1
|
Sousa JA, Callejas BE, Wang A, Higgins E, Herik A, Andonian N, Yousuf M, Colarusso P, Raman M, McKay DM. GPx1 deficiency confers increased susceptibility to ferroptosis in macrophages from individuals with active Crohn's disease. Cell Death Dis 2024; 15:903. [PMID: 39695083 DOI: 10.1038/s41419-024-07289-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/28/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024]
Abstract
Intestinal cell death is a defining feature of Crohn's disease (CD), a major form of inflammatory bowel disease. The focus on this aspect of enteric inflammation has mainly been on epithelial cells, while other cell types such as stromal and myeloid cells have received less attention. Hypothesising that decreased macrophage viability in an oxidative environment could be a contributing factor to the pathophysiology of CD, we found that monocyte-derived macrophages from individuals with active CD (but not those in clinical disease remission) have increased sensitivity to cell death induced by H2O2. Molecular biology and pharmacological studies ruled out apoptosis and necroptosis, while increased lipid peroxidation and surface expression of the transferrin receptor implicated ferroptosis as the mechanism of the H2O2-induced cell death: this was supported by suppression of H2O2-cytotoxicity by liproxstatin-1, a pharmacological inhibitor of ferroptosis. Selenoproteins are important antioxidants, and selenium deficiency can be a feature of CD. Despite normal dietary intake of selenium, monocyte-derived macrophages and intestinal macrophages in individuals with CD had decreased protein and/or mRNA expression of the selenoprotein, glutathione peroxidase (GPx)-1. Knockdown of GPx1 in macrophages from healthy volunteers resulted in increased H2O2-induced cell death reminiscent of that observed with macrophages from CD. In summary, monocyte-derived macrophages from individuals with CD have increased susceptibility to H2O2-induced ferroptosis cell death, that may be facilitated, at least in part, by reduced expression of the antioxidant GPx1. We suggest that reduced GPx1 in monocytes recruited to the gut and intestinal macrophages renders these cells vulnerable to reactive oxygen species-evoked ferroptosis cell death and that unraveling the participation of this pathway in Crohn's disease may reveal novel therapeutic approaches to this chronic condition.
Collapse
Affiliation(s)
- James A Sousa
- Gastrointestinal Research Group, Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Live Cell Imaging Laboratory, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Blanca E Callejas
- Gastrointestinal Research Group, Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Arthur Wang
- Gastrointestinal Research Group, Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Eve Higgins
- Gastrointestinal Research Group, Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Aydin Herik
- Gastrointestinal Research Group, Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Natalie Andonian
- Gastrointestinal Research Group, Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Munazza Yousuf
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Pina Colarusso
- Live Cell Imaging Laboratory, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Maitreyi Raman
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Department of Community Health Science, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Derek M McKay
- Gastrointestinal Research Group, Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
2
|
Hong D, Kim HK, Yang W, Yoon C, Kim M, Yang CS, Yoon S. Integrative analysis of single-cell RNA-seq and gut microbiome metabarcoding data elucidates macrophage dysfunction in mice with DSS-induced ulcerative colitis. Commun Biol 2024; 7:731. [PMID: 38879692 PMCID: PMC11180211 DOI: 10.1038/s42003-024-06409-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 06/03/2024] [Indexed: 06/19/2024] Open
Abstract
Ulcerative colitis (UC) is a significant inflammatory bowel disease caused by an abnormal immune response to gut microbes. However, there are still gaps in our understanding of how immune and metabolic changes specifically contribute to this disease. Our research aims to address this gap by examining mouse colons after inducing ulcerative colitis-like symptoms. Employing single-cell RNA-seq and 16 s rRNA amplicon sequencing to analyze distinct cell clusters and microbiomes in the mouse colon at different time points after induction with dextran sodium sulfate. We observe a significant reduction in epithelial populations during acute colitis, indicating tissue damage, with a partial recovery observed in chronic inflammation. Analyses of cell-cell interactions demonstrate shifts in networking patterns among different cell types during disease progression. Notably, macrophage phenotypes exhibit diversity, with a pronounced polarization towards the pro-inflammatory M1 phenotype in chronic conditions, suggesting the role of macrophage heterogeneity in disease severity. Increased expression of Nampt and NOX2 complex subunits in chronic UC macrophages contributes to the inflammatory processes. The chronic UC microbiome exhibits reduced taxonomic diversity compared to healthy conditions and acute UC. The study also highlights the role of T cell differentiation in the context of dysbiosis and its implications in colitis progression, emphasizing the need for targeted interventions to modulate the inflammatory response and immune balance in colitis.
Collapse
Affiliation(s)
- Dawon Hong
- RNA Cell Biology Laboratory, Graduate Department of Bioconvergence Engineering, Dankook University, Yongin, Republic of Korea
| | - Hyo Keun Kim
- Dept of Molecular and Life Science and Center for Bionano Intelligence Education and Research, Hanyang University, Ansan-si, Korea
| | - Wonhee Yang
- Department of AI-based Convergence, Dankook University, Yongin, Republic of Korea
| | - Chanjin Yoon
- Dept of Molecular and Life Science and Institute of Natural Science and Technology, Hanyang University, Ansan-si, Korea
| | - Minsoo Kim
- Department of Computer Science, College of SW Convergence, Dankook University, Yongin, Republic of Korea
| | - Chul-Su Yang
- Dept of Medicinal and Life Science and Center for Bionano Intelligence Education and Research, Hanyang University, Ansan-si, Korea.
| | - Seokhyun Yoon
- Department of Electronics & Electrical Engineering, College of Engineering, Dankook University, Yongin, Republic of Korea.
| |
Collapse
|
3
|
Jones AN, Scheurlen KM, Macleod A, Simon HL, Galandiuk S. Obesity and Inflammatory Factors in the Progression of Early-Onset Colorectal Cancer. Cancers (Basel) 2024; 16:1403. [PMID: 38611081 PMCID: PMC11010915 DOI: 10.3390/cancers16071403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/27/2024] [Accepted: 04/01/2024] [Indexed: 04/14/2024] Open
Abstract
Metabolic dysfunction associated with obesity leads to a chronic pro-inflammatory state with systemic effects, including the alteration of macrophage metabolism. Tumor-associated macrophages have been linked to the formation of cancer through the production of metabolites such as itaconate. Itaconate downregulates peroxisome proliferator-activated receptor gamma as a tumor-suppressing factor and upregulates anti-inflammatory cytokines in M2-like macrophages. Similarly, leptin and adiponectin also influence macrophage cytokine expression and contribute to the progression of colorectal cancer via changes in gene expression within the PI3K/AKT pathway. This pathway influences cell proliferation, differentiation, and tumorigenesis. This work provides a review of obesity-related hormones and inflammatory mechanisms leading to the development and progression of early-onset colorectal cancer (EOCRC). A literature search was performed using the PubMed and Cochrane databases to identify studies related to obesity and EOCRC, with keywords including 'EOCRC', 'obesity', 'obesity-related hormones', 'itaconate', 'adiponectin', 'leptin', 'M2a macrophage', and 'microbiome'. With this concept of pro-inflammatory markers contributing to EOCRC, increased use of chemo-preventative agents such as aspirin may have a protective effect. Elucidating this association between obesity-related, hormone/cytokine-driven inflammatory effects with EOCRC may help lead to new therapeutic targets in preventing and treating EOCRC.
Collapse
Affiliation(s)
- Alexandra N. Jones
- Price Institute of Surgical Research, University of Louisville, Louisville, KY 40202, USA; (A.N.J.); (A.M.); (H.L.S.)
| | - Katharina M. Scheurlen
- Price Institute of Surgical Research, University of Louisville, Louisville, KY 40202, USA; (A.N.J.); (A.M.); (H.L.S.)
| | - Anne Macleod
- Price Institute of Surgical Research, University of Louisville, Louisville, KY 40202, USA; (A.N.J.); (A.M.); (H.L.S.)
| | - Hillary L. Simon
- Price Institute of Surgical Research, University of Louisville, Louisville, KY 40202, USA; (A.N.J.); (A.M.); (H.L.S.)
- Division of Colon and Rectal Surgery, Hiram C. Polk Jr. MD Department of Surgery, University of Louisville, Louisville, KY 40202, USA
| | - Susan Galandiuk
- Price Institute of Surgical Research, University of Louisville, Louisville, KY 40202, USA; (A.N.J.); (A.M.); (H.L.S.)
- Division of Colon and Rectal Surgery, Hiram C. Polk Jr. MD Department of Surgery, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
4
|
Selvakumar B, Sekar P, Samsudin AR. Intestinal macrophages in pathogenesis and treatment of gut leakage: current strategies and future perspectives. J Leukoc Biol 2024; 115:607-619. [PMID: 38198217 DOI: 10.1093/jleuko/qiad165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 11/13/2023] [Accepted: 12/08/2023] [Indexed: 01/12/2024] Open
Abstract
Macrophages play key roles in tissue homeostasis, defense, disease, and repair. Macrophages are highly plastic and exhibit distinct functional phenotypes based on micro-environmental stimuli. In spite of several advancements in understanding macrophage biology and their different functional phenotypes in various physiological and pathological conditions, currently available treatment strategies targeting macrophages are limited. Macrophages' high plasticity and diverse functional roles-including tissue injury and wound healing mechanisms-mark them as potential targets to mine for efficient therapeutics to treat diseases. Despite mounting evidence on association of gut leakage with several extraintestinal diseases, there is no targeted standard therapy to treat gut leakage. Therefore, there is an urgent need to develop therapeutic strategies to treat this condition. Macrophages are the cells that play the largest role in interacting with the gut microbiota in the intestinal compartment and exert their intended functions in injury and repair mechanisms. In this review, we have summarized the current knowledge on the origins and phenotypes of macrophages. The specific role of macrophages in intestinal barrier function, their role in tissue repair mechanisms, and their association with gut microbiota are discussed. In addition, currently available therapies and the putative tissue repair mediators of macrophages for treating microbiota dysbiosis induced gut leakage are also discussed. The overall aim of this review is to convey the intense need to screen for microbiota induced macrophage-released prorepair mediators, which could lead to the identification of potential candidates that could be developed for treating the leaky gut and associated diseases.
Collapse
Affiliation(s)
- Balachandar Selvakumar
- Department of Microbiota, Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - Priyadharshini Sekar
- Department of Microbiota, Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - A Rani Samsudin
- Department of Microbiota, Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| |
Collapse
|
5
|
Zogorean R, Wirtz S. The yin and yang of B cells in a constant state of battle: intestinal inflammation and inflammatory bowel disease. Front Immunol 2023; 14:1260266. [PMID: 37849749 PMCID: PMC10577428 DOI: 10.3389/fimmu.2023.1260266] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/18/2023] [Indexed: 10/19/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease of the gastrointestinal tract, defined by a clinical relapse-remitting course. Affecting people worldwide, the origin of IBD is still undefined, arising as a consequence of the interaction between genes, environment, and microbiota. Although the root cause is difficult to identify, data clearly indicate that dysbiosis and pathogenic microbial taxa are connected with the establishment and clinical course of IBD. The composition of the microbiota is shaped by plasma cell IgA secretion and binding, while cytokines such as IL10 or IFN-γ are important fine-tuners of the immune response in the gastrointestinal environment. B cells may also influence the course of inflammation by promoting either an anti-inflammatory or a pro-inflammatory milieu. Here, we discuss IgA-producing B regulatory cells as an anti-inflammatory factor in intestinal inflammation. Moreover, we specify the context of IgA and IgG as players that can potentially participate in mucosal inflammation. Finally, we discuss the role of B cells in mouse infection models where IL10, IgA, or IgG contribute to the outcome of the infection.
Collapse
Affiliation(s)
- Roxana Zogorean
- Medizinische Klinik 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stefan Wirtz
- Medizinische Klinik 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, Erlangen, Bavaria, Germany
| |
Collapse
|
6
|
Ma C, Zhang Z, Li T, Tao Y, Zhu G, Xu L, Ju Y, Huang X, Zhai J, Wang X. Colonic expression of glutathione S-transferase alpha 4 and 4-hydroxynonenal adducts is correlated with the pathology of murine colitis-associated cancer. Heliyon 2023; 9:e19815. [PMID: 37810110 PMCID: PMC10559223 DOI: 10.1016/j.heliyon.2023.e19815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/30/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023] Open
Abstract
Chronic inflammation-induced oxidative stress is an important driving force for developing colitis-associated cancer (CAC). 4-hydroxynonenal (4-HNE) is a highly reactive aldehyde derived from lipid peroxidation of ω-6 polyunsaturated fatty acids that contributes to colorectal carcinogenesis. Glutathione S-transferase alpha 4 (Gsta4) specifically conjugates glutathione to 4-HNE and thereby detoxifies 4-HNE. The correlation of these oxidative biomarkers with the pathological changes in CAC is, however, unclear. In this study, we investigated the expression of Gsta4 and 4-HNE adducts in azoxymethane/dextran sulfate sodium (AOM/DSS)-induced murine CAC, and analyzed the correlations of 4-HNE and Gsta4 with inflammatory cytokines and the pathological scores in the colon biopsies. Real-time quantitative PCR showed that expression of IL6, TNFα, and Gsta4 sequentially increased in colon tissues for mice treated with DSS for 1, 2, and 3 cycles, respectively. Moreover, immunohistochemical staining showed remarkably increased expression of 4-HNE adducts, Gsta4, TNFα, and IL6 in the colon biopsies after 3 cycles of DSS treatment. Correlation analysis demonstrated that 4-HNE adducts in the colon biopsies were positively correlated with Gsta4 expression. Additionally, the expression of Gsta4 and 4-HNE adducts were strongly correlated with the pathological changes of colon, as well as the expression of TNFα and IL6 in colon tissues. These results provide evidence for the association of oxidative biomarkers Gsta4 and 4-HNE with the pathological changes of CAC and may help developing novel histopathological biomarkers and prevention targets for CAC.
Collapse
Affiliation(s)
- Chunhua Ma
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Zhanhu Zhang
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Tianqi Li
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Yumei Tao
- Department of Pathology, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Guoxiang Zhu
- Department of Pathology, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Lili Xu
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Yuanyuan Ju
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Xu Huang
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Jinyun Zhai
- Department of Medical Experimental Technology, Nantong University Xinglin College, Nantong, China
| | - Xingmin Wang
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| |
Collapse
|
7
|
Camaya I, O’Brien B, Donnelly S. How do parasitic worms prevent diabetes? An exploration of their influence on macrophage and β-cell crosstalk. Front Endocrinol (Lausanne) 2023; 14:1205219. [PMID: 37564976 PMCID: PMC10411736 DOI: 10.3389/fendo.2023.1205219] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/10/2023] [Indexed: 08/12/2023] Open
Abstract
Diabetes is the fastest growing chronic disease globally, with prevalence increasing at a faster rate than heart disease and cancer. While the disease presents clinically as chronic hyperglycaemia, two distinct subtypes have been recognised. Type 1 diabetes (T1D) is characterised as an autoimmune disease in which the insulin-producing pancreatic β-cells are destroyed, and type 2 diabetes (T2D) arises due to metabolic insufficiency, in which inadequate amounts of insulin are produced, and/or the actions of insulin are diminished. It is now apparent that pro-inflammatory responses cause a loss of functional β-cell mass, and this is the common underlying mechanism of both T1D and T2D. Macrophages are the central immune cells in the pathogenesis of both diseases and play a major role in the initiation and perpetuation of the proinflammatory responses that compromise β-cell function. Furthermore, it is the crosstalk between macrophages and β-cells that orchestrates the inflammatory response and ensuing β-cell dysfunction/destruction. Conversely, this crosstalk can induce immune tolerance and preservation of β-cell mass and function. Thus, specifically targeting the intercellular communication between macrophages and β-cells offers a unique strategy to prevent/halt the islet inflammatory events underpinning T1D and T2D. Due to their potent ability to regulate mammalian immune responses, parasitic worms (helminths), and their excretory/secretory products, have been examined for their potential as therapeutic agents for both T1D and T2D. This research has yielded positive results in disease prevention, both clinically and in animal models. However, the focus of research has been on the modulation of immune cells and their effectors. This approach has ignored the direct effects of helminths and their products on β-cells, and the modulation of signal exchange between macrophages and β-cells. This review explores how the alterations to macrophages induced by helminths, and their products, influence the crosstalk with β-cells to promote their function and survival. In addition, the evidence that parasite-derived products interact directly with endocrine cells to influence their communication with macrophages to prevent β-cell death and enhance function is discussed. This new paradigm of two-way metabolic conversations between endocrine cells and macrophages opens new avenues for the treatment of immune-mediated metabolic disease.
Collapse
Affiliation(s)
| | | | - Sheila Donnelly
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| |
Collapse
|
8
|
El-Sayed A, Aleya L, Kamel M. Epigenetics and the role of nutraceuticals in health and disease. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:28480-28505. [PMID: 36694069 DOI: 10.1007/s11356-023-25236-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 01/05/2023] [Indexed: 06/17/2023]
Abstract
In the post-genomic era, the data provided by complete genome sequencing could not answer several fundamental questions about the causes of many noninfectious diseases, diagnostic biomarkers, and novel therapeutic approaches. The rapidly expanding understanding of epigenetic mechanisms, as well as widespread acceptance of their hypothesized role in disease induction, facilitated the development of a number of novel diagnostic markers and therapeutic concepts. Epigenetic aberrations are reversible in nature, which enables the treatment of serious incurable diseases. Therefore, the interest in epigenetic modulatory effects has increased over the last decade, so about 60,000 publications discussing the expression of epigenetics could be detected in the PubMed database. Out of these, 58,442 were published alone in the last 10 years, including 17,672 reviews (69 historical articles), 314 clinical trials, 202 case reports, 197 meta-analyses, 156 letters to the editor, 108 randomized controlled trials, 87 observation studies, 40 book chapters, 22 published lectures, and 2 clinical trial protocols. The remaining publications are either miscellaneous or a mixture of the previously mentioned items. According to the species and gender, the publications included 44,589 human studies (17,106 females, 14,509 males, and the gender is not mentioned in the remaining papers) and 30,253 animal studies. In the present work, the role of epigenetic modulations in health and disease and the influencing factors in epigenetics are discussed.
Collapse
Affiliation(s)
- Amr El-Sayed
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, 25030, Besançon Cedex, France
| | - Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
| |
Collapse
|
9
|
Zheng Z, Hou X, Bian Z, Jia W, Zhao L. Gut microbiota and colorectal cancer metastasis. Cancer Lett 2023; 555:216039. [PMID: 36528182 DOI: 10.1016/j.canlet.2022.216039] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/03/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Gut microbiota play critical roles in the development of colorectal cancer (CRC) metastasis, but the underlying mechanisms remain elusive. This review discusses the molecular mechanisms by which the gut microbiota contribute to a tumor-permissive microenvironment and facilitate malignant transformation and dissemination of tumor cells, thereby mediating CRC metastasis.
Collapse
Affiliation(s)
- Zongmei Zheng
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinxin Hou
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhaoxiang Bian
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Wei Jia
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Ling Zhao
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
10
|
Dougherty MW, Jobin C. Intestinal bacteria and colorectal cancer: etiology and treatment. Gut Microbes 2023; 15:2185028. [PMID: 36927206 PMCID: PMC10026918 DOI: 10.1080/19490976.2023.2185028] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 02/17/2023] [Indexed: 03/18/2023] Open
Abstract
The etiology of colorectal cancer (CRC) is influenced by bacterial communities that colonize the gastrointestinal tract. These microorganisms derive essential nutrients from indigestible dietary or host-derived compounds and activate molecular signaling pathways necessary for normal tissue and immune function. Associative and mechanistic studies have identified bacterial species whose presence may increase CRC risk, including notable examples such as Fusobacterium nucleatum, Enterotoxigenic Bacteroides fragilis, and pks+ E. coli. In recent years this work has expanded in scope to include aspects of host mutational status, intra-tumoral microbial heterogeneity, transient infection, and the cumulative influence of multiple carcinogenic bacteria after sequential or co-colonization. In this review, we will provide an updated overview of how host-bacteria interactions influence CRC development, how this knowledge may be utilized to diagnose or prevent CRC, and how the gut microbiome influences CRC treatment efficacy.
Collapse
Affiliation(s)
- Michael W. Dougherty
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Christian Jobin
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
- Department of Infectious Diseases and Immunology, University of Florida College of Medicine, Gainesville, FL, USA
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
11
|
Feng N, Wang S, Liu C, Xu Z, Song Z, Li K, Yu Z. A network meta-analysis to evaluate the efficacy of traditional Chinese medicine on intestinal flora in patients with gastrointestinal cancer. Front Genet 2022; 13:1069780. [DOI: 10.3389/fgene.2022.1069780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/10/2022] [Indexed: 11/29/2022] Open
Abstract
Background and Purpose: Traditional Chinese medicine (TCM) can regulate intestinal flora so as to affect the occurrence, progression, and prognosis of gastrointestinal cancer. According to clinical studies, TCM oral administration, TCM external treatment, and TCM injections, can adjust intestinal flora disorders in patients with gastrointestinal cancer. This network meta-analysis aims to evaluate the effect of three treatments on the intestinal flora in gastrointestinal cancer patients.Methods: This meta-analysis was registered in PROSPERO (CRD42022332553). Six electronic databases, namely CNKI, Wanfang, CSTJ, PubMed, Cochrane Library, and EMBASE, were searched from their inception to 1 April 2022. We identified randomized controlled trials (RCT) used to compare the efficacy of three TCM treatment methods—oral administration, external therapy and injections—on the intestinal flora in gastrointestinal cancer patients. The main outcome indicators were Bifidobacteria, Lactobacilli, Escherichia coli, and Enterococci. Stata (15.1) and the Cochrane risk of bias assessment tool were employed.Results: We identified 20 eligible RCTs with a total of 1,774 patients. According to network meta-analysis results, TCM injection plus common treatment (CT) or oral administration of TCM plus CT was superior to CT alone for supporting Bifidobacterium. In supporting Lactobacillus, TCM injection plus CT demonstrated more obvious effect relative to oral administration of TCM plus CT; TCM injection plus CT was more effective than CT only; and oral administration of TCM plus CT was superior to CT only.The inhibitory effect of TCM injection plus CT on Escherichia coli was better compared with CT only. In terms of inhibiting Enterococci, oral administration of TCM plus CT was superior to CT only.The difference in efficacy among the above treatments was statistically significant. In the SUCRA probability ranking, TCM injection plus CT had the best ranking curve among the three treatments and was the most effective in supporting Bifidobacteria (Sucra = 90.08%), Lactobacilli (Sucra = 96.4%), and regulating Escherichia coli (Sucra = 86.1%) and Enterococci (Sucra = 87.1%).Conclusion: TCM injections plus CT is the most effective therapy in balancing the intestinal flora of gastrointestinal cancer patients. However, the current results deserve further validation through high-quality research.Systematic Review Registration: http://www.prisma-statement.org/, identifier 10.1136/bmj.n71.
Collapse
|
12
|
Qin D, Ma Y, Wang Y, Hou X, Yu L. Contribution of Lactobacilli on Intestinal Mucosal Barrier and Diseases: Perspectives and Challenges of Lactobacillus casei. LIFE (BASEL, SWITZERLAND) 2022; 12:life12111910. [PMID: 36431045 PMCID: PMC9696601 DOI: 10.3390/life12111910] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022]
Abstract
The intestine barrier, the front line of normal body defense, relies on its structural integrity, microbial composition and barrier immunity. The intestinal mucosal surface is continuously exposed to a complex and dynamic community of microorganisms. Although it occupies a relatively small proportion of the intestinal microbiota, Lactobacilli has been discovered to have a significant impact on the intestine tract in previous studies. It is undeniable that some Lactobacillus strains present probiotic properties through maintaining the micro-ecological balance via different mechanisms, such as mucosal barrier function and barrier immunity, to prevent infection and even to solve some neurology issues by microbiota-gut-brain/liver/lung axis communication. Notably, not only living cells but also Lactobacillus derivatives (postbiotics: soluble secreted products and para-probiotics: cell structural components) may exert antipathogenic effects and beneficial functions for the gut mucosal barrier. However, substantial research on specific effects, safety and action mechanisms in vivo should be done. In clinical application of humans and animals, there are still doubts about the precise evaluation of Lactobacilli's safety, therapeutic effect, dosage and other aspects. Therefore, we provide an overview of central issues on the impacts of Lactobacillus casei (L. casei) and their products on the intestinal mucosal barrier and some diseases and highlight the urgent need for further studies.
Collapse
Affiliation(s)
- Da Qin
- Heilongjiang Provincial Key Laboratory of Environmental Microbiology and Recycling of Argo-Waste in Cold Region, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yixuan Ma
- Heilongjiang Provincial Key Laboratory of Environmental Microbiology and Recycling of Argo-Waste in Cold Region, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yanhong Wang
- Heilongjiang Provincial Key Laboratory of Environmental Microbiology and Recycling of Argo-Waste in Cold Region, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Xilin Hou
- Colleges of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
- Correspondence: (X.H.); (L.Y.); Tel.: +86-4596-819-290 (X.H. & L.Y.); Fax: +86-4596-819-292 (X.H. & L.Y.)
| | - Liyun Yu
- Heilongjiang Provincial Key Laboratory of Environmental Microbiology and Recycling of Argo-Waste in Cold Region, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
- Colleges of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
- Correspondence: (X.H.); (L.Y.); Tel.: +86-4596-819-290 (X.H. & L.Y.); Fax: +86-4596-819-292 (X.H. & L.Y.)
| |
Collapse
|
13
|
Cellular Carcinogenesis: Role of Polarized Macrophages in Cancer Initiation. Cancers (Basel) 2022; 14:cancers14112811. [PMID: 35681791 PMCID: PMC9179569 DOI: 10.3390/cancers14112811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/26/2022] [Accepted: 06/02/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Inflammation is a hallmark of many cancers. Macrophages are key participants in innate immunity and important drivers of inflammation. When chronically polarized beyond normal homeostatic responses to infection, injury, or aging, macrophages can express several pro-carcinogenic phenotypes. In this review, evidence supporting polarized macrophages as endogenous sources of carcinogenesis is discussed. In addition, the depletion or modulation of macrophages by small molecule inhibitors and probiotics are reviewed as emerging strategies in cancer prevention. Abstract Inflammation is an essential hallmark of cancer. Macrophages are key innate immune effector cells in chronic inflammation, parainflammation, and inflammaging. Parainflammation is a form of subclinical inflammation associated with a persistent DNA damage response. Inflammaging represents low-grade inflammation due to the dysregulation of innate and adaptive immune responses that occur with aging. Whether induced by infection, injury, or aging, immune dysregulation and chronic macrophage polarization contributes to cancer initiation through the production of proinflammatory chemokines/cytokines and genotoxins and by modulating immune surveillance. This review presents pre-clinical and clinical evidence for polarized macrophages as endogenous cellular carcinogens in the context of chronic inflammation, parainflammation, and inflammaging. Emerging strategies for cancer prevention, including small molecule inhibitors and probiotic approaches, that target macrophage function and phenotype are also discussed.
Collapse
|
14
|
Amamou A, O’Mahony C, Leboutte M, Savoye G, Ghosh S, Marion-Letellier R. Gut Microbiota, Macrophages and Diet: An Intriguing New Triangle in Intestinal Fibrosis. Microorganisms 2022; 10:490. [PMID: 35336066 PMCID: PMC8952309 DOI: 10.3390/microorganisms10030490] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 02/07/2023] Open
Abstract
Intestinal fibrosis is a common complication in inflammatory bowel disease (IBD) without specific treatment. As macrophages are the key actors in inflammatory responses and the wound healing process, they have been extensively studied in chronic diseases these past decades. By their exceptional ability to integrate diverse stimuli in their surrounding environment, macrophages display a multitude of phenotypes to underpin a broad spectrum of functions, from the initiation to the resolution of inflammation following injury. The hypothesis that distinct macrophage subtypes could be involved in fibrogenesis and wound healing is emerging and could open up new therapeutic perspectives in the treatment of intestinal fibrosis. Gut microbiota and diet are two key factors capable of modifying intestinal macrophage profiles, shaping their specific function. Defects in macrophage polarisation, inadequate dietary habits, and alteration of microbiota composition may contribute to the development of intestinal fibrosis. In this review, we describe the intriguing triangle between intestinal macrophages, diet, and gut microbiota in homeostasis and how the perturbation of this discreet balance may lead to a pro-fibrotic environment and influence fibrogenesis in the gut.
Collapse
Affiliation(s)
- Asma Amamou
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, T12 YT20 Cork, Ireland; (C.O.); (S.G.)
| | - Cian O’Mahony
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, T12 YT20 Cork, Ireland; (C.O.); (S.G.)
| | - Mathilde Leboutte
- INSERM UMR 1073 “Nutrition, Inflammation and Gut-Brain Axis”, Normandy University, 76183 Rouen, France; (M.L.); (R.M.-L.)
| | - Guillaume Savoye
- Department of Gastroenterology, Rouen University Hospital, 76031 Rouen, France;
| | - Subrata Ghosh
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, T12 YT20 Cork, Ireland; (C.O.); (S.G.)
| | - Rachel Marion-Letellier
- INSERM UMR 1073 “Nutrition, Inflammation and Gut-Brain Axis”, Normandy University, 76183 Rouen, France; (M.L.); (R.M.-L.)
| |
Collapse
|
15
|
A bioanalytical screening method for Enterococcus faecalis RNPP-type quorum sensing peptides in murine feces. Bioanalysis 2022; 14:151-167. [PMID: 35014887 DOI: 10.4155/bio-2021-0225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: Bacteria coordinate their behavior as a group via communication with their peers, known as 'quorum sensing'. Enterococcus faecalis employs quorum sensing via RNPP-peptides which were not yet reported to be present in mammalian biofluids. Results: Solid phase extraction of murine feces was performed, followed by ultra high performance liquid chromatography (UHPLC-MS/MS) in multiple reaction monitoring (MRM) mode (in total <90 min/sample) for the nine known RNPP peptides. Limits of detection ranged between 0.045 and 52 nM. Adequate identification criteria allowed detection of RNPP quorum sensing peptides in 2/20 wild-type murine feces samples (i.e., cAM373 and cOB1). Conclusion: A fit-for-purpose UHPLC-MS/MS method detected these RNPP peptides in wild-type murine feces samples.
Collapse
|
16
|
Yang X, Guo Y, Chen C, Shao B, Zhao L, Zhou Q, Liu J, Wang G, Yuan W, Sun Z. Interaction between intestinal microbiota and tumour immunity in the tumour microenvironment. Immunology 2021; 164:476-493. [PMID: 34322877 PMCID: PMC8517597 DOI: 10.1111/imm.13397] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/18/2021] [Accepted: 07/20/2021] [Indexed: 11/27/2022] Open
Abstract
In recent years, an increasing number of studies have reported that intestinal microbiota have an important effect on tumour immunity by affecting the tumour microenvironment (TME). The intestinal microbiota are closely associated with various immune cells, such as T lymphocytes, natural killer cells (NK cells) and macrophages. Some bacteria, such as Akkermansia muciniphila (A. muciniphila) and Lactobacillus reuteri (L. reuteri), have been shown to improve the effect of tumour immunity. Furthermore, microbial imbalance, such as the increased abundance of Fusobacterium nucleatum (F. nucleatum) and Helicobacter hepaticus (H. hepaticus), generally causes tumour formation and progression. In addition, some microbiota also play important roles in tumour immunotherapy, especially PD-L1-related therapies. Therefore, what is the relationship between these processes and how do they affect each other? In this review, we summarize the interactions and corresponding mechanisms among the intestinal microbiota, immune system and TME to facilitate the research and development of new targeted drugs and provide new approaches to tumour therapy.
Collapse
Affiliation(s)
- Xiuxiu Yang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- School of MedicineZhengzhou UniversityZhengzhouChina
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Yaxin Guo
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
- Department of Basic MedicalAcademy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Academy of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Chen Chen
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
- School of Life SciencesZhengzhou UniversityZhengzhouChina
| | - Bo Shao
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Luyang Zhao
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
- Department of Basic MedicalAcademy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Academy of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Quanbo Zhou
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jinbo Liu
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Guixian Wang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Weitang Yuan
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhenqiang Sun
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| |
Collapse
|
17
|
Wang X, Undi RB, Ali N, Huycke MM. It takes a village: microbiota, parainflammation, paligenosis and bystander effects in colorectal cancer initiation. Dis Model Mech 2021; 14:dmm048793. [PMID: 33969420 PMCID: PMC10621663 DOI: 10.1242/dmm.048793] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Sporadic colorectal cancer (CRC) is a leading cause of worldwide cancer mortality. It arises from a complex milieu of host and environmental factors, including genetic and epigenetic changes in colon epithelial cells that undergo mutation, selection, clonal expansion, and transformation. The gut microbiota has recently gained increasing recognition as an additional important factor contributing to CRC. Several gut bacteria are known to initiate CRC in animal models and have been associated with human CRC. In this Review, we discuss the factors that contribute to CRC and the role of the gut microbiota, focusing on a recently described mechanism for cancer initiation, the so-called microbiota-induced bystander effect (MIBE). In this cancer mechanism, microbiota-driven parainflammation is believed to act as a source of endogenous mutation, epigenetic change and induced pluripotency, leading to the cancerous transformation of colon epithelial cells. This theory links the gut microbiota to key risk factors and common histologic features of sporadic CRC. MIBE is analogous to the well-characterized radiation-induced bystander effect. Both phenomena drive DNA damage, chromosomal instability, stress response signaling, altered gene expression, epigenetic modification and cellular proliferation in bystander cells. Myeloid-derived cells are important effectors in both phenomena. A better understanding of the interactions between the gut microbiota and mucosal immune effector cells that generate bystander effects can potentially identify triggers for parainflammation, and gain new insights into CRC prevention.
Collapse
Affiliation(s)
- Xingmin Wang
- Nantong Institute of Genetics and Reproductive Medicine, Nantong Maternity and Child Healthcare Hospital, Nantong University, Nantong, Jiangsu 226018, China
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Ram Babu Undi
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Naushad Ali
- Department of Internal Medicine, Section of Digestive Diseases and Nutrition, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Mark M. Huycke
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
18
|
Li C, Pi G, Li F. The Role of Intestinal Flora in the Regulation of Bone Homeostasis. Front Cell Infect Microbiol 2021; 11:579323. [PMID: 33777828 PMCID: PMC7994858 DOI: 10.3389/fcimb.2021.579323] [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: 07/06/2020] [Accepted: 02/09/2021] [Indexed: 12/16/2022] Open
Abstract
Intestinal flora located within the intestinal tract comprises a large number of cells, which are referred to as the second gene pool of the human body and form a complex symbiotic relationship with the host. The knowledge of the complex interaction between the intestinal flora and various life activities of the host is a novel and rapidly expanding field. Recently, many studies are being conducted on the relationship between the intestinal flora and bone homeostasis and indicate that the intestinal flora can regulate bone homeostasis via the host immune, metabolic, and endocrine systems. What’s more, based on several clinical and preclinical pieces of evidence, changing the composition and function of the host intestinal flora through the application of probiotics, prebiotics, and fecal microbiota transplantation is being considered to be a potential novel target for the regulation of bone homeostasis. Here, we searched relevant literature and reviewed the role of the intestinal flora in the regulation of bone homeostasis and its modulating interventions.
Collapse
Affiliation(s)
- Chengxiang Li
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guofu Pi
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Feng Li
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
19
|
Chattopadhyay I, Dhar R, Pethusamy K, Seethy A, Srivastava T, Sah R, Sharma J, Karmakar S. Exploring the Role of Gut Microbiome in Colon Cancer. Appl Biochem Biotechnol 2021; 193:1780-1799. [PMID: 33492552 DOI: 10.1007/s12010-021-03498-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/07/2021] [Indexed: 02/06/2023]
Abstract
Dysbiosis of the gut microbiome has been associated with the development of colorectal cancer (CRC). Gut microbiota is involved in the metabolic transformations of dietary components into oncometabolites and tumor-suppressive metabolites that in turn affect CRC development. In a healthy colon, the major of microbial metabolism is saccharolytic fermentation pathways. The alpha-bug hypothesis suggested that oncogenic bacteria such as enterotoxigenic Bacteroides fragilis (ETBF) induce the development of CRC through direct interactions with colonic epithelial cells and alterations of microbiota composition at the colorectal site. Escherichia coli, E. faecalis, F. nucleatum, and Streptococcus gallolyticus showed higher abundance whereas Bifidobacterium, Clostridium, Faecalibacterium, and Roseburia showed reduced abundance in CRC patients. The alterations of gut microbiota may be used as potential therapeutic approaches to prevent or treat CRC. Probiotics such as Lactobacillus and Bifidobacterium inhibit the growth of CRC through inhibiting inflammation and angiogenesis and enhancing the function of the intestinal barrier through the secretion of short-chain fatty acids (SCFAs). Crosstalk between lifestyle, host genetics, and gut microbiota is well documented in the prevention and treatment of CRC. Future studies are required to understand the interaction between gut microbiota and host to the influence and prevention of CRC. However, a better understanding of bacterial dysbiosis in the heterogeneity of CRC tumors should also be considered. Metatranscriptomic and metaproteomic studies are considered a powerful omic tool to understand the anti-cancer properties of certain bacterial strains. The clinical benefits of probiotics in the CRC context remain to be determined. Metagenomic approaches along with metabolomics and immunology will open a new avenue for the treatment of CRC shortly. Dietary interventions may be suitable to modulate the growth of beneficial microbiota in the gut.
Collapse
Affiliation(s)
- Indranil Chattopadhyay
- Department of Life Sciences, Central University of Tamil Nadu, Thiruvarur, Tamil Nadu, 610005, India
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Ansari Nagar, New Delhi, India
| | - Karthikeyan Pethusamy
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Ansari Nagar, New Delhi, India
| | - Ashikh Seethy
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Ansari Nagar, New Delhi, India
| | - Tryambak Srivastava
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Ansari Nagar, New Delhi, India
| | - Ramkishor Sah
- Rajendra Prasad Center for Opthalmic Sciences, AIIMS, Ansari Nagar, New Delhi, USA
| | - Jyoti Sharma
- Department of Surgical Oncology, NCI AIIMS, Jhajjar, Haryana, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Ansari Nagar, New Delhi, India.
| |
Collapse
|
20
|
Cao X, Tang L, Zeng Z, Wang B, Zhou Y, Wang Q, Zou P, Li W. Effects of Probiotics BaSC06 on Intestinal Digestion and Absorption, Antioxidant Capacity, Microbiota Composition, and Macrophage Polarization in Pigs for Fattening. Front Vet Sci 2020; 7:570593. [PMID: 33240950 PMCID: PMC7677304 DOI: 10.3389/fvets.2020.570593] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/29/2020] [Indexed: 01/24/2023] Open
Abstract
This study aimed to compare the effects of BaSC06 and antibiotics on growth, digestive functions, antioxidant capacity, macrophage polarization, and intestinal microbiota of pigs for fattening. A total of 117 pigs for fattening with similar weight and genetic basis were divided into 3 groups: Anti group (containing 40 g/t Kitasamycin in the diet), Anti+Ba group (containing 20 g/t Kitasamycin and 0.5 × 108 CFU/kg BaSC06 in the diet) and Ba group (containing 1 × 108 cfu/Kg BaSC06 in the diet without any antibiotics). Each treatment was performed in three replicates with 13 pigs per replicate. Results showed that BaSC06 replacement significantly improved the ADG (P < 0.05), intestinal digestion and absorption function by increasing the activity of intestinal digestive enzymes and the expression of glucose transporters SGLT1 (P < 0.05) and small peptide transporters PEPT1 (P < 0.05). Besides, BaSC06 supplementation enhanced intestinal and body antioxidant capacity by activating the Nrf2/Keap1 antioxidant signaling pathway due to the increased expression of p-Nrf2 (P < 0.05). Notably, BaSC06 alleviated intestinal inflammation by inhibiting the production of pro-inflammatory cytokines, IL-8, IL-6, and MCP1 (P < 0.05), and simultaneously increasing the expression of M1 macrophage marker protein iNOS (P < 0.05) and M2 macrophage marker protein Arg (P < 0.05) in the intestinal mucosa. Moreover, BaSC06 promoted the polarization of macrophages to M2 phenotype by stimulating the STAT3 signaling pathway. It was also noted that BaSC06 improved microbiota composition by enhancing the proportion of Firmicutes, and reducing that of Bacteroidetes and Proteobacteria. Taken together, our results indicate that dietary supplementation of BaSC06 in pigs for fattening improves the growth, mucosal structure, antioxidative capacity, immune functions (including increasing M1 and M2 polarization of macrophage) and composition of intestinal microbiota, which is much better than antibiotics, suggesting that it is an effective alternative to antibiotics in the preparation of pig feed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Weifen Li
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Institute of Feed Science, College of Animal Sciences, Zhejiang University, and Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
| |
Collapse
|
21
|
The Macrophages-Microbiota Interplay in Colorectal Cancer (CRC)-Related Inflammation: Prognostic and Therapeutic Significance. Int J Mol Sci 2020; 21:ijms21186866. [PMID: 32962159 PMCID: PMC7558485 DOI: 10.3390/ijms21186866] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023] Open
Abstract
Tumor-associated macrophages (TAMs) are the main population of myeloid cells infiltrating solid tumors and the pivotal orchestrators of cancer-promoting inflammation. However, due to their exceptional plasticity, macrophages can be also key effector cells and powerful activators of adaptive anti-tumor immunity. This functional heterogeneity is emerging in human tumors, colorectal cancer (CRC) in particular, where the dynamic co-existence of different macrophage subtypes influences tumor development, outcome, and response to therapies. Intestinal macrophages are in close interaction with enteric microbiota, which contributes to carcinogenesis and affects treatment outcomes. This interplay may be particularly relevant in CRC, one of the most prevalent and lethal cancer types in the world. Therefore, both macrophages and intestinal microbiota are considered promising prognostic indicators and valuable targets for new therapeutic approaches. Here, we discuss the current understanding of the molecular circuits underlying the interplay between macrophages and microbiota in CRC development, progression, and response to both conventional therapies and immunotherapies.
Collapse
|
22
|
Wang S, Liu W, Wang J, Bai X. Curculigoside inhibits ferroptosis in ulcerative colitis through the induction of GPX4. Life Sci 2020; 259:118356. [PMID: 32861798 DOI: 10.1016/j.lfs.2020.118356] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/17/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022]
Abstract
Curculigoside (CUR) is natural ingredient from Curculigo orchioides Gaertn with multiple biological activities. However, whether CUR protects from ulcerative colitis (UC) and underlying mechanisms are unclear. Herein, mice challenged with dextran sulfate sodium (DSS) were established and administrated with CUR for 7 days. Then histological pathologies and ferroptosis regulators were determined in vivo. The ferroptotic IEC-6 cells were prepared to investigate the underlying mechanism of CUR. Results showed that CUR inhibited the disease activity index, histological damage and cell death in mice with colitis. We also found that ferroptosis was induced in mice with colitis, as evidenced by iron overload, GSH depletion, ROS and MDA production, accompanied by decreased expression of SOD and GPX4. CUR treatment significantly reversed these alterations of ferroptotic features in DSS-induced mice. Furthermore, similar effects of CUR on ferroptosis were observed in IEC-6 cells under the combined treatment of H2O2 and iron chloride hexahydrate. Interestingly, we found that CUR could increase the selenium sensitivity and promote GPX4 transcription level in IEC-6 cells. Knockdown of GPX4 significantly blocked the protective effects of CUR on cell death, GSH and MDA contents as well as LDH activity in ferroptotic IEC-6 cells. Taken together, these findings suggest that CUR protects against ferroptosis in UC by the induction of GPX4, which presents a potential agent for UC treatment.
Collapse
Affiliation(s)
- Shujun Wang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China.
| | - Wei Liu
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Jin Wang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Xia Bai
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| |
Collapse
|
23
|
Watson KM, Gaulke CA, Tsikitis VL. Understanding the microbiome: a primer on the role of the microbiome in colorectal neoplasia. Ann Gastroenterol 2020; 33:223-236. [PMID: 32382225 PMCID: PMC7196612 DOI: 10.20524/aog.2020.0467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/24/2020] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer is a leading cause of cancer-related death internationally, with mounting evidence pointing to the role of the microbiome in adenoma and cancer development. This article aims to provide clinicians with a foundation for understanding the field of research into the microbiome. We also illustrate the various ways in which the microbiota have been linked to colorectal cancer, with a specific focus on microbiota with identified virulence factors, and also on the ways that byproducts of microbiota metabolism may result in oncogenesis. We also review strategies for manipulating the microbiome for therapeutic effects.
Collapse
Affiliation(s)
- Katherine M. Watson
- Department of Surgery, Oregon Health & Science University, Portland, OR (Katherine M. Watson, Vassiliki Liana Tsikitis)
| | | | - Vassiliki Liana Tsikitis
- Department of Surgery, Oregon Health & Science University, Portland, OR (Katherine M. Watson, Vassiliki Liana Tsikitis)
| |
Collapse
|
24
|
Barrett M, Hand CK, Shanahan F, Murphy T, O'Toole PW. Mutagenesis by Microbe: the Role of the Microbiota in Shaping the Cancer Genome. Trends Cancer 2020; 6:277-287. [PMID: 32209443 DOI: 10.1016/j.trecan.2020.01.019] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 02/06/2023]
Abstract
Cancers arise through the process of somatic evolution fueled by the inception of somatic mutations. We lack a complete understanding of the sources of these somatic mutations. Humans host a vast repertoire of microbes collectively known as the microbiota. The microbiota plays a role in altering the tumor microenvironment and proliferation. In addition, microbes have been shown to elicit DNA damage which provides the driver for somatic mutations. An understanding of microbiota-driven mutational mechanisms would contribute to a more complete understanding of the origins of the cancer genome. Here, we review the modes by which microbes stimulate DNA damage and the effect of these phenomena upon the cancer genomic architecture, specifically in the form of mutational spectra and mutational signatures.
Collapse
Affiliation(s)
- Maurice Barrett
- APC Microbiome Ireland, University College Cork, National University of Ireland, Cork, Ireland; School of Microbiology, University College Cork, National University of Ireland, Cork, Ireland
| | - Collette K Hand
- Department of Pathology, University College Cork, Cork, Ireland
| | - Fergus Shanahan
- APC Microbiome Ireland, University College Cork, National University of Ireland, Cork, Ireland; Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland
| | - Thomas Murphy
- Department of Surgery, Mercy University Hospital, Cork, Ireland
| | - Paul W O'Toole
- APC Microbiome Ireland, University College Cork, National University of Ireland, Cork, Ireland; School of Microbiology, University College Cork, National University of Ireland, Cork, Ireland.
| |
Collapse
|
25
|
Pan HW, Du LT, Li W, Yang YM, Zhang Y, Wang CX. Biodiversity and richness shifts of mucosa-associated gut microbiota with progression of colorectal cancer. Res Microbiol 2020; 171:107-114. [PMID: 31982498 DOI: 10.1016/j.resmic.2020.01.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 01/14/2020] [Accepted: 01/14/2020] [Indexed: 12/19/2022]
Abstract
The host-associated gut microbiota is considered critical for the occurrence and progression of colorectal cancer (CRC); however, systematic evaluations of the changes in the biodiversity and richness of mucosa-associated gut microbiota with the development of CRC have been limited. Twenty-three paired samples from colorectal tumor sites and the surrounding non-tumor tissues were collected from stage I to IV CRC patients. The microbial compositions of the samples were analyzed by Illumina MiSeq sequencing of the V4 region of the 16S rRNA gene. Gut bacterial alterations at the tumor sites and surrounding healthy tissue sites collected from the different stages of CRC patients were analyzed. No significant differences were observed in the overall microbial richness and biodiversity between the CRC tissue and surrounding non-CRC tissue samples, however, composition and community segregation of the gut microbiota with the progression of CRC were observed. A general increasing trend of Bacteroidetes, Firmicutes, and Fusobacteria and decreasing trend of Proteobacteria were observed at the phylum level with the development of CRC. Further analysis revealed that thirty-four taxa differed significantly with the progression of CRC. Conclusively, our findings provide a comprehensive view of the human mucosa-associated gut microbiota, in association with the different stages of CRC.
Collapse
Affiliation(s)
- Hong-Wei Pan
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Lu-Tao Du
- Department of Clinical Laboratory, Second Hospital of Shandong University, Jinan, 250033, Shandong Province, China
| | - Wei Li
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Yong-Mei Yang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Yi Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Chuan-Xin Wang
- Department of Clinical Laboratory, Second Hospital of Shandong University, Jinan, 250033, Shandong Province, China.
| |
Collapse
|
26
|
Macrophage Polarization Induced by Probiotic Bacteria: a Concise Review. Probiotics Antimicrob Proteins 2019; 12:798-808. [DOI: 10.1007/s12602-019-09612-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
27
|
Tarashi S, Siadat SD, Ahmadi Badi S, Zali M, Biassoni R, Ponzoni M, Moshiri A. Gut Bacteria and their Metabolites: Which One Is the Defendant for Colorectal Cancer? Microorganisms 2019; 7:E561. [PMID: 31766208 PMCID: PMC6920974 DOI: 10.3390/microorganisms7110561] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/22/2019] [Accepted: 09/04/2019] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is a worldwide health concern which requires efficient therapeutic strategies. The mechanisms underlying CRC remain an essential subject of investigations in the cancer biology field. The evaluation of human microbiota can be critical in this regard, since the disruption of the normal community of gut bacteria is an important issue in the development of CRC. However, several studies have already evaluated the different aspects of the association between microbiota and CRC. The current study aimed at reviewing and summarizing most of the studies on the modifications of gut bacteria detected in stool and tissue samples of CRC cases. In addition, the importance of metabolites derived from gut bacteria, their relationship with the microbiota, and epigenetic modifications have been evaluated.
Collapse
Affiliation(s)
- Samira Tarashi
- Microbiology Research Center, Pasteur Institute of Iran, 1316943551 Tehran, Iran; (S.T.); (S.D.S.); (S.A.B.)
- Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, 1316943551 Tehran, Iran
| | - Seyed Davar Siadat
- Microbiology Research Center, Pasteur Institute of Iran, 1316943551 Tehran, Iran; (S.T.); (S.D.S.); (S.A.B.)
- Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, 1316943551 Tehran, Iran
| | - Sara Ahmadi Badi
- Microbiology Research Center, Pasteur Institute of Iran, 1316943551 Tehran, Iran; (S.T.); (S.D.S.); (S.A.B.)
- Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, 1316943551 Tehran, Iran
| | - Mohammadreza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, 19857-17411 Tehran, Iran;
| | - Roberto Biassoni
- Laboratory of Molecular Medicine, IRCCS Instituto Giannina Gaslini, 16147 Genova, Italy;
| | - Mirco Ponzoni
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Arfa Moshiri
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, 19857-17411 Tehran, Iran;
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| |
Collapse
|
28
|
Alhinai EA, Walton GE, Commane DM. The Role of the Gut Microbiota in Colorectal Cancer Causation. Int J Mol Sci 2019; 20:ijms20215295. [PMID: 31653078 PMCID: PMC6862640 DOI: 10.3390/ijms20215295] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/18/2019] [Accepted: 10/23/2019] [Indexed: 12/16/2022] Open
Abstract
Here, we reviewed emerging evidence on the role of the microbial community in colorectal carcinogenesis. A healthy gut microbiota promotes intestinal homeostasis and can exert anti-cancer effects; however, this microbiota also produces a variety of metabolites that are genotoxic and which can negatively influence epithelial cell behaviour. Disturbances in the normal microbial balance, known as dysbiosis, are frequently observed in colorectal cancer (CRC) patients. Microbial species linked to CRC include certain strains of Bacteroides fragilis, Escherichia coli, Streptococcus gallolyticus, Enterococcus faecalis and Fusobacterium nucleatum, amongst others. Whether these microbes are merely passive dwellers exploiting the tumour environment, or rather, active protagonists in the carcinogenic process is the subject of much research. The incidence of chemically-induced tumours in mice models varies, depending upon the presence or absence of these microorganisms, thus strongly suggesting influences on disease causation. Putative mechanistic explanations differentially link these strains to DNA damage, inflammation, aberrant cell behaviour and immune suppression. In the future, modulating the composition and metabolic activity of this microbial community may have a role in prevention and therapy.
Collapse
Affiliation(s)
- Eiman A Alhinai
- Dietetics Department, Al Nahdha Hospital, Ministry of Health, Muscat, PO Box 937, Ruwi, Muscat PC 112, Oman.
| | - Gemma E Walton
- Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6UA, UK.
| | - Daniel M Commane
- Department of Applied and Health Sciences, University of Northumbria, Newcastle Upon Tyne NE1 8ST, UK.
| |
Collapse
|
29
|
Teeuwssen M, Fodde R. Cell Heterogeneity and Phenotypic Plasticity in Metastasis Formation: The Case of Colon Cancer. Cancers (Basel) 2019; 11:cancers11091368. [PMID: 31540068 PMCID: PMC6770401 DOI: 10.3390/cancers11091368] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/09/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022] Open
Abstract
The adenoma-to-carcinoma progression in colon cancer is driven by a sequential accumulation of genetic alterations at specific tumor suppressors and oncogenes. In contrast, the multistage route from the primary site to metastasis formation is underlined by phenotypic plasticity, i.e., the capacity of disseminated tumor cells to undergo transiently and reversible transformations in order to adapt to the ever-changing environmental contexts. Notwithstanding the considerable body of evidence in support of the role played by epithelial-to-mesenchymal transition (EMT)/mesenchymal-to-epithelial transition (MET) in metastasis, its rate-limiting function, the detailed underlying cellular and molecular mechanisms, and the extension of the necessary morphologic and epigenetic changes are still a matter of debate. Rather than leading to a complete epithelial or mesenchymal state, the EMT/MET-program generates migrating cancer cells displaying intermediate phenotypes featuring both epithelial and mesenchymal characteristics. In this review, we will address the role of colon cancer heterogeneity and phenotypic plasticity in metastasis formation and the contribution of EMT to these processes. The alleged role of hybrid epithelial/mesenchymal (E/M) in collective and/or single-cell migration during local dissemination at the primary site and more systemic spreading will also be highlighted.
Collapse
Affiliation(s)
- Miriam Teeuwssen
- Department of Pathology, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands.
| | - Riccardo Fodde
- Department of Pathology, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands.
| |
Collapse
|
30
|
Geravand M, Fallah P, Yaghoobi MH, Soleimanifar F, Farid M, Zinatizadeh N, Yaslianifard S. INVESTIGATION OF ENTEROCOCCUS FAECALIS POPULATION IN PATIENTS WITH POLYP AND COLORECTAL CANCER IN COMPARISON OF HEALTHY INDIVIDUALS. ARQUIVOS DE GASTROENTEROLOGIA 2019; 56:141-145. [PMID: 31460576 DOI: 10.1590/s0004-2803.201900000-28] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 06/05/2019] [Indexed: 01/05/2023]
Abstract
BACKGROUND Colorectal cancer is one of the most commonly diagnosed cancers around the world. One of the factors involved in the development of colorectal cancer is the changes in the normal flora of the intestine. OBJECTIVE In this study, the mean copy number of Enterococcus faecalis in people with polyps and people with colorectal cancer has been evaluated in comparison with healthy controls. METHODS In this study, 25 patients with colorectal cancer and 28 patients with intestinal polyps were selected and stool specimens were taken. In addition, 24 healthy individuals were selected as control group. Extraction of bacterial DNA from the stool sample were performed. The molecular methods of PCR for confirmation of standard strain and absolute Real Time PCR (qRT-PCR) method were used to evaluate the number of Enterococcus faecalis in the studied groups. RESULTS The results of this study indicate that the mean copy number of Enterococcus faecalis in patients with colorectal cancer was 11.2x109 per gram of stool, and in patients with polyps was 9.4x108 per gram of stool. In healthy people, this number was 9x108 per gram of stool. There was a significant difference between the implicit copy numbers in the three groups. (P<0.05). CONCLUSION Enterococcus faecalis in faecal flora of people with colorectal cancer was significantly higher than those with polyps and healthy people. This could potentially signify the ability of this bacterium to induce colorectal cancer. More studies are needed to prove this theory.
Collapse
Affiliation(s)
- Maryam Geravand
- Alborz University of Medical Sciences, School of Medicine, Student Research Committee, Karaj, Iran
| | - Parviz Fallah
- Alborz University of Medical Sciences, School of Paramedical Sciences, Department of Hematology, Karaj, Iran
| | - Mojtaba Hedayat Yaghoobi
- Alborz University of Medical Sciences, School of Medicine, Department of Infectious Disease, Karaj, Iran
| | - Fatemeh Soleimanifar
- Alborz University of Medical Sciences, Dietary Supplements and Probiotic Research Center, Karaj, Iran
| | - Malihe Farid
- Alborz University of Medical Sciences, Social Determinants of Health Research Center, Karaj, IR Iran.,Alborz University of Medical Sciences, Faculty of Medicine, Karaj, IR Iran
| | - Nazi Zinatizadeh
- Alborz University of Medical Sciences, School of Medicine, Student Research Committee, Karaj, Iran
| | - Somayeh Yaslianifard
- Alborz University of Medical Sciences, Dietary Supplements and Probiotic Research Center, Karaj, Iran.,Alborz University of Medical Sciences, School of Medicine, Department of Microbiology, Karaj, Iran
| |
Collapse
|
31
|
Li L, Rao S, Cheng Y, Zhuo X, Deng C, Xu N, Zhang H, Yang L. Microbial osteoporosis: The interplay between the gut microbiota and bones via host metabolism and immunity. Microbiologyopen 2019; 8:e00810. [PMID: 31001921 PMCID: PMC6692530 DOI: 10.1002/mbo3.810] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 01/09/2019] [Accepted: 01/11/2019] [Indexed: 01/15/2023] Open
Abstract
The complex relationship between intestinal microbiota and host is a novel field in recent years. A large number of studies are being conducted on the relationship between intestinal microbiota and bone metabolism. Bone metabolism consisted of bone absorption and formation exists in the whole process of human growth and development. The nutrient components, inflammatory factors, and hormone environment play important roles in bone metabolism. Recently, intestinal microbiota has been found to influence bone metabolism via influencing the host metabolism, immune function, and hormone secretion. Here, we searched relevant literature on Pubmed and reviewed the effect of intestinal microbiota on bone metabolism through the three aspects, which may provide new ideas and targets for the clinical treatment of osteoporosis.
Collapse
Affiliation(s)
- Lishan Li
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Shitao Rao
- School of Biomedical SciencesCUHKShatin, N.THong Kong SARChina
| | - Yanzhen Cheng
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Xiaoyun Zhuo
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Caihong Deng
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ningning Xu
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Hua Zhang
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Li Yang
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
32
|
Liu L, Liang L, Liang H, Wang M, Lu B, Xue M, Deng J, Chen Y. Fusobacterium nucleatum Aggravates the Progression of Colitis by Regulating M1 Macrophage Polarization via AKT2 Pathway. Front Immunol 2019; 10:1324. [PMID: 31249571 PMCID: PMC6582778 DOI: 10.3389/fimmu.2019.01324] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/24/2019] [Indexed: 12/15/2022] Open
Abstract
Disordered intestinal flora and discordant immune response are associated with the development of ulcerative colitis (UC). Recent work has described the ability of macrophages to undergo repolarization toward a proinflammatory M1 or anti-inflammatory M2 phenotype in response to particular bacterium-derived signals. Fusobacterium nucleatum (F. nucleatum, Fn) is a species of intestinal commensal bacteria with potential pathogenicity, but its association with UC and how it may contribute to progression of UC is largely unknown. In this study, we provide new evidence that F. nucleatum accumulated heavily in the intestine of UC patients and was accompanied by the secretion of IFN-γ and the skewing of M1 macrophages. Mechanistically, our data showed that F. nucleatum aggravated dextran sodium sulfate (DSS)-induced colitis in the production of Th1-related cytokines IFN-γ through the AKT2 signaling pathway in vitro and in vivo. To further confirm the disease-relevance of these shifts in macrophage repolarization in response to F. nucleatum, stimulated bone marrow-derived macrophages (BMDMs) were transferred into recipient mice with DSS colitis. This transfer resulted in increased disease activity and inflammatory cytokine production. Taken together, we show clearly that F. nucleatum can promote the progression of UC via proinflammatory M1 macrophage skewing, and targeting F. nucleatum or AKT2 signaling may be a viable means of blocking development of UC.
Collapse
Affiliation(s)
- Le Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, State Key Laboratory of Organ Failure Research, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liping Liang
- Guangdong Provincial Key Laboratory of Gastroenterology, State Key Laboratory of Organ Failure Research, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Huifen Liang
- Guangdong Provincial Key Laboratory of Gastroenterology, State Key Laboratory of Organ Failure Research, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mingming Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, State Key Laboratory of Organ Failure Research, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bingyun Lu
- Guangdong Provincial Key Laboratory of Gastroenterology, State Key Laboratory of Organ Failure Research, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Meng Xue
- Guangdong Provincial Key Laboratory of Gastroenterology, State Key Laboratory of Organ Failure Research, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Deng
- Guangdong Provincial Key Laboratory of Gastroenterology, State Key Laboratory of Organ Failure Research, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Chen
- Guangdong Provincial Key Laboratory of Gastroenterology, State Key Laboratory of Organ Failure Research, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
33
|
Alterations in intestinal microbiota of colorectal cancer patients receiving radical surgery combined with adjuvant CapeOx therapy. SCIENCE CHINA-LIFE SCIENCES 2019; 62:1178-1193. [PMID: 30796721 DOI: 10.1007/s11427-018-9456-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/12/2018] [Indexed: 01/20/2023]
Abstract
An intricate relationship exists and interactions occur between gut microbiota and colorectal cancer (CRC). Radical surgery combined with adjuvant chemotherapy (AC) serves as the mainstream therapeutic scheme for most CRC patients. The current research was conducted to assess the effect of surgery or chemotherapy on gut microbiota. Forty-three CRC patients who received radical surgery and AC were enrolled. Fecal samples were collected preoperatively, postoperatively, and after the first to fifth cycles of postoperative chemotherapy. The microbial community of each sample was analyzed using high throughput 16S rRNA amplicon sequencing. Compared with preoperative samples, fecal samples collected postoperatively exhibited a significant decrease of obligate anaerobes, tumor-related bacteria, and butyric acid-producing bacteria. However, a significant increase of some conditional pathogens was observed. In addition, the AC regimen (CapeOx) was found to alter intestinal microbiota dramatically. In particular, several changes were observed after chemotherapy including an increase of pathogenic bacteria, the "rebound effect" of chemotherapy-adapted bacteria, the shift of lactate-utilizing microbiota from Veillonella to Butyricimonas and Butyricicoccus, as well as the decrease of probiotics. Both radical surgery and CapeOx chemotherapy exert a non-negligible effect on the gut microbiota of CRC patients. Microbiota-based intervention may be beneficial for patients during postoperative clinical management.
Collapse
|
34
|
Hakozaki T, Okuma Y, Omori M, Hosomi Y. Impact of prior antibiotic use on the efficacy of nivolumab for non-small cell lung cancer. Oncol Lett 2019; 17:2946-2952. [PMID: 30854072 DOI: 10.3892/ol.2019.9899] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 10/18/2018] [Indexed: 12/26/2022] Open
Abstract
Gut microbiota serves an important role in shaping systemic immune responses. Antibiotics cause changes in the gut microbiota that may influence the efficacy of cancer immunotherapy. In the present study, a retrospective analysis of the data from 90 patients treated with nivolumab for non-small cell lung cancer (NSCLC) was conducted. A total of 13 patients were treated with antibiotics prior to nivolumab therapy. The median progression-free survival time in patients treated with antibiotics was 1.2 months [95% confidence interval (CI), 0.5-5.8], while the time for patients who were not treated with antibiotics was 4.4 months (95% CI, 2.5-7.4). The median overall survival time in patients treated with antibiotics was 8.8 months, while it was not reached in those not treated with antibiotics, respectively. The differences between the survival curves with regard to PFS and OS were statistically significant (P=0.04 and P=0.037, respectively). However, in multivariate analysis, no statistically significant association was indicated between survival and prior antibiotic use, although a certain trend concerning the negative influence of antibiotic use was conveyed.
Collapse
Affiliation(s)
- Taiki Hakozaki
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo 113-8677, Japan
| | - Yusuke Okuma
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo 113-8677, Japan.,Division of Oncology, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Miwako Omori
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Yukio Hosomi
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo 113-8677, Japan
| |
Collapse
|
35
|
Mezouar S, Chantran Y, Michel J, Fabre A, Dubus JC, Leone M, Sereme Y, Mège JL, Ranque S, Desnues B, Chanez P, Vitte J. Microbiome and the immune system: From a healthy steady-state to allergy associated disruption. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.humic.2018.10.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
36
|
Stein K, Lysson M, Schumak B, Vilz T, Specht S, Heesemann J, Roers A, Kalff JC, Wehner S. Leukocyte-Derived Interleukin-10 Aggravates Postoperative Ileus. Front Immunol 2018; 9:2599. [PMID: 30581430 PMCID: PMC6294129 DOI: 10.3389/fimmu.2018.02599] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 10/22/2018] [Indexed: 01/29/2023] Open
Abstract
Objective: Postoperative ileus (POI) is an inflammation-mediated complication of abdominal surgery, characterized by intestinal dysmotility and leukocyte infiltration into the muscularis externa (ME). Previous studies indicated that interleukin (IL)-10 is crucial for the resolution of a variety of inflammation-driven diseases. Herein, we investigated how IL-10 affects the postoperative ME inflammation and found an unforeseen role of IL-10 in POI. Design: POI was induced by a standardized intestinal manipulation (IM) in C57BL/6 and multiple transgenic mouse strain including C-C motif chemokine receptor 2-/-, IL-10-/-, and LysMcre/IL-10fl/fl mice. Leukocyte infiltration, gene and protein expression of cytokines, chemokines, and macrophage differentiation markers as well as intestinal motility were analyzed. IL-10 serum levels in surgical patients were determined by ELISA. Results: IL-10 serum levels were increased in patient after abdominal surgery. In mice, a complete or leucocyte-restricted IL-10 deficiency ameliorated POI and reduced the postoperative ME neutrophil infiltration. Infiltrating monocytes were identified as main IL-10 producers and undergo IL-10-dependent M2 polarization. Interestingly, M2 polarization is not crucial to POI development as abrogation of monocyte infiltration did not prevent POI due to a compensation of the IL-10 loss by resident macrophages and neutrophils. Organ culture studies demonstrated that IL-10 deficiency impeded neutrophil migration toward the surgically traumatized ME. This mechanism is mediated by reduction of neutrophil attracting chemokines. Conclusion: Monocyte-derived macrophages are the major IL-10 source during POI. An IL-10 deficiency decreases the postoperative expression of neutrophil-recruiting chemokines, consequently reduces the neutrophil extravasation into the postsurgical bowel wall, and finally protects mice from POI.
Collapse
Affiliation(s)
- Kathy Stein
- Department of Surgery, University of Bonn, Bonn, Germany
| | - Mariola Lysson
- Department of Surgery, University of Bonn, Bonn, Germany
| | - Beatrix Schumak
- Institute for Medical Microbiology, Immunology and Parasitology, University of Bonn, Bonn, Germany
| | - Tim Vilz
- Department of Surgery, University of Bonn, Bonn, Germany
| | - Sabine Specht
- Institute for Medical Microbiology, Immunology and Parasitology, University of Bonn, Bonn, Germany
| | - Jürgen Heesemann
- Department of Bacteriology, Max von Pettenkofer Institute, Munich, Germany
| | - Axel Roers
- Institute for Immunology, Medical Faculty, Technical University Dresden, Dresden, Germany
| | - Jörg C Kalff
- Department of Surgery, University of Bonn, Bonn, Germany
| | - Sven Wehner
- Department of Surgery, University of Bonn, Bonn, Germany
| |
Collapse
|
37
|
Loss of Forkhead Box O3 Facilitates Inflammatory Colon Cancer: Transcriptome Profiling of the Immune Landscape and Novel Targets. Cell Mol Gastroenterol Hepatol 2018; 7:391-408. [PMID: 30718226 PMCID: PMC6360252 DOI: 10.1016/j.jcmgh.2018.10.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 10/03/2018] [Accepted: 10/09/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Diminished forkhead box O3 (FOXO3) function drives inflammation and cancer growth; however, mechanisms fostering these pathobiologies are unclear. Here, we aimed to identify in colon loss of FOXO3-dependent cellular and molecular changes that facilitate inflammation-mediated tumor growth. METHODS FOXO3 knockout (KO) and wild-type (WT) mice were used in the AOM/DSS model of inflammation-mediated colon cancer. Bioinformatics were used for profiling of mRNA sequencing data from human and mouse colon and tumors; specific targets were validated in human colon cancer cells (shFOXO3). RESULTS In mice, FOXO3 deficiency led to significantly elevated colonic tumor burden (incidence and size) compared with WT (P < .05). In FOXO3 KO colon, activated molecular pathways overlapped with those associated with mouse and human colonic inflammation and cancer, especially human colonic tumors with inflammatory microsatellite instability (false discovery rate < 0.05). FOXO3 KO colon, similar to tumors, had increased neutrophils, macrophages, B cells, T cells, and decreased natural killer cells (false discovery rate < 0.05). Moreover, in KO colon differentially expressed transcripts were linked to activation of inflammatory nuclear factor kappa B, tumorigenic cMyc, and bacterial Toll-like receptor signaling. Among differentially expressed transcripts, we validated altered expression of integrin subunit alpha 2 (ITGA2), ADAM metallopeptidase with thrombospondin type 1 motif 12, and ST8 alpha-N-acetyl-neuraminide alpha-2,8-sialyltransferase 5 in mouse WT and FOXO3 KO colon and tumors (P < .05). Similarly, their altered expression was found in human inflammatory bowel disease and colon cancer tissues and linked to poor patient survival. Ultimately, in human colon cancer cells, FOXO3 knockdown (shFOXO3) led to significantly increased ITGA2, and silencing ITGA2 (siRNA) alone diminished cell growth. CONCLUSIONS We identified the loss of FOXO3-mediated immune landscape, pathways, and transcripts that could serve as biomarkers and new targets for inflammatory colon cancer treatment.
Collapse
|
38
|
Druzhinin VG, Matskova LV, Fucic A. Induction and modulation of genotoxicity by the bacteriome in mammals. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2018; 776:70-77. [PMID: 29807578 DOI: 10.1016/j.mrrev.2018.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 04/19/2018] [Accepted: 04/20/2018] [Indexed: 02/07/2023]
Abstract
The living environment is a multilevel physical and chemical xenobiotic complex with potentially mutagenic effects and health risks. In addition to inorganic exposures, all terrestrial and aquatic living forms interact with microbiota as selectively established communities of bacteria, viruses and fungi. Along these lines, the human organism should then be considered a "meta-organism" with complex dynamics of interaction between the environment and microbiome. Bacterial communities within the microbiome, bacteriome, by its mass, symbiotic or competitive position and composition are in a fragile balance with the host organisms and have a crucial impact on their homeostasis. Bacteriome taxonomic composition is modulated by age, sex and host genetic profile and may be changed by adverse environmental exposures and life style factors such as diet or drug intake. A changed and/or misbalanced bacteriome has genotoxic potential with significant impact on the pathogenesis of acute, chronic and neoplastic diseases in the host organism. Bacteria may produce genotoxins, express a variety of pathways in which they generate free radicals or affect DNA repair causing genome damage, cell cycle arrest and apoptosis, modulate immune response and launch carcinogenesis in the host organism. Future investigations should focus on the interplay between exposure to xenobiotics and bacteriome composition, immunomodulation caused by misbalanced bacteriome, impact of the environment on bacteriome composition in children and its lifelong effect on health risks.
Collapse
Affiliation(s)
- V G Druzhinin
- Department of Genetics, Kemerovo State University, Kemerovo. Russia; Federal Research Center of Coal and Coal Chemistry of Siberian Branch of the Russian Academy of Sciences, Kemerovo, Russia
| | - L V Matskova
- Department of Microbiology and Tumor Biology, Karolinska Institute, Stockholm. Sweden
| | - A Fucic
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| |
Collapse
|
39
|
Gaines S, Williamson AJ, Hyman N, Kandel J. How the microbiome is shaping our understanding of cancer biology and its treatment. SEMINARS IN COLON AND RECTAL SURGERY 2018. [DOI: 10.1053/j.scrs.2017.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
40
|
Xue L, Williamson A, Gaines S, Andolfi C, Paul-Olson T, Neerukonda A, Steinhagen E, Smith R, Cannon LM, Polite B, Umanskiy K, Hyman N. An Update on Colorectal Cancer. Curr Probl Surg 2018; 55:76-116. [PMID: 29631699 DOI: 10.1067/j.cpsurg.2018.02.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Lai Xue
- Department of Surgery, University of Chicago Medicine, Chicago, IL
| | | | - Sara Gaines
- Department of Surgery, University of Chicago Medicine, Chicago, IL
| | - Ciro Andolfi
- Department of Surgery, University of Chicago Medicine, Chicago, IL
| | - Terrah Paul-Olson
- Department of Surgery, Emory University School of Medicine, Atlanta, GA
| | - Anu Neerukonda
- Department of Medicine, University of Chicago Medicine, Chicago, IL
| | - Emily Steinhagen
- Department of Surgery, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Radhika Smith
- Department of Surgery, University of Chicago Medicine, Chicago, IL
| | - Lisa M Cannon
- Department of Surgery, University of Chicago Medicine, Chicago, IL
| | - Blasé Polite
- Department of Medicine, University of Chicago Medicine, Chicago, IL
| | | | - Neil Hyman
- Department of Surgery, University of Chicago Medicine, Chicago, IL.
| |
Collapse
|
41
|
Bader JE, Enos RT, Velázquez KT, Carson MS, Nagarkatti M, Nagarkatti PS, Chatzistamou I, Davis JM, Carson JA, Robinson CM, Murphy EA. Macrophage depletion using clodronate liposomes decreases tumorigenesis and alters gut microbiota in the AOM/DSS mouse model of colon cancer. Am J Physiol Gastrointest Liver Physiol 2018; 314:G22-G31. [PMID: 29025731 PMCID: PMC5866374 DOI: 10.1152/ajpgi.00229.2017] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/11/2017] [Accepted: 09/12/2017] [Indexed: 01/31/2023]
Abstract
We examined the role of macrophages in inflammation associated with colorectal cancer (CRC). Given the emerging evidence on immune-microbiota interactions in CRC, we also sought to examine the interaction between macrophages and gut microbiota. To induce CRC, male C57BL/6 mice ( n = 32) received a single injection of azoxymethane (AOM), followed by three cycles of dextran sodium sulfate (DSS)-supplemented water in weeks 1, 4, and 7. Prior to the final DSS cycle ( week 7) and twice weekly until euthanasia, mice ( n = 16/group) received either 200 μl ip of clodronate-filled liposomes (CLD) or phosphate-buffered saline (PBS) encapsulated liposomes to deplete macrophages. Colon tissue was analyzed for polyp burden, macrophage markers, transcription factors, and inflammatory mediators. Stool samples were collected, and DNA was isolated and subsequently sequenced for 16S rRNA. Clodronate liposomes decreased tumor number by ∼36% and specifically large (≥1 mm) tumors by ∼36% ( P < 0.05). This was consistent with a decrease in gene expression of EMR1 in the colon tissue and polyp tissue as well as expression of select markers associated with M1 (IL-6) and M2 macrophages (IL-13, IL-10, TGFβ, CCL17) in the colon tissue ( P < 0.05). Similarly, there was a decrease in STAT3 and p38 MAPK and ERK signaling in colon tissue. Clodronate liposomes increased the relative abundance of the Firmicutes phylum ( P < 0.05) and specifically Lactobacillaceae and Clostridiaceae families, which have been associated with reduced CRC risk. Overall, these data support the development of therapeutic strategies to target macrophages in CRC and provide support for further evaluation of immune-microbiota interactions in CRC. NEW & NOTEWORTHY We found that macrophage depletion during late-stage tumorigenesis is effective at reducing tumor growth. This was associated with a decrease in macrophage markers and chemokines in the colon tissue and a decrease in transcription factors that are linked to colorectal cancer. The macrophage-depleted group was found to have an increased abundance of Firmicutes, a phylum with documented anti-tumorigenic effects. Overall, these data support the development of therapeutic strategies to target macrophages in colorectal cancer.
Collapse
Affiliation(s)
- Jackie E Bader
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Reilly T Enos
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Kandy T Velázquez
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
- Department of Exercise Science, School of Public Health, University of South Carolina , Columbia, South Carolina
| | - Meredith S Carson
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Prakash S Nagarkatti
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - J Mark Davis
- Department of Exercise Science, School of Public Health, University of South Carolina , Columbia, South Carolina
| | - James A Carson
- Department of Exercise Science, School of Public Health, University of South Carolina , Columbia, South Carolina
- Center for Colon Cancer Research, University of South Carolina , Columbia, South Carolina
| | - Cory M Robinson
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University , Morgantown, West Virginia
| | - E Angela Murphy
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
- Center for Colon Cancer Research, University of South Carolina , Columbia, South Carolina
| |
Collapse
|
42
|
Rubio CA, Langner C, Schmidt PT. Partial to complete abrogation of the subepithelial macrophage barrier against the gut microbiota in patients with ulcerative colitis and Crohn's colitis. Histopathology 2017; 72:580-587. [PMID: 29023984 DOI: 10.1111/his.13417] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 10/08/2017] [Indexed: 02/06/2023]
Abstract
AIMS The integrity of the band of indigenous macrophages in the subepithelial layer of the lamina propria (SLP) is crucial in preventing the commensal gut microbiota from attacking the host. The breakdown of the SLP macrophage barrier results in microbiota inflow and improper immune responses; this might lead to inflammatory bowel disease (IBD). During inflammation, the SLP macrophage barrier is reinforced by inflammation-elicited macrophages (IEMs), which are derived from blood-circulating monocytes. The aim was to explore the characteristics of the SLP macrophage band in a cohort of biopsies without inflammation, in patients with ulcerative colitis in remission (UCre), and in patients with right-sided Crohn's colitis (RCC). METHODS AND RESULTS Endoscopic biopsies were taken from endoscopically normal descending colon in 247 patients; 80 with IBD (27 UCre and 53 RCC), and 167 without IBD [90 had colonic diarrhoea, 63 were enrolled in a colorectal cancer (CRC) surveillance programme, seven had microscopic colitis in remission, and seven had miscellaneous colonic ailments]. Sections showed no inflammatory changes; they were immunostained with CD68. Among patients with UCre and RCC, the SLP band of CD68+ macrophages was fragmented or minute in 59% (47/80) and negative in 9% (7/80). In contrast, only 31% (51/167) of the biopsies from control patients had a fragmented/minute SLP band of CD68+ macrophages, and none had a negative SLP band of CD68+ macrophages (IBD versus controls, P < 0.05). CONCLUSIONS The finding that the SLP macrophage barrier was fragmented to totally abrogated in UCre and RCC patients suggests a longlasting defect in the SLP CD68+ macrophage barrier in these patients. The lack of ongoing inflammation in colonic biopsies should rule out the participation of bone marrow-derived IEMs in the abrogation of the SLP macrophage barrier reported here.
Collapse
Affiliation(s)
- Carlos A Rubio
- Department of Pathology, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Cord Langner
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Peter T Schmidt
- Department of Medicine, Karolinska Institute, Centre for Digestive Diseases, University Hospital, Stockholm, Sweden
| |
Collapse
|
43
|
Wang X, Yang Y, Huycke MM. Commensal-infected macrophages induce dedifferentiation and reprogramming of epithelial cells during colorectal carcinogenesis. Oncotarget 2017; 8:102176-102190. [PMID: 29254234 PMCID: PMC5731944 DOI: 10.18632/oncotarget.22250] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 09/29/2017] [Indexed: 01/01/2023] Open
Abstract
The colonic microbiome contributes to the initiation of colorectal cancer through poorly characterized mechanisms. We have shown that commensal-polarized macrophages induce gene mutation, chromosomal instability, and endogenous transformation through microbiome-induced bystander effects (MIBE). In this study we show that MIBE activates Wnt/β-catenin signaling and pluripotent transcription factors associated with dedifferentiation, reprogramming, and the development of colorectal cancer stem cells (CSCs). Exposure of murine primary colon epithelial cells (YAMC) to Enterococcus faecalis-infected macrophages increased Wnt3α expression while suppressing Wnt inhibitor factor 1 (Wif1). Wnt/β-catenin activation was confirmed by increased active β-catenin and Tcf4. in vivo, active β-catenin was evident in colon biopsies from E. faecalis-colonized Il10 knockout mice compared to sham-colonized mice. This effect was mediated, in part, by 4-hydroxy-2-nonenal and tumor necrosis factor α. MIBE also activated pluripotent transcription factors c-Myc, Klf4, Oct4, and Sox2 in YAMC cells and colons from E. faecalis-colonized Il10 knockout mice. These transcription factors are associated with cellular reprogramming, dedifferentiation, and induction of colorectal CSC progenitors. In support of this was an increase in the expression of Dclk1 and CD44, two colorectal CSC markers, in YAMC cells that were exposed to MIBE. Finally, compared to normal colon biopsies and hyperplastic polyps, DCLK1 expression increased in human tubular adenomas and invasive colorectal cancers. Blocking β-catenin/TCF4 signaling using FH535 and CTNNB1-specific small interfering RNA decreased DCLK1 expression in HCT116 human colon cancer cells. These findings provide mechanism for microbiome-induced colorectal cancer and identify new potential targets for colorectal cancer prevention.
Collapse
Affiliation(s)
- Xingmin Wang
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.,The Muchmore Laboratories for Infectious Diseases Research, Oklahoma City VA Health Care System, Oklahoma City, OK 73104, USA
| | - Yonghong Yang
- Gansu Province Children's Hospital, Lanzhou, Gansu 730030, China.,Key Laboratory of Gastrointestinal Cancer, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Mark M Huycke
- The Muchmore Laboratories for Infectious Diseases Research, Oklahoma City VA Health Care System, Oklahoma City, OK 73104, USA.,Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73126, USA
| |
Collapse
|
44
|
Dong T, Feng Q, Liu F, Chang LK, Zhou X, Han M, Tian X, Zhong N, Liu S. Alteration of stomach microbiota compositions in the progression of gastritis induces nitric oxide in gastric cell. Exp Ther Med 2017; 13:2793-2800. [PMID: 28587343 PMCID: PMC5450739 DOI: 10.3892/etm.2017.4373] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 03/29/2017] [Indexed: 12/14/2022] Open
Abstract
Atrophic gastritis is considered to be an antecedent to intestinal metaplasia and gastric cancer. A previous study identified that Helicobacter pylori was absent at the severe atrophic gastritis stage, and alterations in the gastric microbial composition resembled those in gastric cancer. To explore the role of the bacteria absence of H. pylori in gastric carcinogenesis, in the current study, we compared the microbiota of clinically collected H. pylori-free gastric fluids from 30 patients with non-atrophic gastritis (N) and 22 patients with severe atrophic gastritis (S). We estimated the bacterial loads in the N and S groups by colony counting in culture agar as well as by measuring the concentration of the extracted DNA. The results showed a significant increase in bacterial load in patients with atrophic gastritis in comparison to non-atrophic gastritis. Then, we analyzed the microbial communities of the gastric fluids from all 52 patients using high-throughput sequencing of 16S rRNA amplicons. The Chao 1, Shannon and Simpson diversity indexes demonstrated that the bacterial richness and diversity were not significantly different between the N and S groups. Moreover, principal component analysis illustrated that the microbiomes from the S group were more scattered. Microbiota composition analysis showed that the entire dataset was clustered into 27 phyla, 61 classes, 106 orders, 177 families, 292 genera and 121 species. At the genus level, only the abundance of Prevotella was significantly different between the N and S groups. Further analysis showed that all the higher taxonomic categories were significantly different between the N and S groups. To assess the effects of the metabolic products of Prevotella spp. on gastric cell physiology, we treated the human gastric epithelial cell line AGS with acetic acid and monitored nitric oxide (NO) production. The results showed that acetic acid at low concentrations (0.5 and 5 µM) significantly inhibited AGS cells to secrete NO compared to phosphate buffer saline-treated control cells. These results suggest that the microbiota in non-atrophic gastritis may influence gastric epithelial cell physiology.
Collapse
Affiliation(s)
- Tianyi Dong
- Department of Medical Microbiology, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China.,Department of Breast Thyroid Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Qiang Feng
- Department of Human Microbiome, School of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, Shandong 250021, P.R. China
| | - Fengyan Liu
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Lap Kam Chang
- Department of Medical Microbiology, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xiangyu Zhou
- Department of Medical Microbiology, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Mingyong Han
- Department of Breast Thyroid Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xingsong Tian
- Department of Breast Thyroid Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Ning Zhong
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Shili Liu
- Department of Medical Microbiology, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
45
|
Rezasoltani S, Asadzadeh-Aghdaei H, Nazemalhosseini-Mojarad E, Dabiri H, Ghanbari R, Zali MR. Gut microbiota, epigenetic modification and colorectal cancer. IRANIAN JOURNAL OF MICROBIOLOGY 2017; 9:55-63. [PMID: 29213996 PMCID: PMC5715278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Micro-organisms contain 90% of cells in human body and trillions foreign genes versus less than 30 thousand of their own. The human colon host various species of microorganisms, appraised at more than 1014 microbiota and contained of over a thousand species. Although each one's profile is separable, the relative abundance and distribution of bacterial species is the same between healthy ones, causing conservation of each person's overall health. Germline DNA mutations have been attributed to the less than 5% of CRC occurrence while more than 90% is associated with the epigenetic regulation. The most ubiquitous environmental factor in epigenetic modification is gut microbiota. Disruptive changes in the gut microbiome strongly contributed to the improvement of colorectal cancer. Gut microbiota may play critical role in progression of CRC via their metabolite or their structural component interacting with host intestinal epithelial cell (IEC). Herein we discuss the mechanism of epigenetic modification and its implication in CRC development, progression even metastasis by gut microbiota induction.
Collapse
Affiliation(s)
- Sama Rezasoltani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 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, Iran
| | - Ehsan Nazemalhosseini-Mojarad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Corresponding author: Ehsan Nazemalhosseini-Mojarad, PhD, Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran. Tel: +98-21-22432516
| | - Hossein Dabiri
- Department of Medical Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Ghanbari
- Digestive Oncology Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Wang X, Yang Y, Huycke MM. Microbiome-driven carcinogenesis in colorectal cancer: Models and mechanisms. Free Radic Biol Med 2017; 105:3-15. [PMID: 27810411 DOI: 10.1016/j.freeradbiomed.2016.10.504] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/19/2016] [Accepted: 10/25/2016] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer death and archetype for cancer as a genetic disease. However, the mechanisms for genetic change and their interactions with environmental risk factors have been difficult to unravel. New hypotheses, models, and methods are being used to investigate a complex web of risk factors that includes the intestinal microbiome. Recent research has clarified how the microbiome can generate genomic change in CRC. Several phenotypes among a small group of selected commensals have helped us better understand how mutations and chromosomal instability (CIN) are induced in CRC (e.g., toxin production, metabolite formation, radical generation, and immune modulation leading to a bystander effect). This review discusses recent hypotheses, models, and mechanisms by which the intestinal microbiome contributes to the initiation and progression of sporadic and colitis-associated forms of CRC. Overall, it appears the microbiome can initiate and/or promote CRC at all stages of tumorigenesis by acting as an inducer of DNA damage and CIN, regulating cell growth and death, generating epigenetic changes, and modulating host immune responses. Understanding how the microbiome interacts with other risk factors to define colorectal carcinogenesis will ultimately lead to more accurate risk prediction. A deeper understanding of CRC etiology will also help identify new targets for prevention and treatment and help accelerate the decline in mortality for this common cancer.
Collapse
Affiliation(s)
- Xingmin Wang
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, USA; Muchmore Laboratories for Infectious Diseases Research, Oklahoma City VA Health Care System, USA
| | - Yonghong Yang
- Gansu Province Children's Hospital, Lanzhou, China; Key Laboratory of Gastrointestinal Cancer, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Mark M Huycke
- Muchmore Laboratories for Infectious Diseases Research, Oklahoma City VA Health Care System, USA; Department of Internal Medicine, PO Box 26901, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73126-0901, USA.
| |
Collapse
|
47
|
Botticelli A, Zizzari I, Mazzuca F, Ascierto PA, Putignani L, Marchetti L, Napoletano C, Nuti M, Marchetti P. Cross-talk between microbiota and immune fitness to steer and control response to anti PD-1/PDL-1 treatment. Oncotarget 2017; 8:8890-8899. [PMID: 27806346 PMCID: PMC5352451 DOI: 10.18632/oncotarget.12985] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 10/13/2016] [Indexed: 12/22/2022] Open
Abstract
Immune Checkpoint Inhibitors (ICIs) are improving the survival of cancer patients, however only the 20-30% of treated patients present clinical benefits. Toxicity represents the major cause of reduced dosage, delayed drug administration and therapy discontinuation. Hence in the context of multiple treatment possibilities, the identification of predictive markers of response and toxicity is a challenging approach for drug selection in order to obtain the best clinical benefit while minimizing the side effects. The loss of the protective function of intestinal barriers that interacts with the environment measured as increased intestinal permeability and the changes occurring in the microbiota composition have been proposed as a mechanism potentially explaining the pathogenesis of immune related toxicity.In this review we discuss the new perspectives on the involvement of PD-1 and PDL-1 in the cross talk between gut microbiota and immune fitness and how gut microbiota impacts on the efficacy of anti-PD-1 and anti-PDL-1 treatments in cancer.
Collapse
Affiliation(s)
- Andrea Botticelli
- Department of Clinical and Molecular Medicine, SantAndrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Ilaria Zizzari
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Federica Mazzuca
- Department of Clinical and Molecular Medicine, SantAndrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Paolo Antonio Ascierto
- Melanoma, Cancer Immunotherapy, and Innovative Therapy, Istituto nazionale Tumori Fondazione G Pascale, Napoli, Italy
| | - Lorenza Putignani
- Units of Parasitology and Human Microbiome, Bambino Ges Childrens Hospital and Research Institute, Rome, Italy
| | - Luca Marchetti
- Department of Clinical Oncology, Policlinico Umberto I, University of Rome Sapienza, Rome, Italy
| | - Chiara Napoletano
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Marianna Nuti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, SantAndrea Hospital, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
48
|
Gagnaire A, Nadel B, Raoult D, Neefjes J, Gorvel JP. Collateral damage: insights into bacterial mechanisms that predispose host cells to cancer. Nat Rev Microbiol 2017; 15:109-128. [DOI: 10.1038/nrmicro.2016.171] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
49
|
Gastrointestinal microbiome and breast cancer: correlations, mechanisms and potential clinical implications. Breast Cancer 2016; 24:220-228. [PMID: 27709424 DOI: 10.1007/s12282-016-0734-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 09/21/2016] [Indexed: 02/05/2023]
Abstract
Gastrointestinal microbiome plays as a symbiont which provides protection effect against invading pathogens, aids in the immune system development, nutrient reclamation and absorption as well as molecule breakdown. And it may avert carcinogenesis through these biological activities. By now, studies have been carried out to elaborate the association between gastrointestinal microbiome and breast cancer. It has been implicated that breast cancer was substantially associated with estrogen-dependent and estrogen-independent functions of gastrointestinal microbiome. Evidence from animal experiments also confirmed mammary tumor-related changes in microbial community. The possible mechanisms involve estrogen metabolism, immune regulation, obese status and so forth. Based on the current evidence, cues on future management strategies of breast cancer such as antibiotics and dietary interventions are proposed. In conclusion, large-scale clinical studies and bench-based researches are needed to validate the associations and elaborate the mechanisms, so as to reduce the risk of breast cancer and improve the outcomes of those already diagnosed.
Collapse
|
50
|
Current Hypothesis for the Relationship between Dietary Rice Bran Intake, the Intestinal Microbiota and Colorectal Cancer Prevention. Nutrients 2016; 8:nu8090569. [PMID: 27649240 PMCID: PMC5037554 DOI: 10.3390/nu8090569] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/07/2016] [Accepted: 09/08/2016] [Indexed: 12/17/2022] Open
Abstract
Globally, colorectal cancer (CRC) is the third most common form of cancer. The development of effective chemopreventive strategies to reduce CRC incidence is therefore of paramount importance. Over the past decade, research has indicated the potential of rice bran, a byproduct of rice milling, in CRC chemoprevention. This was recently suggested to be partly attributable to modification in the composition of intestinal microbiota when rice bran was ingested. Indeed, previous studies have reported changes in the population size of certain bacterial species, or microbial dysbiosis, in the intestines of CRC patients and animal models. Rice bran intake was shown to reverse such changes through the manipulation of the population of health-promoting bacteria in the intestine. The present review first provides an overview of evidence on the link between microbial dysbiosis and CRC carcinogenesis and describes the molecular events associated with that link. Thereafter, there is a summary of current data on the effect of rice bran intake on the composition of intestinal microbiota in human and animal models. The article also highlights the need for further studies on the inter-relationship between rice bran intake, the composition of intestinal microbiota and CRC prevention.
Collapse
|