause intestinal inflammation but also invade the bloodstream through dysbiosis and gut barrier dysfunction, thereby leading to systemic inflammation, namely bacterial translocation. The involvement of bacterial translocation in the carcinogenesis of gastrointestinal cancers and their prognosis is increasingly being recognized. The Toll-like receptor signaling pathways plays an important role in the carcinogenesis of such cancers. In addition, bacterial translocation influences the treatment of cancers such as surgery and chemotherapy. In this review, we outline the concept of bacterial translocation, summarize the current knowledge on the relationship between gut bacteria and gastrointestinal cancer, and provide future perspectives of this field.
Collapse
|
558
|
pH-taxis drives aerobic bacteria in duodenum to migrate into the pancreas with tumors. Sci Rep 2022; 12:1783. [PMID: 35110595 PMCID: PMC8810860 DOI: 10.1038/s41598-022-05554-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/13/2022] [Indexed: 01/07/2023] Open
Abstract
As oral or intestinal bacteria have been found in pancreatic cystic fluid and tumors, understanding bacterial migration from the duodenum into the pancreas via hepato-pancreatic duct is critical. Mathematical models of migration of aerobic bacteria from the duodenum to the pancreas with tumors were developed. Additionally, the bacterial distributions under the pH gradient and those under flow were measured in double-layer flow based microfluidic device and T-shaped cylinders. Migration of aerobic bacteria from the duodenum into pancreas is counteracted by bile and pancreatic juice flow but facilitated by pH-taxis from acidic duodenum fluid toward more favorable slightly alkaline pH in pancreatic juice. Additionally, the reduced flow velocity in cancer patients, due to compressed pancreatic duct by solid tumor, facilitates migration. Moreover, measured distribution of GFP E. coli under the pH gradient in a microfluidic device validated pH-tactic behaviors. Furthermore, Pseudomonas fluorescens in hydrochloride solution, but not in bicarbonate solution, migrated upstream against bicarbonate flow of > 20 μm/s, with an advancement at approximately 50 μm/s.
Collapse
|
559
|
Jiang J, Mei J, Ma Y, Jiang S, Zhang J, Yi S, Feng C, Liu Y, Liu Y. Tumor hijacks macrophages and microbiota through extracellular vesicles. EXPLORATION (BEIJING, CHINA) 2022; 2:20210144. [PMID: 37324578 PMCID: PMC10190998 DOI: 10.1002/exp.20210144] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/16/2021] [Indexed: 06/17/2023]
Abstract
The tumor microenvironment (TME) is a biological system with sophisticated constituents. In addition to tumor cells, tumor-associated macrophages (TAMs) and microbiota are also dominant components. The phenotypic and functional changes of TAMs are widely considered to be related to most tumor progressions. The chronic colonization of pathogenic microbes and opportunistic pathogens accounts for the generation and development of tumors. As messengers of cell-to-cell communication, tumor-derived extracellular vesicles (TDEVs) can transfer various malignant factors, regulating physiological and pathological changes in the recipients and affecting TAMs and microbes in the TME. Despite the new insights into tumorigenesis and progress brought by the above factors, the crosstalk among tumor cells, macrophages, and microbiota remain elusive, and few studies have focused on how TDEVs act as an intermediary. We reviewed how tumor cells recruit and domesticate macrophages and microbes through extracellular vehicles and how hijacked macrophages and microbiota interact with tumor-promoting feedback, achieving a reciprocal coexistence under the TME and working together to facilitate tumor progression. It is significant to seek evidence to clarify those specific interactions and reveal therapeutic targets to curb tumor progression and improve prognosis.
Collapse
Affiliation(s)
- Jipeng Jiang
- Postgraduate School Medical School of Chinese PLA Beijing P. R. China
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Jie Mei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Beijing P. R. China
- University of Chinese Academy of Science Beijing P. R. China
| | - Yongfu Ma
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Shasha Jiang
- Postgraduate School Medical School of Chinese PLA Beijing P. R. China
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Jian Zhang
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Shaoqiong Yi
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Changjiang Feng
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Yang Liu
- Postgraduate School Medical School of Chinese PLA Beijing P. R. China
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Beijing P. R. China
- GBA National Institute for Nanotechnology Innovation Guangdong P. R. China
| |
Collapse
|
560
|
Newsome RC, Yang Y, Jobin C. The microbiome, gastrointestinal cancer, and immunotherapy. J Gastroenterol Hepatol 2022; 37:263-272. [PMID: 34820895 PMCID: PMC9922516 DOI: 10.1111/jgh.15742] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022]
Abstract
The gastrointestinal tract greatly contributes to global cancer burden and cancer-related deaths. The microbiota represents the population of microorganisms that live in and around the body, located primarily in the gastrointestinal tract. The microbiota has been implicated in colorectal cancer development and progression, but its role in cancer therapy for the gastrointestinal tract is less defined, especially for extra-intestinal cancers. In this review, we discuss the past 5 years of research into microbial involvement in immune-related therapies for colorectal, pancreatic, hepatic, and gastric cancers, with the goal of highlighting recent advances and new areas for investigation in this field.
Collapse
Affiliation(s)
- Rachel C Newsome
- Departments of Medicine, University of Florida, Gainesville, Florida, USA
| | - Ye Yang
- Departments of Medicine, University of Florida, Gainesville, Florida, USA
| | - Christian Jobin
- Departments of Medicine, University of Florida, Gainesville, Florida, USA
- Infectious Diseases and Immunology, University of Florida, Gainesville, Florida, USA
- Anatomy and Cell Biology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
561
|
Sholl J, Sepich-Poore GD, Knight R, Pradeu T. Redrawing therapeutic boundaries: microbiota and cancer. Trends Cancer 2022; 8:87-97. [PMID: 34844910 PMCID: PMC8770609 DOI: 10.1016/j.trecan.2021.10.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/28/2021] [Indexed: 02/07/2023]
Abstract
The unexpected roles of the microbiota in cancer challenge explanations of carcinogenesis that focus on tumor-intrinsic properties. Most tumors contain bacteria and viruses, and the host's proximal and distal microbiota influence both cancer incidence and therapeutic responsiveness. Continuing the history of cancer-microbe research, these findings raise a key question: to what extent is the microbiota relevant for clinical oncology? We approach this by critically evaluating three issues: how the microbiota provides a predictive biomarker of cancer growth and therapeutic responsiveness, the microbiota's causal role(s) in cancer development, and how therapeutic manipulations of the microbiota improve patient outcomes in cancer. Clarifying the conceptual and empirical aspects of the cancer-associated microbiota can orient future research and guide its implementation in clinical oncology.
Collapse
Affiliation(s)
- Jonathan Sholl
- University of Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, 33000 Bordeaux, France.
| | | | - Rob Knight
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA; Department of Pediatrics, University of California San Diego, La Jolla, CA, USA; Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA; Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Thomas Pradeu
- University of Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, 33000 Bordeaux, France.
| |
Collapse
|
562
|
Kohi S, Macgregor-Das A, Dbouk M, Yoshida T, Chuidian M, Abe T, Borges M, Lennon AM, Shin EJ, Canto MI, Goggins M. Alterations in the Duodenal Fluid Microbiome of Patients With Pancreatic Cancer. Clin Gastroenterol Hepatol 2022; 20:e196-e227. [PMID: 33161160 PMCID: PMC8120597 DOI: 10.1016/j.cgh.2020.11.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/28/2020] [Accepted: 11/01/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS The tumor microbiome of patients with pancreas ductal adenocarcinoma (PDAC) includes bacteria normally present in the upper gastrointestinal tract. If the predominant source of intratumoral bacteria in patients with PDAC is retrograde migration from the duodenum, duodenal fluid could be a representative biospecimen for determining microbiome profiles of patients with PDAC or at risk of developing PDAC. METHODS We performed a case-control study comparing bacterial and fungal (16S and 18S rRNA) profiles of secretin-stimulated duodenal fluid collections from 308 patients undergoing duodenal endoscopy including 134 normal pancreas control subjects, 98 patients with pancreatic cyst(s) and 74 patients with PDAC. RESULTS Alterations in duodenal fluid microbiomes with diminished alpha diversity were significantly associated with age >70 and proton pump inhibitor use. Patients with PDAC had significantly decreased duodenal microbial alpha diversity compared with age-matched control subjects with normal pancreata and those with pancreatic cyst(s). There was evidence of enrichment of Bifidobacterium genera in the duodenal fluid of patients with PDAC compared with control subjects and those with pancreatic cyst(s). There were also enrichment of duodenal fluid Fusobacteria and Rothia bacteria among patients with PDAC with short-term survival. Duodenal fluid microbiome profiles were not significantly different between control subjects and patients with pancreatic cyst(s). CONCLUSION Patients with PDAC have alterations in their duodenal fluid microbiome profiles compared with patients with pancreatic cysts and those with normal pancreata. ClinicalTrials.gov, Number: NCT02000089.
Collapse
Affiliation(s)
- Shiro Kohi
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Anne Macgregor-Das
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Mohamad Dbouk
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Takeichi Yoshida
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Miguel Chuidian
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Toshiya Abe
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Michael Borges
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Anne Marie Lennon
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Eun Ji Shin
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD.,Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Marcia Irene Canto
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD.,Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Michael Goggins
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland; Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, Maryland; Department of Oncology, the Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland.
| |
Collapse
|
563
|
Golčić M, Simetić L, Majnarić T, Golčić G, Herceg D. Could fecal microbial transplantation offer a new potential in the treatment of metastatic pancreatic ductal adenocarcinoma? Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
564
|
Huang Y, Zhu N, Zheng X, Liu Y, Lu H, Yin X, Hao H, Tan Y, Wang D, Hu H, Liang Y, Li X, Hu Z, Yin Y. Intratumor Microbiome Analysis Identifies Positive Association Between Megasphaera and Survival of Chinese Patients With Pancreatic Ductal Adenocarcinomas. Front Immunol 2022; 13:785422. [PMID: 35145519 PMCID: PMC8821101 DOI: 10.3389/fimmu.2022.785422] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/03/2022] [Indexed: 12/24/2022] Open
Abstract
Human tumors harbor a plethora of microbiota. It has been shown that the composition and diversity of intratumor microbiome are significantly associated with the survival of patients with pancreatic ductal adenocarcinoma (PDAC). However, the association in Chinese patients as well as the effect of different microorganisms on inhibiting tumor growth are unclear. In this study, we collected tumor samples resected from long-term and short-term PDAC survivors and performed 16S rRNA amplicon sequencing. We found that the microbiome in samples with different survival time were significantly different, and the differential bacterial composition was associated with the metabolic pathways in the tumor microenvironment. Furthermore, administration of Megasphaera, one of the differential bacteria, induced a better tumor growth inhibition effect when combined with the immune checkpoint inhibitor anti-programmed cell death-1 (anti-PD-1) treatment in mice bearing 4T1 tumor. These results indicate that specific intratumor microbiome can enhance the anti-tumor effect in the host, laying a foundation for further clarifying the underlying detailed mechanism.
Collapse
Affiliation(s)
- Yu Huang
- Department of General Surgery, No.903 Hospital of People’s Liberation Army Joint Logistic Support Forcel, Hangzhou, China
| | - Ning Zhu
- Department of Research and Development, Shenzhen Xbiome Biotech Co. Ltd., Shenzhen, China
| | - Xing Zheng
- Department of Research and Development, Shenzhen Xbiome Biotech Co. Ltd., Shenzhen, China
| | - Yanhong Liu
- Department of Research and Development, Shenzhen Xbiome Biotech Co. Ltd., Shenzhen, China
| | - Haopeng Lu
- Department of Research and Development, Shenzhen Xbiome Biotech Co. Ltd., Shenzhen, China
| | - Xiaochen Yin
- Department of Research and Development, Shenzhen Xbiome Biotech Co. Ltd., Shenzhen, China
| | - Huaijie Hao
- Department of Research and Development, Shenzhen Xbiome Biotech Co. Ltd., Shenzhen, China
| | - Yan Tan
- Department of Research and Development, Shenzhen Xbiome Biotech Co. Ltd., Shenzhen, China
| | - Dongjie Wang
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Han Hu
- Department of Research and Development, Shenzhen Xbiome Biotech Co. Ltd., Shenzhen, China
| | - Yong Liang
- Department of Research and Development, Shenzhen Xbiome Biotech Co. Ltd., Shenzhen, China
| | - Xinxing Li
- Department of General Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiqian Hu
- Department of General Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of General Surgery, Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Yiming Yin
- Department of Research and Development, Shenzhen Xbiome Biotech Co. Ltd., Shenzhen, China
| |
Collapse
|
565
|
Abstract
Chronic inflammation increases the risk of several cancers, including gastric, colon, and hepatic cancers. Conversely, tumors, similar to tissue injury, trigger an inflammatory response coordinated by the innate immune system. Cellular and molecular mediators of inflammation modulate tumor growth directly and by influencing the adaptive immune response. Depending on the balance of immune cell types and signals within the tumor microenvironment, inflammation can support or restrain the tumor. Adding to the complexity, research from the past two decades has revealed that innate immune cells are highly heterogeneous and plastic, with variable phenotypes depending on tumor type, stage, and treatment. The field is now on the cusp of being able to harness this wealth of data to (a) classify tumors on the basis of their immune makeup, with implications for prognosis, treatment choice, and clinical outcome, and (b) design therapeutic strategies that activate antitumor immune responses by targeting innate immune cells.
Collapse
Affiliation(s)
- Laura Maiorino
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA;
| | | | - Lijuan Sun
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA; , ,
| | - Mikala Egeblad
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA; , ,
| |
Collapse
|
566
|
Gut microbiome alteration as a diagnostic tool and associated with inflammatory response marker in primary liver cancer. Hepatol Int 2022; 16:99-111. [DOI: 10.1007/s12072-021-10279-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/12/2021] [Indexed: 12/14/2022]
|
567
|
Havasi A, Sur D, Cainap SS, Lungulescu CV, Gavrilas LI, Cainap C, Vlad C, Balacescu O. Current and New Challenges in the Management of Pancreatic Neuroendocrine Tumors: The Role of miRNA-Based Approaches as New Reliable Biomarkers. Int J Mol Sci 2022; 23:1109. [PMID: 35163032 PMCID: PMC8834851 DOI: 10.3390/ijms23031109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/10/2022] [Accepted: 01/18/2022] [Indexed: 12/17/2022] Open
Abstract
Pancreatic neuroendocrine tumors (PanNETs) are rare tumors; however, their incidence greatly increases with age, and they occur more frequently among the elderly. They represent 5% of all pancreatic tumors, and despite the fact that low-grade tumors often have an indolent evolution, they portend a poor prognosis in an advanced stages and undifferentiated tumors. Additionally, functional pancreatic neuroendocrine tumors greatly impact quality of life due to the various clinical syndromes that result from abnormal hormonal secretion. With limited therapeutic and diagnostic options, patient stratification and selection of optimal therapeutic strategies should be the main focus. Modest improvements in the management of pancreatic neuroendocrine tumors have been achieved in the last years. Therefore, it is imperative to find new biomarkers and therapeutic strategies to improve patient survival and quality of life, limiting the disease burden. MicroRNAs (miRNAs) are small endogenous molecules that modulate the expression of thousands of genes and control numerous critical processes involved in tumor development and progression. New data also suggest the implication of miRNAs in treatment resistance and their potential as prognostic or diagnostic biomarkers and therapeutic targets. In this review, we discusses the current and new challenges in the management of PanNETs, including genetic and epigenetic approaches. Furthermore, we summarize the available data on miRNAs as potential prognostic, predictive, or diagnostic biomarkers and discuss their function as future therapeutic targets.
Collapse
Affiliation(s)
- Andrei Havasi
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania; (A.H.); (C.C.)
- 11th Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400015 Cluj-Napoca, Romania;
- MedEuropa Radiotherapy Center, 410191 Oradea, Romania
| | - Daniel Sur
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania; (A.H.); (C.C.)
- 11th Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400015 Cluj-Napoca, Romania;
| | - Simona Sorana Cainap
- Department of Mother and Child, Pediatric Cardiology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400015 Cluj-Napoca, Romania;
| | | | - Laura-Ioana Gavrilas
- Department of Bromatology, Hygiene, Nutrition, University of Medicine and Pharmacy “Iuliu Hatieganu”, 23 Marinescu Street, 400337 Cluj-Napoca, Romania;
| | - Calin Cainap
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania; (A.H.); (C.C.)
- 11th Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400015 Cluj-Napoca, Romania;
| | - Catalin Vlad
- Department of Surgery, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 34–36, Republicii Street, 400015 Cluj-Napoca, Romania;
- Department of Oncology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8, Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Ovidiu Balacescu
- 11th Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400015 Cluj-Napoca, Romania;
- Department of Genetics, Genomics and Experimental Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuta’’, 400015 Cluj-Napoca, Romania
| |
Collapse
|
568
|
Hanahan D. Hallmarks of Cancer: New Dimensions. Cancer Discov 2022; 12:31-46. [PMID: 35022204 DOI: 10.1158/2159-8290.cd-21-1059] [Citation(s) in RCA: 4564] [Impact Index Per Article: 1521.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 02/06/2023]
Abstract
The hallmarks of cancer conceptualization is a heuristic tool for distilling the vast complexity of cancer phenotypes and genotypes into a provisional set of underlying principles. As knowledge of cancer mechanisms has progressed, other facets of the disease have emerged as potential refinements. Herein, the prospect is raised that phenotypic plasticity and disrupted differentiation is a discrete hallmark capability, and that nonmutational epigenetic reprogramming and polymorphic microbiomes both constitute distinctive enabling characteristics that facilitate the acquisition of hallmark capabilities. Additionally, senescent cells, of varying origins, may be added to the roster of functionally important cell types in the tumor microenvironment. SIGNIFICANCE: Cancer is daunting in the breadth and scope of its diversity, spanning genetics, cell and tissue biology, pathology, and response to therapy. Ever more powerful experimental and computational tools and technologies are providing an avalanche of "big data" about the myriad manifestations of the diseases that cancer encompasses. The integrative concept embodied in the hallmarks of cancer is helping to distill this complexity into an increasingly logical science, and the provisional new dimensions presented in this perspective may add value to that endeavor, to more fully understand mechanisms of cancer development and malignant progression, and apply that knowledge to cancer medicine.
Collapse
Affiliation(s)
- Douglas Hanahan
- Ludwig Institute for Cancer Research - Lausanne Branch, Lausanne, Switzerland. The Swiss Institute for Experimental Cancer Research (ISREC) within the School of Life Sciences at the Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland. The Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland.
| |
Collapse
|
569
|
Xu F, Pushalkar S, Lin Z, Thomas SC, Persaud JK, Sierra MA, Vardhan M, Vasconcelos R, Akapo A, Guo Y, Gordon T, Corby PM, Kamer AR, Li X, Saxena D. Electronic cigarette use enriches periodontal pathogens. Mol Oral Microbiol 2022; 37:63-76. [PMID: 34997976 DOI: 10.1111/omi.12361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/03/2022] [Accepted: 01/06/2022] [Indexed: 11/28/2022]
Abstract
The effect of electronic cigarette (e-cigarette) smoking, especially its long-term impact on oral health, is poorly understood. Here, we conducted a longitudinal clinical study with two study visits, 6 months apart, to investigate the effect of e-cigarette use on the bacterial community structure in the saliva of 101 periodontitis patients. Our data demonstrated that e-cigarette use altered the oral microbiome in periodontitis patients, enriching members of the Filifactor, Treponema, and Fusobacterium taxa. For patients at the same periodontal disease stage, cigarette smokers and e-cigarette smokers shared more similarities in their oral bacterial composition. E-cigarette smoking may have a similar potential as cigarette smoking at altering the bacterial composition of saliva over time, leading to an increase in the relative abundance of periodontal disease-associated pathogens such as Porphyromonas gingivalis and Fusobacterium nucleatum. The correlation analysis showed that certain genera, such as Dialister, Selenomonas, and Leptotrichia in the e-cigarette smoking group, were positively correlated with the levels of proinflammatory cytokines, including IFN-γ, IL-1β, and TNF-α. E-cigarette use was also associated with elevated levels of proinflammatory cytokines such as IFN-γ and TNF-α, which contribute to oral microbiome dysbiosis and advanced disease state. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Fangxi Xu
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Smruti Pushalkar
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Ziyan Lin
- Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Scott C Thomas
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Julia Kishanie Persaud
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Maria A Sierra
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Mridula Vardhan
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Rebeca Vasconcelos
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Adenike Akapo
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Yuqi Guo
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Terry Gordon
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, United States
| | - Patricia M Corby
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Angela R Kamer
- Department of Periodontology and Implant Dentistry, New York University College of Dentistry, New York, NY, United States
| | - Xin Li
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Deepak Saxena
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| |
Collapse
|
570
|
Chu CS, Yang CY, Yeh CC, Lin RT, Chen CC, Bai LY, Hung MC, Lin CC, Wu CY, Lin JT. Endoscopic ultrasound-guided fine-needle biopsy as a tool for studying the intra-tumoral microbiome in pancreatic ductal adenocarcinoma: a pilot study. Sci Rep 2022; 12:107. [PMID: 34997106 PMCID: PMC8741880 DOI: 10.1038/s41598-021-04095-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/15/2021] [Indexed: 12/04/2022] Open
Abstract
A new approach by investigating the intra-tumoral microbiome raised great interest because they may influence the host immune response and natural history of the disease. However, previous studies on the intra-tumoral microbiome of pancreatic ductal adenocarcinoma (PDAC) were mostly based on examining the formalin-fixed paraffin-embedded tumor specimens. This study aims to investigate the feasibility of using endoscopic ultrasound-guided fine-needle biopsy (EUS-FNB) as a complementary procedure of surgical biopsy to obtain adequate fresh pancreatic cancer tissue for intra-tumoral microbial research. This was a prospective pilot study performed at a single tertiary referral center. We obtained pancreatic cancer tissue by EUS-FNB and surgical biopsy, respectively. We amplified the V3-V4 hyper-variable region of bacterial 16S ribosomal ribonucleic acid (rRNA) genes, constructed a pair-end library, and performed high-throughput sequencing. From August 2020 to November 2020, nine eligible patients with PDAC were enrolled in this study. The intra-tumoral microbiome profile was successfully generated from the PDAC cancer tissue obtained by EUS-FNB as well as by surgical biopsy. There was no significant difference in intra-tumoral alpha-diversity or bacterial taxonomic composition between tissues obtained by EUS-FNB and by surgical biopsy. EUS-FNB can collect sufficient fresh cancer tissue for microbiome analyses without complication. The intra-tumoral microbiome profile in tissues obtained by EUS-FNB had similar alpha-diversity and taxonomic profiles with those obtained by surgical biopsy. It implicated, except for surgical biopsy, EUS-FNB can be another valid and valuable tool for studying intra-tumoral microbiome in patients with resectable and unresectable PDAC.
Collapse
Affiliation(s)
- Chia-Sheng Chu
- Digestive Medicine Center, China Medical University Hospital, 2 Yuh-Der Road, Taichung, 404, Taiwan
| | - Chi-Ying Yang
- Digestive Medicine Center, China Medical University Hospital, 2 Yuh-Der Road, Taichung, 404, Taiwan
| | - Chun-Chieh Yeh
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Ro-Ting Lin
- College of Public Health, China Medical University, Taichung, Taiwan
| | - Chi-Ching Chen
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Li-Yuan Bai
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan.,School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Mien-Chie Hung
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.,The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chun-Che Lin
- Digestive Medicine Center, China Medical University Hospital, 2 Yuh-Der Road, Taichung, 404, Taiwan.,School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Chun-Ying Wu
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jaw-Town Lin
- Digestive Medicine Center, China Medical University Hospital, 2 Yuh-Der Road, Taichung, 404, Taiwan.
| |
Collapse
|
571
|
Current Status and Future Therapeutic Options for Fecal Microbiota Transplantation. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58010084. [PMID: 35056392 PMCID: PMC8780626 DOI: 10.3390/medicina58010084] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/23/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022]
Abstract
The intestinal microbiota plays an important role in maintaining human health, and its alteration is now associated with the development of various gastrointestinal (ulcerative colitis, irritable bowel syndrome, constipation, etc.) and extraintestinal diseases, such as cancer, metabolic syndrome, neuropsychiatric diseases. In this context, it is not surprising that gut microbiota modification methods may constitute a therapy whose potential has not yet been fully investigated. In this regard, the most interesting method is thought to be fecal microbiota transplantation, which consists of the simultaneous replacement of the intestinal microbiota of a sick recipient with fecal material from a healthy donor. This review summarizes the most interesting findings on the application of fecal microbiota transplantation in gastrointestinal and extraintestinal pathologies.
Collapse
|
572
|
Michaud D, Mirlekar B, Steward C, Bishop G, Pylayeva-Gupta Y. B Cell Receptor Signaling and Protein Kinase D2 Support Regulatory B Cell Function in Pancreatic Cancer. Front Immunol 2022; 12:745873. [PMID: 35046933 PMCID: PMC8761795 DOI: 10.3389/fimmu.2021.745873] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 12/07/2021] [Indexed: 12/24/2022] Open
Abstract
B cells can act as potent suppressors of anti-tumor T cell immunity, presenting a mechanism of resistance to immunotherapy. In pancreatic ductal adenocarcinoma, B cells can display a T cell-suppressive or regulatory phenotype centered on the expression of the cytokine Interleukin 35 (IL-35). While B cell-mediated immunosuppression presents a barrier to anti-tumorigenic T cell function, it is not clear how regulatory B cell function could be targeted, and the signals that promote this suppressive phenotype in B cells are not well understood. Here we use a novel IL-35 reporter model to understand which signaling pathways are important for immunosuppressive properties in B cells. In vitro analysis of IL-35 reporter B cells revealed a synergy between the BCR and TLR4 signaling pathways is sufficient to induce IL-35 expression. However, in vivo, B cell receptor activation, as opposed to MyD88 signaling in B cells, is central to B cell-mediated suppression and promotion of pancreatic cancer growth. Further analysis identified protein kinase D2 (PKD2) as being a key downstream regulator of IL-35 expression in B cells. Regulatory B cells with an inactivating mutation in PKD2 failed to produce IL-35 or fully suppress effector T cell function in vitro. Furthermore, inhibition of PKD in B cells decreased tumor growth and promoted effector T cell function upon adoptive transfer into B cell-deficient mice. Collectively, these data provide insight into how regulatory B cell function is promoted in pancreatic cancer and identify potential therapeutic targets to restrain this function.
Collapse
Affiliation(s)
- Daniel Michaud
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| | - Bhalchandra Mirlekar
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| | - Colleen Steward
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| | - Gail Bishop
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
- Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Yuliya Pylayeva-Gupta
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
- Department of Genetics, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| |
Collapse
|
573
|
Microbiota in relation to cancer. Cancer 2022. [DOI: 10.1016/b978-0-323-91904-3.00007-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
574
|
Jiang J, Mei J, Yi S, Feng C, Ma Y, Liu Y, Liu Y, Chen C. Tumor associated macrophage and microbe: The potential targets of tumor vaccine delivery. Adv Drug Deliv Rev 2022; 180:114046. [PMID: 34767863 DOI: 10.1016/j.addr.2021.114046] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 10/29/2021] [Accepted: 11/04/2021] [Indexed: 02/08/2023]
Abstract
The occurrence and development of tumors depend on the tumor microenvironment (TME), which is made of various immune cells, activated fibroblasts, basement membrane, capillaries, and extracellular matrix. Tumor associated macrophages (TAMs) and microbes are important components in TME. Tumor cells can recruit and educate TAMs and microbes, and the hijacked TAMs and microbes can promote the progression of tumor reciprocally. Tumor vaccine delivery remodeling TME by targeting TAM and microbes can not only enhance the specificity and immunogenicity of antigens, but also contribute to the regulation of TME. Tumor vaccine design benefits from nanotechnology which is a suitable platform for antigen and adjuvant delivery to catalyze new candidate vaccines applying to clinical therapy at unparalleled speed. In view of the characteristics and mechanisms of TME development, vaccine delivery targeting and breaking the malignant interactions among tumor cells, TAMs, and microbes may serve as a novel strategy for tumor therapy.
Collapse
|
575
|
Jiang Q, Liu X, Yang Q, Chen L, Yang D. Salivary Microbiome in Adenoid Cystic Carcinoma Detected by 16S rRNA Sequencing and Shotgun Metagenomics. Front Cell Infect Microbiol 2022; 11:774453. [PMID: 34970508 PMCID: PMC8712576 DOI: 10.3389/fcimb.2021.774453] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 11/25/2021] [Indexed: 01/14/2023] Open
Abstract
Microorganisms are confirmed to be closely related to the occurrence and development of cancers in human beings. However, there has been no published report detailing relationships between the oral microbiota and salivary adenoid cystic carcinoma (SACC). In this study, unstimulated saliva was collected from 13 SACC patients and 10 healthy controls. The microbial diversities, compositions and functions were comprehensively analyzed after 16S rRNA sequencing and whole-genome shotgun metagenomic sequencing. The alpha diversity showed no significant difference between SACC patients and healthy controls, while beta diversity showed a separation trend. The SACC patients showed higher abundances of Streptococcus and Rothia, while Prevotella and Alloprevotella were more abundant in healthy controls. The prevalent KEGG pathways, carbohydrate-active enzymes, antibiotic resistances and virulence factors as well as the biomarkers in SACC were determined by functional gene analysis. Our study preliminarily investigated the salivary microbiome of SACC patients compared with healthy controls and might be the basis for further studies on novel diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Qian Jiang
- Stomatological Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Xing Liu
- Stomatological Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Qifen Yang
- Stomatological Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Liang Chen
- Stomatological Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Deqin Yang
- Stomatological Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
576
|
Mahapatra S, Mohanty S, Mishra R, Prasad P. An overview of cancer and the human microbiome. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 191:83-139. [DOI: 10.1016/bs.pmbts.2022.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
577
|
Li H, Xie J, Guo X, Yang G, Cai B, Liu J, Yue M, Tang Y, Wang G, Chen S, Guo J, Qi X, Wang D, Zheng H, Liu W, Yu H, Wang C, Zhu SJ, Guo F. Bifidobacterium spp. and their metabolite lactate protect against acute pancreatitis via inhibition of pancreatic and systemic inflammatory responses. Gut Microbes 2022; 14:2127456. [PMID: 36195972 PMCID: PMC9542615 DOI: 10.1080/19490976.2022.2127456] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 08/16/2022] [Accepted: 09/16/2022] [Indexed: 02/04/2023] Open
Abstract
Severe acute pancreatitis (SAP) is a critical illness characterized by a severe systemic inflammatory response resulting in persistent multiple organ failure and sepsis. The intestinal microbiome is increasingly appreciated to play a crucial role in modulation of AP disease outcome, but limited information is available about the identity and mechanism of action for specific commensal bacteria involved in AP-associated inflammation. Here we show that Bifidobacteria, particularly B. animalis, can protect against AP by regulating pancreatic and systemic inflammation in germ-free (GF) and oral antibiotic-treated (Abx) mouse models. Colonization by B. animalis and administration of its metabolite lactate protected Abx and GF mice from AP by reducing serum amylase concentration, ameliorating pancreatic lesions and improving survival rate after retrograde injection of sodium taurocholate. B. animalis relieved macrophage-associated local and systemic inflammation of AP in a TLR4/MyD88- and NLRP3/Caspase1-dependent manner through its metabolite lactate. Supporting our findings from the mouse study, clinical AP patients exhibited a decreased fecal abundance of Bifidobacteria that was inversely correlated with the severity of systemic inflammatory responses. These results may shed light on the heterogeneity of clinical outcomes and drive the development of more efficacious therapeutic interventions for AP, and potentially for other inflammatory disorders.
Collapse
Affiliation(s)
- Han Li
- Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, China
| | - Jinyan Xie
- Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, China
| | - Xiuliu Guo
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guilian Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
| | - Bin Cai
- Department of Quality Management, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Central Laboratory of Medicine, Shaoxing People’s Hospital, Shaoxing, China
| | - Jingtianyi Liu
- Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, China
| | - Mengjia Yue
- Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, China
| | - Yixin Tang
- Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, China
| | - Gan Wang
- Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, China
| | - Shuxian Chen
- Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, China
| | - Jialin Guo
- Institute of Land and Food Systems, University of British Columbia, Vancouver, BC, Canada
| | - Xuchen Qi
- Central Laboratory of Medicine, Shaoxing People’s Hospital, Shaoxing, China
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Donghai Wang
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huijun Zheng
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Hong Yu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chunfeng Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
| | - Shu Jeffrey Zhu
- Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, China
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Central Laboratory of Medicine, Shaoxing People’s Hospital, Shaoxing, China
| | - Feng Guo
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
578
|
Itoyama S, Noda E, Takamatsu S, Kondo J, Kawaguchi R, Shimosaka M, Fukuoka T, Motooka D, Nakamura S, Tanemura M, Mitsufuji S, Iwagami Y, Akita H, Tobe T, Kamada Y, Eguchi H, Miyoshi E. Enterococcus spp. have higher fitness for survival, in a pH-dependent manner, in pancreatic juice among duodenal bacterial flora. JGH Open 2022; 6:85-90. [PMID: 35071793 PMCID: PMC8762619 DOI: 10.1002/jgh3.12703] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 12/15/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND AIM Bacterial infection is involved in the progression of many gastrointestinal diseases, including those of pancreas; however, how and which bacteria colonize in pancreatic juice and tissue have yet to be elucidated. Recently, we reported that Enterococcus faecalis exists in the pancreatic juice and tissues of patients with chronic pancreatic disease. Here, we investigated the survival of E. faecalis in duodenal juice with different pH conditions. METHODS Pancreatic juice samples from 62 patients with cancers of the duodeno-pancreato-biliary region were evaluated for the presence of E. faecalis. 16S ribosomal RNA polymerase chain reaction and 16S-based metagenome analyses were performed to determine the bacterial composition. The survival of E. faecalis in various pancreatic juice conditions was evaluated. RESULTS Of 62 samples, 27% (17/62) were positive for Enterococcus spp., among which 71% (12/17) contained E. faecalis. Enterococcus spp. showed the highest fitness for survival in alkaline pancreatic juice among various bacterial species. The microbiome of pancreatic juice from patients with pancreatic and bile duct cancer showed diversity, but Enterococcus spp. were enriched among duodenal tumors and intraductal papillary mucinous neoplasms. CONCLUSIONS Alkalinity is one of the important factors for the selective survival of E. faecalis among microbiota. E. faecalis can colonize the pancreatic duct when the pancreatic juice condition is altered.
Collapse
Affiliation(s)
- Saki Itoyama
- Department of Molecular Biochemistry and Clinical InvestigationOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Emika Noda
- Department of Molecular Biochemistry and Clinical InvestigationOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Shinji Takamatsu
- Department of Molecular Biochemistry and Clinical InvestigationOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Jumpei Kondo
- Department of Molecular Biochemistry and Clinical InvestigationOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Rui Kawaguchi
- Department of Molecular Biochemistry and Clinical InvestigationOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Munefumi Shimosaka
- Department of Molecular Biochemistry and Clinical InvestigationOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Tomoya Fukuoka
- Department of Molecular Biochemistry and Clinical InvestigationOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Daisuke Motooka
- Research Institute for Microbial DiseasesOsaka UniversitySuitaOsakaJapan
| | - Shota Nakamura
- Research Institute for Microbial DiseasesOsaka UniversitySuitaOsakaJapan
| | - Masahiro Tanemura
- Department of SurgeryRinku General Medical CenterIzumisanoOsakaJapan
| | - Suguru Mitsufuji
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Yoshifumi Iwagami
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Hirofumi Akita
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Toru Tobe
- Laboratory of Molecular Medical MicrobiologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Yoshihiro Kamada
- Department of Advanced Metabolic HepatologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Hidetoshi Eguchi
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry and Clinical InvestigationOsaka University Graduate School of MedicineSuitaOsakaJapan
| |
Collapse
|
579
|
Sharma V, Malla MA, Kori RK, Yadav RS, Azam Z. Applications of Metagenomics for Unrevealing the Extended Horizons of Microbiota Prevalence from Soil to Human Health. Open Microbiol J 2021. [DOI: 10.2174/1874285802115010177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Phylogenetic analysis of different ecosystems has shown that the number of microbial communities in a single sample exceeds their cultured counterparts. Microbes have been found throughout nature and can thrive in adverse conditions. Besides inhabiting diverse environments, they also play a key role in the maintenance of the ecosystem. Most of these microbes are either unculturable or difficult to culture with conventional culturing methods. Metagenomics is an emerging field of science that has been in the light for a decade and offers a potential way to assess microbial diversity. The development of metagenomics opens new ways to study genetic material directly from the environmental samples. DNA sequencing and synthesis technologies are making it possible to read and write entire genomes. The huge amount of data obtained from genome sequencing inevitably requires bioinformatics tools to handle and further process them for analysis. Advances in DNA sequencing and high-performance computing have brought about exemplar improvement in metagenomics, allowing in-depth study of the largely unexplored frontier of microbial life. This culture-independent method provides extensive information regarding the structure, composition, and function of the diverse assemblages of the environmental microbes. The current review presents an overview of the technical aspects of metagenomics along with its diverse applications.
Collapse
|
580
|
Schepis T, De Lucia SS, Nista EC, Manilla V, Pignataro G, Ojetti V, Piccioni A, Gasbarrini A, Franceschi F, Candelli M. Microbiota in Pancreatic Diseases: A Review of the Literature. J Clin Med 2021; 10:5920. [PMID: 34945216 PMCID: PMC8704740 DOI: 10.3390/jcm10245920] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 02/05/2023] Open
Abstract
The gut microbiota is a critical element in the balance between human health and disease. Its impairment, defined as dysbiosis, is associated with gastroenterological and systemic diseases. Pancreatic secretions are involved in the composition and changes of the gut microbiota, and the gut microbiota may colonize the pancreatic parenchyma and be associated with the occurrence of diseases. The gut microbiota and the pancreas influence each other, resulting in a "gut microbiota-pancreas axis". Moreover, the gut microbiota may be involved in pancreatic diseases, both through direct bacterial colonization and an indirect effect of small molecules and toxins derived from dysbiosis. Pancreatic diseases such as acute pancreatitis, chronic pancreatitis, autoimmune pancreatitis, and pancreatic cancer are common gastroenterological diseases associated with high morbidity and mortality. The involvement of the microbiota in pancreatic diseases is increasingly recognized. Therefore, modifying the intestinal bacterial flora could have important therapeutic implications on these pathologies. The aim of this study is to review the literature to evaluate the alterations of the gut microbiota in pancreatic diseases, and the role of the microbiota in the treatment of these diseases.
Collapse
Affiliation(s)
- Tommaso Schepis
- Medical and Surgical Science Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy; (T.S.); (S.S.D.L.); (E.C.N.); (V.M.); (A.G.)
| | - Sara S. De Lucia
- Medical and Surgical Science Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy; (T.S.); (S.S.D.L.); (E.C.N.); (V.M.); (A.G.)
| | - Enrico C. Nista
- Medical and Surgical Science Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy; (T.S.); (S.S.D.L.); (E.C.N.); (V.M.); (A.G.)
| | - Vittoria Manilla
- Medical and Surgical Science Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy; (T.S.); (S.S.D.L.); (E.C.N.); (V.M.); (A.G.)
| | - Giulia Pignataro
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy; (G.P.); (V.O.); (A.P.); (F.F.)
| | - Veronica Ojetti
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy; (G.P.); (V.O.); (A.P.); (F.F.)
| | - Andrea Piccioni
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy; (G.P.); (V.O.); (A.P.); (F.F.)
| | - Antonio Gasbarrini
- Medical and Surgical Science Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy; (T.S.); (S.S.D.L.); (E.C.N.); (V.M.); (A.G.)
| | - Francesco Franceschi
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy; (G.P.); (V.O.); (A.P.); (F.F.)
| | - Marcello Candelli
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy; (G.P.); (V.O.); (A.P.); (F.F.)
| |
Collapse
|
581
|
Xu F, Yang C, Tang M, Wang M, Cheng Z, Chen D, Chen X, Liu K. The Role of Gut Microbiota and Genetic Susceptibility in the Pathogenesis of Pancreatitis. Gut Liver 2021; 16:686-696. [PMID: 34911043 PMCID: PMC9474482 DOI: 10.5009/gnl210362] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/06/2021] [Accepted: 09/17/2021] [Indexed: 11/22/2022] Open
Abstract
Pancreatitis is one of the most common inflammatory diseases of the pancreas caused by autodigestion induced by excessive premature protease activation. However, recognition of novel pathophysiological mechanisms remains a still challenge. Both genetic and environmental factors contribute to the pathogenesis of pancreatitis, and the gut microbiota is a potential source of an environmental effect. In recent years, several new frontiers in gut microbiota and genetic risk assessment research have emerged and improved the understanding of the disease. These investigations showed that the disease progression of pancreatitis could be regulated by the gut microbiome, either through a translocation influence or in a host immune response manner. Meanwhile, the onset of the disease is also associated with the heritage of a pathogenic mutation, and the disease progression could be modified by genetic risk factors. In this review, we focused on the recent advances in the role of gut microbiota in the pathogenesis of pancreatitis, and the genetic susceptibility in pancreatitis.
Collapse
Affiliation(s)
- Fumin Xu
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Chunmei Yang
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Mingcheng Tang
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Ming Wang
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhenhao Cheng
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Dongfeng Chen
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiao Chen
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Kaijun Liu
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
582
|
Sędzikowska A, Szablewski L. Human Gut Microbiota in Health and Selected Cancers. Int J Mol Sci 2021; 22:13440. [PMID: 34948234 PMCID: PMC8708499 DOI: 10.3390/ijms222413440] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 12/24/2022] Open
Abstract
The majority of the epithelial surfaces of our body, and the digestive tract, respiratory and urogenital systems, are colonized by a vast number of bacteria, archaea, fungi, protozoans, and viruses. These microbiota, particularly those of the intestines, play an important, beneficial role in digestion, metabolism, and the synthesis of vitamins. Their metabolites stimulate cytokine production by the human host, which are used against potential pathogens. The composition of the microbiota is influenced by several internal and external factors, including diet, age, disease, and lifestyle. Such changes, called dysbiosis, may be involved in the development of various conditions, such as metabolic diseases, including metabolic syndrome, type 2 diabetes mellitus, Hashimoto's thyroidis and Graves' disease; they can also play a role in nervous system disturbances, such as multiple sclerosis, Alzheimer's disease, Parkinson's disease, and depression. An association has also been found between gut microbiota dysbiosis and cancer. Our health is closely associated with the state of our microbiota, and their homeostasis. The aim of this review is to describe the associations between human gut microbiota and cancer, and examine the potential role of gut microbiota in anticancer therapy.
Collapse
Affiliation(s)
| | - Leszek Szablewski
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, ul. Chalubinskiego 5, 02-004 Warsaw, Poland;
| |
Collapse
|
583
|
Wang Y, Du J, Wu X, Abdelrehem A, Ren Y, Liu C, Zhou X, Wang S. Crosstalk between autophagy and microbiota in cancer progression. Mol Cancer 2021; 20:163. [PMID: 34895252 PMCID: PMC8665582 DOI: 10.1186/s12943-021-01461-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/16/2021] [Indexed: 12/18/2022] Open
Abstract
Autophagy is a highly conserved catabolic process seen in eukaryotes and is essentially a lysosome-dependent protein degradation pathway. The dysregulation of autophagy is often associated with the pathogenesis of numerous types of cancers, and can not only promote the survival of cancer but also trigger the tumor cell death. During cancer development, the microbial community might predispose cells to tumorigenesis by promoting mucosal inflammation, causing systemic disorders, and may also regulate the immune response to cancer. The complex relationship between autophagy and microorganisms can protect the body by activating the immune system. In addition, autophagy and microorganisms can crosstalk with each other in multifaceted ways to influence various physiological and pathological responses involved in cancer progression. Various molecular mechanisms, correlating the microbiota disorders and autophagy activation, control the outcomes of protumor or antitumor responses, which depend on the cancer type, tumor microenvironment and disease stage. In this review, we mainly emphasize the leading role of autophagy during the interaction between pathogenic microorganisms and human cancers and investigate the various molecular mechanisms by which autophagy modulates such complicated biological processes. Moreover, we also highlight the possibility of curing cancers with multiple molecular agents targeting the microbiota/autophagy axis. Finally, we summarize the emerging clinical trials investigating the therapeutic potential of targeting either autophagy or microbiota as anticancer strategies, although the crosstalk between them has not been explored thoroughly.
Collapse
Affiliation(s)
- Yu Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, Tianjin, 300060 China
- National Clinical Research Center of Cancer, Tianjin, 300060 China
| | - Jiang Du
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, Tianjin, 300060 China
- National Clinical Research Center of Cancer, Tianjin, 300060 China
| | - Xuemei Wu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Gastroenterology and Hepatology Institute, Tianjin Medical University, Tianjin, 300052 China
- Key Laboratory of Immune Microenvironment and Disease, Tianjin Medical University, Ministry of Education, Tianjin, 300070 China
| | - Ahmed Abdelrehem
- Department of Craniomaxillofacial and Plastic Surgery, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| | - Yu Ren
- Tianjin Research Center of Basic Medical Science, Tianjin Medical University, Tianjin, 300070 China
| | - Chao Liu
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, Tianjin, 300060 China
- National Clinical Research Center of Cancer, Tianjin, 300060 China
| | - Xuan Zhou
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, Tianjin, 300060 China
- National Clinical Research Center of Cancer, Tianjin, 300060 China
| | - Sinan Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Gastroenterology and Hepatology Institute, Tianjin Medical University, Tianjin, 300052 China
- Key Laboratory of Immune Microenvironment and Disease, Tianjin Medical University, Ministry of Education, Tianjin, 300070 China
| |
Collapse
|
584
|
Chen Y, Wang C, Song J, Xu R, Ruze R, Zhao Y. S100A2 Is a Prognostic Biomarker Involved in Immune Infiltration and Predict Immunotherapy Response in Pancreatic Cancer. Front Immunol 2021; 12:758004. [PMID: 34887861 PMCID: PMC8650155 DOI: 10.3389/fimmu.2021.758004] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/02/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer (PC) is a highly fatal and aggressive disease with its incidence and mortality quite discouraging. It is of great significance to construct an effective prognostic signature of PC and find the novel biomarker for the optimization of the clinical decision-making. Due to the crucial role of immunity in tumor development, a prognostic model based on nine immune-related genes was constructed, which was proved to be effective in The Cancer Genome Atlas (TCGA) training set, TCGA testing set, TCGA entire set, GSE78229 set, and GSE62452 set. Furthermore, S100A2 (S100 Calcium Binding Protein A2) was identified as the gene occupying the most paramount position in risk model. Gene set enrichment analysis (GSEA), ESTIMATE and CIBERSORT algorithm revealed that S100A2 was closely associated with the immune status in PC microenvironment, mainly related to lower proportion of CD8+T cells and activated NK cells and higher proportion of M0 macrophages. Meanwhile, patients with high S100A2 expression might get more benefit from immunotherapy according to immunophenoscore algorithm. Afterwards, our independent cohort was also used to demonstrate S100A2 was an unfavorable marker of PC, as well as its remarkably positive correlation with the expression of PD-L1. In conclusion, our results demonstrate S100A2 might be responsible for the preservation of immune-suppressive status in PC microenvironment, which was identified with significant potentiality in predicting prognosis and immunotherapy response in PC patients.
Collapse
Affiliation(s)
- Yuan Chen
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Chengcheng Wang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jianlu Song
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Ruiyuan Xu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Rexiati Ruze
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yupei Zhao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
585
|
Pancreatic Cancer and Gut Microbiome-Related Aspects: A Comprehensive Review and Dietary Recommendations. Nutrients 2021; 13:nu13124425. [PMID: 34959977 PMCID: PMC8709322 DOI: 10.3390/nu13124425] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/04/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota plays a significant role in the human body providing many beneficial effects on the host. However, its dysbiotic alterations may affect the tumorigenic pathway and then trigger the development of pancreatic cancer. This dysbiosis can also modulate the aggressiveness of the tumor, influencing the microenvironment. Because pancreatic cancer is still one of the most lethal cancers worldwide with surgery as the only method that influences prognosis and has curative potential, there is a need to search for other strategies which will enhance the efficiency of standard therapy and improve patients' quality of life. The administration of prebiotics, probiotics, next-generation probiotics (Faecalibacterium prausnitzii, Akkermansia muciniphila), synbiotics, postbiotics, and fecal microbiota transplantation through multiple mechanisms affects the composition of the gut microbiota and may restore its balance. Despite limited data, some studies indicate that the aforementioned methods may allow to achieve better effect of pancreatic cancer treatment and improve therapeutic strategies for pancreatic cancer patients.
Collapse
|
586
|
Opitz FV, Haeberle L, Daum A, Esposito I. Tumor Microenvironment in Pancreatic Intraepithelial Neoplasia. Cancers (Basel) 2021; 13:cancers13246188. [PMID: 34944807 PMCID: PMC8699458 DOI: 10.3390/cancers13246188] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) is a very aggressive neoplasm with a poor survival rate. This is mainly due to late detection, which substantially limits therapy options. A better understanding of the early phases of pancreatic carcinogenesis is fundamental for improving patient prognosis in the future. In this article, we focused on the tumor microenvironment (TME), which provides the biological niche for the development of PDAC from its most common precursor lesions, PanIN (pancreatic intraepithelial neoplasias). Abstract Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive tumors with a poor prognosis. A characteristic of PDAC is the formation of an immunosuppressive tumor microenvironment (TME) that facilitates bypassing of the immune surveillance. The TME consists of a desmoplastic stroma, largely composed of cancer-associated fibroblasts (CAFs), immunosuppressive immune cells, immunoregulatory soluble factors, neural network cells, and endothelial cells with complex interactions. PDAC develops from various precursor lesions such as pancreatic intraepithelial neoplasia (PanIN), intraductal papillary mucinous neoplasms (IPMN), mucinous cystic neoplasms (MCN), and possibly, atypical flat lesions (AFL). In this review, we focus on the composition of the TME in PanINs to reveal detailed insights into the complex restructuring of the TME at early time points in PDAC progression and to explore ways of modifying the TME to slow or even halt tumor progression.
Collapse
|
587
|
Huang L, Liu B, Liu Z, Feng W, Liu M, Wang Y, Peng D, Fu X, Zhu H, Cui Z, Xie L, Ma Y. Gut Microbiota Exceeds Cervical Microbiota for Early Diagnosis of Endometriosis. Front Cell Infect Microbiol 2021; 11:788836. [PMID: 34950610 PMCID: PMC8688745 DOI: 10.3389/fcimb.2021.788836] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
The diagnosis of endometriosis is typically delayed by years for the unexclusive symptom and the traumatic diagnostic method. Several studies have demonstrated that gut microbiota and cervical mucus potentially can be used as auxiliary diagnostic biomarkers. However, none of the previous studies has compared the robustness of endometriosis classifiers based on microbiota of different body sites or demonstrated the correlation among microbiota of gut, cervical mucus, and peritoneal fluid of endometriosis, searching for alternative diagnostic approaches. Herein, we enrolled 41 women (control, n = 20; endometriosis, n = 21) and collected 122 well-matched samples, derived from feces, cervical mucus, and peritoneal fluid, to explore the nature of microbiome of endometriosis patients. Our results indicated that microbial composition is remarkably distinguished between three body sites, with 19 overlapped taxa. Moreover, endometriosis patients harbor distinct microbial communities versus control group especially in feces and peritoneal fluid, with increased abundance of pathogens in peritoneal fluid and depletion of protective microbes in feces. Particularly, genera of Ruminococcus and Pseudomonas were identified as potential biomarkers in gut and peritoneal fluid, respectively. Furthermore, novel endometriosis classifiers were constructed based on taxa selected by a robust machine learning method. These results demonstrated that gut microbiota exceeds cervical microbiota in diagnosing endometriosis. Collectively, this study reveals important insights into the microbial profiling in different body sites of endometriosis, which warrant future exploration into the role of microbiota in endometriosis and highlighted values on gut microbiota in early diagnosis of endometriosis.
Collapse
Affiliation(s)
- Liujing Huang
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Bingdong Liu
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zhihong Liu
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Wanqin Feng
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Minjuan Liu
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yifeng Wang
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Dongxian Peng
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiafei Fu
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Honglei Zhu
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zongbin Cui
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Liwei Xie
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
- College of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Ying Ma
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
588
|
Ibragimova S, Ramachandran R, Ali FR, Lipovich L, Ho SB. Dietary Patterns and Associated Microbiome Changes that Promote Oncogenesis. Front Cell Dev Biol 2021; 9:725821. [PMID: 34869313 PMCID: PMC8633417 DOI: 10.3389/fcell.2021.725821] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022] Open
Abstract
The recent increases in cancer incidences have been linked to lifestyle changes that result in obesity and metabolic syndrome. It is now evident that these trends are associated with the profound changes that occur in the intestinal microbiome, producing altered microbial population signatures that interact, directly or indirectly, with potentially pro-carcinogenic molecular pathways of transcription, proliferation, and inflammation. The effects of the entire gut microbial population on overall health are complex, but individual bacteria are known to play important and definable roles. Recent detailed examinations of a large number of subjects show a tight correlation between habitual diets, fecal microbiome signatures, and markers of metabolic health. Diets that score higher in healthfulness or diversity such as plant-based diets, have altered ratios of specific bacteria, including an increase in short-chain fatty acid producers, which in turn have been linked to improved metabolic markers and lowered cancer risk. Contrarily, numerous studies have implicated less healthy, lower-scoring diets such as the Western diet with reduced intestinal epithelial defenses and promotion of specific bacteria that affect carcinogenic pathways. In this review, we will describe how different dietary patterns affect microbial populations in the gut and illustrate the subsequent impact of bacterial products and metabolites on molecular pathways of cancer development, both locally in the gut and systemically in distant organs.
Collapse
Affiliation(s)
- Shakhzada Ibragimova
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, UAE
| | - Revathy Ramachandran
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, UAE
| | - Fahad R Ali
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, UAE
| | - Leonard Lipovich
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, UAE
| | - Samuel B Ho
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, UAE.,Department of Medicine, Mediclinic City Hospital, Dubai Healthcare City, Dubai, UAE
| |
Collapse
|
589
|
Chavkin TA, Pham LD, Kostic A. E. coli Nissle 1917 modulates host glucose metabolism without directly acting on glucose. Sci Rep 2021; 11:23230. [PMID: 34853343 PMCID: PMC8636602 DOI: 10.1038/s41598-021-02431-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/08/2021] [Indexed: 01/07/2023] Open
Abstract
Managing postprandial glycemic response, or the increase in blood sugar following a meal, is a crucial component to maintaining healthy blood sugar in patients with diabetes. To test whether oral probiotics can impact postprandial glycemic response, E. coli Nissle 1917 (EcN) was evaluated in an oral glucose tolerance test. Oral gavage of EcN concurrent with a glucose bolus reduced the post-gavage glycemic response in mice. However, there was no difference in glycemic response when comparing EcN to a mutant deficient in glucose metabolism. This suggests that while EcN can alter glycemic response to a glucose bolus, this effect is not mediated by direct uptake of glucose. Of the possible indirect effects EcN could have, gastric emptying rate was highlighted as a likely cause, but EcN had no effect on gastric emptying rate in mice. This leaves many more possible indirect explanations for the interaction between EcN and host glucose metabolism to be explored in future work.
Collapse
Affiliation(s)
- Theodore A Chavkin
- Section on Pathophysiology and Molecular Pharmacology, Joslin Diabetes Center, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Loc-Duyen Pham
- Section on Pathophysiology and Molecular Pharmacology, Joslin Diabetes Center, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Aleksandar Kostic
- Section on Pathophysiology and Molecular Pharmacology, Joslin Diabetes Center, Boston, MA, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
590
|
Hester R, Mazur PK, McAllister F. Immunotherapy in Pancreatic Adenocarcinoma: Beyond "Copy/Paste". Clin Cancer Res 2021; 27:6287-6297. [PMID: 34193514 PMCID: PMC8639640 DOI: 10.1158/1078-0432.ccr-18-0900] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/26/2021] [Accepted: 06/29/2021] [Indexed: 01/07/2023]
Abstract
Immunotherapy has dramatically changed the cancer treatment landscape during the past decade, but very limited efficacy has been reported against pancreatic cancer. Several factors unique to pancreatic cancer may explain the resistance: the well-recognized suppressive elements in the tumor microenvironment, the functional and structural barrier imposed by the stroma components, T-cell exhaustion, the choice of perhaps the wrong immune targets, and microbial factors including gut dysbiosis and the unexpected presence of tumor microbes. Furthermore, we discuss various strategies to overcome these barriers.
Collapse
Affiliation(s)
- Robert Hester
- Division of Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pawel K. Mazur
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Corresponding Author: Florencia McAllister, Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, 1155 Pressler Street, CPB6.3500, Houston, TX 77030. E-mail:
| |
Collapse
|
591
|
The intratumoral microbiome: Characterization methods and functional impact. Cancer Lett 2021; 522:63-79. [PMID: 34517085 DOI: 10.1016/j.canlet.2021.09.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/01/2021] [Accepted: 09/06/2021] [Indexed: 12/24/2022]
Abstract
Live-pathogenic bacteria, which were identified inside tumors hundreds year ago, are key elements in modern cancer research. As they have a relatively accessible genome, they offer a multitude of metabolic engineering opportunities, useful in several clinical fields. Better understanding of the tumor microenvironment and its associated microbiome would help conceptualize new metabolically engineered species, triggering efficient therapeutic responses against cancer. Unfortunately, given the low microbial biomass nature of tumors, characterizing the tumor microbiome remains a challenge. Tumors have a high host versus bacterial DNA ratio, making it extremely complex to identify tumor-associated bacteria. Nevertheless, with the improvements in next-generation analytic tools, recent studies demonstrated the existence of intratumor bacteria inside defined tumors. It is now proven that each cancer subtype has a unique microbiome, characterized by bacterial communities with specific metabolic functions. This review provides a brief overview of the main approaches used to characterize the tumor microbiome, and of the recently proposed functions of intracellular bacteria identified in oncological entities. The therapeutic aspects of live-pathogenic microbes are also discussed, regarding the tumor microenvironment of each cancer type.
Collapse
|
592
|
Zhang J, Dai Z, Yan C, Zhang W, Wang D, Tang D. A new biological triangle in cancer: intestinal microbiota, immune checkpoint inhibitors and antibiotics. Clin Transl Oncol 2021; 23:2415-2430. [PMID: 34125407 PMCID: PMC8557192 DOI: 10.1007/s12094-021-02659-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023]
Abstract
Cancer immunotherapy has revolutionized the treatment of many malignant tumors. Although immune checkpoint inhibitors (ICIs) can reactivate the anti-tumor activity of immune cells, sensitivity to immune checkpoint inhibitor therapy depends on the complex tumor immune processes. In recent years, numerous researches have demonstrated the role of intestinal microbiota in immunity and metabolism of the tumor microenvironment, as well as the efficacy of immunotherapy. Epidemiological studies have further demonstrated the efficacy of antibiotic therapy on the probability of patients' response to ICIs and predictability of the short-term survival of cancer patients. Disturbance to the intestinal microbiota significantly affects ICIs-mediated immune reconstitution and is considered a possible mechanism underlying the development of adverse effects during antibiotic-based ICIs treatment. Intestinal microbiota, antibiotics, and ICIs have gradually become important considerations for the titer of immunotherapy. In the case of immunotherapy, the rational use of antibiotics and intestinal microbiota is expected to yield a better prognosis for patients with malignant tumors.
Collapse
Affiliation(s)
- Jie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Zhujiang Dai
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Cheng Yan
- Dalian Medical University, Dalian, China
| | - Wenjie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu Province Hospital, Yangzhou University, Yangzhou, 225001, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu Province Hospital, Yangzhou University, Yangzhou, 225001, China.
| |
Collapse
|
593
|
Current status of intratumour microbiome in cancer and engineered exogenous microbiota as a promising therapeutic strategy. Biomed Pharmacother 2021; 145:112443. [PMID: 34847476 DOI: 10.1016/j.biopha.2021.112443] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 02/06/2023] Open
Abstract
Research on the relationship between microbiome and cancer has made significant progress in the past few decades. It is now known that the gut microbiome has multiple effects on tumour biology. However, the relationship between intratumoral bacteria and cancers remains unclear. Growing evidence suggests that intratumoral bacteria are important components of the microenvironment in several types of cancers. Furthermore, several studies have demonstrated that intratumoral bacteria may directly influence tumorigenesis, progression and responses to treatment. Limited studies have been conducted on intratumoral bacteria, and using intratumoral bacteria to treat tumours remains a challenge. Bacteria have been studied as anticancer therapeutics since the 19th century when William B. Coley successfully treated patients with inoperable sarcomas using Streptococcus pyogenes. With the development of synthetic biological approaches, several bacterial species have been genetically engineered to increase their applicability for cancer treatment. Genetically engineered bacteria for cancer therapy have unique properties compared to other treatment methods. They can specifically accumulate within tumours and inhibit cancer growth. In addition, genetically engineered bacteria may be used as a vector to deliver antitumour agents or combined with radiation and chemotherapy to synergise the effectiveness of cancer treatment. However, various problems in treating tumours with genetically engineered bacteria need to be addressed. In this review, we focus on the role of intratumoral bacteria on tumour initiation, progression and responses to chemotherapy or immunotherapy. Moreover, we summarised the recent progress in the treatment of tumours with genetically engineered bacteria.
Collapse
|
594
|
Huang X, Li M, Hou S, Tian B. Role of the microbiome in systemic therapy for pancreatic ductal adenocarcinoma (Review). Int J Oncol 2021; 59:101. [PMID: 34738624 PMCID: PMC8577795 DOI: 10.3892/ijo.2021.5281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/18/2021] [Indexed: 02/05/2023] Open
Abstract
A large body of evidence has revealed that the microbiome serves a role in all aspects of cancer, particularly cancer treatment. To date, studies investigating the relationship between the microbiome and systemic therapy for pancreatic ductal adenocarcinoma (PDAC) are lacking. PDAC is a high‑mortality malignancy (5‑year survival rate; <9% for all stages). Systemic therapy is one of the most important treatment choices for all patients; however, resistance or toxicity can affect its efficacy. Studies have supported the hypothesis that the microbiome is closely associated with the response to systemic therapy in PDAC, including the induction of drug resistance, or toxicity and therapy‑related changes in microbiota composition. The present review comprehensively summarized the role of the microbiome in systemic therapy for PDAC and the associated molecular mechanisms in an attempt to provide a novel direction for the improvement of treatment response and proposed potential directions for in‑depth research.
Collapse
Affiliation(s)
| | | | - Shengzhong Hou
- Department of Pancreatic Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Bole Tian
- Department of Pancreatic Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
595
|
Sammallahti H, Kokkola A, Rezasoltani S, Ghanbari R, Asadzadeh Aghdaei H, Knuutila S, Puolakkainen P, Sarhadi VK. Microbiota Alterations and Their Association with Oncogenomic Changes in Pancreatic Cancer Patients. Int J Mol Sci 2021; 22:12978. [PMID: 34884776 PMCID: PMC8658013 DOI: 10.3390/ijms222312978] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) is an aggressive disease with a high mortality and poor prognosis. The human microbiome is a key factor in many malignancies, having the ability to alter host metabolism and immune responses and participate in tumorigenesis. Gut microbes have an influence on physiological functions of the healthy pancreas and are themselves controlled by pancreatic secretions. An altered oral microbiota may colonize the pancreas and cause local inflammation by the action of its metabolites, which may lead to carcinogenesis. The mechanisms behind dysbiosis and PC development are not completely clear. Herein, we review the complex interactions between PC tumorigenesis and the microbiota, and especially the question, whether and how an altered microbiota induces oncogenomic changes, or vice versa, whether cancer mutations have an impact on microbiota composition. In addition, the role of the microbiota in drug efficacy in PC chemo- and immunotherapies is discussed. Possible future scenarios are the intentional manipulation of the gut microbiota in combination with therapy or the utilization of microbial profiles for the noninvasive screening and monitoring of PC.
Collapse
Affiliation(s)
- Heidelinde Sammallahti
- Department of Pathology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
- Department of Surgery, Abdominal Center, Helsinki University Hospital and University of Helsinki, 00290 Helsinki, Finland; (A.K.); (P.P.)
| | - Arto Kokkola
- Department of Surgery, Abdominal Center, Helsinki University Hospital and University of Helsinki, 00290 Helsinki, Finland; (A.K.); (P.P.)
| | - Sama Rezasoltani
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran P.O. Box 1985717411, Iran;
| | - Reza Ghanbari
- Digestive Oncology Research Center, Digestive Diseases Research Institute, Tehran University of Medical Science, Tehran P.O. Box 1411713135, Iran;
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran P.O. Box 1985717411, Iran;
| | - Sakari Knuutila
- Department of Pathology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
| | - Pauli Puolakkainen
- Department of Surgery, Abdominal Center, Helsinki University Hospital and University of Helsinki, 00290 Helsinki, Finland; (A.K.); (P.P.)
| | - Virinder Kaur Sarhadi
- Department of Oral and Maxillofacial Diseases, Helsinki University Hospital and University of Helsinki, 00290 Helsinki, Finland;
| |
Collapse
|
596
|
Daniluk J, Daniluk U, Rogalski P, Dabrowski A, Swidnicka-Siergiejko A. Microbiome-Friend or Foe of Pancreatic Cancer? J Clin Med 2021; 10:5624. [PMID: 34884327 PMCID: PMC8658245 DOI: 10.3390/jcm10235624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/15/2021] [Accepted: 11/27/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma is one of the deadliest human neoplasms. Despite the development of new surgical and adjuvant therapies, the prognosis remains very poor, with the overall survival rate not exceeding 9%. There is now increasing evidence that the human microbiome, which is involved in many physiological functions, including the regulation of metabolic processes and the modulation of the immune system, is possibly linked to pancreatic oncogenesis. However, the exact mechanisms of action are poorly understood. Our review summarizes the current understanding of how the microbiome affects pancreatic cancer development and progression. We discuss potential pathways of microbe translocation to the pancreas, as well as the mechanism of their action. We describe the role of the microbiome as a potential marker of pancreatic cancer diagnosis, progression, and survival. Finally, we discuss the possibilities of modifying the microbiome to improve treatment effectiveness for this deadly disease.
Collapse
Affiliation(s)
- Jaroslaw Daniluk
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland; (P.R.); (A.D.); (A.S.-S.)
| | - Urszula Daniluk
- Department of Pediatrics, Gastroenterology, Hepatology, Nutrition and Allergology, Medical University of Bialystok, 15-274 Bialystok, Poland;
| | - Pawel Rogalski
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland; (P.R.); (A.D.); (A.S.-S.)
| | - Andrzej Dabrowski
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland; (P.R.); (A.D.); (A.S.-S.)
| | - Agnieszka Swidnicka-Siergiejko
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland; (P.R.); (A.D.); (A.S.-S.)
| |
Collapse
|
597
|
Orlacchio A, Mazzone P. The Role of Toll-like Receptors (TLRs) Mediated Inflammation in Pancreatic Cancer Pathophysiology. Int J Mol Sci 2021; 22:12743. [PMID: 34884547 PMCID: PMC8657588 DOI: 10.3390/ijms222312743] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most lethal forms of cancer, characterized by its aggressiveness and metastatic potential. Despite significant improvements in PC treatment and management, the complexity of the molecular pathways underlying its development has severely limited the available therapeutic opportunities. Toll-like receptors (TLRs) play a pivotal role in inflammation and immune response, as they are involved in pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs). Activation of TLRs initiates a signaling cascade, which in turn, leads to the transcription of several genes involved in inflammation and anti-microbial defense. TLRs are also deregulated in several cancers and can be used as prognostic markers and potential targets for cancer-targeted therapy. In this review we discuss the current knowledge about the role of TLRs in PC progression, focusing on the available TLRs-targeting compounds and their possible use in PC therapy.
Collapse
Affiliation(s)
- Arturo Orlacchio
- NYU Grossman School of Medicine, NYU Langone Health, New York, NY 10016, USA
| | - Pellegrino Mazzone
- Biogem Scarl, Istituto di Ricerche Genetiche Gaetano Salvatore, 83031 Ariano Irpino, Italy
| |
Collapse
|
598
|
Zhou H, Yuan Y, Wang H, Xiang W, Li S, Zheng H, Wen Y, Ming Y, Chen L, Zhou J. Gut Microbiota: A Potential Target for Cancer Interventions. Cancer Manag Res 2021; 13:8281-8296. [PMID: 34764691 PMCID: PMC8572730 DOI: 10.2147/cmar.s328249] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota plays a crucial role in many physiological processes in the human body. Dysbiosis can disrupt the intestinal barrier and alter metabolism and immune responses, leading to the development of diseases. Over the past few decades, evidence has accumulated linking changes in the composition of the gut microbiota to dozens of seemingly unrelated conditions, including cancer. Overall, the gut microbiota mainly affects the occurrence and development of cancer by damaging host DNA, forming and maintaining a pro-inflammatory environment, and affecting host immune responses. In addition, the gut microbiota can also affect the efficacy and toxicity of chemotherapy, radiotherapy, and immunotherapy. Scientists attempt to improve the efficacy and decrease the toxicity of these treatment modalities by fine-tuning the gut microbiota. The aim of this review is to assist researchers and clinicians in developing new strategies for the detection and treatment of tumors by providing the latest information on the intestinal microbiome and cancer, as well as exploring potential application prospects and mechanisms of action.
Collapse
Affiliation(s)
- Hu Zhou
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, People's Republic of China
| | - Yuan Yuan
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, People's Republic of China.,Department of Operation and Anaesthesia, Yibin First People's Hospital, Yibin, Sichuan, People's Republic of China
| | - Haorun Wang
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, People's Republic of China
| | - Wei Xiang
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, People's Republic of China
| | - Shenjie Li
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, People's Republic of China
| | - Haowen Zheng
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, People's Republic of China
| | - Yuqi Wen
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, People's Republic of China
| | - Yang Ming
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, People's Republic of China
| | - Ligang Chen
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, People's Republic of China.,Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, People's Republic of China.,Neurological Diseases and Brain Function Laboratory, Luzhou, Sichuan, People's Republic of China
| | - Jie Zhou
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, People's Republic of China.,Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, People's Republic of China.,Neurological Diseases and Brain Function Laboratory, Luzhou, Sichuan, People's Republic of China
| |
Collapse
|
599
|
Liao G, Wu J, Peng X, Li Y, Tang L, Xu X, Deng D, Zhou X. Visualized analysis of trends and hotspots in global oral microbiome research: A bibliometric study. MedComm (Beijing) 2021; 1:351-361. [PMID: 34766127 PMCID: PMC8491219 DOI: 10.1002/mco2.47] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 02/05/2023] Open
Abstract
The oral microbiome contains numerous bacteria, which directly or indirectly participate in various human functions and continuously exchange signals and substances with the human body, significantly affecting human life cycle, health, and disease. This study aimed to conduct bibliometric studies on the scientific outputs of global oral microbiome research by Citespace software. The data were obtained from the Thomson Reuters' Web of Science Core Collection (WoSCC), from the first relevant literature published until December 31st, 2019, and a total of 2225 articles and reviews were identified. The top country and institutions are the United States and Harvard University. Keywords analysis showed that periodontal disease, oral microbes, and dental plaque are research hotspots. The burst word analysis indicates that early childhood caries, squamous cell carcinoma, gut microbiome, Helicobacter pylori, Candida albicans, and dysbiosis are likely to become the research hotspots of the next era. We also recommend the use of knowledge mapping methods to track specific knowledge areas efficiently and objectively regularly, which can accurately identify hotspots and frontiers and provide valuable information for practitioners in the field, including related scientists, students, journals, and editors.
Collapse
Affiliation(s)
- Ga Liao
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology Sichuan University Chengdu China.,Medical Big Data Center Sichuan University Chengdu China.,Department of Information Management Department of Stomatology Informatics, West China Hospital of Stomatology Sichuan University Chengdu China
| | - Jinyun Wu
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology Sichuan University Chengdu China.,Department of Cariology and Endodontics, West China Hospital of Stomatology Sichuan University Chengdu China
| | - Xian Peng
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology Sichuan University Chengdu China
| | - Yuqing Li
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology Sichuan University Chengdu China
| | - Li Tang
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology Sichuan University Chengdu China.,Department of Cariology and Endodontics, West China Hospital of Stomatology Sichuan University Chengdu China
| | - Xin Xu
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology Sichuan University Chengdu China.,Department of Cariology and Endodontics, West China Hospital of Stomatology Sichuan University Chengdu China
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Center for Dentistry Amsterdam (ACTA) University of Amsterdam and VU University Amsterdam Amsterdam Netherlands
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology Sichuan University Chengdu China.,Department of Cariology and Endodontics, West China Hospital of Stomatology Sichuan University Chengdu China
| |
Collapse
|
600
|
Wan X, Song M, Wang A, Zhao Y, Wei Z, Lu Y. Microbiome Crosstalk in Immunotherapy and Antiangiogenesis Therapy. Front Immunol 2021; 12:747914. [PMID: 34745119 PMCID: PMC8566949 DOI: 10.3389/fimmu.2021.747914] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/28/2021] [Indexed: 12/30/2022] Open
Abstract
The human body and its microbiome constitute a highly delicate system. The gut microbiome participates in the absorption of the host's nutrients and metabolism, maintains the microcirculation, and modulates the immune response. Increasing evidence shows that gut microbiome dysbiosis in the body not only affects the occurrence and development of tumors but also tumor prognosis and treatment. Microbiome have been implicated in tumor control in patients undergoing anti- angiogenesis therapy and immunotherapy. In cases with unsatisfactory responses to chemotherapy, radiotherapy, and targeted therapy, appropriate adjustment of microbes abundance is considered to enhance the treatment response. Here, we review the current research progress in cancer immunotherapy and anti- angiogenesis therapy, as well as the unlimited potential of their combination, especially focusing on how the interaction between intestinal microbiota and the immune system affects cancer pathogenesis and treatment. In addition, we discuss the effects of microbiota on anti-cancer immune response and anti- angiogenesis therapy, and the potential value of these interactions in promoting further research in this field.
Collapse
Affiliation(s)
- Xueting Wan
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing, China
| | - Mengyao Song
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing, China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing, China.,Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Yang Zhao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing, China.,Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhonghong Wei
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing, China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing, China.,Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| |
Collapse
|