1
|
Wang K, Wang X, Song L. Unraveling the complex role of neutrophils in lymphoma: From pathogenesis to therapeutic approaches (Review). Mol Clin Oncol 2024; 21:85. [PMID: 39347476 PMCID: PMC11428085 DOI: 10.3892/mco.2024.2783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/21/2024] [Indexed: 10/01/2024] Open
Abstract
Lymphoma, a malignancy of the lymphatic system, which is critical for maintaining the body's immune defenses, has become a focal point in recent research due to its intricate interplay with neutrophils-white blood cells essential for combating infections and inflammation. Unlike prior perceptions associating neutrophils only with tumor support, contemporary studies underscore their intricate and multifaceted involvement in the immune response to lymphoma. Recognizing the nuanced participation of neutrophils in lymphoma is crucial for developing innovative treatments to improve patient outcomes.
Collapse
Affiliation(s)
- Ke Wang
- Department of Cell Engineering, School of Life Sciences and Biotechnology, Sanquan College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Xiao Wang
- Reproduction Medicine Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, Guangdong 524002, P.R. China
| | - Li Song
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Sanquan College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| |
Collapse
|
2
|
Yu Y, Zhou M, Sadiq FA, Hu P, Gao F, Wang J, Liu A, Liu Y, Wu H, Zhang G. Comparison of the effects of three sourdough postbiotics on high-fat diet-induced intestinal damage. Food Funct 2024; 15:9053-9069. [PMID: 39162079 DOI: 10.1039/d4fo02948h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
There is significant interest in using postbiotics as an intervention strategy to address obesity. This study assesses the efficacy of postbiotics derived from different sourdough strains (Lactiplantibacillus plantarum LP1, LP25, and Pediococcus pentosaceus PP18) in mitigating intestinal injury in zebrafish fed on a high-fat diet. We screened postbiotics for their anti-colon cancer cell effects and compared various preparation methods applied to live bacterial strains, including heat-killing at different temperatures, pH adjustments, and ultraviolet radiation exposure. Heat-killing at 120 °C proved to be the most effective preparation method. A marked variation in health effects was observed in the heat-killed microbial cells, as evidenced by their hydrophobicity and self-aggregation ability. A five-week high-fat dietary intervention study in zebrafish demonstrated that diets supplemented with 108 CFU g-1 K-LP25 significantly attenuated weight gain and body fat, along with reductions in FASN, Leptin, and SREBF1 mRNA expression. However, diets supplemented with 107 CFU g-1 K-PP18 only reduced Leptin and SREBF1 mRNA expression. K-PP18 was more effective at mitigating gut barrier damage, promoting colonic Occludin, ZO-1, and Claudin-1 levels. Additionally, K-LP25 supplementation markedly downregulated the pro-inflammatory cytokines TNF-α, IL-6, and IL-1β, reducing intestinal inflammation. Supplementation with K-LP1 and K-PP18 increased the abundance of Acinetobacter spp., whereas K-LP25 increased the abundance of Cetobacterium and Plesiomonas. Collectively, these findings suggest that inactivated strains confer protective effects against high-fat diet-induced intestinal damage in zebrafish, with variation observed across different species. Studying the effects of sourdough-derived postbiotics on gut health may open new avenues for dietary interventions to manage gut-related diseases.
Collapse
Affiliation(s)
- Yujuan Yu
- School of Life Science, Shanxi University, Taiyuan, 030006, PR China.
| | - Min Zhou
- School of Life Science, Shanxi University, Taiyuan, 030006, PR China.
| | - Faizan Ahmed Sadiq
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK
| | - Pengli Hu
- School of Life Science, Shanxi University, Taiyuan, 030006, PR China.
| | - Feng Gao
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Juanxia Wang
- School of Life Science, Shanxi University, Taiyuan, 030006, PR China.
| | - Aowen Liu
- School of Life Science, Shanxi University, Taiyuan, 030006, PR China.
| | - Yue Liu
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Haili Wu
- School of Life Science, Shanxi University, Taiyuan, 030006, PR China.
| | - Guohua Zhang
- School of Life Science, Shanxi University, Taiyuan, 030006, PR China.
| |
Collapse
|
3
|
Tang Y, Lin TC, Yang H, Zhou Y, Sibeko L, Liu Z. High-fat diet during early life reshapes the gut microbiome and is associated with the disrupted mammary microenvironment in later life in mice. Nutr Res 2024; 127:1-12. [PMID: 38763113 DOI: 10.1016/j.nutres.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/22/2024] [Accepted: 04/27/2024] [Indexed: 05/21/2024]
Abstract
The influence of gut microbiota on gut health is well-documented, but it remains obscure for extraintestinal diseases such as breast cancer. Moreover, it is entirely unknown how gut dysbiosis during early life contributes to breast tumorigenesis later in life. In this study, we hypothesized that a high-fat diet during early life leads to alterations in the gut microbiome and is associated with disruptions in the mammary microenvironment. Female C57BL/6 mice were fed a low-fat diet (10% kcal fat) or a high-fat diet (HF, 60% kcal fat) for 8 weeks from the age of 4 to 12 weeks, which is equivalent to human childhood and adolescence. Twelve mice were sacrificed immediately after the 8-week feeding, the remainder were euthanized after switching to a normal lifecycle-supporting diet for an additional 12 weeks; the gut microbiome was then sequenced. The 8-week HF diet feeding altered the beta-diversity (Bray & Jaccard P < .01), and the difference remained significant after switching the diet (Bray & Jaccard P < .05). Immediately after HF feeding, a greater number of microbial taxa (>50) were altered, and about half of the taxa (25) remained significantly changed after switching the diet. The abundance of Alistipes, Bilophila, and Rikenellaceae stood out as significantly associated with multiple metabolic and inflammatory biomarkers in mammary tissue, including aromatase, Ccl2, and Cox2. In conclusion, an 8-week early-life HF feeding reshaped the gut microbiome, which connected with disrupted mammary microenvironments.
Collapse
Affiliation(s)
- Ying Tang
- School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, USA; Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Ting-Chun Lin
- School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, USA
| | - Hong Yang
- Department of Oncology and Pathology, Hunan Provincial People's Hospital, Changsha, China
| | - Yanjiao Zhou
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, USA; Hunan, China
| | - Lindiwe Sibeko
- School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, USA
| | - Zhenhua Liu
- School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, USA; UMass Cancer Center, University of Massachusetts Chan Medical Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
4
|
Wu Z, Zhuang X, Liang M, Sheng L, Huang L, Li Y, Ke Y. Identification of an inflammatory response-related gene prognostic signature and immune microenvironment for cervical cancer. Front Mol Biosci 2024; 11:1394902. [PMID: 38903179 PMCID: PMC11187284 DOI: 10.3389/fmolb.2024.1394902] [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: 03/02/2024] [Accepted: 05/20/2024] [Indexed: 06/22/2024] Open
Abstract
Background: Cervical cancer (CC) is the fourth most common cancer among women worldwide. As part of the brisk cross-talk between the host and the tumor, prognosis can be affected through inflammatory responses or the tumor microenvironment. However, further exploration of the inflammatory response-related genes that have prognostic value, microenvironment infiltration, and chemotherapeutic therapies in CC is needed. Methods: The clinical data and mRNA expression profiles of CC patients were downloaded from a public database for this study. In the TCGA cohort, a multigene prognostic signature was constructed by least absolute shrinkage and selection operator (LASSO) and Cox analyses. CC patients from the GEO cohort were used for validation. K‒M analysis was used to compare overall survival (OS) between the high- and low-risk groups. Univariate and multivariate Cox analyses were applied to determine the independent predictors of OS. The immune cell infiltration and immune-related functional score were calculated by single-sample gene set enrichment analysis (GSEA). Immunohistochemistry was utilized to validate the protein expression of prognostic genes in CC tissues. Results: A genetic signature model associated with the inflammatory response was built by LASSO Cox regression analysis. Patients in the high-risk group had a significantly lower OS rate. The predictive ability of the prognostic genes was evaluated by means of receiver operating characteristic (ROC) curve analysis. The risk score was confirmed to be an independent predictor of OS by univariate and multivariate Cox analyses. The immune status differed between the high-risk and low-risk groups, and the cancer-related pathways were enriched in the high-risk group according to functional analysis. The risk score was significantly related to tumor stage and immune infiltration type. The expression levels of five prognostic genes (LCK, GCH1, TNFRSF9, ITGA5, and SLC7A1) were positively related to sensitivity to antitumor drugs. Additionally, the expression of prognostic genes was significantly different between CC tissues and myoma patient cervix (non-tumorous) tissues in the separate sample cohort. Conclusion: A model consisting of 5 inflammation-related genes can be used to predict prognosis and influence immune status in CC patients. Furthermore, the inhibition or enhancement of these genes may become a novel alternative therapy.
Collapse
Affiliation(s)
- Zhuna Wu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Xuanxuan Zhuang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Meili Liang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Liying Sheng
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Li Huang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Yanting Li
- Department of Gynecology and Obstetrics, Anhai Hospital of Jinjiang, Quanzhou, Fujian, China
| | - Yumin Ke
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
5
|
Bani Saeid A, De Rubis G, Williams KA, Yeung S, Chellappan DK, Singh SK, Gupta G, Hansbro PM, Shahbazi MA, Gulati M, Kaur IP, Santos HA, Paudel KR, Dua K. Revolutionizing lung health: Exploring the latest breakthroughs and future prospects of synbiotic nanostructures in lung diseases. Chem Biol Interact 2024; 395:111009. [PMID: 38641145 DOI: 10.1016/j.cbi.2024.111009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/04/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
The escalating prevalence of lung diseases underscores the need for innovative therapies. Dysbiosis in human body microbiome has emerged as a significant factor in these diseases, indicating a potential role for synbiotics in restoring microbial equilibrium. However, effective delivery of synbiotics to the target site remains challenging. Here, we aim to explore suitable nanoparticles for encapsulating synbiotics tailored for applications in lung diseases. Nanoencapsulation has emerged as a prominent strategy to address the delivery challenges of synbiotics in this context. Through a comprehensive review, we assess the potential of nanoparticles in facilitating synbiotic delivery and their structural adaptability for this purpose. Our review reveals that nanoparticles such as nanocellulose, starch, and chitosan exhibit high potential for synbiotic encapsulation. These offer flexibility in structure design and synthesis, making them promising candidates for addressing delivery challenges in lung diseases. Furthermore, our analysis highlights that synbiotics, when compared to probiotics alone, demonstrate superior anti-inflammatory, antioxidant, antibacterial and anticancer activities. This review underscores the promising role of nanoparticle-encapsulated synbiotics as a targeted and effective therapeutic approach for lung diseases, contributing valuable insights into the potential of nanomedicine in revolutionizing treatment strategies for respiratory conditions, ultimately paving the way for future advancements in this field.
Collapse
Affiliation(s)
- Ayeh Bani Saeid
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Kylie A Williams
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Stewart Yeung
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Sachin Kumar Singh
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, 144411, India
| | - Gaurav Gupta
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, India; Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - Philip M Hansbro
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Mohammad-Ali Shahbazi
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, AV, 9713, Groningen, the Netherlands
| | - Monica Gulati
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Indu Pal Kaur
- University Institute of Pharmaceutical Sciences, Punjab University Chandigarh, India
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, AV, 9713, Groningen, the Netherlands; Drug Research Program Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India.
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India.
| |
Collapse
|
6
|
Feng K, Ren F, Shang Q, Wang X, Wang X. Association between oral microbiome and breast cancer in the east Asian population: A Mendelian randomization and case-control study. Thorac Cancer 2024; 15:974-986. [PMID: 38485288 PMCID: PMC11045337 DOI: 10.1111/1759-7714.15280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND The causal relationship between breast cancer (BC) and the oral microbiome remains unclear. In this case-control study, using two-sample Mendelian randomization (MR), we thoroughly explored the relationship between the oral microbiome and BC in the East Asian population. METHODS Genetic summary data related to oral microbiota and BC were collected from genome-wide association studies involving participants of East Asian descent. MR estimates were generated by conducting various analyses. Sequencing data from a case-control study were used to verify the validity of these findings. RESULTS MR analysis revealed that 30 tongue and 37 salivary bacterial species were significantly associated with BC. Interestingly, in both tongue and salivary microbiomes, we observed the causal effect of six genera, namely, Aggregatibacter, Streptococcus, Prevotella, Haemophilus, Lachnospiraceae, Oribacterium, and Solobacterium, on BC. Our case-control study findings suggest differences in specific bacteria between patients with BC and healthy controls. Moreover, sequencing data confirmed the MR analysis results, demonstrating that compared with the healthy control group, the BC group had a higher relative abundance of Pasteurellaceae and Streptococcaceae but a lower relative abundance of Bacteroidaceae. CONCLUSIONS Our MR analysis suggests that the oral microbiome exerts a causative effect on BC risk, supported by the sequencing data of a case-control study. In the future, studies should be undertaken to comprehensively understand the complex interaction mechanisms between the oral microbiota and BC.
Collapse
Affiliation(s)
- Kexin Feng
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Fei Ren
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Qingyao Shang
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xin Wang
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiang Wang
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
7
|
Lee C, Lee S, Yoo W. Metabolic Interaction Between Host and the Gut Microbiota During High-Fat Diet-Induced Colorectal Cancer. J Microbiol 2024; 62:153-165. [PMID: 38625645 DOI: 10.1007/s12275-024-00123-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/07/2024] [Accepted: 02/16/2024] [Indexed: 04/17/2024]
Abstract
Colorectal cancer (CRC) is the second-highest cause of cancer-associated mortality among both men and women worldwide. One of the risk factors for CRC is obesity, which is correlated with a high-fat diet prevalent in Western dietary habits. The association between an obesogenic high-fat diet and CRC has been established for several decades; however, the mechanisms by which a high-fat diet increases the risk of CRC remain unclear. Recent studies indicate that gut microbiota strongly influence the pathogenesis of both high-fat diet-induced obesity and CRC. The gut microbiota is composed of hundreds of bacterial species, some of which are implicated in CRC. In particular, the expansion of facultative anaerobic Enterobacteriaceae, which is considered a microbial signature of intestinal microbiota functional imbalance (dysbiosis), is associated with both high-fat diet-induced obesity and CRC. Here, we review the interaction between the gut microbiome and its metabolic byproducts in the context of colorectal cancer (CRC) during high-fat diet-induced obesity. In addition, we will cover how a high-fat diet can drive the expansion of genotoxin-producing Escherichia coli by altering intestinal epithelial cell metabolism during gut inflammation conditions.
Collapse
Affiliation(s)
- Chaeeun Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Seungrin Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Woongjae Yoo
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea.
| |
Collapse
|
8
|
Zhang J, He X, Tang BZ. Aggregation-Induced Emission-Armored Living Bacteriophage-DNA Nanobioconjugates for Targeting, Imaging, and Efficient Elimination of Intracellular Bacterial Infection. ACS NANO 2024; 18:3199-3213. [PMID: 38227824 DOI: 10.1021/acsnano.3c09695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Intracellular bacterial infections bring a considerable risk to human life and health due to their capability to elude immune defenses and exhibit significant drug resistance. As a result, confronting and managing these infections present substantial challenges. In this study, we developed a multifunctional living phage nanoconjugate by integrating aggregation-induced emission luminogen (AIEgen) photosensitizers and nucleic acids onto a bacteriophage framework (forming MS2-DNA-AIEgen bioconjugates). These nanoconjugates can rapidly penetrate mammalian cells and specifically identify intracellular bacteria while concurrently producing a detectable fluorescent signal. By harnessing the photodynamic property of AIEgen photosensitizer and the bacteriophage's inherent targeting and lysis capability, the intracellular bacteria can be effectively eliminated and the activity of the infected cells can be restored. Moreover, our engineered phage nanoconjugates were able to expedite the healing process in bacterially infected wounds observed in diabetic mice models while simultaneously enhancing immune activity within infected cells and in vivo, without displaying noticeable toxicity. We envision that these multifunctional phage nanoconjugates, which utilize AIEgen photosensitizers and spherical nucleic acids, may present a groundbreaking strategy for combating intracellular bacteria and offer powerful avenues for theranostic applications in intracellular bacterial infection-associated diseases.
Collapse
Affiliation(s)
- Jing Zhang
- The Key Lab of Health Chemistry and Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, China
| | - Xuewen He
- The Key Lab of Health Chemistry and Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, China
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
| |
Collapse
|
9
|
Zeber-Lubecka N, Kulecka M, Jagiełło-Gruszfeld A, Dąbrowska M, Kluska A, Piątkowska M, Bagińska K, Głowienka M, Surynt P, Tenderenda M, Mikula M, Ostrowski J. Breast cancer but not the menopausal status is associated with small changes of the gut microbiota. Front Oncol 2024; 14:1279132. [PMID: 38327745 PMCID: PMC10848918 DOI: 10.3389/fonc.2024.1279132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/03/2024] [Indexed: 02/09/2024] Open
Abstract
Background Possible relationships between gut dysbiosis and breast cancer (BC) development and progression have been previously reported. However, the results of these metagenomics studies are inconsistent. Our study involved 88 patients diagnosed with breast cancer and 86 cancer-free control women. Participants were divided into groups based on their menopausal status. Fecal samples were collected from 47 and 41 pre- and postmenopausal newly diagnosed breast cancer patients and 51 and 35 pre- and postmenopausal controls, respectively. In this study, we performed shotgun metagenomic analyses to compare the gut microbial community between pre- and postmenopausal BC patients and the corresponding controls. Results Firstly, we identified 12, 64, 158, and 455 bacterial taxa on the taxonomy level of phyla, families, genera, and species, respectively. Insignificant differences of the Shannon index and β-diversity were found at the genus and species levels between pre- and postmenopausal controls; the differences concerned only the Chao index at the species level. No differences in α-diversity indexes were found between pre- and postmenopausal BC patients, although β-diversity differed these subgroups at the genus and species levels. Consistently, only the abundance of single taxa differed between pre- and postmenopausal controls and cases, while the abundances of 14 and 23 taxa differed or tended to differ between premenopausal cases and controls, and between postmenopausal cases and controls, respectively. There were similar differences in the distribution of enterotypes. Of 460 bacterial MetaCyc pathways discovered, no pathways differentiated pre- and postmenopausal controls or BC patients, while two and one pathways differentiated cases from controls in the pre- and postmenopausal subgroups, respectively. Conclusion While our findings did not reveal an association of changes in the overall microbiota composition and selected taxa with the menopausal status in cases and controls, they confirmed differences of the gut microbiota between pre- and postmenopausal BC patients and the corresponding controls. However, these differences were less extensive than those described previously.
Collapse
Affiliation(s)
- Natalia Zeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Maria Kulecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Agnieszka Jagiełło-Gruszfeld
- Department of Breast Cancer & Reconstructive Surgery, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Michalina Dąbrowska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Anna Kluska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Magdalena Piątkowska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Katarzyna Bagińska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Maria Głowienka
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Piotr Surynt
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Michał Tenderenda
- Department of Oncological Surgery and Neuroendocrine Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Michał Mikula
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Jerzy Ostrowski
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| |
Collapse
|
10
|
Wu J, Zhang P, Mei W, Zeng C. Intratumoral microbiota: implications for cancer onset, progression, and therapy. Front Immunol 2024; 14:1301506. [PMID: 38292482 PMCID: PMC10824977 DOI: 10.3389/fimmu.2023.1301506] [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: 09/25/2023] [Accepted: 12/28/2023] [Indexed: 02/01/2024] Open
Abstract
Significant advancements have been made in comprehending the interactions between the microbiome and cancer. However, prevailing research predominantly directs its focus toward the gut microbiome, affording limited consideration to the interactions of intratumoral microbiota and tumors. Within the tumor microenvironment (TME), the intratumoral microbiome and its associated products wield regulatory influence, directing the modulation of cancer cell properties and impacting immune system functionality. However, to grasp a more profound insight into the intratumoral microbiota in cancer, further research into its underlying mechanisms is necessary. In this review, we delve into the intricate associations between intratumoral microbiota and cancer, with a specific focus on elucidating the significant contribution of intratumoral microbiota to the onset and advancement of cancer. Notably, we provide a detailed exploration of therapeutic advances facilitated by intratumoral microbiota, offering insights into recent developments in this burgeoning field.
Collapse
Affiliation(s)
- Jinmei Wu
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Pengfei Zhang
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Wuxuan Mei
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, China
| |
Collapse
|
11
|
Agrawal A, Anjankar A. Alterations of Gastrointestinal Microbe Composition in Various Human Diseases and Its Significance in the Early Diagnosis of Diseases. Cureus 2024; 16:e52435. [PMID: 38371166 PMCID: PMC10870805 DOI: 10.7759/cureus.52435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 01/16/2024] [Indexed: 02/20/2024] Open
Abstract
A 100 trillion bacteria, viruses, fungi, and archaea make up the human gut microbe. It has co-evolved with its human host and carries out essential tasks that improve general health. The relationship between gastrointestinal microbes and human health has been a growing field of interest and research in recent times. The gastrointestinal microbes are connected by complex networks and connections, and the host has given birth to the gut-microbe-brain axis, which shows the crucial effect that this circumstance could have on the health and diseases of the brain and spinal cord (or the central nervous system [CNS]). The microbe and the CNS interact bi-directionally via autonomic, neuroendocrine, gastrointestinal, and immune system pathways. The gut microbe has been connected to a range of gastrointestinal and extra-gastrointestinal diseases. The recent investigation supports the suspicion that the gut-microbe-brain axis could play a role in neuropsychiatric disorders including depression, dementia, post-traumatic stress disorder, anxiousness, bipolar disorder, schizophrenia, and obsessive-compulsive disorder, alongside chronic host illnesses such as obesity, diabetes, and inflammation. Studies point to gut microorganisms as possible biomarkers for a wide range of mental health issues. Changes in the gut microbe may be a crucial factor in the onset and advancement of non-alcoholic fatty liver damage. Gut microbes have been seen to influence microglia's response to the CNS's regional signals and thus to pain and inflammation. Data suggest that altering the gut microbe in those with chronic pain may be a successful method for reducing pain. Numerous investigations have documented alterations in the gut microbes made in Alzheimer patients and schizophrenic patients. The risk of breast cancer can be reduced by restoring gut microbe homeostasis and reducing systemic estrogen levels.
Collapse
Affiliation(s)
- Aman Agrawal
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Ashish Anjankar
- Biochemistry, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
12
|
Abd El-Aal AAA, Jayakumar FA, Reginald K. Dual-action potential of cationic cryptides against infections and cancers. Drug Discov Today 2023; 28:103764. [PMID: 37689179 DOI: 10.1016/j.drudis.2023.103764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/18/2023] [Accepted: 09/04/2023] [Indexed: 09/11/2023]
Abstract
Cryptides are a subfamily of bioactive peptides embedded latently in their parent proteins and have multiple biological functions. Cationic cryptides could be used as modern drugs in both infectious diseases and cancers because their mechanism of action is less likely to be affected by genetic mutations in the treated cells, therefore addressing a current unmet need in these two areas of medicine. In this review, we present the current understanding of cryptides, methods to mine them sustainably using available online databases and prediction tools, with a particular focus on their antimicrobial and anticancer potential, and their potential applicability in a clinical setting.
Collapse
Affiliation(s)
- Amr A A Abd El-Aal
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500 Selangor, Malaysia
| | - Fairen A Jayakumar
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500 Selangor, Malaysia
| | - Kavita Reginald
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500 Selangor, Malaysia.
| |
Collapse
|
13
|
Lee CC, Yang HW, Liu CJ, Lee F, Ko WC, Chang YC, Yang PS. Unraveling the connections between gut microbiota, stress, and quality of life for holistic care in newly diagnosed breast cancer patients. Sci Rep 2023; 13:17916. [PMID: 37864098 PMCID: PMC10589294 DOI: 10.1038/s41598-023-45123-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023] Open
Abstract
There is little research about the stress, quality of life (QOL) and gut microbiota in newly diagnosed breast cancer patients. In this study addressing the dearth of research on stress, quality of life (QOL), and gut microbiota in newly diagnosed breast cancer patients, 82 individuals were prospectively observed. Utilizing the Functional Assessment of Chronic Illness Therapy (FACT)-Breast questionnaire to assess health-related quality of life (HRQOL) and the Distress Thermometer (DT) to gauge distress levels, the findings revealed a mean FACT-B score of 104.5, underscoring HRQOL's varied impact. Significantly, 53.7% reported moderate to severe distress, with a mean DT score of 4.43. Further exploration uncovered compelling links between distress levels, FACT-B domains, and microbial composition. Notably, Alcaligenaceae and Sutterella were more abundant in individuals with higher DT scores at the family and genus levels (p = 0.017), while Streptococcaceae at the family level and Streptococcus at the genus level were prevalent in those with lower DT scores (p = 0.028 and p = 0.023, respectively). This study illuminates the intricate interplay of stress, QOL, and gut microbiota in newly diagnosed breast cancer patients, offering valuable insights for potential interventions of biomarker or probiotics aimed at alleviating stress and enhancing QOL in this patient cohort.
Collapse
Affiliation(s)
- Chi-Chan Lee
- Department of General Surgery, MacKay Memorial Hospital, Taipei, Taiwan
| | - Horng-Woei Yang
- Division of Molecular Medicine, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chih-Ju Liu
- Department of Nursing, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Nursing, MacKay Medical College, New Taipei, Taiwan
| | - Fang Lee
- Department of General Surgery, MacKay Memorial Hospital, Taipei, Taiwan
| | - Wen-Ching Ko
- Department of General Surgery, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yuan-Ching Chang
- Department of General Surgery, MacKay Memorial Hospital, Taipei, Taiwan
| | - Po-Sheng Yang
- Department of General Surgery, MacKay Memorial Hospital, Taipei, Taiwan.
- Department of Medicine, MacKay Medical College, New Taipei, Taiwan.
| |
Collapse
|
14
|
Liu RH, Sun AQ, Liao Y, Tang ZX, Zhang SH, Shan X, Hu JT. Lactiplantibacillus plantarum Regulated Intestinal Microbial Community and Cytokines to Inhibit Salmonella typhimurium Infection. Probiotics Antimicrob Proteins 2023; 15:1355-1370. [PMID: 36074298 DOI: 10.1007/s12602-022-09987-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2022] [Indexed: 11/25/2022]
Abstract
Lactic acid bacteria (LAB) are recognized as food-grade safe microorganisms and have many beneficial effects. LAB could maintain the host intestinal homeostasis and regulate intestinal microbial community to exert antibacterial effects. In this study, Lactiplantibacillus plantarum (L. plantarum, Lp01) strain isolated from pig intestine was orally administered to C57BL/6 mice, and mice were then infected with Salmonella typhimurium (ATCC14028). The protective effects of L. plantarum were evaluated by monitoring body weight loss, survival rates, bacterial loads in tissue, colon histopathology analysis, and cytokine secretion. 16S rRNA gene sequencing was also utilized to detect the dynamics of the blind gut microbial community in mice. We found that L. plantarum could significantly reduce the body weight loss and improve the survival rates. The survival rate in the L. P-Sty group was up to 67.5%, which was much higher than that in the STY group (25%). Counting of bacterial loads displayed that the colony-forming unit (CFU) of S. typhimurium in the spleen (p < 0.05) and the liver (p < 0.05) from L. P-Sty group both decreased, compared with STY group. Intestinal histopathology showed that it alleviated the intestinal injury caused by Salmonella, inhibited the secretion of pro-inflammatory cytokines, and promoted anti-inflammatory cytokines (p < 0. 01). In addition, L. plantarum also significantly ameliorated the intestinal gut microbiome disturbance caused by Salmonella. It displayed an obvious increase of beneficial bacteria including Lactobacillus and Bacteroidetes and reduction of pathogenic bacteria like Proteobacteria. In conclusion, L. plantarum could regulate microbial community to inhibit Salmonella typhimurium infection.
Collapse
Affiliation(s)
- Rui-Han Liu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - An-Qi Sun
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Ye Liao
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Zheng-Xu Tang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Shi-Han Zhang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xin Shan
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jing-Tao Hu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China.
| |
Collapse
|
15
|
Jiang Y, Gong W, Xian Z, Xu W, Hu J, Ma Z, Dong H, Lin C, Fu S, Chen X. 16S full-length gene sequencing analysis of intestinal flora in breast cancer patients in Hainan Province. Mol Cell Probes 2023; 71:101927. [PMID: 37595804 DOI: 10.1016/j.mcp.2023.101927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/07/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
Breast cancer has become the number one cancer in the world, and intestinal flora may be closely linked to it. Geographic location also has an important impact on human intestinal flora. We conducted the first study on the intestinal flora of breast cancer patients and non-breast cancer patients in a tropical region - Hainan Province in China. At the same time, Pacbio platform based on third-generation sequencing was used for the first time to conduct 16S full-length sequencing of fecal microorganism DNA. We completed the species diversity analysis and differential species analysis of the intestinal flora between the two groups, inferred their functional genetic composition and performed functional difference analysis. There were statistically significant differences in alpha diversity between the two groups in Hainan Province. By species composition difference analysis, at the phylum level, Bacteroidales (P = 0.006) and Firmicutes (P = 0.002) was differed between the two groups, and at the genus level, 17 breast cancer-related differential species such as Bacteroides were screened. According to the five grouping methods including ER level, PR level, HER2 status, Ki67 index and histological grade of breast cancer patients, 4, 1, 9, 6, 5 differential microbiota were screened out respectively, which were in total 25 (P < 0.05 for all subgroups) . The functional prediction and difference analysis revealed two functional metabolisms with significant differences between the two groups of microbes (P < 0.05). These results suggest that breast cancer is associated with changes in the composition and function of intestinal flora. These microflora and functional differences may become biomarkers or new targets for diagnosis and treatment of breast cancer.
Collapse
Affiliation(s)
- Yonglan Jiang
- Department of Clinical Laboratory, Changsha County People's Hospital (Xingsha Campus of Hunan Provincial People's Hospital), Changsha, Hunan, China
| | - Wei Gong
- Department of Clinical Laboratory, Affiliated Cancer Hospital of Hainan Medical University, Hainan Cancer Hospital, Haikou, Hainan, China; Hainan University, Haikou, Hainan, China
| | - Zhenyong Xian
- Department of Clinical Laboratory, Lingao County Traditional Chinese Medicine Hospital, Lingao, Hainan, China
| | - Weihua Xu
- Department of Clinical Laboratory, Affiliated Cancer Hospital of Hainan Medical University, Hainan Cancer Hospital, Haikou, Hainan, China; Hainan Tropical Tumor Institute, Haikou, Hainan, China; Academician Innovation Platform of Hainan Province, Haikou, Hainan, China
| | - Junjie Hu
- Department of Clinical Laboratory, Affiliated Cancer Hospital of Hainan Medical University, Hainan Cancer Hospital, Haikou, Hainan, China; Academician Innovation Platform of Hainan Province, Haikou, Hainan, China
| | - Zhichao Ma
- Department of Clinical Laboratory, Affiliated Cancer Hospital of Hainan Medical University, Hainan Cancer Hospital, Haikou, Hainan, China; Academician Innovation Platform of Hainan Province, Haikou, Hainan, China
| | - Huaying Dong
- Department of Breast Surgery, Hainan General Hospital, Haikou, Hainan, China
| | - Chong Lin
- Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Shengmiao Fu
- Academician Innovation Platform of Hainan Province, Haikou, Hainan, China.
| | - Xinping Chen
- Department of Clinical Laboratory, Affiliated Cancer Hospital of Hainan Medical University, Hainan Cancer Hospital, Haikou, Hainan, China; Hainan Tropical Tumor Institute, Haikou, Hainan, China; Hainan University, Haikou, Hainan, China; Academician Innovation Platform of Hainan Province, Haikou, Hainan, China.
| |
Collapse
|
16
|
Pajewska M, Partyka O, Czerw A, Deptała A, Cipora E, Gąska I, Wojtaszek M, Sygit K, Sygit M, Krzych-Fałta E, Schneider-Matyka D, Cybulska AM, Grochans E, Asendrych-Woźniak A, Romanowicz A, Drobnik J, Bandurska E, Ciećko W, Maciuszek-Bartkowska B, Curyło M, Wróbel K, Kozłowski R, Marczak M. Management of Metastatic Pancreatic Cancer-Comparison of Global Guidelines over the Last 5 Years. Cancers (Basel) 2023; 15:4400. [PMID: 37686675 PMCID: PMC10486352 DOI: 10.3390/cancers15174400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Pancreatic cancer (PC) is usually diagnosed at an advanced stage of its development, which results in lower overall survival (OS). Prognosis is also poor even with curative-intent surgery. Approximately 80% of patients with localized PDAC have micrometastases at the time of diagnosis, which leads to a worse prognosis than in other cancers. The objective of this study is to present the progress in the treatment of metastatic pancreatic cancer based on the recommendations of oncological scientific societies, such as ESMO, NCCN, ASCO, NICE and SEOM, over the last 5 years. Combined FOLFIRINOX therapy is mostly a recommended therapy among patients with good performance statuses, while gemcitabine is recommended for more fragile patients as a first-line treatment. The newest guidelines suggest that molecular profiling of the tumor should be the first step in determining the course of treatment. The use of modern molecular therapies in patients with specific gene mutations should extend the survival of patients with this disease.
Collapse
Affiliation(s)
- Monika Pajewska
- Department of Health Economics and Medical Law, Medical University of Warsaw, 01-445 Warsaw, Poland; (M.P.)
- Department of Economic and System Analyses, National Institute of Public Health NIH-National Research Institute, 00-791 Warsaw, Poland
| | - Olga Partyka
- Department of Health Economics and Medical Law, Medical University of Warsaw, 01-445 Warsaw, Poland; (M.P.)
- Department of Economic and System Analyses, National Institute of Public Health NIH-National Research Institute, 00-791 Warsaw, Poland
| | - Aleksandra Czerw
- Department of Health Economics and Medical Law, Medical University of Warsaw, 01-445 Warsaw, Poland; (M.P.)
- Department of Economic and System Analyses, National Institute of Public Health NIH-National Research Institute, 00-791 Warsaw, Poland
| | - Andrzej Deptała
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Elżbieta Cipora
- Medical Institute, Jan Grodek State University in Sanok, 38-500 Sanok, Poland
| | - Izabela Gąska
- Medical Institute, Jan Grodek State University in Sanok, 38-500 Sanok, Poland
| | - Marek Wojtaszek
- Medical Institute, Jan Grodek State University in Sanok, 38-500 Sanok, Poland
| | - Katarzyna Sygit
- Faculty of Health Sciences, Calisia University, 62-800 Kalisz, Poland
| | - Marian Sygit
- Faculty of Health Sciences, Calisia University, 62-800 Kalisz, Poland
| | - Edyta Krzych-Fałta
- Department of Basic of Nursing, Faculty of Health Sciences, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Daria Schneider-Matyka
- Department of Nursing, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 71-210 Szczecin, Poland
| | - Anna M. Cybulska
- Department of Nursing, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 71-210 Szczecin, Poland
| | - Elżbieta Grochans
- Department of Nursing, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 71-210 Szczecin, Poland
| | - Alicja Asendrych-Woźniak
- Clinical Department of Oncology, The National Institute of Medicine of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| | - Agnieszka Romanowicz
- Clinical Department of Oncology, The National Institute of Medicine of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| | - Jarosław Drobnik
- Department of Family Medicine, Faculty of Medicine, Wroclaw Medical University, 51-141 Wroclaw, Poland
| | - Ewa Bandurska
- Center for Competence Development, Integrated Care and e-Health, Medical University of Gdansk, 80-204 Gdansk, Poland
| | - Weronika Ciećko
- Center for Competence Development, Integrated Care and e-Health, Medical University of Gdansk, 80-204 Gdansk, Poland
| | | | - Mateusz Curyło
- Department of Internal Medicine, Rehabilitation and Physical Medicine, Medical University of Lodz, 90-647 Lodz, Poland
- Medical Rehabilitation Department, The Ministry of the Interior and Administration Hospital, 30-053 Cracow, Poland
| | - Kacper Wróbel
- Department of Management and Logistics in Healthcare, Medical University of Lodz, 90-131 Lodz, Poland
| | - Remigiusz Kozłowski
- Center for Security Technologies in Logistics, Faculty of Management, University of Lodz, 90-237 Lodz, Poland
| | - Michał Marczak
- Collegium of Management, WSB Merito University in Warsaw, 03-204 Warszawa, Poland
| |
Collapse
|
17
|
Zhao W, Shen X, Hua Q, Yang L, Zhou R, Zhou C, Xu P. Red cell distribution width-a potential prognostic indicator for colorectal cancer patients after radical resection in China. J Gastrointest Oncol 2023; 14:1746-1758. [PMID: 37720452 PMCID: PMC10502564 DOI: 10.21037/jgo-23-54] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 06/26/2023] [Indexed: 09/19/2023] Open
Abstract
Background Red cell distribution width (RDW) can signal poor prognosis in inflammatory medical conditions. The purpose of the study was to investigate the relationship between preoperative RDW and colorectal cancer (CRC) in a large cohort of patients. Methods A total of 6,224 CRC patients who underwent radical resection at the Fudan University Shanghai Cancer Center were evaluated retrospectively. The prognostic significance of RDW for overall survival (OS) and disease-free survival (DFS) was analyzed using Cox proportional hazards models and Kaplan-Meier method. Propensity score matching (PSM) was used based on survival confounding factors. Results The mean age of the study participants was 59.5±12.0 years and the study cohort was 44% female. The overall median and mean RDW values were 13.3% and 14.0%, respectively. Patients were stratified into three groups based on their RDW value (≤13.3%, 13.4-14.0%, and >14.0%). OS and DFS were shown to significantly deteriorate with increasing RDW category. In the PSM population, OS and DFS were significantly lower in the high RDW group compared with matched controls. However, the differences vanished in the comparisons between the middle RDW group and the control group. Conclusions Our findings demonstrate that preoperative RDW may represent a simple and powerful prognostic factor for CRC patients after radical resection. Integrating RDW into clinical practice may better inform the prognosis and optimize therapeutic approaches for patients with CRC.
Collapse
Affiliation(s)
- Weiwei Zhao
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xuefang Shen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qing Hua
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Liu Yang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ru Zhou
- Department of General Surgery, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changming Zhou
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Cancer Prevention, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Pingbo Xu
- Department of Anesthesiology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| |
Collapse
|
18
|
Zhang Z, Shan J, Shi B, Dong B, Wu Q, Zhang Z. SeNPs alleviates BDE-209-induced intestinal damage by affecting necroptosis, inflammation, intestinal barrier and intestinal flora in layer chickens. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 262:115336. [PMID: 37567103 DOI: 10.1016/j.ecoenv.2023.115336] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/30/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
As environmental pollutants, polybrominated diphenyl ethers (PBDEs) can have toxic effects on living organisms and has a bioaccumulative effect. Low doses of selenium nanoparticles (SeNPs) can exert antioxidant, anti-inflammatory and anti-toxin functions on the organism. This experiment evaluated SeNPs' ability to prevent chicken's intestinal damage from decabromodiphenyl ether (BDE-209) exposure. Sixty layer chickens were separated into four groups at randomly and equally: Control group, SeNPs group (1 mg/kg SeNPs), BDE-209 group (400 mg/kg BDE-209), and BDE-209 +SeNPs group (400 mg/kg BDE-209 and 1 mg/kg SeNPs), for 42 days. The results showed that BDE-209 increased MDA content, decreased the activities of T-SOD, T-AOC, GSH and iNOS, up-regulated the expression of TNF-α, RIPK1, RIPK3 and MLKL, promoted the production of inflammatory factors, reduced the levels of tight junction proteins (Claudin-1, Occludin, ZO-1). SeNPs attenuated intestinal oxidative stress, necroptosis, inflammation and intestinal barrier damage caused by BDE-209. This protective effect is associated with the MAPK/NF-κB signaling pathway. Moreover, SeNPs restores flora alpha and beta diversity, improves intestinal flora composition and its abundance. It shifts the dysbiosis of intestinal flora caused by BDE-209 to normal. Overall, SeNPs can alleviate BDE-209-induced intestinal barrier damage and intestinal flora disorders, which are associated with intestinal oxidative stress, necroptosis and inflammation.
Collapse
Affiliation(s)
- Zhuoqi Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jianhua Shan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Bendong Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Bowen Dong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Qiong Wu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 100096, PR China.
| | - Ziwei Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, PR China.
| |
Collapse
|
19
|
Garg A, Ellis LB, Love RL, Grewal K, Bowden S, Bennett PR, Kyrgiou M. Vaginal microbiome in obesity and its impact on reproduction. Best Pract Res Clin Obstet Gynaecol 2023; 90:102365. [PMID: 37399714 DOI: 10.1016/j.bpobgyn.2023.102365] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/25/2023] [Accepted: 06/04/2023] [Indexed: 07/05/2023]
Abstract
A number of reproductive outcomes have been increasingly found to be affected by the vaginal microbiota. Obesity has become a global epidemic, affecting increasing numbers of reproductive-age women, and has been shown to be a risk factor for a number of adverse female health outcomes. A healthy vaginal microbiome is characterized by Lactobacillus-dominance, in particular Lactobacillus crispatus; obesity has been found to be associated with higher diversity and a lower likelihood of Lactobacillus-dominance. In this review, we summarize the evidence on the vaginal microbiome in obese women and the impact on reproductive outcomes such as conception rates, early pregnancy, and preterm birth. We further explore the mechanisms by which obesity may result in an altered microbial composition and highlight future avenues for therapeutic targeting of the vaginal microbiota.
Collapse
Affiliation(s)
- Akanksha Garg
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital, W12 0NN, London, UK; Imperial College Healthcare NHS Trust, London, UK
| | - Laura Burney Ellis
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital, W12 0NN, London, UK; Imperial College Healthcare NHS Trust, London, UK
| | - Ryan Laurence Love
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital, W12 0NN, London, UK; Imperial College Healthcare NHS Trust, London, UK
| | - Karen Grewal
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital, W12 0NN, London, UK; Imperial College Healthcare NHS Trust, London, UK
| | - Sarah Bowden
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital, W12 0NN, London, UK
| | - Phillip R Bennett
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital, W12 0NN, London, UK
| | - Maria Kyrgiou
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital, W12 0NN, London, UK; Imperial College Healthcare NHS Trust, London, UK.
| |
Collapse
|
20
|
Ruze R, Song J, Yin X, Chen Y, Xu R, Wang C, Zhao Y. Mechanisms of obesity- and diabetes mellitus-related pancreatic carcinogenesis: a comprehensive and systematic review. Signal Transduct Target Ther 2023; 8:139. [PMID: 36964133 PMCID: PMC10039087 DOI: 10.1038/s41392-023-01376-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 01/31/2023] [Accepted: 02/15/2023] [Indexed: 03/26/2023] Open
Abstract
Research on obesity- and diabetes mellitus (DM)-related carcinogenesis has expanded exponentially since these two diseases were recognized as important risk factors for cancers. The growing interest in this area is prominently actuated by the increasing obesity and DM prevalence, which is partially responsible for the slight but constant increase in pancreatic cancer (PC) occurrence. PC is a highly lethal malignancy characterized by its insidious symptoms, delayed diagnosis, and devastating prognosis. The intricate process of obesity and DM promoting pancreatic carcinogenesis involves their local impact on the pancreas and concurrent whole-body systemic changes that are suitable for cancer initiation. The main mechanisms involved in this process include the excessive accumulation of various nutrients and metabolites promoting carcinogenesis directly while also aggravating mutagenic and carcinogenic metabolic disorders by affecting multiple pathways. Detrimental alterations in gastrointestinal and sex hormone levels and microbiome dysfunction further compromise immunometabolic regulation and contribute to the establishment of an immunosuppressive tumor microenvironment (TME) for carcinogenesis, which can be exacerbated by several crucial pathophysiological processes and TME components, such as autophagy, endoplasmic reticulum stress, oxidative stress, epithelial-mesenchymal transition, and exosome secretion. This review provides a comprehensive and critical analysis of the immunometabolic mechanisms of obesity- and DM-related pancreatic carcinogenesis and dissects how metabolic disorders impair anticancer immunity and influence pathophysiological processes to favor cancer initiation.
Collapse
Affiliation(s)
- Rexiati Ruze
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Jianlu Song
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Xinpeng Yin
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Yuan Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Ruiyuan Xu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Chengcheng Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China.
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China.
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China.
| |
Collapse
|
21
|
Candido MF, Medeiros M, Veronez LC, Bastos D, Oliveira KL, Pezuk JA, Valera ET, Brassesco MS. Drugging Hijacked Kinase Pathways in Pediatric Oncology: Opportunities and Current Scenario. Pharmaceutics 2023; 15:pharmaceutics15020664. [PMID: 36839989 PMCID: PMC9966033 DOI: 10.3390/pharmaceutics15020664] [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: 12/15/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Childhood cancer is considered rare, corresponding to ~3% of all malignant neoplasms in the human population. The World Health Organization (WHO) reports a universal occurrence of more than 15 cases per 100,000 inhabitants around the globe, and despite improvements in diagnosis, treatment and supportive care, one child dies of cancer every 3 min. Consequently, more efficient, selective and affordable therapeutics are still needed in order to improve outcomes and avoid long-term sequelae. Alterations in kinases' functionality is a trademark of cancer and the concept of exploiting them as drug targets has burgeoned in academia and in the pharmaceutical industry of the 21st century. Consequently, an increasing plethora of inhibitors has emerged. In the present study, the expression patterns of a selected group of kinases (including tyrosine receptors, members of the PI3K/AKT/mTOR and MAPK pathways, coordinators of cell cycle progression, and chromosome segregation) and their correlation with clinical outcomes in pediatric solid tumors were accessed through the R2: Genomics Analysis and Visualization Platform and by a thorough search of published literature. To further illustrate the importance of kinase dysregulation in the pathophysiology of pediatric cancer, we analyzed the vulnerability of different cancer cell lines against their inhibition through the Cancer Dependency Map portal, and performed a search for kinase-targeted compounds with approval and clinical applicability through the CanSAR knowledgebase. Finally, we provide a detailed literature review of a considerable set of small molecules that mitigate kinase activity under experimental testing and clinical trials for the treatment of pediatric tumors, while discuss critical challenges that must be overcome before translation into clinical options, including the absence of compounds designed specifically for childhood tumors which often show differential mutational burdens, intrinsic and acquired resistance, lack of selectivity and adverse effects on a growing organism.
Collapse
Affiliation(s)
- Marina Ferreira Candido
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Mariana Medeiros
- Regional Blood Center, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Luciana Chain Veronez
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - David Bastos
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| | - Karla Laissa Oliveira
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| | - Julia Alejandra Pezuk
- Departament of Biotechnology and Innovation, Anhanguera University of São Paulo, UNIAN/SP, São Paulo 04119-001, SP, Brazil
| | - Elvis Terci Valera
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - María Sol Brassesco
- Departament of Biotechnology and Innovation, Anhanguera University of São Paulo, UNIAN/SP, São Paulo 04119-001, SP, Brazil
- Correspondence: ; Tel.: +55-16-3315-9144; Fax: +55-16-3315-4886
| |
Collapse
|
22
|
Butyrate limits human natural killer cell effector function. Sci Rep 2023; 13:2715. [PMID: 36792800 PMCID: PMC9932090 DOI: 10.1038/s41598-023-29731-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
The gut microbiota regulates chronic inflammation and has been implicated in the pathogenesis of a broad spectrum of disease including autoimmunity and cancer. Microbial short-chain fatty acids (SCFAs) e.g., butyrate have demonstrated immunomodulatory effects and are thought to be key mediators of the host-microbiome interaction. Here, we investigated the effect of butyrate on effector functions of blood derived human NK cells stimulated for 18 h with a combination of IL-12/IL-15, a potent mix of cytokines that drive NK cell activation. We show that butyrate has a strong anti-inflammatory effect on NK cells. NK cells cultured in the presence of butyrate expressed lower levels of activating receptors (TRAIL, NKp30, NKp44) and produced lower levels of cytokines (IFNγ, TNF-α, IL-22, granzyme B, granzyme A, perforin) in response to IL-12/IL-15. Butyrate restricted NK cell function by downregulation of mTORC1 activity, c-Myc mRNA expression and metabolism. Using a shotgun proteomic approach, we confirmed the effect of butyrate on NK cell cytokine signaling and metabolism and identified BRD2, MAT2A and EHD1 as downstream mediators of these effects. This insight into the immunomodulatory activity of butyrate on human NK cell function might help to develop new ways to limit NK cell function during chronic inflammation.
Collapse
|
23
|
Cassotta M, Cianciosi D, De Giuseppe R, Navarro-Hortal MD, Armas Diaz Y, Forbes-Hernández TY, Pifarre KT, Pascual Barrera AE, Grosso G, Xiao J, Battino M, Giampieri F. Possible role of nutrition in the prevention of inflammatory bowel disease-related colorectal cancer: A focus on human studies. Nutrition 2023; 110:111980. [PMID: 36965240 DOI: 10.1016/j.nut.2023.111980] [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: 05/25/2022] [Revised: 01/10/2023] [Accepted: 01/22/2023] [Indexed: 02/05/2023]
Abstract
Patients with inflammatory bowel disease (IBD) are at substantially high risk for colorectal cancer (CRC). IBD-associated CRC accounts for roughly 10% to 15% of the annual mortality in patients with IBD. IBD-related CRC also affects younger patients compared with sporadic CRC, with a 5-y survival rate of 50%. Regardless of medical therapies, the persistent inflammatory state characterizing IBD raises the risk for precancerous changes and CRC, with additional input from several elements, including genetic and environmental risk factors, IBD-associated comorbidities, intestinal barrier dysfunction, and gut microbiota modifications. It is well known that nutritional habits and dietary bioactive compounds can influence IBD-associated inflammation, microbiome abundance and composition, oxidative stress balance, and gut permeability. Additionally, in recent years, results from broad epidemiologic and experimental studies have associated certain foods or nutritional patterns with the risk for colorectal neoplasia. The present study aimed to review the possible role of nutrition in preventing IBD-related CRC, focusing specifically on human studies. It emerges that nutritional interventions based on healthy, nutrient-dense dietary patterns characterized by a high intake of fiber, vegetables, fruit, ω-3 polyunsaturated fatty acids, and a low amount of animal proteins, processed foods, and alcohol, combined with probiotic supplementation have the potential of reducing IBD-activity and preventing the risk of IBD-related CRC through different mechanisms, suggesting that targeted nutritional interventions may represent a novel promising approach for the prevention and management of IBD-associated CRC.
Collapse
Affiliation(s)
- Manuela Cassotta
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander, Spain
| | - Danila Cianciosi
- Department of Clinical Sciences, Faculty of Medicine, Polytechnic University of Marche, Ancona, Italy
| | - Rachele De Giuseppe
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy; NBFC, National Biodiversity Future Center, Palermo 90133, Italy
| | - Maria Dolores Navarro-Hortal
- Biomedical Research Centre, Institute of Nutrition and Food Technology "José Mataix Verdú," Department of Physiology, Faculty of Pharmacy, University of Granada, Armilla, Granada, Spain
| | - Yasmany Armas Diaz
- Department of Clinical Sciences, Faculty of Medicine, Polytechnic University of Marche, Ancona, Italy
| | - Tamara Yuliett Forbes-Hernández
- Biomedical Research Centre, Institute of Nutrition and Food Technology "José Mataix Verdú," Department of Physiology, Faculty of Pharmacy, University of Granada, Armilla, Granada, Spain
| | - Kilian Tutusaus Pifarre
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander, Spain; Project Department, Universidade Internacional do Cuanza, Cuito, Bié, Angola
| | - Alina Eugenia Pascual Barrera
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander, Spain; Department of Project Management, Universidad Internacional Iberoamericana, Campeche, Mexico
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Jianbo Xiao
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, Universidade de Vigo - Ourense Campus, Ourense, Spain
| | - Maurizio Battino
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander, Spain; Department of Clinical Sciences, Faculty of Medicine, Polytechnic University of Marche, Ancona, Italy; International Joint Research Laboratory of Intelligent Agriculture and Agri-products Processing, Jiangsu University, Zhenjiang, China
| | - Francesca Giampieri
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander, Spain.
| |
Collapse
|
24
|
Mahmood R, Voisin A, Olof H, Khorasaniha R, Lawal SA, Armstrong HK. Host Microbiomes Influence the Effects of Diet on Inflammation and Cancer. Cancers (Basel) 2023; 15:521. [PMID: 36672469 PMCID: PMC9857231 DOI: 10.3390/cancers15020521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Cancer is the second leading cause of death globally, and there is a growing appreciation for the complex involvement of diet, microbiomes, and inflammatory processes culminating in tumorigenesis. Although research has significantly improved our understanding of the various factors involved in different cancers, the underlying mechanisms through which these factors influence tumor cells and their microenvironment remain to be completely understood. In particular, interactions between the different microbiomes, specific dietary factors, and host cells mediate both local and systemic immune responses, thereby influencing inflammation and tumorigenesis. Developing an improved understanding of how different microbiomes, beyond just the colonic microbiome, can interact with dietary factors to influence inflammatory processes and tumorigenesis will support our ability to better understand the potential for microbe-altering and dietary interventions for these patients in future.
Collapse
Affiliation(s)
- Ramsha Mahmood
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Athalia Voisin
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Hana Olof
- Department of Immunology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Reihane Khorasaniha
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Samuel A. Lawal
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Heather K. Armstrong
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Immunology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
25
|
Shrode RL, Knobbe JE, Cady N, Yadav M, Hoang J, Cherwin C, Curry M, Garje R, Vikas P, Sugg S, Phadke S, Filardo E, Mangalam AK. Breast cancer patients from the Midwest region of the United States have reduced levels of short-chain fatty acid-producing gut bacteria. Sci Rep 2023; 13:526. [PMID: 36631533 PMCID: PMC9834383 DOI: 10.1038/s41598-023-27436-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/02/2023] [Indexed: 01/13/2023] Open
Abstract
As geographical location can impact the gut microbiome, it is important to study region-specific microbiome signatures of various diseases. Therefore, we profiled the gut microbiome of breast cancer (BC) patients of the Midwestern region of the United States. The bacterial component of the gut microbiome was profiled utilizing 16S ribosomal RNA sequencing. Additionally, a gene pathway analysis was performed to assess the functional capabilities of the bacterial microbiome. Alpha diversity was not significantly different between BC and healthy controls (HC), however beta diversity revealed distinct clustering between the two groups at the species and genera level. Wilcoxon Rank Sum test revealed modulation of several gut bacteria in BC specifically reduced abundance of those linked with beneficial effects such as Faecalibacterium prausnitzii. Machine learning analysis confirmed the significance of several of the modulated bacteria found by the univariate analysis. The functional analysis showed a decreased abundance of SCFA (propionate) production in BC compared to HC. In conclusion, we observed gut dysbiosis in BC with the depletion of SCFA-producing gut bacteria suggesting their role in the pathobiology of breast cancer. Mechanistic understanding of gut bacterial dysbiosis in breast cancer could lead to refined prevention and treatment.
Collapse
Affiliation(s)
- Rachel L. Shrode
- grid.214572.70000 0004 1936 8294Department of Informatics, University of Iowa, Iowa City, IA 52242 USA
| | - Jessica E. Knobbe
- grid.214572.70000 0004 1936 8294Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242 USA ,grid.214572.70000 0004 1936 8294Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA 52242 USA ,grid.214572.70000 0004 1936 8294Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Nicole Cady
- grid.214572.70000 0004 1936 8294Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA ,grid.214458.e0000000086837370Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109 USA
| | - Meeta Yadav
- grid.214572.70000 0004 1936 8294Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA ,grid.214572.70000 0004 1936 8294College of Dentistry, University of Iowa, Iowa City, IA 52242 USA
| | - Jemmie Hoang
- grid.214572.70000 0004 1936 8294College of Nursing, University of Iowa, Iowa City, IA 52242 USA
| | - Catherine Cherwin
- grid.214572.70000 0004 1936 8294College of Nursing, University of Iowa, Iowa City, IA 52242 USA
| | - Melissa Curry
- grid.412584.e0000 0004 0434 9816Holden Comprehensive Cancer Center, University of Iowa Hospital and Clinics, Iowa City, IA 52242 USA
| | - Rohan Garje
- grid.214572.70000 0004 1936 8294Department of Internal Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Praveen Vikas
- grid.214572.70000 0004 1936 8294Department of Internal Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Sonia Sugg
- grid.214572.70000 0004 1936 8294Department of Surgery, University of Iowa, Iowa City, IA 52242 USA
| | - Sneha Phadke
- grid.214572.70000 0004 1936 8294Division of Hematology, Oncology, and Blood and Marrow Transplantation, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242 USA
| | | | - Ashutosh K. Mangalam
- grid.214572.70000 0004 1936 8294Department of Informatics, University of Iowa, Iowa City, IA 52242 USA ,grid.214572.70000 0004 1936 8294Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242 USA ,grid.214572.70000 0004 1936 8294Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA ,grid.214572.70000 0004 1936 8294College of Dentistry, University of Iowa, Iowa City, IA 52242 USA ,grid.214572.70000 0004 1936 8294University of Iowa, 25 S Grand Ave, 1080-ML, Iowa City, IA 52246 USA
| |
Collapse
|
26
|
Van Pee T, Hogervorst J, Dockx Y, Witters K, Thijs S, Wang C, Bongaerts E, Van Hamme JD, Vangronsveld J, Ameloot M, Raes J, Nawrot TS. Accumulation of Black Carbon Particles in Placenta, Cord Blood, and Childhood Urine in Association with the Intestinal Microbiome Diversity and Composition in Four- to Six-Year-Old Children in the ENVIR ONAGE Birth Cohort. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:17010. [PMID: 36719212 PMCID: PMC9888258 DOI: 10.1289/ehp11257] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
BACKGROUND The gut microbiome plays an essential role in human health. Despite the link between air pollution exposure and various diseases, its association with the gut microbiome during susceptible life periods remains scarce. OBJECTIVES In this study, we examined the association between black carbon particles quantified in prenatal and postnatal biological matrices and bacterial richness and diversity measures, and bacterial families. METHODS A total of 85 stool samples were collected from 4- to 6-y-old children enrolled in the ENVIRonmental influence ON early AGEing birth cohort. We performed 16S rRNA gene sequencing to calculate bacterial richness and diversity indices (Chao1 richness, Shannon diversity, and Simpson diversity) and the relative abundance of bacterial families. Black carbon particles were quantified via white light generation under femtosecond pulsed laser illumination in placental tissue and cord blood, employed as prenatal exposure biomarkers, and in urine, used as a post-natal exposure biomarker. We used robust multivariable-adjusted linear models to examine the associations between quantified black carbon loads and measures of richness (Chao1 index) and diversity (Shannon and Simpson indices), adjusting for parity, season of delivery, sequencing batch, age, sex, weight and height of the child, and maternal education. Additionally, we performed a differential relative abundance analysis of bacterial families with a correction for sampling fraction bias. Results are expressed as percentage difference for a doubling in black carbon loads with 95% confidence interval (CI). RESULTS Two diversity indices were negatively associated with placental black carbon [Shannon: -4.38% (95% CI: -8.31%, -0.28%); Simpson: -0.90% (95% CI: -1.76%, -0.04%)], cord blood black carbon [Shannon: -3.38% (95% CI: -5.66%, -0.84%); Simpson: -0.91 (95% CI: -1.66%, -0.16%)], and urinary black carbon [Shannon: -3.39% (95% CI: -5.77%, -0.94%); Simpson: -0.89% (95% CI: -1.37%, -0.40%)]. The explained variance of black carbon on the above indices varied from 6.1% to 16.6%. No statistically significant associations were found between black carbon load and the Chao1 richness index. After multiple testing correction, placental black carbon was negatively associated with relative abundance of the bacterial families Defluviitaleaceae and Marinifilaceae, and urinary black carbon with Christensenellaceae and Coriobacteriaceae; associations with cord blood black carbon were not statistically significant after correction. CONCLUSION Black carbon particles quantified in prenatal and postnatal biological matrices were associated with the composition and diversity of the childhood intestinal microbiome. These findings address the influential role of exposure to air pollution during pregnancy and early life in human health. https://doi.org/10.1289/EHP11257.
Collapse
Affiliation(s)
- Thessa Van Pee
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Janneke Hogervorst
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Yinthe Dockx
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Katrien Witters
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Sofie Thijs
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Congrong Wang
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Eva Bongaerts
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Jonathan D Van Hamme
- Department of Biological Sciences, Thompson Rivers University, Kamloops, British Columbia, Canada
| | - Jaco Vangronsveld
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
- Department of Plant Physiology and Biophysics, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Lublin, Poland
| | - Marcel Ameloot
- Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Jeroen Raes
- Department of Microbiology and Immunology, Rega Instituut, KU Leuven-University of Leuven, Leuven, Belgium
- Center for Microbiology, VIB, Leuven, Belgium
| | - Tim S Nawrot
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
- Department of Public Health and Primary Care, Leuven University, Leuven, Belgium
| |
Collapse
|
27
|
Khorasaniha R, Olof H, Voisin A, Armstrong K, Wine E, Vasanthan T, Armstrong H. Diversity of fibers in common foods: Key to advancing dietary research. Food Hydrocoll 2023. [DOI: 10.1016/j.foodhyd.2023.108495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
28
|
Elkafas H, Walls M, Al-Hendy A, Ismail N. Gut and genital tract microbiomes: Dysbiosis and link to gynecological disorders. Front Cell Infect Microbiol 2022; 12:1059825. [PMID: 36590579 PMCID: PMC9800796 DOI: 10.3389/fcimb.2022.1059825] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
Every year, millions of women are affected by genital tract disorders, such as bacterial vaginosis (BV), endometrial cancer, polycystic ovary syndrome (PCOS), endometriosis, and uterine fibroids (UFs). These disorders pose a significant economic burden on healthcare systems and have serious implications for health and fertility outcomes. This review explores the relationships between gut, vaginal, and uterine dysbiosis and the pathogenesis of various diseases of the female genital tract. In recent years, reproductive health clinicians and scientists have focused on the microbiome to investigate its role in the pathogenesis and prevention of such diseases. Recent studies of the gut, vaginal, and uterine microbiomes have identified patterns in bacterial composition and changes across individuals' lives associated with specific healthy and diseased states, particularly regarding the effects of the estrogen-gut microbiome axis on estrogen-driven disorders (such as endometrial cancer, endometriosis, and UFs) and disorders associated with estrogen deficiency (such as PCOS). Furthermore, this review discusses the contribution of vitamin D deficiency to gut dysbiosis and altered estrogen metabolism as well as how these changes play key roles in the pathogenesis of UFs. More research on the microbiome influences on reproductive health and fertility is vital.
Collapse
Affiliation(s)
- Hoda Elkafas
- Department of Pharmacology and Toxicology, Egyptian Drug Authority [EDA; formerly The National Organization for Drug Control and Research (NODCAR)], Cairo, Egypt
| | - Melinique Walls
- Pritzker School of Medicine, University of Chicago, Chicago, IL, United States
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, United States
| | - Nahed Ismail
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
29
|
Devoy C, Flores Bueso Y, Tangney M. Understanding and harnessing triple-negative breast cancer-related microbiota in oncology. Front Oncol 2022; 12:1020121. [PMID: 36505861 PMCID: PMC9730816 DOI: 10.3389/fonc.2022.1020121] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/31/2022] [Indexed: 11/27/2022] Open
Abstract
Bacterial inhabitants of the body have the potential to play a role in various stages of cancer initiation, progression, and treatment. These bacteria may be distal to the primary tumour, such as gut microbiota, or local to the tissue, before or after tumour growth. Breast cancer is well studied in this context. Amongst breast cancer types, Triple Negative Breast Cancer (TNBC) is more aggressive, has fewer treatment options than receptor-positive breast cancers, has an overall worse prognosis and higher rates of reoccurrence. Thus, an in-depth understanding of the bacterial influence on TNBC progression and treatment is of high value. In this regard, the Gut Microbiota (GM) can be involved in various stages of tumour progression. It may suppress or promote carcinogenesis through the release of carcinogenic metabolites, sustenance of proinflammatory environments and/or the promotion of epigenetic changes in our genome. It can also mediate metastasis and reoccurrence through interactions with the immune system and has been recently shown to influence chemo-, radio-, and immune-therapies. Furthermore, bacteria have also been found to reside in normal and malignant breast tissue. Several studies have now described the breast and breast tumour microbiome, with the tumour microbiota of TNBC having the least taxonomic diversity among all breast cancer types. Here, specific conditions of the tumour microenvironment (TME) - low O2, leaky vasculature and immune suppression - are supportive of tumour selective bacterial growth. This innate bacterial ability could enable their use as delivery agents for various therapeutics or as diagnostics. This review aims to examine the current knowledge on bacterial relevance to TNBC and potential uses while examining some of the remaining unanswered questions regarding mechanisms underpinning observed effects.
Collapse
Affiliation(s)
- Ciaran Devoy
- Cancer Research@UCC, College of Medicine and Health, University College Cork, Cork, Ireland,SynBio Center, University College Cork, Cork, Ireland,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Yensi Flores Bueso
- Cancer Research@UCC, College of Medicine and Health, University College Cork, Cork, Ireland,SynBio Center, University College Cork, Cork, Ireland,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Mark Tangney
- Cancer Research@UCC, College of Medicine and Health, University College Cork, Cork, Ireland,SynBio Center, University College Cork, Cork, Ireland,APC Microbiome Ireland, University College Cork, Cork, Ireland,School of Pharmacy, College of Medicine and Health, University College Cork, Cork, Ireland,*Correspondence: Mark Tangney,
| |
Collapse
|
30
|
Hinshaw DC, Swain CA, Chen D, Hanna A, Molina PA, Maynard CL, Lee G, McFarland BC, Samant RS, Shevde LA. Hedgehog blockade remodels the gut microbiota and the intestinal effector CD8 + T cells in a mouse model of mammary carcinoma. J Transl Med 2022; 102:1236-1244. [PMID: 36775449 DOI: 10.1038/s41374-022-00828-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/01/2022] [Accepted: 07/06/2022] [Indexed: 01/07/2023] Open
Abstract
Given the gut microbiome's rise as a potential frontier in cancer pathogenesis and therapy, leveraging microbial analyses in the study of breast tumor progression and treatment could unveil novel interactions between commensal bacteria and disease outcomes. In breast cancer, the Hedgehog (Hh) signaling pathway is a potential target for treatment due to its aberrant activation leading to poorer prognoses and drug resistance. There are limited studies that have investigated the influences of orally administered cancer therapeutics, such as Vismodegib (a pharmacological, clinically used Hh inhibitor) on the gut microbiota. Using a 4T1 mammary carcinoma mouse model and 16 S rRNA sequencing, we longitudinally mapped alterations in immunomodulating gut microbes during mammary tumor development. Next, we identified changes in the abundance of commensal microbiota in response to Vismodegib treatment of 4T1 mammary tumor-bearing mice. In addition to remodeling gut microbiota, Vismodegib treatment elicited an increase in proliferative CD8+ T cells in the colonic immune network, without any remarkable gastrointestinal-associated side effects. To our knowledge, this is the first study to assess longitudinal changes in the gut microbiome during mammary tumor development and progression. Our study also pioneers an investigation of the dynamic effects of an orally delivered Hh inhibitor on the gut microbiome and the gut-associated immune-regulatory adaptive effector CD8+ T cells. These findings inform future comprehensive studies on the consortium of altered microbes that can impact potential systemic immunomodulatory roles of Vismodegib.
Collapse
Affiliation(s)
- Dominique C Hinshaw
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Courtney A Swain
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dongquan Chen
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Division of Preventive Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.,Center for Clinical and Translational Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ann Hanna
- Vanderbilt Center for Immunobiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Patrick A Molina
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Craig L Maynard
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Goo Lee
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Braden C McFarland
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rajeev S Samant
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Birmingham VA Medical Center, Birmingham, AL, USA
| | - Lalita A Shevde
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA. .,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
31
|
Zaidi AH, Pratama MY, Omstead AN, Gorbonova A, Mansoor R, Melton-Kreft R, Jobe BA, Wagner PL, Kelly RJ, Goel A. A blood-based circulating microbial metagenomic panel for early diagnosis and prognosis of oesophageal adenocarcinoma. Br J Cancer 2022; 127:2016-2024. [PMID: 36097175 PMCID: PMC9681745 DOI: 10.1038/s41416-022-01974-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Emerging evidence indicates the potential clinical significance of specific microbial signatures as diagnostic and prognostic biomarkers, in multiple cancers. However, to date, no studies have systematically interrogated circulating metagenome profiling in oesophageal adenocarcinoma (EAC) patients, particularly as novel non-invasive, early detection, surveillance and prognostic classifiers. METHODS Metagenome sequencing was performed on 81 serum specimens collected across EAC spectrum, with sequencing reads classified using Bracken and MetaPhlAn3. Followed by the Linear Discriminant Analysis effect size (LEfSe) method to identify microbial profiles between groups. Logistic regression and Kaplan-Meier analyses were used to build classifiers. RESULTS A significant loss of alpha and beta diversity was identified in serum specimens from EAC patients. We observed a shift in microbial taxa between each group-at the phylum, genus, and species level-with Lactobacillus sakei as the most prominent species in gastroesophageal reflux (GERD) vs other patient groups. Interestingly, LEfSe analysis identified a complete loss of Lactobacillus (L. Sakei and L. Curvatus), Collinsella stercoris and Bacteroides stercoris but conversely a significant increase in Escherichia coli in patients with EAC. Finally, we developed a metagenome panel that discriminated EAC from GERD patients with an AUC value of 0.89 (95% CI: 0.78-0.95; P < 0.001) and this panel in conjunction with the TNM stage was a robust predictor of overall survival (≥24 months; AUC = 0.84 (95% CI: 0.66-0.92; P = 0.006)). CONCLUSION This study firstly describes unique blood-based microbial profiles in patients across EAC carcinogenesis, that are further utilised to establish a novel circulating diagnostic and prognostic metagenomic signature for EAC. TRANSLATIONAL RELEVANCE Accumulating data indicates the clinical relevance of specific microbial signatures as diagnostic and prognostic biomarkers, in multiple cancers. However, to date, no studies have systematically interrogated circulating metagenome profiling in patients with oesophageal adenocarcinoma (EAC). Herein, we performed metagenome sequencing in serum specimens from EAC patients 81 collected across EAC spectrum and observed a significant loss of alpha and beta diversity, with a shift in microbial taxa between each group-at the phylum, genus, and species level-with Lactobacillus sakei as the most prominent species in gastroesophageal reflux (GERD) vs other patient groups. Interestingly, LEfSe analysis identified a complete loss of Lactobacillus (L. Sakei and L. Curvatus), Collinsella stercoris and Bacteroides stercoris but conversely a significant increase in Escherichia coli in patients with EAC. Finally, we developed a metagenome panel that discriminated EAC from GERD patients with an AUC value of 0.89 and this panel, in conjunction with the TNM stage, was a robust predictor of overall survival. This study for the first time describes unique blood-based microbial profiles in patients across EAC carcinogenesis, that are further utilised to establish a novel circulating diagnostic and prognostic metagenomic signature for EAC.
Collapse
Affiliation(s)
- Ali H Zaidi
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Muhammad Yogi Pratama
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Monrovia, CA, USA
| | - Ashten N Omstead
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Anastasia Gorbonova
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Rubab Mansoor
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Rachael Melton-Kreft
- The Allegheny Health Network, Center of Excellence in Biofilm Research, Pittsburgh, PA, USA
| | - Blair A Jobe
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Patrick L Wagner
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Ronan J Kelly
- The Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Monrovia, CA, USA.
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| |
Collapse
|
32
|
Hou Y, Zhao W, Yang Z, Zhang B. Serum amyloid A (SAA) and Interleukin-6 (IL-6) as the potential biomarkers for gastric cancer. Medicine (Baltimore) 2022; 101:e31514. [PMID: 36316846 PMCID: PMC9622617 DOI: 10.1097/md.0000000000031514] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
To explore serum amyloid A (SAA) and interleukin-6 (IL-6) as potential diagnostic biomarkers for gastric cancer (GCa) and the application value of the combined diagnosis of SAA, IL6, and Cancer embryonic antigen. Serum samples were collected before the initial surgery from 159 patients comprising samples from 122 patients with GCa and 37 patients with benign gastric disease. All patients were hospitalized at Beijing Aerospace General Hospital in China between 2018 and 2020. The IL-6 and SAA levels were assessed using standard laboratory protocols. The levels of SAA and IL-6 were significantly higher in patients with GCa than in controls. Compared with the healthy group, the concentration of SAA and IL-6 in FIGO III-IV group were significantly higher and the difference were statistically significant. In addition, significant differences were observed between the FIGO III-IV group and FIGO I-II groups. The Receiver operating characteristic (ROC) curve for the combined detection of SAA, IL-6, and Cancer embryonic antigen showed an area under the curve (AUC) of 0.948, sensitivity of 91.0%, and specificity of 89.2%. Spearman's correlation analysis indicated obvious correlations among the levels of serum SAA, IL-6, advanced FIGO stage, lymphatic invasion, and distant metastasis. AA and IL-6 may serve as useful biomarkers for poor prognosis of GCa. Clinical diagnosis combined with SAA and IL-6 may help assess therapeutic outcomes.
Collapse
Affiliation(s)
- Yongwang Hou
- Clinical Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou City, Hebei Province, China
- * Correspondence: Yongwang Hou, Clinical Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou City, Hebei Province, China (e-mail: )
| | - Weidong Zhao
- Beijing Aerospace General Hospital, Beijing, China
| | - Zhicong Yang
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou City, Hebei Province, China
| | - Bin Zhang
- Clinical Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou City, Hebei Province, China
| |
Collapse
|
33
|
Boulaka A, Mantellou P, Stanc GM, Souka E, Valavanis C, Saxami G, Mitsou E, Koutrotsios G, Zervakis GI, Kyriacou A, Pletsa V, Georgiadis P. Genoprotective activity of the Pleurotus eryngii mushrooms following their in vitro and in vivo fermentation by fecal microbiota. Front Nutr 2022; 9:988517. [PMID: 36082029 PMCID: PMC9445615 DOI: 10.3389/fnut.2022.988517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/08/2022] [Indexed: 11/19/2022] Open
Abstract
Pleurotus eryngii mushrooms are commercially cultivated and widely consumed due to their organoleptic properties, and the low caloric and high nutritional value. In addition, they contain various biologically active and health-promoting compounds; very recently, their genoprotective effect in Caco-2 cells after their fermentation by the human fecal microbiota was also documented. In the current study, the effect of P. eryngii pre- and post-fermentation supernatants in micronuclei formation was evaluated in human lymphocytes. In addition, the genoprotective properties of increasing concentrations of aqueous extracts from P. eryngii mushrooms (150, 300, 600 mg/kg) against the cyclophosphamide-induced DNA damage were studied in young and elderly female and male mice in bone marrow and whole blood cells. The ability of the highest dose (600 mg/kg) to regulate the main cellular signaling pathways was also evaluated in gut and liver tissues of female animals by quantifying the mRNA expression of NrF2, Nfkβ, DNMT1, and IL-22 genes. P. eryngii post-fermentation, but not pre-fermentation, supernatants were able to protect human lymphocytes from the mitomycin C-induced DNA damage in a dose-dependent manner. Similarly, genoprotection was also observed in bone marrow cells of mice treated by gavage with P. eryngii extract. The effect was observed in all the experimental groups of mice (young and elderly, male and female) and was more potent in young female mice. Overexpression of all genes examined was observed in both tissues, mainly among the elderly animals. In conclusion, P. eryngii mushrooms were shown to maintain genome integrity through protecting cells from genotoxic insults. These beneficial effects can be attributed to their antioxidant and immunomodulatory properties, as well as their ability to regulate the cell's epigenetic mechanisms and maintain cell homeostasis.
Collapse
Affiliation(s)
- Athina Boulaka
- Laboratory of Environment and Health, Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Panagiota Mantellou
- Laboratory of Environment and Health, Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
| | - Gabriela-Monica Stanc
- Department of Pathology, Molecular Pathology Unit, Metaxa Cancer Hospital, Piraeus, Greece
| | - Efthymia Souka
- Department of Pathology, Molecular Pathology Unit, Metaxa Cancer Hospital, Piraeus, Greece
| | - Christoς Valavanis
- Department of Pathology, Molecular Pathology Unit, Metaxa Cancer Hospital, Piraeus, Greece
| | - Georgia Saxami
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - Evdokia Mitsou
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - Georgios Koutrotsios
- Laboratory of General and Agricultural Microbiology, Agricultural University of Athens, Athens, Greece
| | - Georgios I. Zervakis
- Laboratory of General and Agricultural Microbiology, Agricultural University of Athens, Athens, Greece
| | | | - Vasiliki Pletsa
- Laboratory of Environment and Health, Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Panagiotis Georgiadis
- Laboratory of Environment and Health, Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| |
Collapse
|
34
|
Circulating and non-circulating proteins and nucleic acids as biomarkers and therapeutic molecules in ovarian cancer. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
35
|
Niemiec MJ, Kapitan M, Himmel M, Döll K, Krüger T, Köllner TG, Auge I, Kage F, Alteri CJ, Mobley HL, Monsen T, Linde S, Nietzsche S, Kniemeyer O, Brakhage AA, Jacobsen ID. Augmented Enterocyte Damage During Candida albicans and Proteus mirabilis Coinfection. Front Cell Infect Microbiol 2022; 12:866416. [PMID: 35651758 PMCID: PMC9149288 DOI: 10.3389/fcimb.2022.866416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/15/2022] [Indexed: 12/24/2022] Open
Abstract
The human gut acts as the main reservoir of microbes and a relevant source of life-threatening infections, especially in immunocompromised patients. There, the opportunistic fungal pathogen Candida albicans adapts to the host environment and additionally interacts with residing bacteria. We investigated fungal-bacterial interactions by coinfecting enterocytes with the yeast Candida albicans and the Gram-negative bacterium Proteus mirabilis resulting in enhanced host cell damage. This synergistic effect was conserved across different P. mirabilis isolates and occurred also with non-albicans Candida species and C. albicans mutants defective in filamentation or candidalysin production. Using bacterial deletion mutants, we identified the P. mirabilis hemolysin HpmA to be the key effector for host cell destruction. Spatially separated coinfections demonstrated that synergism between Candida and Proteus is induced by contact, but also by soluble factors. Specifically, we identified Candida-mediated glucose consumption and farnesol production as potential triggers for Proteus virulence. In summary, our study demonstrates that coinfection of enterocytes with C. albicans and P. mirabilis can result in increased host cell damage which is mediated by bacterial virulence factors as a result of fungal niche modification via nutrient consumption and production of soluble factors. This supports the notion that certain fungal-bacterial combinations have the potential to result in enhanced virulence in niches such as the gut and might therefore promote translocation and dissemination.
Collapse
Affiliation(s)
- Maria Joanna Niemiec
- Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
- Center for Sepsis Control and Care, Jena, Germany
| | - Mario Kapitan
- Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
- Center for Sepsis Control and Care, Jena, Germany
| | - Maximilian Himmel
- Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Kristina Döll
- Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Thomas Krüger
- Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Tobias G. Köllner
- Department of Biochemistry, Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Isabel Auge
- Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Franziska Kage
- Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Christopher J. Alteri
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, MI, United States
| | - Harry L.T. Mobley
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Tor Monsen
- Department Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Susanne Linde
- Center for Electron Microscopy, University Hospital, Jena, Germany
| | - Sandor Nietzsche
- Center for Electron Microscopy, University Hospital, Jena, Germany
| | - Olaf Kniemeyer
- Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Axel A. Brakhage
- Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| | - Ilse D. Jacobsen
- Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
- Center for Sepsis Control and Care, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
- *Correspondence: Ilse D. Jacobsen,
| |
Collapse
|
36
|
Periodontitis, Metabolic and Gastrointestinal Tract Diseases: Current Perspectives on Possible Pathogenic Connections. J Pers Med 2022; 12:jpm12030341. [PMID: 35330341 PMCID: PMC8955434 DOI: 10.3390/jpm12030341] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 12/24/2022] Open
Abstract
Comprehensive research conducted over the past decades has shown that there is a definite connection between periodontal and systemic conditions, leading to the development and consolidation of the “periodontal medicine” concept. The 2018 classification of periodontal conditions uses this concept as a key element of the precise diagnosis of and individualized therapeutical protocols for periodontitis patients. The topic of this review is the pathogenic connections that exist between periodontal disease and metabolic/digestive tract conditions. It is important to remember that the oral cavity is a key element of the digestive tract and that any conditions affecting its integrity and function (such as periodontitis or oral cancer) can have a significant impact on the metabolic and gastrointestinal status of a patient. Thus, significant diseases with links to metabolic or digestive disruptions were chosen for inclusion in the review, such as diabetes mellitus, hepatic conditions and gastric cancers. Periodontal pathogenic mechanisms share several significant elements with these conditions, including mutual pro-inflammatory mediators, bacterial elements and genetic predisposition. Consequently, periodontal screening should be recommended for affected patients, and conversely, periodontitis patients should be considered for careful monitoring of their metabolic and digestive status.
Collapse
|
37
|
A Comprehensive Review of the Current and Future Role of the Microbiome in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14041020. [PMID: 35205769 PMCID: PMC8870349 DOI: 10.3390/cancers14041020] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary This review summarizes the current literature related to the microbiome and pancreatic ductal adenocarcinoma (PDAC). The aim of this review is to explore the current role of the microbiome in the disease process, screening/diagnostics and to postulate the future role with regards to therapeutic strategies including chemotherapy, immunotherapy and surgery. We further explore the future of microbiome modulation (faecal microbiome transplants, bacterial consortiums, anti-microbials and probiotics), their applications and how we can improve the future of microbiome modulation in a bid to improve PDAC outcomes. Abstract Pancreatic ductal adenocarcinoma (PDAC) is expected to become the second most common cause of cancer death in the USA by 2030, yet progress continues to lag behind that of other cancers, with only 9% of patients surviving beyond 5 years. Long-term survivorship of PDAC and improving survival has, until recently, escaped our understanding. One recent frontier in the cancer field is the microbiome. The microbiome collectively refers to the extensive community of bacteria and fungi that colonise us. It is estimated that there is one to ten prokaryotic cells for each human somatic cell, yet, the significance of this community in health and disease has, until recently, been overlooked. This review examines the role of the microbiome in PDAC and how it may alter survival outcomes. We evaluate the possibility of employing microbiomic signatures as biomarkers of PDAC. Ultimately this review analyses whether the microbiome may be amenable to targeting and consequently altering the natural history of PDAC.
Collapse
|
38
|
Riedel S, Pheiffer C, Johnson R, Louw J, Muller CJF. Intestinal Barrier Function and Immune Homeostasis Are Missing Links in Obesity and Type 2 Diabetes Development. Front Endocrinol (Lausanne) 2022; 12:833544. [PMID: 35145486 PMCID: PMC8821109 DOI: 10.3389/fendo.2021.833544] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
Noncommunicable diseases, such as type 2 diabetes (T2D), place a burden on healthcare systems worldwide. The rising prevalence of obesity, a major risk factor for T2D, is mainly attributed to the adoption of Westernized diets and lifestyle, which cause metabolic dysfunction and insulin resistance. Moreover, diet may also induce changes in the microbiota composition, thereby affecting intestinal immunity. The critical role of intestinal immunity and intestinal barrier function in the development of T2D is increasingly acknowledged, however, limited studies have investigated the link between intestinal function and metabolic disease. In this review, studies reporting specific roles of the intestinal immune system and intestinal epithelial cells (IECs) in metabolic disease are highlighted. Innate chemokine signaling, eosinophils, immunoglobulin A (IgA), T helper (Th) 17 cells and their cytokines were associated with obesity and/or dysregulated glucose homeostasis. Intestinal epithelial cells (IECs) emerged as critical modulators of obesity and glucose homeostasis through their effect on lipopolysaccharide (LPS) signaling and decontamination. Furthermore, IECs create a link between microbial metabolites and whole-body metabolic function. Future in depth studies of the intestinal immune system and IECs may provide new opportunities and targets to develop treatments and prevention strategies for obesity and T2D.
Collapse
Affiliation(s)
- Sylvia Riedel
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
| | - Carmen Pheiffer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
- Department of Obstetrics and Gynaecology, University of Pretoria, Pretoria, South Africa
| | - Rabia Johnson
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
| | - Johan Louw
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | - Christo J. F. Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| |
Collapse
|
39
|
Archna, Chawla PA, Teli G, Pathania S, Singh S, Srivastava V. Exploration of Antioxidant, Anti-inflammatory and Anticancer Potential of Substituted 4-Thiazolidinone Derivatives: Synthesis, Biological Evaluation and Docking Studies. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2021.2019796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Archna
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Pooja A. Chawla
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Ghanshyam Teli
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Shelly Pathania
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Shamsher Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | | |
Collapse
|
40
|
He TT, Xiao H, Wusiman M, Yishake D, Fang AP, Luo Y, Liu XZ, Liu ZY, Zhu HL. Dietary intake of one-carbon metabolism-related nutrients and hepatocellular carcinoma survival in the Guangdong Liver Cancer Cohort. Food Funct 2022; 13:8081-8090. [DOI: 10.1039/d2fo00943a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Dietary intake of one-carbon metabolism-related nutrients has been linked to cancer-related outcomes, but their effects on hepatocellular carcinoma (HCC) mortality are still unknown. The objective was to assess whether pre-diagnostic...
Collapse
|
41
|
The uninvited guests of our microbiome: Helicobacter pylori and Epstein-Barr virus and their role in gastric cancerogenesis. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
It is well established that human body is an ecosystem for numerous microorganisms: bacteria, fungi, eukaryotic parasites, and viruses. They form a “microbiome” that under conditions of homeostasis remains in a friendly mutual relationship with the host. However, the composition and diversity of this microbe community is dynamic and can be changed under the influence of environmental factors, such as diet, antibiotic therapy, lifestyle, and the host’s genotype and immunity. The result of gut microbiome dysbiosis can lead even to cancer. The aim of this review is the description of the healthy gastrointestinal microbiome and the role of two infectious agents: Gram-negative bacteria Helicobacter pylori and Epstein-Barr virus in the development of gastric cancer in terms of gut dysbiosis. H. pylori is the most important pathogen of gastric microbiome with clear impact on its diversity. Coinfection with Epstein-Barr virus causes chronic gastritis, and the inflammatory process is significantly increased. The process of carcinogenesis begins with chronic inflammation that causes atrophic gastritis, intestinal metaplasia, dysplasia, and finally cancer. It has been proven that chronic inflammatory infection caused by infectious agents increases the risk of stomach cancer. Molecular methods that are progressively used to explore the human microbiome provide hope that this knowledge will be used for future diagnoses and therapy in the state of its dysbiosis and in cases of gastric cancer.
Collapse
|
42
|
Kossai M, Radosevic-Robin N, Penault-Llorca F. Refining patient selection for breast cancer immunotherapy: beyond PD-L1. ESMO Open 2021; 6:100257. [PMID: 34487970 PMCID: PMC8426207 DOI: 10.1016/j.esmoop.2021.100257] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023] Open
Abstract
Therapies that modulate immune response to cancer, such as immune checkpoint inhibitors, began an intense development a few years ago; however, in breast cancer (BC), the results have been relatively disappointing so far. Finding biomarkers for better selection of BC patients for various immunotherapies remains a significant unmet medical need. At present, only tumour tissue programmed death-ligand 1 (PD-L1) and mismatch repair deficiency status are approved as theranostic biomarkers for programmed cell death-1 (PD-1)/PD-L1 inhibitors in BC. However, due to the complexity of tumour microenvironment (TME) and cancer response to immunomodulators, none of them is a perfect selector. Therefore, an intense quest is ongoing for complementary tumour- or host-related predictive biomarkers in breast immuno-oncology. Among the upcoming biomarkers, quantity, immunophenotype and spatial distribution of tumour-infiltrating lymphocytes and other TME cells as well as immune gene signatures emerge as most promising and are being increasingly tested in clinical trials. Biomarkers or strategies allowing dynamic assessment of BC response to immunotherapy, such as circulating/exosomal PD-L1, quantity of white/immune blood cell subpopulations and molecular imaging are particularly suitable for immunotreatment monitoring. Finally, host-related factors, such as microbiome and lifestyle, should also be taken into account when planning integration of immunomodulating therapies into BC management. As none of the biomarkers taken separately is accurate enough, the solution could come from composite biomarkers, which would combine clinical, molecular and immunological features of the disease, possibly powered by artificial intelligence. At present, immune checkpoint inhibitors (ICIs) are the only approved immunotherapy drugs in BC. Tumour PD-L1 and microsatellite status are current companion biomarkers for ICIs in BC; however, these need improvement. Evaluation of tumour immune contexture and the dynamics of circulating immune cell counts are promising novel approaches. Development of noninvasive monitoring and composite biomarkers will facilitate cancer immunotherapy, including in BC.
Collapse
Affiliation(s)
- M Kossai
- Department of Pathology, University Clermont Auvergne, INSERM U1240, Centre Jean Perrin, Clermont-Ferrand, France
| | - N Radosevic-Robin
- Department of Pathology, University Clermont Auvergne, INSERM U1240, Centre Jean Perrin, Clermont-Ferrand, France.
| | - F Penault-Llorca
- Department of Pathology, University Clermont Auvergne, INSERM U1240, Centre Jean Perrin, Clermont-Ferrand, France
| |
Collapse
|
43
|
Crosstalk between Environmental Inflammatory Stimuli and Non-Coding RNA in Cancer Occurrence and Development. Cancers (Basel) 2021; 13:cancers13174436. [PMID: 34503246 PMCID: PMC8430834 DOI: 10.3390/cancers13174436] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 08/20/2021] [Accepted: 08/31/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Increasing evidence has indicated that chronic inflammatory processes have an influence on tumor occurrence and all stages of tumor development. A dramatic increase of studies into non-coding RNAs (ncRNAs) biology has shown that ncRNAs act as oncogenic drivers and tumor suppressors in various inflammation-induced cancers. Thus, this complex network of inflammation-associated cancers and ncRNAs offers targets for prevention from the malignant transformation from inflammation and treatment of malignant diseases. Abstract There is a clear relationship between inflammatory response and different stages of tumor development. Common inflammation-related carcinogens include viruses, bacteria, and environmental mutagens, such as air pollutants, toxic metals, and ultraviolet light. The expression pattern of ncRNA changes in a variety of disease conditions, including inflammation and cancer. Non-coding RNAs (ncRNAs) have a causative role in enhancing inflammatory stimulation and evading immune responses, which are particularly important in persistent pathogen infection and inflammation-to-cancer transformation. In this review, we investigated the mechanism of ncRNA expression imbalance in inflammation-related cancers. A better understanding of the function of inflammation-associated ncRNAs may help to reveal the potential of ncRNAs as a new therapeutic strategy.
Collapse
|
44
|
El-Sayed A, Aleya L, Kamel M. Microbiota and epigenetics: promising therapeutic approaches? ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:49343-49361. [PMID: 34319520 PMCID: PMC8316543 DOI: 10.1007/s11356-021-15623-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/20/2021] [Indexed: 04/15/2023]
Abstract
The direct/indirect responsibility of the gut microbiome in disease induction in and outside the digestive tract is well studied. These results are usually from the overpopulation of certain species on the cost of others, interaction with beneficial microflora, interference with normal epigenetic control mechanisms, or suppression of the immune system. Consequently, it is theoretically possible to cure such disorders by rebalancing the microbiome inside our bodies. This can be achieved by changing the lifestyle pattern and diet or by supplementation with beneficial bacteria or their metabolites. Various approaches have been explored to manipulate the normal microbial inhabitants, including nutraceutical, supplementations with prebiotics, probiotics, postbiotics, synbiotics, and antibiotics, or through microbiome transplantation (fecal, skin, or vaginal microbiome transplantation). In the present review, the interaction between the microbiome and epigenetics and their role in disease induction is discussed. Possible future therapeutic approaches via the reestablishment of equilibrium in our internal micro-ecosystem are also highlighted.
Collapse
Affiliation(s)
- Amr El-Sayed
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, F-25030, Besançon Cedex, France
| | - Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt.
| |
Collapse
|
45
|
Park JM, Lee WH, Seo H, Oh JY, Lee DY, Kim SJ, Hahm KB. Microbiota changes with fermented kimchi contributed to either the amelioration or rejuvenation of Helicobacter pylori-associated chronic atrophic gastritis. J Clin Biochem Nutr 2021; 69:98-110. [PMID: 34376919 PMCID: PMC8325762 DOI: 10.3164/jcbn.20-123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/25/2020] [Indexed: 12/21/2022] Open
Abstract
Korean fermented kimchi is probiotic food preventing Helicobacter pylori (H. pylori)-associated atrophic gastritis in both animal and human trial. In order to reveal the effect of fermented kimchi against H. pylori infection, we performed clinical trial to document the changes of fecal microbiota in 32 volunteers (H. pylori (-) chronic superficial gastritis (CSG), H. pylori (+) CSG, and H. pylori (+) chronic atrophic gastritis (CAG) with 10 weeks kimchi. Each amplicon is sequenced on MiSeq of Illumina and the sequence reads were clustered into operational taxonomic units using VSEARCH and the Chao, Simpson, and Shannon Indices. Though significant difference in α- or β-diversity was not seen in three groups, kimchi intake led to significant diversity of fecal microbiome. As results, Klebsiella, Enterococcus, Ruminococcaceae, Streptococcus, Roseburia, and Clostirdiumsensu were significantly increased in H. pylori (+) CAG, while Akkermansia, Citrobacter, and Lactobacillus were significantly decreased in H. pylori (+) CAG. With 10 weeks of kimchi administration, Bifidobacterium, Lactobacillus, and Ruminococcus were significantly increased in H. pylori (+) CAG, whereas Bacteroides, Subdoligranulum, and Eubacterium coprostanolines were significantly decreased in H. pylori (-) CAG. 10 weeks of kimchi intake significantly improved pepsinogen I/II ratio (p<0.01) with significant decreases in interleukin-1β. Conclusively, fermented kimchi significantly changed fecal microbiota to mitigate H. pylori-associated atrophic gastritis.
Collapse
Affiliation(s)
- Jong Min Park
- Daejeon University School of Oriental Medicine, Daehak-ro 62, Dong-gu, Daejeon 34520, Korea
| | | | | | | | | | - Seong Jin Kim
- Medpacto Research Institute, Medpacto, Myungdal-ro 92, Seocho-gu, Seoul 06668, Korea
| | - Ki Baik Hahm
- Medpacto Research Institute, Medpacto, Myungdal-ro 92, Seocho-gu, Seoul 06668, Korea
- CHA Cancer Preventive Research Center, CHA Bio Complex, 330 Pangyo-ro, Bundang-gu, Seongnam 13497, Korea
| |
Collapse
|
46
|
Jiang Z, Li L, Chen J, Wei G, Ji Y, Chen X, Liu J, Huo J. Human gut-microbiome interplay: Analysis of clinical studies for the emerging roles of diagnostic microbiology in inflammation, oncogenesis and cancer management. INFECTION GENETICS AND EVOLUTION 2021; 93:104946. [PMID: 34052417 DOI: 10.1016/j.meegid.2021.104946] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 04/21/2021] [Accepted: 05/26/2021] [Indexed: 12/24/2022]
Abstract
Microorganisms have been known to coexist in various parts of human body including the gut. The interactions between microbes and the surrounding tissues of the host are critical for fine fettle of the gut. The incidence of such microorganisms tends to vary among specific type of cancer affected individuals. Such microbial communities of specific tumor sites in cancer affected individuals could plausibly be used as prognostic and/or diagnostic markers for tumors associated with that specific site. Microorganisms of intestinal and non-intestinal origins including Helicobacter pylori can target several organs, act as carcinogens and promote cancer. It is interesting to note that diets causing inflammation can also increase the cancer risk. Yet, dietary supplementation with prebiotics and probiotics can reduce the incidence of cancer. Therefore, both diet and microbial community of the gut have dual roles of prevention and oncogenesis. Hence, this review intends to summarize certain important details related to gut microbiome and cancer.
Collapse
Affiliation(s)
- Ziyu Jiang
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Lingchang Li
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Jianan Chen
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China
| | - Guoli Wei
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Yi Ji
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Xi Chen
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Jingbing Liu
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China.
| | - Jiege Huo
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China.
| |
Collapse
|
47
|
Bonde A, Daly S, Kirsten J, Kondapaneni S, Mellnick V, Menias CO, Katabathina VS. Human Gut Microbiota-associated Gastrointestinal Malignancies: A Comprehensive Review. Radiographics 2021; 41:1103-1122. [PMID: 33989072 DOI: 10.1148/rg.2021200168] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The human gastrointestinal tract houses trillions of microbes. The gut and various types of microorganisms, including bacteria, viruses, fungi, and archaea, form a complex ecosystem known as the gut microbiota, and the whole genome of the gut microbiota is referred to as the gut microbiome. The gut microbiota is essential for homeostasis and the overall well-being of a person and is increasingly considered an adjunct "virtual organ," with a complexity level comparable to that of the other organ systems. The gut microbiota plays an essential role in nutrition, local mucosal homeostasis, inflammation, and the mucosal immune system. An imbalanced state of the gut microbiota, known as dysbiosis, can predispose to development of various gastrointestinal malignancies through three speculated pathogenic mechanisms: (a) direct cytotoxic effects with damage to the host DNA, (b) disproportionate proinflammatory signaling inducing inflammation, and (c) activation of tumorigenic pathways or suppression of tumor-suppressing pathways. Several microorganisms, including Helicobacter pylori, Epstein-Barr virus, human papillomavirus, Mycoplasma species, Escherichia coli, and Streptococcus bovis, are associated with gastrointestinal malignancies such as esophageal adenocarcinoma, gastric adenocarcinoma, gastric mucosa-associated lymphoid tissue lymphoma, colorectal adenocarcinoma, and anal squamous cell carcinoma. Imaging plays a pivotal role in diagnosis and management of microbiota-associated gastrointestinal malignancies. Appropriate use of probiotics, fecal microbiota transplantation, and overall promotion of the healthy gut are ongoing areas of research for prevention and treatment of malignancies. Online supplemental material is available for this article. ©RSNA, 2021.
Collapse
Affiliation(s)
- Apurva Bonde
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (A.B., S.D., J.K., V.S.K.); University of Texas at Austin, Austin, Tex (S.K.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.); and Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M.)
| | - Sean Daly
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (A.B., S.D., J.K., V.S.K.); University of Texas at Austin, Austin, Tex (S.K.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.); and Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M.)
| | - Julia Kirsten
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (A.B., S.D., J.K., V.S.K.); University of Texas at Austin, Austin, Tex (S.K.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.); and Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M.)
| | - Sainath Kondapaneni
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (A.B., S.D., J.K., V.S.K.); University of Texas at Austin, Austin, Tex (S.K.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.); and Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M.)
| | - Vincent Mellnick
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (A.B., S.D., J.K., V.S.K.); University of Texas at Austin, Austin, Tex (S.K.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.); and Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M.)
| | - Christine O Menias
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (A.B., S.D., J.K., V.S.K.); University of Texas at Austin, Austin, Tex (S.K.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.); and Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M.)
| | - Venkata S Katabathina
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (A.B., S.D., J.K., V.S.K.); University of Texas at Austin, Austin, Tex (S.K.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.); and Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M.)
| |
Collapse
|
48
|
Zhu R, Lang T, Yan W, Zhu X, Huang X, Yin Q, Li Y. Gut Microbiota: Influence on Carcinogenesis and Modulation Strategies by Drug Delivery Systems to Improve Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003542. [PMID: 34026439 PMCID: PMC8132165 DOI: 10.1002/advs.202003542] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/05/2021] [Indexed: 05/05/2023]
Abstract
Gut microbiota have close interactions with the host. It can affect cancer progression and the outcomes of cancer therapy, including chemotherapy, immunotherapy, and radiotherapy. Therefore, approaches toward the modulation of gut microbiota will enhance cancer prevention and treatment. Modern drug delivery systems (DDS) are emerging as rational and promising tools for microbiota intervention. These delivery systems have compensated for the obstacles associated with traditional treatments. In this review, the essential roles of gut microbiota in carcinogenesis, cancer progression, and various cancer therapies are first introduced. Next, advances in DDS that are aimed at enhancing the efficacy of cancer therapy by modulating or engineering gut microbiota are highlighted. Finally, the challenges and opportunities associated with the application of DDS targeting gut microbiota for cancer prevention and treatment are briefly discussed.
Collapse
Affiliation(s)
- Runqi Zhu
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Tianqun Lang
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
- Yantai Key Laboratory of Nanomedicine and Advanced PreparationsYantai Institute of Materia MedicaYantai264000China
| | - Wenlu Yan
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Xiao Zhu
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Xin Huang
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Qi Yin
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
- Yantai Key Laboratory of Nanomedicine and Advanced PreparationsYantai Institute of Materia MedicaYantai264000China
| | - Yaping Li
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
- Yantai Key Laboratory of Nanomedicine and Advanced PreparationsYantai Institute of Materia MedicaYantai264000China
- School of PharmacyYantai UniversityYantai264005China
| |
Collapse
|
49
|
Armstrong H, Bording-Jorgensen M, Wine E. The Multifaceted Roles of Diet, Microbes, and Metabolites in Cancer. Cancers (Basel) 2021; 13:cancers13040767. [PMID: 33673140 PMCID: PMC7917909 DOI: 10.3390/cancers13040767] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Many studies performed to date have implicated select microbes and dietary factors in a variety of cancers, yet the complexity of both these diseases and the relationship between these factors has limited the ability to translate findings into therapies and preventative guidelines. Here we begin by discussing recently published studies relating to dietary factors, such as vitamins and chemical compounds used as ingredients, and their contribution to cancer development. We further review recent studies, which display evidence of the microbial-diet interaction in the context of cancer. The field continues to advance our understanding of the development of select cancers and how dietary factors are related to the development, prevention, and treatment of these cancers. Finally, we highlight the science available in the discussion of common misconceptions with regards to cancer and diet. We conclude this review with thoughts on where we believe future research should focus in order to provide the greatest impact towards human health and preventative medicine.
Collapse
Affiliation(s)
- Heather Armstrong
- CEGIIR, University of Alberta, Edmonton, AB T6G 2X8, Canada;
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 1C9, Canada
- Correspondence: (H.A.); (E.W.)
| | - Michael Bording-Jorgensen
- CEGIIR, University of Alberta, Edmonton, AB T6G 2X8, Canada;
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Eytan Wine
- CEGIIR, University of Alberta, Edmonton, AB T6G 2X8, Canada;
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 1C9, Canada
- Department of Physiology, University of Alberta, Edmonton, AB T6G 1C9, Canada
- Correspondence: (H.A.); (E.W.)
| |
Collapse
|
50
|
Abstract
ABSTRACT Microorganisms can help maintain homeostasis in humans by providing nutrition, maintaining hormone balance, and regulating inflammatory responses. In the case of imbalances, these microbes can cause various diseases, even malignancy. Pancreatic cancer (PC) is characterized by high tumor invasiveness, distant metastasis, and insensitivity to traditional chemotherapeutic drugs, and it is confirmed that PC is closely related to microorganisms. Recently, most studies based on clinical samples or case reports discussed the positive or negative relationships between microorganisms and PC. However, the specific mechanisms are blurry, especially the involved immunological pathways, and the roles of beneficial flora have usually been ignored. We reviewed studies published through September 2020 as identified using PubMed, MEDLINE, and Web of Science. We mainly introduced the traits of oral, gastrointestinal, and intratumoral microbes in PC and summarized the roles of these microbes in tumorigenesis and tumoral development through immunological pathways, in addition to illustrating the relationships between metabolic diseases with PC by microorganism. In addition, we identified microorganisms as biomarkers for early diagnosis and immunotherapy. This review will be significant for greater understanding the effect of microorganisms in PC and provide more meaningful guidance for future clinical applications.
Collapse
Affiliation(s)
- Xin Wei
- From the Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun
| | - Chunlei Mei
- Institute of Reproductive Health, Huazhong University of Science and Technology, Wuhan, China
| | - Xixi Li
- From the Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun
| | - Yingjun Xie
- From the Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun
| |
Collapse
|