101
|
Vaginom- und „Endometriom“-Diagnostik bei Kinderwunsch. GYNAKOLOGISCHE ENDOKRINOLOGIE 2022. [DOI: 10.1007/s10304-022-00466-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
ZusammenfassungDie molekulare Diagnostik der Vaginal- und Endometriumflora ermöglicht neue Einblicke in die physiologische Besiedlung des weiblichen Genitaltrakts. Wesentlich ist hierbei eine Dominanz von bestimmten Laktobazillenarten bei gleichzeitig geringer Artenvielfalt. Die Bildung von D‑Laktat ist eine entscheidende Voraussetzung für die Abwehr von sexuell übertragbaren Erkrankungen, den Erfolg einer In-vitro-Fertilisation sowie einen ungestörten Schwangerschaftsverlauf. Eine Störung der Flora durch pathogene Bakterienarten mit der Folge des Auftretens einer bakteriellen Vaginose oder einer chronischen Endometritis kann zu Implantationsversagen, Aborten und Frühgeburtsbestrebungen führen. Bei wiederholtem Implantationsversagen ermöglicht eine Mikrobiomdiagnostik – im Gegensatz zur klassischen Kultur, den Amsel-Kriterien oder dem Nugent-Score – einen sehr viel detaillierteren Einblick in die Pathophysiologie, da sich entscheidende Bakterienarten nur schwer oder gar nicht anzüchten lassen. Dies erlaubt eine sehr viel bessere Planung der Therapie zur Wiederherstellung physiologischer Verhältnisse.
Collapse
|
102
|
Hu K, Li C, Yu T, Guo H, Sun H, Mao S, Zhou Z, Jin W, Liu K, Xie L, Wang G, Liang Y. Global analysis of qualitative and quantitative metabolism of Notoginsenoside R1 in rat liver-brain-gut axis based on LC-IT-TOF/MS combing mMDF strategy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154261. [PMID: 35793598 DOI: 10.1016/j.phymed.2022.154261] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/28/2022] [Accepted: 06/07/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND The metabolism study of active components for traditional Chinese medicine (TCM) in target organs is conducive to clarify the authentic active ingredients. Notoginsenoside R1 (NG-R1), one of the bioactive components of Panax notoginsenoside (PNS), is commonly acknowledged as the characteristic marker of PNS. However, the metabolism of NG-R1 in target organs has not been clarified yet due to the lack of robust technique and approach. PURPOSE The present study aimed to develop a reliable and efficient strategy and technology for revealing the qualitative and quantitative metabolism of active components of TCMs in target organs, and to clarify the biotransformation of NG-R1 in liver-brain-intestinal axis. METHODS The metabolic transformation of NG-R1 in the brain gut axis was investigated in the in vitro incubation system of fresh rat brain, liver homogenate, and intestinal flora. To quickly lock the target metabolites, we set the mass defect filter (MDF) in different ranges to screen metabolites with different molecular weight (MW). This strategy was defined as multi-stage MDF (mMDF). In addition, we performed relative quantitative analysis on all metabolites according to the peak area acquired by LC-IT-TOF/MS to overcome the challenge that metabolites are difficult to be quantified due to the lack of standards. RESULTS When MDF was set at 0.50 to 0.65 to screen metabolites with MW of 900 to 1200 Da, 6 novel metabolites were quickly found, and then identified as glucuronic acid binding, oxidation, dehydrogenation, methylation and hydrogenation products according to their LC and MS characteristics. When setting MDF at 0.42 - 0.52, 6 metabolites with MW of 600 to 900 Da were effectively screened and identified as Rg1, NG-R2, Rh1, Rg1+CH2+2H and Rg1+CH2. To screen the metabolites with MW of 300 to 600 Da, MDF was set at 0.25 - 0.42, and 4 novel metabolites were screened rapidly. The results of quantitative metabolism suggested that intestinal flora was the main metabolic site of NG-R1 in rat, and more than 60% of NG-R1 was converted to Rg1 by deglycosylation in the intestinal flora. CONCLUSION The mMDF strategy can significantly improve the research efficiency of qualitative metabolism of saponins. Although NG-R1 could be transformed into a variety of metabolites in rat liver and brain homogenate, it still existed mainly in prototype form. In the rat flora, NG-R1 mainly existed in the form of deglycosylated metabolite Rg1.
Collapse
Affiliation(s)
- Kangrui Hu
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, PR China
| | - Changjian Li
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, PR China
| | - Tengjie Yu
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, PR China
| | - Huimin Guo
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, PR China
| | - Hong Sun
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, PR China
| | - Shuying Mao
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, PR China
| | - Zhihao Zhou
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, PR China
| | - Wei Jin
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, PR China
| | - Keanqi Liu
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, PR China
| | - Lin Xie
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, PR China
| | - Guangji Wang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, PR China.
| | - Yan Liang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, PR China.
| |
Collapse
|
103
|
Kafeenah H, Kuo CM, Chang TY, Jen HH, Yang JH, Shen YS, Wu CH, Chen SH. Label-free and de-conjugation-free workflow to simultaneously quantify trace amount of free/conjugated and protein-bound estrogen metabolites in human serum. Anal Chim Acta 2022; 1232:340457. [DOI: 10.1016/j.aca.2022.340457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/18/2022] [Accepted: 09/26/2022] [Indexed: 11/27/2022]
|
104
|
In-Silico Characterization of Estrogen Reactivating β-Glucuronidase Enzyme in GIT Associated Microbiota of Normal Human and Breast Cancer Patients. Genes (Basel) 2022; 13:genes13091545. [PMID: 36140713 PMCID: PMC9498756 DOI: 10.3390/genes13091545] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/30/2022] Open
Abstract
Estrogen circulating in blood has been proved to be a strong biomarker for breast cancer. A β-glucuronidase enzyme (GUS) from human gastrointestinal tract (GIT) microbiota including probiotics has significant involvement in enhancing the estrogen concentration in blood through deconjugation of glucuronidated estrogens. The present project has been designed to explore GIT microbiome-encoded GUS enzymes (GUSOME) repertoire in normal human and breast cancer patients. For this purpose, a total of nineteen GUS enzymes from human GIT microbes, i.e., seven from healthy and twelve from breast cancer patients have been focused on. Protein sequences of enzymes retrieved from UniProt database were subjected to ProtParam, CELLO2GO, SOPMA (secondary structure prediction method), PDBsum (Protein Database summaries), PHYRE2 (Protein Homology/AnalogY Recognition Engine), SAVES v6.0 (Structure Validation Server), MEME version 5.4.1 (Multiple Em for Motif Elicitation), Caver Web server v 1.1, Interproscan and Predicted Antigenic Peptides tool. Analysis revealed the number of amino acids, isoelectric point, extinction coefficient, instability index and aliphatic index of GUS enzymes in the range of 586−795, 4.91−8.92, 89,980−155,075, 25.88−40.93 and 71.01−88.10, respectively. Sub-cellular localization of enzyme was restricted to cytoplasm and inner-membrane in case of breast cancer patients’ bacteria as compared to periplasmic space, outer membrane and extracellular space in normal GIT bacteria. The 2-D structure analysis showed α helix, extended strand, β turn and random coil in the range of 27.42−22.66%, 22.04−25.91%, 5.39−8.30% and 41.75−47.70%, respectively. The druggability score was found to be 0.05−0.45 and 0.06−0.80 in normal and breast cancer patients GIT, respectively. The radius, length and curvature of catalytic sites were observed to be 1.1−2.8 Å, 1.4−15.9 Å and 0.65−1.4, respectively. Ten conserved protein motifs with p < 0.05 and width 25−50 were found. Antigenic propensity-associated sequences were 20−29. Present study findings hint about the use of the bacterial GUS enzymes against breast cancer tumors after modifications via site-directed mutagenesis of catalytic sites involved in the activation of estrogens and through destabilization of these enzymes.
Collapse
|
105
|
Wu H, Ganguly S, Tollefsbol TO. Modulating Microbiota as a New Strategy for Breast Cancer Prevention and Treatment. Microorganisms 2022; 10:microorganisms10091727. [PMID: 36144329 PMCID: PMC9503838 DOI: 10.3390/microorganisms10091727] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/18/2022] Open
Abstract
Breast cancer (BC) is the most common cancer in women in the United States. There has been an increasing incidence and decreasing mortality rate of BC cases over the past several decades. Many risk factors are associated with BC, such as diet, aging, personal and family history, obesity, and some environmental factors. Recent studies have shown that healthy individuals and BC patients have different microbiota composition, indicating that microbiome is a new risk factor for BC. Gut and breast microbiota alterations are associated with BC prognosis. This review will evaluate altered microbiota populations in gut, breast tissue, and milk of BC patients, as well as mechanisms of interactions between microbiota modulation and BC. Probiotics and prebiotics are commercially available dietary supplements to alleviate side-effects of cancer therapies. They also shape the population of human gut microbiome. This review evaluates novel means of modulating microbiota by nutritional treatment with probiotics and prebiotics as emerging and promising strategies for prevention and treatment of BC. The mechanistic role of probiotic and prebiotics partially depend on alterations in estrogen metabolism, systematic immune regulation, and epigenetics regulation.
Collapse
Affiliation(s)
- Huixin Wu
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA
| | - Sebanti Ganguly
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA
| | - Trygve O. Tollefsbol
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA
- Integrative Center for Aging Research, University of Alabama Birmingham, 1530 3rd Avenue South, Birmingham, AL 35294, USA
- O’Neal Comprehensive Cancer Center, University of Alabama Birmingham, 1802 6th Avenue South, Birmingham, AL 35294, USA
- Nutrition Obesity Research Center, University of Alabama Birmingham, 1675 University Boulevard, Birmingham, AL 35294, USA
- Comprehensive Diabetes Center, University of Alabama Birmingham, 1825 University Boulevard, Birmingham, AL 35294, USA
- University Wide Microbiome Center, University of Alabama Birmingham, 845 19th Street South, Birmingham, AL 35294, USA
- Correspondence: ; Tel.: +1-205-934-4573; Fax: +1-205-975-6097
| |
Collapse
|
106
|
Chen Y, Kim M, Paye S, Benayoun BA. Sex as a Biological Variable in Nutrition Research: From Human Studies to Animal Models. Annu Rev Nutr 2022; 42:227-250. [PMID: 35417195 PMCID: PMC9398923 DOI: 10.1146/annurev-nutr-062220-105852] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Biological sex is a fundamental source of phenotypic variability across species. Males and females have different nutritional needs and exhibit differences in nutrient digestion and utilization, leading to different health outcomes throughout life. With personalized nutrition gaining popularity in scientific research and clinical practice, it is important to understand the fundamentals of sex differences in nutrition research. Here, we review key studies that investigate sex dimorphism in nutrition research: sex differences in nutrient intake and metabolism, sex-dimorphic response in nutrient-restricted conditions, and sex differences in diet and gut microbiome interactions. Within each area above, factors from sex chromosomes, sex hormones, and sex-specific loci are highlighted.
Collapse
Affiliation(s)
- Yilin Chen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA;
| | - Minhoo Kim
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA;
| | - Sanjana Paye
- Department of Molecular and Computational Biology, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California, USA
| | - Bérénice A Benayoun
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA;
- Department of Molecular and Computational Biology, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Epigenetics and Gene Regulation Program, USC Norris Comprehensive Cancer Center, Los Angeles, California, USA
- USC Stem Cell Initiative, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
107
|
Li Y, Wang K, Ding J, Sun S, Ni Z, Yu C. Influence of the gut microbiota on endometriosis: Potential role of chenodeoxycholic acid and its derivatives. Front Pharmacol 2022; 13:954684. [PMID: 36071850 PMCID: PMC9442031 DOI: 10.3389/fphar.2022.954684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/27/2022] [Indexed: 11/28/2022] Open
Abstract
The gut microbiota (GM) has received extensive attention in recent years, and its key role in the establishment and maintenance of health and in the development of diseases has been confirmed. A strong correlation between the GM and the progression of endometriosis (EMS) has been observed in emerging research. Alterations in the composition and function of the GM have been described in many studies on EMS. In contrast, the GM in the environment of EMS, especially the GM metabolites, such as bile acids and short-chain fatty acids that are related to the pathogenesis of EMS, can promote disease progression. Chenodeoxycholic acid (CDCA), as one of the primary bile acids produced in the liver, is metabolized by various enzymes derived from the GM and is critically important in maintaining intestinal homeostasis and regulating lipid and carbohydrate metabolism and innate immunity. Given that the complexity of CDCA as a signalling molecule and the interaction between the GM and EMS have not been clarified, the role of the CDCA and GM in EMS should be understood from a novel perspective. However, few articles on the relationship between CDCA and EMS have been reviewed. Therefore, we review the available and possible potential links between CDCA, the GM and EMS and put forward the hypothesis that CDCA and its derivative obeticholic acid can improve the symptoms of EMS through the GM.
Collapse
Affiliation(s)
- Yangshuo Li
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Kaili Wang
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jie Ding
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Shuai Sun
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zhexin Ni
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
- *Correspondence: Zhexin Ni, ; Chaoqin Yu,
| | - Chaoqin Yu
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- *Correspondence: Zhexin Ni, ; Chaoqin Yu,
| |
Collapse
|
108
|
Peters BA, Santoro N, Kaplan RC, Qi Q. Spotlight on the Gut Microbiome in Menopause: Current Insights. Int J Womens Health 2022; 14:1059-1072. [PMID: 35983178 PMCID: PMC9379122 DOI: 10.2147/ijwh.s340491] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/05/2022] [Indexed: 11/23/2022] Open
Abstract
The gut microbiome is an important contributor to human health, shaped by many endogenous and exogenous factors. The gut microbiome displays sexual dimorphism, suggesting influence of sex hormones, and also has been shown to change with aging. Yet, little is known regarding the influence of menopause - a pivotal event of reproductive aging in women - on the gut microbiome. Here, we summarize what is known regarding the interrelationships of female sex hormones and the gut microbiome, and review the available literature on menopause, female sex hormones, and the gut microbiome in humans. Taken together, research suggests that menopause is associated with lower gut microbiome diversity and a shift toward greater similarity to the male gut microbiome, however more research is needed in large study populations to identify replicable patterns in taxa impacted by menopause. Many gaps in knowledge remain, including the role the gut microbiome may play in menopause-related disease risks, and whether menopausal hormone therapy modifies menopause-related change in the gut microbiome. Given the modifiable nature of the gut microbiome, better understanding of its role in menopause-related health will be critical to identify novel opportunities for improvement of peri- and post-menopausal health and well-being.
Collapse
Affiliation(s)
- Brandilyn A Peters
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nanette Santoro
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
109
|
Walker ME, Simpson JB, Redinbo MR. A structural metagenomics pipeline for examining the gut microbiome. Curr Opin Struct Biol 2022; 75:102416. [PMID: 35841748 PMCID: PMC10039758 DOI: 10.1016/j.sbi.2022.102416] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 04/25/2022] [Accepted: 05/18/2022] [Indexed: 12/13/2022]
Abstract
Metagenomic sequencing data provide a rich resource from which to expand our understanding of differential protein functions involved in human health. Here, we outline a pipeline that combines microbial whole genome sequencing with protein structure data to yield a structural metagenomics-informed atlas of microbial enzyme families of interest. Visualizing metagenomics data through a structural lens facilitates downstream studies including targeted inhibition and probe-based proteomics to define at the molecular level how different enzyme orthologs impact in vivo function. Application of this pipeline to gut microbial enzymes like glucuronidases, TMA lyases, and bile salt hydrolases is expected to pinpoint their involvement in health and disease and may aid in the development of therapeutics that target specific enzymes within the microbiome.
Collapse
Affiliation(s)
- Morgan E Walker
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Joshua B Simpson
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matthew R Redinbo
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Integrated Program for Biological and Genome Sciences, And Departments of Biochemistry and Microbiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
110
|
Lephart ED, Naftolin F. Estrogen Action and Gut Microbiome Metabolism in Dermal Health. Dermatol Ther (Heidelb) 2022; 12:1535-1550. [PMID: 35752663 PMCID: PMC9276867 DOI: 10.1007/s13555-022-00759-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/08/2022] [Indexed: 01/14/2023] Open
Abstract
Emerging scientific advances in microbial research linking estrogens and the gut-skin microbiome in reference to dermal health are featured in this narrative review of journal reports and reviews from January 2018 through February 2022. Background information on advances in microbial research along with defining the microbiota and microbiome is presented in brief. The development of and factors that influence the gut microbiome in health and disease as well as the intrinsic and extrinsic factors influencing the skin microbiome and skin aging are summarized. New information on the development and changes of organ microbiomes have exposed similarities between skin and gut structure/function, microbial components/diversity/taxonomy and how they impact the immune response for combating disease and enhancing wellness. Estrogens promote health and support homeostasis in general and directly impact dermal health. Moreover, the gut, based upon the level of the microbial enzyme β-glucuronidase, which regulates estrogen's enterohepatic recirculation, constitutes a gut-skin microbial axis. This axis revolves around the systemically available estrogen to support immune function, counteract inflammation and oxidative stress, and decrease the risk of hormone-dependent skin cancers. These data support the direct effect of estrogens on skin health and the interaction of diet on dermal health via effects on the gut microflora. Finally, the potential for bioactive botanicals containing phytoestrogens or selective estrogen receptor modulators (SERMs) to evade the effects of gut β-glucuronidase expressing flora is proposed that may have a positive impact on skin.
Collapse
Affiliation(s)
- Edwin D. Lephart
- Department of Cell Biology, Physiology and The Neuroscience Center LS 4005, College of Life Sciences, Brigham Young University, Provo, UT 84602 USA
| | | |
Collapse
|
111
|
Xu Q, Li D, Chen J, Yang J, Yan J, Xia Y, Zhang F, Wang X, Cao H. Crosstalk between the gut microbiota and postmenopausal osteoporosis: Mechanisms and applications. Int Immunopharmacol 2022; 110:108998. [PMID: 35785728 DOI: 10.1016/j.intimp.2022.108998] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/08/2022] [Accepted: 06/21/2022] [Indexed: 12/14/2022]
Abstract
Postmenopausal osteoporosis (PMO) results from a reduction in bone mass and microarchitectural deterioration in bone tissue due to estrogen deficiency, which may increase the incidence of fragility fractures. The number of people suffering from PMO has increased over the years because of the rapidly aging population worldwide. However, several pharmacological agents for the treatment of PMO have many safety risks and impose a heavy financial burden to patients and society. In recent years, the "gut-bone" axis has been proposed as a new approach in the prevention and treatment of PMO. This paper reviews the relationship between the gut microbiota and PMO, which mainly includes the underlying mechanisms between hormones, immunity, nutrient metabolism, metabolites of the gut microbiota and intestinal permeability, and explores the possible role of the gut microbiota in these processes. Finally, we discuss the therapeutic effects of diet, prebiotics, probiotics, and fecal microbiota transplantation on the gut microbiota.
Collapse
Affiliation(s)
- Qin Xu
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Dan Li
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Jing Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Nursing Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Ju Yang
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Jiai Yan
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Yanping Xia
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Feng Zhang
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Xuesong Wang
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Department of Orthopedics, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Hong Cao
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Department of Endocrinology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China.
| |
Collapse
|
112
|
Menopause Is Associated with an Altered Gut Microbiome and Estrobolome, with Implications for Adverse Cardiometabolic Risk in the Hispanic Community Health Study/Study of Latinos. mSystems 2022; 7:e0027322. [PMID: 35675542 PMCID: PMC9239235 DOI: 10.1128/msystems.00273-22] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Menopause is a pivotal period during which loss of ovarian hormones increases cardiometabolic risk and may also influence the gut microbiome. However, the menopause-microbiome relationship has not been examined in a large study, and its implications for cardiometabolic disease are unknown. In the Hispanic Community Health Study/Study of Latinos, a population with high burden of cardiometabolic risk factors, shotgun metagenomic sequencing was performed on stool from 2,300 participants (295 premenopausal women, 1,027 postmenopausal women, and 978 men), and serum metabolomics was available on a subset. Postmenopausal women trended toward lower gut microbiome diversity and altered overall composition compared to premenopausal women, while differing less from men, in models adjusted for age and other demographic/behavioral covariates. Differentially abundant taxa for post- versus premenopausal women included Bacteroides sp. strain Ga6A1, Prevotella marshii, and Sutterella wadsworthensis (enriched in postmenopause) and Escherichia coli-Shigella spp., Oscillibacter sp. strain KLE1745, Akkermansia muciniphila, Clostridium lactatifermentans, Parabacteroides johnsonii, and Veillonella seminalis (depleted in postmenopause); these taxa similarly differed between men and women. Postmenopausal women had higher abundance of the microbial sulfate transport system and decreased abundance of microbial β-glucuronidase; these functions correlated with serum progestin metabolites, suggesting involvement of postmenopausal gut microbes in sex hormone retention. In postmenopausal women, menopause-related microbiome alterations were associated with adverse cardiometabolic profiles. In summary, in a large U.S. Hispanic/Latino population, menopause is associated with a gut microbiome more similar to that of men, perhaps related to the common condition of a low estrogen/progesterone state. Future work should examine similarity of results in other racial/ethnic groups. IMPORTANCE The menopausal transition, marked by declining ovarian hormones, is recognized as a pivotal period of cardiometabolic risk. Gut microbiota metabolically interact with sex hormones, but large population studies associating menopause with the gut microbiome are lacking. Our results from a large study of Hispanic/Latino women and men suggest that the postmenopausal gut microbiome in women is slightly more similar to the gut microbiome in men and that menopause depletes specific gut pathogens and decreases the hormone-related metabolic potential of the gut microbiome. At the same time, gut microbes may participate in sex hormone reactivation and retention in postmenopausal women. Menopause-related gut microbiome changes were associated with adverse cardiometabolic risk in postmenopausal women, indicating that the gut microbiome contributes to changes in cardiometabolic health during menopause.
Collapse
|
113
|
Jiang FR, Hang L, Zhou Y, Feng Y, Yuan JY. Estrogen-gut microbiota interactions and irritable bowel syndrome. Shijie Huaren Xiaohua Zazhi 2022; 30:511-520. [DOI: 10.11569/wcjd.v30.i12.511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a common functional gastrointestinal disorder with a complex pathogenesis that has a serious impact on the quality of life of patients. Abnormal visceral sensation, disordered gut motility, dysregulated immunity, and damaged intestinal barrier are thought to be involved in the pathogenesis of IBS. Female predisposition to IBS strongly suggests that sex hormones such as estrogen are involved in the development of IBS. In addition, dysbiosis of the intestinal flora is closely related to IBS. The interaction between estrogen and gut microbiota in IBS has not been fully elucidated. This review summarizes and evaluates the progress of related studies. Based on the new findings and shortcomings of current studies, we discuss the directions and issues that need to be resolved in future research.
Collapse
Affiliation(s)
- Feng-Ru Jiang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Lu Hang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yan Zhou
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Ya Feng
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jian-Ye Yuan
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| |
Collapse
|
114
|
Diviccaro S, FitzGerald JA, Cioffi L, Falvo E, Crispie F, Cotter PD, O’Mahony SM, Giatti S, Caruso D, Melcangi RC. Gut Steroids and Microbiota: Effect of Gonadectomy and Sex. Biomolecules 2022; 12:biom12060767. [PMID: 35740892 PMCID: PMC9220917 DOI: 10.3390/biom12060767] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 05/27/2022] [Accepted: 05/29/2022] [Indexed: 01/02/2023] Open
Abstract
Sex steroids, derived mainly from gonads, can shape microbiota composition; however, the impact of gonadectomy and sex on steroid production in the gut (i.e., gut steroids), and its interaction with microbiota composition, needs to be clarified. In this study, steroid environment and gut steroidogenesis were analysed by liquid chromatography tandem mass spectrometry and expression analyses. Gut microbiota composition as branched- and short-chain fatty acids were determined by 16S rRNA gene sequence analysis and gas chromatography flame ionisation detection, respectively. Here, we first demonstrated that levels of pregnenolone (PREG), progesterone (PROG), and isoallopregnanolone (ISOALLO) were higher in the female rat colon, whereas the level of testosterone (T) was higher in males. Sexual dimorphism on gut steroidogenesis is also reported after gonadectomy. Sex, and more significantly, gonadectomy, affects microbiota composition. We noted that a number of taxa and inferred metabolic pathways were associated with gut steroids, such as positive associations between Blautia with T, dihydroprogesterone (DHP), and allopregnanolone (ALLO), whereas negative associations were noted between Roseburia and T, ALLO, PREG, ISOALLO, DHP, and PROG. In conclusion, this study highlights the novel sex-specific association between microbiota and gut steroids with possible relevance for the gut-brain axis.
Collapse
Affiliation(s)
- Silvia Diviccaro
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133 Milano, Italy; (S.D.); (L.C.); (E.F.); (S.G.); (D.C.)
| | - Jamie A. FitzGerald
- Teagasc Food Research Centre, Moorepark, P61 C996 Cork, Ireland; (J.A.F.); (F.C.); (P.D.C.)
- APC Microbiome Ireland, University College Cork, College Road, T12 YT20 Cork, Ireland;
| | - Lucia Cioffi
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133 Milano, Italy; (S.D.); (L.C.); (E.F.); (S.G.); (D.C.)
| | - Eva Falvo
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133 Milano, Italy; (S.D.); (L.C.); (E.F.); (S.G.); (D.C.)
| | - Fiona Crispie
- Teagasc Food Research Centre, Moorepark, P61 C996 Cork, Ireland; (J.A.F.); (F.C.); (P.D.C.)
- APC Microbiome Ireland, University College Cork, College Road, T12 YT20 Cork, Ireland;
| | - Paul D. Cotter
- Teagasc Food Research Centre, Moorepark, P61 C996 Cork, Ireland; (J.A.F.); (F.C.); (P.D.C.)
- APC Microbiome Ireland, University College Cork, College Road, T12 YT20 Cork, Ireland;
| | - Siobhain M. O’Mahony
- APC Microbiome Ireland, University College Cork, College Road, T12 YT20 Cork, Ireland;
| | - Silvia Giatti
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133 Milano, Italy; (S.D.); (L.C.); (E.F.); (S.G.); (D.C.)
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133 Milano, Italy; (S.D.); (L.C.); (E.F.); (S.G.); (D.C.)
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133 Milano, Italy; (S.D.); (L.C.); (E.F.); (S.G.); (D.C.)
- Correspondence:
| |
Collapse
|
115
|
Sobstyl M, Brecht P, Sobstyl A, Mertowska P, Grywalska E. The Role of Microbiota in the Immunopathogenesis of Endometrial Cancer. Int J Mol Sci 2022; 23:ijms23105756. [PMID: 35628566 PMCID: PMC9143279 DOI: 10.3390/ijms23105756] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 02/01/2023] Open
Abstract
The female reproductive tract hosts a specific microbiome, which plays a crucial role in sustaining equilibrium and good health. In the majority of reproductive women, the microbiota (all bacteria, viruses, fungi, and other single-celled organisms within the human body) of the vaginal and cervical microenvironment are dominated by Lactobacillus species, which benefit the host through symbiotic relationships, in comparison to the uterus, fallopian tubes, and ovaries, which may contain a low-biomass microbiome with a diverse mixture of microorganisms. Although disruption to the balance of the microbiota develops, the altered immune and metabolic signaling may cause an impact on diseases such as cancer. These pathophysiological modifications in the gut–uterus axis may spark gynecological cancers. New information displays that gynecological and gastrointestinal tract dysbiosis (disruption of the microbiota homeostasis) can play an active role in the advancement and metastasis of gynecological neoplasms, such as cervical, endometrial, and ovarian cancers. Understanding the relationship between microbiota and endometrial cancer is critical for prognosis, diagnosis, prevention, and the development of innovative treatments. Identifying a specific microbiome may become an effective method for characterization of the specific microbiota involved in endometrial carcinogenesis. The aim of this study was to summarize the current state of knowledge that describes the correlation of microbiota with endometrial cancer with regard to the formation of immunological pathologies.
Collapse
Affiliation(s)
- Małgorzata Sobstyl
- Department of Gynecology and Gynecological Endocrinology, Medical University of Lublin, 20-037 Lublin, Poland;
| | - Peet Brecht
- Department of Experimental Immunology, Medical University of Lublin, Chodźki 4a St., 20-093 Lublin, Poland; (P.B.); (A.S.)
| | - Anna Sobstyl
- Department of Experimental Immunology, Medical University of Lublin, Chodźki 4a St., 20-093 Lublin, Poland; (P.B.); (A.S.)
| | - Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, Chodźki 4a St., 20-093 Lublin, Poland; (P.B.); (A.S.)
- Correspondence: (P.M.); (E.G.)
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, Chodźki 4a St., 20-093 Lublin, Poland; (P.B.); (A.S.)
- Correspondence: (P.M.); (E.G.)
| |
Collapse
|
116
|
Personalized Nutrition in the Management of Female Infertility: New Insights on Chronic Low-Grade Inflammation. Nutrients 2022; 14:nu14091918. [PMID: 35565885 PMCID: PMC9105997 DOI: 10.3390/nu14091918] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/19/2022] [Accepted: 04/29/2022] [Indexed: 02/01/2023] Open
Abstract
Increasing evidence on the significance of nutrition in reproduction is emerging from both animal and human studies, suggesting a mutual association between nutrition and female fertility. Different “fertile” dietary patterns have been studied; however, in humans, conflicting results or weak correlations are often reported, probably because of the individual variations in genome, proteome, metabolome, and microbiome and the extent of exposure to different environmental conditions. In this scenario, “precision nutrition”, namely personalized dietary patterns based on deep phenotyping and on metabolomics, microbiome, and nutrigenetics of each case, might be more efficient for infertile patients than applying a generic nutritional approach. In this review, we report on new insights into the nutritional management of infertile patients, discussing the main nutrigenetic, nutrigenomic, and microbiomic aspects that should be investigated to achieve effective personalized nutritional interventions. Specifically, we will focus on the management of low-grade chronic inflammation, which is associated with several infertility-related diseases.
Collapse
|
117
|
Läsche M, Gallwas J, Gründker C. Like Brothers in Arms: How Hormonal Stimuli and Changes in the Metabolism Signaling Cooperate, Leading HPV Infection to Drive the Onset of Cervical Cancer. Int J Mol Sci 2022; 23:5050. [PMID: 35563441 PMCID: PMC9103757 DOI: 10.3390/ijms23095050] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 02/06/2023] Open
Abstract
Despite all precautionary actions and the possibility of using vaccinations to counteract infections caused by human papillomaviruses (HPVs), HPV-related cancers still account for approximately 5% of all carcinomas. Worldwide, many women are still excluded from adequate health care due to their social position and origin. Therefore, immense efforts in research and therapy are still required to counteract the challenges that this disease entails. The special thing about an HPV infection is that it is not only able to trick the immune system in a sophisticated way, but also, through genetic integration into the host genome, to use all the resources available to the host cells to complete the replication cycle of the virus without activating the alarm mechanisms of immune recognition and elimination. The mechanisms utilized by the virus are the metabolic, immune, and hormonal signaling pathways that it manipulates. Since the virus is dependent on replication enzymes of the host cells, it also intervenes in the cell cycle of the differentiating keratinocytes and shifts their terminal differentiation to the uppermost layers of the squamocolumnar transformation zone (TZ) of the cervix. The individual signaling pathways are closely related and equally important not only for the successful replication of the virus but also for the onset of cervical cancer. We will therefore analyze the effects of HPV infection on metabolic signaling, as well as changes in hormonal and immune signaling in the tumor and its microenvironment to understand how each level of signaling interacts to promote tumorigenesis of cervical cancer.
Collapse
Affiliation(s)
| | | | - Carsten Gründker
- Department of Gynecology and Obstetrics, University Medicine Göttingen, 37075 Göttingen, Germany; (M.L.); (J.G.)
| |
Collapse
|
118
|
Edwinson AL, Yang L, Peters S, Hanning N, Jeraldo P, Jagtap P, Simpson JB, Yang TY, Kumar P, Mehta S, Nair A, Breen-Lyles M, Chikkamenahalli L, Graham RP, De Winter B, Patel R, Dasari S, Kashyap P, Griffin T, Chen J, Farrugia G, Redinbo MR, Grover M. Gut microbial β-glucuronidases regulate host luminal proteases and are depleted in irritable bowel syndrome. Nat Microbiol 2022; 7:680-694. [PMID: 35484230 PMCID: PMC9081267 DOI: 10.1038/s41564-022-01103-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 03/09/2022] [Indexed: 12/13/2022]
Abstract
Intestinal proteases mediate digestion and immune signaling, while increased gut proteolytic activity disrupts the intestinal barrier and generates visceral hypersensitivity, which in common in irritable bowel syndrome (IBS). However, the mechanisms controlling protease function are unclear. Here we show that members of the gut microbiota suppress intestinal proteolytic activity through production of unconjugated bilirubin. This occurs via microbial β-glucuronidase-mediated conversion of bilirubin conjugates. Metagenomic analysis of fecal samples from patients with post-infection IBS (n=52) revealed an altered gut microbiota composition, in particular a reduction in Alistipes taxa, and high gut proteolytic activity driven by specific host serine proteases compared to controls. Germ-free mice showed 10-fold higher proteolytic activity compared with conventional mice. Colonization with microbiota from high proteolytic activity IBS patients failed to suppress proteolytic activity in germ-free mice, but suppression of proteolytic activity was achieved with colonization using microbiota from healthy donors. High proteolytic activity mice had higher intestinal permeability, a higher relative abundance of Bacteroides and a reduction in Alistipes taxa compared with low proteolytic activity mice. High proteolytic activity IBS patients had lower fecal β-glucuronidase activity and end-products of bilirubin deconjugation. Mice treated with unconjugated bilirubin and β-glucuronidase overexpressing E. coli, which significantly reduced proteolytic activity, while inhibitors of microbial β-glucuronidases increased proteolytic activity. Together, these data define a disease-relevant mechanism of host-microbial interaction that maintains protease homeostasis in the gut.
Collapse
Affiliation(s)
- Adam L Edwinson
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Lu Yang
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Stephanie Peters
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Nikita Hanning
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA.,Laboratory of Experimental Medicine and Pediatrics and Infla-Med, research center of excellence, University of Antwerp, Antwerp, Belgium
| | | | - Pratik Jagtap
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Joshua B Simpson
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Tzu-Yi Yang
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Praveen Kumar
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Subina Mehta
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Asha Nair
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | | | | | - Rondell P Graham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Benedicte De Winter
- Laboratory of Experimental Medicine and Pediatrics and Infla-Med, research center of excellence, University of Antwerp, Antwerp, Belgium.,Division of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| | - Robin Patel
- Division of Clinical Microbiology, Mayo Clinic, Rochester, MN, USA
| | - Surendra Dasari
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Purna Kashyap
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Timothy Griffin
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Jun Chen
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Gianrico Farrugia
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Matthew R Redinbo
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA.,Departments of Biochemistry and Biophysics, and Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Madhusudan Grover
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
119
|
Groestlinger J, Spindler V, Pahlke G, Rychlik M, Del Favero G, Marko D. Alternaria alternata Mycotoxins Activate the Aryl Hydrocarbon Receptor and Nrf2-ARE Pathway to Alter the Structure and Immune Response of Colon Epithelial Cells. Chem Res Toxicol 2022; 35:731-749. [PMID: 35405071 PMCID: PMC9115800 DOI: 10.1021/acs.chemrestox.1c00364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
After ingestion of food commodities, the gastrointestinal tract (GIT) poses the first barrier against xenobiotics and pathogens. Therefore, it is regularly confronted with external stressors potentially affecting the inflammatory response and the epithelial barrier. Alternaria mycotoxins such as alternariol (AOH) and altertoxin II (ATX-II) are frequently occurring food and feed contaminants that are described for their immunomodulatory capacities. Hence, this study aimed at exploring the effect of AOH and ATX-II as single compounds or binary mixtures on the immune response and epithelial homeostasis in noncancerous colon epithelial cells HCEC-1CT. Both toxins suppressed mRNA levels of proinflammatory mediators interleukin-8 (IL-8), tumor necrosis factor α (TNF-α), and secretion of IL-8, as well as mRNA levels of the matrix metallopeptidase 2 (MMP-2). Binary combinations of AOH and ATX-II reduced the response of the single toxins. Additionally, AOH and ATX-II modified immunolocalization of transmembrane proteins such as integrin β1, zona occludens 1 (ZO-1), claudin 4 (Cldn 4), and occludin (Ocln), which support colonic tissue homeostasis and intestinal barrier function. Moreover, the cellular distribution of ZO-1 was affected by ATX-II. Mechanistically, these effects could be traced back to the involvement of several transcription factors. AOH activated the nuclear translocation of the aryl hydrocarbon receptor (AhR) and the nuclear factor erythroid 2-related factor 2 (Nrf2), governing cell metabolic competence and structural integrity. This was accompanied by altered distribution of the NF-κB p65 protein, an important regulator of inflammatory response. ATX-II also induced AhR and Nrf2 translocation, albeit failing to substantiate the effect of AOH on the colonic epithelium. Hence, both toxins coherently repress the intestinal immune response on the cytokine transcriptional and protein levels. Furthermore, both mycotoxins affected the colonic epithelial integrity by altering the cell architecture.
Collapse
Affiliation(s)
- Julia Groestlinger
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währinger Straße 38, 1090 Vienna, Austria
| | - Veronika Spindler
- Chair of Food Analytical Chemistry, Technical University of Munich, Maximus-von-Imhof-Forum 2, 85354 Freising, Germany
| | - Gudrun Pahlke
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währinger Straße 38, 1090 Vienna, Austria
| | - Michael Rychlik
- Chair of Food Analytical Chemistry, Technical University of Munich, Maximus-von-Imhof-Forum 2, 85354 Freising, Germany
| | - Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währinger Straße 38, 1090 Vienna, Austria.,Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna, Währinger Straße 38, 1090 Vienna, Austria
| | - Doris Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währinger Straße 38, 1090 Vienna, Austria
| |
Collapse
|
120
|
An J, Kwon H, Lim W, Moon BI. Staphylococcus aureus-Derived Extracellular Vesicles Enhance the Efficacy of Endocrine Therapy in Breast Cancer Cells. J Clin Med 2022; 11:jcm11072030. [PMID: 35407638 PMCID: PMC9000115 DOI: 10.3390/jcm11072030] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 02/04/2023] Open
Abstract
The microbiome involved in the human estrogen metabolism is known as the estrobolome. This study aimed to show that the estrobolome can be used in breast cancer treatment. We first analyzed the blood microbiome composition of healthy controls and patients with breast cancer. In particular, we investigated the bacteria producing β-glucuronidase and/or β-galactosidase, which are involved in estrogen metabolism in the human body. Staphylococcus species were more abundant in healthy controls than in breast cancer patients and therefore were selected for further analyses. The effect of Staphylococcus aureus on endocrine therapy was analyzed by a combination treatment with tamoxifen. Analysis of the microbiome of blood samples showed that species producing β-glucuronidase were more abundant in breast cancer patients than in healthy controls. Further experiments confirmed that the efficacy of tamoxifen increased when administered in conjugation with the extracellular vesicles (EVs) of S. aureus. Based on our results, we deduced that S. aureus EVs could potentially be used as adjuvants for breast cancer treatment in the future.
Collapse
Affiliation(s)
- Jeongshin An
- Department of Surgery, Ewha Womans University Mokdong Hospital, School of Medicine, Ewha Womans University, 1071 Anyangcheon-ro, Yangcheon-gu, Seoul 07985, Korea; (J.A.); (H.K.); (W.L.)
- Institute of Convergence Medicine Research, Ewha Womans University Mokdong Hospital, School of Medicine, Ewha Womans University, 1071 Anyangcheon-ro, Yangcheon-gu, Seoul 07985, Korea
| | - Hyungju Kwon
- Department of Surgery, Ewha Womans University Mokdong Hospital, School of Medicine, Ewha Womans University, 1071 Anyangcheon-ro, Yangcheon-gu, Seoul 07985, Korea; (J.A.); (H.K.); (W.L.)
| | - Woosung Lim
- Department of Surgery, Ewha Womans University Mokdong Hospital, School of Medicine, Ewha Womans University, 1071 Anyangcheon-ro, Yangcheon-gu, Seoul 07985, Korea; (J.A.); (H.K.); (W.L.)
| | - Byung-In Moon
- Department of Surgery, Ewha Womans University Mokdong Hospital, School of Medicine, Ewha Womans University, 1071 Anyangcheon-ro, Yangcheon-gu, Seoul 07985, Korea; (J.A.); (H.K.); (W.L.)
- Correspondence: ; Tel.: +82-2-2650-5584; Fax: +82-2-2644-7984
| |
Collapse
|
121
|
Di Renzo L, De Lorenzo A, Fontanari M, Gualtieri P, Monsignore D, Schifano G, Alfano V, Marchetti M. Immunonutrients involved in the regulation of the inflammatory and oxidative processes: implication for gamete competence. J Assist Reprod Genet 2022; 39:817-846. [PMID: 35353297 PMCID: PMC9050992 DOI: 10.1007/s10815-022-02472-6] [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: 11/17/2021] [Accepted: 03/16/2022] [Indexed: 11/30/2022] Open
Abstract
Purpose The purpose of this umbrella review is to bring together the most recent reviews concerning the role of immunonutrients for male and female infertility. Methods Regarding immunonutrients and fertility, the authors have analyzed reviews, systematic reviews, and meta-analyses published between 2011 and June 2021. All reviews on animal or in vitro studies were excluded. Relevant keywords to term micronutrients were analyzed alone or in association with other terms such as “gamete competence,” “male OR female fertility,” “male OR female infertility,” “fertile, “folliculogenesis,” “spermatogenesis,” “immunomodulation,” “immune system,” “oxidative stress.” Results The primary research has included 108 results, and after screening by title, abstract. and not topic-related, 41 studies have been included by full texts. The results show the molecular mechanisms and the immunonutrients related impact on gamete formation, development. and competence. In particular, this review focused on arginine, glutamine, vitamin C, vitamin D, vitamin E, omega-3, selenium, and zinc. Conclusions Inflammation and oxidative stress significantly impact human reproduction. For this reason, immunonutrients may play an important role in the treatment of infertile patients. However, due to the lack of consistent clinical trials, their application is limited. Therefore, the development of clinical trials is necessary to define the correct supplementation, in case of deficiency.
Collapse
Affiliation(s)
- Laura Di Renzo
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.,School of Specialization in Food Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.,Italian University Network for Sustainable Development (RUS), Food Working Group, University of Tor Vergata, Via Cracovia, 00133, Rome, Italy
| | - Antonino De Lorenzo
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Marco Fontanari
- School of Specialization in Food Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Paola Gualtieri
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.
| | - Diego Monsignore
- School of Specialization in Food Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Giulia Schifano
- School of Specialization in Food Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Valentina Alfano
- School of Specialization in Food Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Marco Marchetti
- PhD School of Applied Medical-Surgical Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | | |
Collapse
|
122
|
He QD, Guo JJ, Zhang Q, Yau YM, Yu Y, Zhong ZH, Tong ZY, Yang ZB, Chen M. Effects of Electroacupuncture on the Gut Microbiome in Cisplatin-Induced Premature Ovarian Failure Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:9352833. [PMID: 35321505 PMCID: PMC8938064 DOI: 10.1155/2022/9352833] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 01/24/2022] [Accepted: 02/08/2022] [Indexed: 12/17/2022]
Abstract
Growing evidence showed that the gut microbiota was associated with premature ovarian failure (POF). Many clinical types of research had shown that electroacupuncture was effective in the treatment of POF. However, there was little research on regulating the gut microbiome of POF mice by electroacupuncture. Therefore, this study attempted to verify whether electroacupuncture could regulate the gut microbiome in POF mice. POF mice were established by being injected intraperitoneally with cisplatin (2 mg/kg) for 2 weeks. Guanyuan (CV4) and Sanyinjiao (SP6) were selected in the electroacupuncture-at-the-acupoints group (EA group). Nonacupoints around CV4 and SP6 were selected in the electroacupuncture-at-the-nonacupoints group (EN group). The EA group and EN group were treated for 3 weeks. The ovarian function was evaluated by histopathological and molecular assays. Meanwhile, the gut microbiome of all mice was detected by 16S rDNA sequencing. The results showed that EA could restore the estrous cycle and reduce the number of atresia follicles in POF mice. The levels of serum follicle-stimulating hormone and luteinizing hormone were decreased by EA. As well, the levels of serum estradiol, anti-Mullerian hormone, and β-glucuronidase were increased by EA. The relative expressions of PI3K, AKT, and mTOR were increased to promote the proliferation of ovarian cells in the EA group. According to the results of 16S rDNA sequencing, the abundance and diversity of the gut microbiome could be regulated by EA. The relative abundance of beneficial bacteria was increased by EA. The KEGG pathway analysis showed that the gut microbiome associated with the estrogen signaling pathway, oocyte maturation, and PI3K-AKT signaling pathway was regulated by EA.
Collapse
Affiliation(s)
- Qi-da He
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
| | - Jing-jing Guo
- Department of Traditional Chinese Medicine, Xiamen University, Xiamen 361000, China
| | - Qi Zhang
- Department of Traditional Chinese Medicine, Xiamen University, Xiamen 361000, China
| | - Yuen-ming Yau
- Department of Traditional Chinese Medicine, Xiamen University, Xiamen 361000, China
| | - Yue Yu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
| | - Zheng-hong Zhong
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
| | - Zi-yan Tong
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
| | - Zong-bao Yang
- Department of Traditional Chinese Medicine, Xiamen University, Xiamen 361000, China
| | - Min Chen
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
| |
Collapse
|
123
|
Candeliere F, Raimondi S, Ranieri R, Musmeci E, Zambon A, Amaretti A, Rossi M. β-Glucuronidase Pattern Predicted From Gut Metagenomes Indicates Potentially Diversified Pharmacomicrobiomics. Front Microbiol 2022; 13:826994. [PMID: 35308380 PMCID: PMC8928169 DOI: 10.3389/fmicb.2022.826994] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/21/2022] [Indexed: 11/16/2022] Open
Abstract
β-glucuronidases (GUS) of intestinal bacteria remove glucuronic acid from glucoronides, reversing phase II metabolism of the liver and affecting the level of active deconjugated metabolites deriving from drugs or xenobiotics. Two hundred seventy-nine non-redundant GUS sequences are known in the gut microbiota, classified in seven structural categories (NL, L1, L2, mL1, mL2, mL1,2, and NC) with different biocatalytic properties. In the present study, the intestinal metagenome of 60 healthy subjects from five geographically different cohorts was assembled, binned, and mined to determine qualitative and quantitative differences in GUS profile, potentially affecting response to drugs and xenobiotics. Each metagenome harbored 4–70 different GUS, altogether accounting for 218. The amount of intestinal bacteria with at least one GUS gene was highly variable, from 0.7 to 82.2%, 25.7% on average. No significant difference among cohorts could be identified, except for the Ethiopia (ETH) cohort where GUS-encoding bacteria were significantly less abundant. The structural categories were differently distributed among the metagenomes, but without any statistical significance related to the cohorts. GUS profiles were generally dominated by the category NL, followed by mL1, L2, and L1. The GUS categories most involved in the hydrolysis of small molecules, including drugs, are L1 and mL1. Bacteria contributing to these categories belonged to Bacteroides ovatus, Bacteroides dorei, Bacteroides fragilis, Escherichia coli, Eubacterium eligens, Faecalibacterium prausnitzii, Parabacteroides merdae, and Ruminococcus gnavus. Bacteria harboring L1 GUS were generally scarcely abundant (<1.3%), except in three metagenomes, where they reached up to 24.3% for the contribution of E. coli and F. prausnitzii. Bacteria harboring mL1 GUS were significantly more abundant (mean = 4.6%), with Bacteroides representing a major contributor. Albeit mL1 enzymes are less active than L1 ones, Bacteroides likely plays a pivotal role in the deglucuronidation, due to its remarkable abundance in the microbiomes. The observed broad interindividual heterogeneity of GUS profiles, particularly of the L1 and mL1 categories, likely represent a major driver of pharmacomicrobiomics variability, affecting drug response and toxicity. Different geographical origins, genetic, nutritional, and lifestyle features of the hosts seemed not to be relevant in the definition of glucuronidase activity, albeit they influenced the richness of the GUS profile.
Collapse
Affiliation(s)
- Francesco Candeliere
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Stefano Raimondi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Raffaella Ranieri
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Eliana Musmeci
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alfonso Zambon
- Department of Chemistry and Geological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alberto Amaretti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Biogest-Siteia, University of Modena and Reggio Emilia, Modena, Italy
| | - Maddalena Rossi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Biogest-Siteia, University of Modena and Reggio Emilia, Modena, Italy
- *Correspondence: Maddalena Rossi,
| |
Collapse
|
124
|
Łaniewski P, Herbst-Kralovetz MM. Connecting microbiome and menopause for healthy ageing. Nat Microbiol 2022; 7:354-358. [PMID: 35246661 PMCID: PMC9977513 DOI: 10.1038/s41564-022-01071-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Understanding the interplay between the microbiome and menopause holds promise for new interventions to alleviate menopausal symptoms and improve quality of life for women.
Collapse
Affiliation(s)
- Paweł Łaniewski
- Department of Basic Medical Sciences, College of Medicine–Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Melissa M. Herbst-Kralovetz
- Department of Basic Medical Sciences, College of Medicine–Phoenix, University of Arizona, Phoenix, AZ, USA.,Department of Obstetrics and Gynecology, College of Medicine–Phoenix, University of Arizona, Phoenix, AZ, USA.,Correspondence:
| |
Collapse
|
125
|
Pretorius L, Van Staden ADP, Van der Merwe JJ, Henning N, Smith C. Alterations to microbial secretome by estrogen may contribute to sex bias in irritable bowel syndrome. Inflammopharmacology 2022; 30:267-281. [PMID: 35022916 DOI: 10.1007/s10787-021-00906-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/30/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Irritable bowel syndrome (IBS) is a female predominant functional gastrointestinal disorder, underpinned by microbial dysbiosis and microinflammation. We suggest that changes in trace amine (TA) load and metabolism may link together diet, inflammation and sex in this context. METHODS The effect of E2 treatment on microbial growth and TA generation was assessed using liquid chromatography and tandem mass spectrometry methodology. To investigate the effects of TAs on the gut, WST-1, prostaglandin E2 and tight junction protein dynamics were investigated in TA treated (HT-29) colon epithelial monolayer cultures. RESULTS Differential E2-dependent alterations of the TA production capabilities of microbes were observed. Significantly, E2 treatment resulted in a 50% increase in tryptamine secretion from a probiotic microbe (p < 0.0001). Moreover, in vitro experiments indicated that TA treatment exerted type-specific effects in the gut, e.g., reducing mitochondrial functionality, even at low doses of tryptamine (p < 0.0001) and ρ-tyramine (p < 0.001). Additionally, prostaglandin E2 levels were significantly increased following ρ-tyramine and agmatine treatment (p < 0.05). In terms of functionality, all investigated TAs resulted in occludin redistribution and loss of zona occludens-1 and occludin co-localization. CONCLUSION Increases in the gastrointestinal TA load may contribute to a relatively pro-inflammatory outcome in the intestine, along with tight junction protein disruption. Additionally, fluctuating levels of endogenous E2 may modulate microbially-derived TA levels, potentially explaining exaggerating gastrointestinal symptomology in females during low E2 phases. Thus, current data warrants subsequent investigations in appropriate in vivo models to fully elucidate the role of the trace aminergic system in the sex bias observed in IBS.
Collapse
Affiliation(s)
- Lesha Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Anton du Preez Van Staden
- Department of Microbiology, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa.,Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Johannes J Van der Merwe
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa.,LabSPACE, Midrand, South Africa
| | - Natasha Henning
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Carine Smith
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa.
| |
Collapse
|
126
|
Zhang J, Walker ME, Sanidad KZ, Zhang H, Liang Y, Zhao E, Chacon-Vargas K, Yeliseyev V, Parsonnet J, Haggerty TD, Wang G, Simpson JB, Jariwala PB, Beaty VV, Yang J, Yang H, Panigrahy A, Minter LM, Kim D, Gibbons JG, Liu L, Li Z, Xiao H, Borlandelli V, Overkleeft HS, Cloer EW, Major MB, Goldfarb D, Cai Z, Redinbo MR, Zhang G. Microbial enzymes induce colitis by reactivating triclosan in the mouse gastrointestinal tract. Nat Commun 2022; 13:136. [PMID: 35013263 PMCID: PMC8748916 DOI: 10.1038/s41467-021-27762-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 11/24/2021] [Indexed: 12/24/2022] Open
Abstract
Emerging research supports that triclosan (TCS), an antimicrobial agent found in thousands of consumer products, exacerbates colitis and colitis-associated colorectal tumorigenesis in animal models. While the intestinal toxicities of TCS require the presence of gut microbiota, the molecular mechanisms involved have not been defined. Here we show that intestinal commensal microbes mediate metabolic activation of TCS in the colon and drive its gut toxicology. Using a range of in vitro, ex vivo, and in vivo approaches, we identify specific microbial β-glucuronidase (GUS) enzymes involved and pinpoint molecular motifs required to metabolically activate TCS in the gut. Finally, we show that targeted inhibition of bacterial GUS enzymes abolishes the colitis-promoting effects of TCS, supporting an essential role of specific microbial proteins in TCS toxicity. Together, our results define a mechanism by which intestinal microbes contribute to the metabolic activation and gut toxicity of TCS, and highlight the importance of considering the contributions of the gut microbiota in evaluating the toxic potential of environmental chemicals.
Collapse
Affiliation(s)
- Jianan Zhang
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Morgan E Walker
- Departments of Chemistry, Biochemistry, Microbiology and Genomics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Hongna Zhang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, SAR, China
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Yanshan Liang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Ermin Zhao
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | | | - Vladimir Yeliseyev
- Massachusetts Host-Microbiota Center, Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Julie Parsonnet
- Department of Medicine and Department of Health Research and Policy, Stanford University, Stanford, CA, USA
| | - Thomas D Haggerty
- Department of Medicine and Department of Health Research and Policy, Stanford University, Stanford, CA, USA
| | - Guangqiang Wang
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Joshua B Simpson
- Departments of Chemistry, Biochemistry, Microbiology and Genomics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Parth B Jariwala
- Departments of Chemistry, Biochemistry, Microbiology and Genomics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Violet V Beaty
- Departments of Chemistry, Biochemistry, Microbiology and Genomics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jun Yang
- Department of Entomology and Nematology, University of California, Davis, CA, USA
| | - Haixia Yang
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Anand Panigrahy
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Lisa M Minter
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Daeyoung Kim
- Department of Mathematics and Statistics, University of Massachusetts, Amherst, MA, USA
| | - John G Gibbons
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - LinShu Liu
- Eastern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, Wyndmoor, PA, USA
| | - Zhengze Li
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Valentina Borlandelli
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Hermen S Overkleeft
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Erica W Cloer
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael B Major
- Department of Cell Biology and Physiology, and Department of Otolaryngology, Washington University, St. Louis, MO, USA
| | - Dennis Goldfarb
- Department of Cell Biology and Physiology, Institute for Informatics, Washington University, St. Louis, MO, USA
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, SAR, China.
| | - Matthew R Redinbo
- Departments of Chemistry, Biochemistry, Microbiology and Genomics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Guodong Zhang
- Department of Food Science, University of Massachusetts, Amherst, MA, USA.
- Department of Food Science and Technology, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
127
|
Simpson JB, Sekela JJ, Graboski AL, Borlandelli VB, Bivins MM, Barker NK, Sorgen AA, Mordant AL, Johnson RL, Bhatt AP, Fodor AA, Herring LE, Overkleeft H, Lee JR, Redinbo MR. Metagenomics combined with activity-based proteomics point to gut bacterial enzymes that reactivate mycophenolate. Gut Microbes 2022; 14:2107289. [PMID: 35953888 PMCID: PMC9377255 DOI: 10.1080/19490976.2022.2107289] [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: 04/28/2022] [Accepted: 07/19/2022] [Indexed: 02/04/2023] Open
Abstract
Mycophenolate mofetil (MMF) is an important immunosuppressant prodrug prescribed to prevent organ transplant rejection and to treat autoimmune diseases. MMF usage, however, is limited by severe gastrointestinal toxicity that is observed in approximately 45% of MMF recipients. The active form of the drug, mycophenolic acid (MPA), undergoes extensive enterohepatic recirculation by bacterial β-glucuronidase (GUS) enzymes, which reactivate MPA from mycophenolate glucuronide (MPAG) within the gastrointestinal tract. GUS enzymes demonstrate distinct substrate preferences based on their structural features, and gut microbial GUS enzymes that reactivate MPA have not been identified. Here, we compare the fecal microbiomes of transplant recipients receiving MMF to healthy individuals using shotgun metagenomic sequencing. We find that neither microbial composition nor the presence of specific structural classes of GUS genes are sufficient to explain the differences in MPA reactivation measured between fecal samples from the two cohorts. We next employed a GUS-specific activity-based chemical probe and targeted metaproteomics to identify and quantify the GUS proteins present in the human fecal samples. The identification of specific GUS enzymes was improved by using the metagenomics data collected from the fecal samples. We found that the presence of GUS enzymes that bind the flavin mononucleotide (FMN) is significantly correlated with efficient MPA reactivation. Furthermore, structural analysis identified motifs unique to these FMN-binding GUS enzymes that provide molecular support for their ability to process this drug glucuronide. These results indicate that FMN-binding GUS enzymes may be responsible for reactivation of MPA and could be a driving force behind MPA-induced GI toxicity.
Collapse
Affiliation(s)
- Joshua B. Simpson
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Josh J. Sekela
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Amanda L. Graboski
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Valentina B. Borlandelli
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Marissa M. Bivins
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Natalie K. Barker
- UNC Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alicia A. Sorgen
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Angie L. Mordant
- UNC Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rebecca L. Johnson
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Aadra P. Bhatt
- Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, and the Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anthony A. Fodor
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Laura E. Herring
- UNC Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hermen Overkleeft
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - John R. Lee
- Department of Medicine, Division of Nephrology and Hypertension, New York, New York, USA
| | - Matthew. R. Redinbo
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, Department of Microbiology and Immunology, and the Institute for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
128
|
Gu Y, Zhou G, Zhou F, Li Y, Wu Q, He H, Zhang Y, Ma C, Ding J, Hua K. Gut and Vaginal Microbiomes in PCOS: Implications for Women's Health. Front Endocrinol (Lausanne) 2022; 13:808508. [PMID: 35282446 PMCID: PMC8905243 DOI: 10.3389/fendo.2022.808508] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 02/01/2022] [Indexed: 12/12/2022] Open
Abstract
PCOS is defined as a kind of endocrine and metabolic disorder which affects females at reproductive ages, is becoming much more common, nowadays. Microbiomes are known as microorganisms that inhabit the body to play a vital role in human health. In recent years, several basic and clinical studies have tried to investigate the correlation between the reproductive health/disorder and microbiomes (gut microbiomes and vaginal microbiomes). However, the mechanism is still unclear. In this review, we reviewed the relationship between PCOS and microbiomes, including gut/vaginal microbiomes compositions in PCOS, mechanism of microbiomes and PCOS, and then collectively focused on the recent findings on the influence of microbiomes on the novel insight regarding the therapeutic strategies for PCOS in the future clinical practice.
Collapse
Affiliation(s)
- Yuanyuan Gu
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, China
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Guannan Zhou
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Fangyue Zhou
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yao Li
- Department of Urology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Qiongwei Wu
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, China
| | - Hongyu He
- Department of Intensive Care Unit, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi Zhang
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, China
| | - Chengbin Ma
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, China
- *Correspondence: Chengbin Ma, ; Jingxin Ding, ; Keqin Hua,
| | - Jingxin Ding
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
- *Correspondence: Chengbin Ma, ; Jingxin Ding, ; Keqin Hua,
| | - Keqin Hua
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
- *Correspondence: Chengbin Ma, ; Jingxin Ding, ; Keqin Hua,
| |
Collapse
|
129
|
Fu C, Yang Z, Yu J, Wei M. The interaction between gut microbiome and anti-tumor drug therapy. Am J Cancer Res 2021; 11:5812-5832. [PMID: 35018227 PMCID: PMC8727820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/04/2021] [Indexed: 06/14/2023] Open
Abstract
A large number of symbiotic gut microbiome exists in the human gastrointestinal micro-ecosystem. The daily diet, lifestyle, and body constitution influence the type and quantity of gut microbiome in the body. Increasing evidence demonstrates that the gut microbiome can affect tumor development and progress. We discuss in this paper how the gut microbiome impacts tumor pathology through DNA damage, production of dietary and microbial metabolites, altered cellular signaling pathways, immune system suppression, and involvement in pro-inflammatory pathways changing gut microbiome composition. The gut microbiome acts on different types of the anti-tumor drug through bacterial translocation, immuno-modulation, metabolic modulation, enzymatic degradation, and reduction of microbial diversity. This article summarized the aforementioned by reviewing recent studies on the interaction among the gut microbiome, tumor development, and antitumor drugs.
Collapse
Affiliation(s)
- Chen Fu
- Department of Pharmacology, School of Pharmacy, China Medical UniversityShenyang 110122, P. R. China
| | - Ziting Yang
- Department of Pharmacology, School of Pharmacy, China Medical UniversityShenyang 110122, P. R. China
| | - Jiankun Yu
- Department of Pharmacology, School of Pharmacy, China Medical UniversityShenyang 110122, P. R. China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical UniversityShenyang 110122, P. R. China
- Liaoning Medical Diagnosis and Treatment CenterShenyang 110000, P. R. China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical UniversityShenyang 110122, P. R. China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical UniversityShenyang 110122, P. R. China
- Liaoning Medical Diagnosis and Treatment CenterShenyang 110000, P. R. China
| |
Collapse
|
130
|
Korpela K, Kallio S, Salonen A, Hero M, Kukkonen AK, Miettinen PJ, Savilahti E, Kohva E, Kariola L, Suutela M, Tarkkanen A, de Vos WM, Raivio T, Kuitunen M. Gut microbiota develop towards an adult profile in a sex-specific manner during puberty. Sci Rep 2021; 11:23297. [PMID: 34857814 PMCID: PMC8640005 DOI: 10.1038/s41598-021-02375-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 11/12/2021] [Indexed: 11/18/2022] Open
Abstract
Accumulating evidence indicates that gut microbiota may regulate sex-hormone levels in the host, with effects on reproductive health. Very little is known about the development of intestinal microbiota during puberty in humans. To assess the connection between pubertal timing and fecal microbiota, and to assess how fecal microbiota develop during puberty in comparison with adult microbiota, we utilized a Finnish allergy-prevention-trial cohort (Flora). Data collected at 13-year follow-up were compared with adult data from a different Finnish cohort. Among the 13-year-old participants we collected questionnaire information, growth data from school-health-system records and fecal samples from 148 participants. Reference adult fecal samples were received from the Health and Early Life Microbiota (HELMi) cohort (n = 840). Fecal microbiota were analyzed using 16S rRNA gene amplicon sequencing; the data were correlated with pubertal timing and compared with data on adult microbiota. Probiotic intervention in the allergy-prevention-trial cohort was considered as a confounding factor only. The main outcome was composition of the microbiota in relation to pubertal timing (time to/from peak growth velocity) in both sexes separately, and similarity to adult microbiota. In girls, fecal microbiota became more adult-like with pubertal progression (p = 0.009). No such development was observed in boys (p = 0.9). Both sexes showed a trend towards increasing relative abundance of estrogen-metabolizing Clostridia and decreasing Bacteroidia with pubertal development, but this was statistically significant in girls only (p = 0.03). In girls, pubertal timing was associated positively with exposure to cephalosporins prior to the age of 10. Our data support the hypothesis that gut microbiota, particularly members of Ruminococcaceae, may affect pubertal timing, possibly via regulating host sex-hormone levels. Trial registration The registration number for the allergy-prevention-trial cohort: ClinicalTrials.gov, NCT00298337, registered 1 March 2006—Retrospectively registered, https://clinicaltrials.gov/show/NCT00298337. The adult-comparison cohort (HELMi) is NCT03996304.
Collapse
Affiliation(s)
- Katri Korpela
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Haartmaninkatu 3, P.O. Box 21, 00014, Helsinki, Finland
| | - Sampo Kallio
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Stenbäckinkatu 9, P.O. Box 347, 00029, Helsinki, Finland.
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Haartmaninkatu 3, P.O. Box 21, 00014, Helsinki, Finland
| | - Matti Hero
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Stenbäckinkatu 9, P.O. Box 347, 00029, Helsinki, Finland
| | - Anna Kaarina Kukkonen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Stenbäckinkatu 9, P.O. Box 347, 00029, Helsinki, Finland
| | - Päivi J Miettinen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Stenbäckinkatu 9, P.O. Box 347, 00029, Helsinki, Finland
| | - Erkki Savilahti
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Stenbäckinkatu 9, P.O. Box 347, 00029, Helsinki, Finland
| | - Ella Kohva
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Stenbäckinkatu 9, P.O. Box 347, 00029, Helsinki, Finland
| | - Laura Kariola
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Stenbäckinkatu 9, P.O. Box 347, 00029, Helsinki, Finland
| | - Maria Suutela
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Stenbäckinkatu 9, P.O. Box 347, 00029, Helsinki, Finland
| | - Annika Tarkkanen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Stenbäckinkatu 9, P.O. Box 347, 00029, Helsinki, Finland
| | - Willem M de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Haartmaninkatu 3, P.O. Box 21, 00014, Helsinki, Finland.,Laboratory of Microbiology, Wageningen University, Stippeneng 4, P.O. Box 8033, 6700 EH, Wageningen, The Netherlands
| | - Taneli Raivio
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Stenbäckinkatu 9, P.O. Box 347, 00029, Helsinki, Finland.,Translational Stem Cell Biology and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Yliopistonkatu 3, P.O. Box 4, 00014, Helsinki, Finland
| | - Mikael Kuitunen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Stenbäckinkatu 9, P.O. Box 347, 00029, Helsinki, Finland
| |
Collapse
|
131
|
Kovács T, Mikó E, Ujlaki G, Yousef H, Csontos V, Uray K, Bai P. The involvement of oncobiosis and bacterial metabolite signaling in metastasis formation in breast cancer. Cancer Metastasis Rev 2021; 40:1223-1249. [PMID: 34967927 PMCID: PMC8825384 DOI: 10.1007/s10555-021-10013-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/15/2021] [Indexed: 12/15/2022]
Abstract
Breast cancer, the most frequent cancer in women, is characterized by pathological changes to the microbiome of breast tissue, the tumor, the gut, and the urinary tract. Changes to the microbiome are determined by the stage, grade, origin (NST/lobular), and receptor status of the tumor. This year is the 50th anniversary of when Hill and colleagues first showed that changes to the gut microbiome can support breast cancer growth, namely that the oncobiome can reactivate excreted estrogens. The currently available human and murine data suggest that oncobiosis is not a cause of breast cancer, but can support its growth. Furthermore, preexisting dysbiosis and the predisposition to cancer are transplantable. The breast's and breast cancer's inherent microbiome and the gut microbiome promote breast cancer growth by reactivating estrogens, rearranging cancer cell metabolism, bringing about a more inflammatory microenvironment, and reducing the number of tumor-infiltrating lymphocytes. Furthermore, the gut microbiome can produce cytostatic metabolites, the production of which decreases or blunts breast cancer. The role of oncobiosis in the urinary tract is largely uncharted. Oncobiosis in breast cancer supports invasion, metastasis, and recurrence by supporting cellular movement, epithelial-to-mesenchymal transition, cancer stem cell function, and diapedesis. Finally, the oncobiome can modify the pharmacokinetics of chemotherapeutic drugs. The microbiome provides novel leverage on breast cancer that should be exploited for better management of the disease.
Collapse
Affiliation(s)
- Tünde Kovács
- Department Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Edit Mikó
- Department Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Gyula Ujlaki
- Department Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Heba Yousef
- Department Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Viktória Csontos
- Department Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Karen Uray
- Department Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Peter Bai
- Department Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary.
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, 4032, Hungary.
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary.
| |
Collapse
|
132
|
Castejón P, Cabas I, Gómez V, Chaves-Pozo E, Cerezo-Ortega I, Moriñigo MÁ, Martínez-Manzanares E, Galindo-Villegas J, García-Ayala A. Vaccination of Gilthead Seabream After Continuous Xenoestrogen Oral Exposure Enhances the Gut Endobolome and Immune Status via GPER1. Front Immunol 2021; 12:742827. [PMID: 34721409 PMCID: PMC8551918 DOI: 10.3389/fimmu.2021.742827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022] Open
Abstract
In fish culture settings, the exogenous input of steroids is a matter of concern. Recently, we unveiled that in the gilthead seabream (Sparus aurata), the G protein-coupled estrogen receptor agonist G-1 (G1) and the endocrine disruptor 17α-ethinylestradiol (EE2) are potent modulators in polyreactive antibody production. However, the integral role of the microbiota upon immunity and antibody processing in response to the effect of EE2 remains largely unexplored. Here, juvenile seabreams continuously exposed for 84 days to oral G1 or EE2 mixed in the fish food were intraperitoneally (i.p.) immune primed on day 42 with the model antigen keyhole limpet hemocyanin (KLH). A critical panel of systemic and mucosal immune markers, serum VTG, and humoral, enzymatic, and bacteriolytic activities were recorded and correlated with gut bacterial metagenomic analysis 1 day post-priming (dpp). Besides, at 15 dpp, animals received a boost to investigate the possible generation of specific anti-KLH antibodies at the systemic and mucosal interphases by the end of the trial. On day 43, EE2 but not G1 induced a significant shift in the serum VTG level of naive fish. Simultaneously, significant changes in some immune enzymatic activities in the serum and gut mucus of the EE2-treated group were recorded. In comparison, the vaccine priming immunization resulted in an attenuated profile of most enzymatic activities in the same group. The gut genes qPCR analysis exhibited a related pattern, only emphasized by a significant shift in the EE2 group's il1b expression. The gut bacterial microbiome status underwent 16S rRNA dynamic changes in alpha diversity indices, only with the exposure to oral G1, supporting functional alterations on cellular processes, signaling, and lipid metabolism in the microbiota. By the same token, the immunization elevated the relative abundance of Fusobacteria only in the control group, while this phylum was depleted in both the treated groups. Remarkably, the immunization also promoted changes in the bacterial class Betaproteobacteria and the estrogen-associated genus Novosphingobium. Furthermore, systemic and mucosal KLH-specific immunoglobulin (Ig)M and IgT levels in the fully vaccinated fish showed only slight changes 84 days post-estrogenic oral administration. In summary, our results highlight the intrinsic relationship among estrogens, their associated receptors, and immunization in the ubiquitous fish immune regulation and the subtle but significant crosstalk with the gut endobolome.
Collapse
Affiliation(s)
- Pablo Castejón
- Department of Cell Biology and Histology, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia, Instituto Murciano de Investigacion Biosanitaria (IMIB), Centro de Investigacion Biomedica en Red Enfermedades Raras (CIBERER), Murcia, Spain
| | - Isabel Cabas
- Department of Cell Biology and Histology, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia, Instituto Murciano de Investigacion Biosanitaria (IMIB), Centro de Investigacion Biomedica en Red Enfermedades Raras (CIBERER), Murcia, Spain
| | - Victoria Gómez
- Department of Cell Biology and Histology, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia, Instituto Murciano de Investigacion Biosanitaria (IMIB), Centro de Investigacion Biomedica en Red Enfermedades Raras (CIBERER), Murcia, Spain
| | - Elena Chaves-Pozo
- Aquaculture Department, Oceanographic Center of Murcia, Spanish Institute of Oceanography (IEO-CSIC), Murcia, Spain
| | - Isabel Cerezo-Ortega
- Department of Microbiology, Faculty of Sciences, University of Malaga, Málaga, Spain
| | - Miguel Ángel Moriñigo
- Department of Microbiology, Faculty of Sciences, University of Malaga, Málaga, Spain
| | | | | | - Alfonsa García-Ayala
- Department of Cell Biology and Histology, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia, Instituto Murciano de Investigacion Biosanitaria (IMIB), Centro de Investigacion Biomedica en Red Enfermedades Raras (CIBERER), Murcia, Spain
| |
Collapse
|
133
|
Salliss ME, Farland LV, Mahnert ND, Herbst-Kralovetz MM. The role of gut and genital microbiota and the estrobolome in endometriosis, infertility and chronic pelvic pain. Hum Reprod Update 2021; 28:92-131. [PMID: 34718567 DOI: 10.1093/humupd/dmab035] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 08/25/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Endometriosis is a chronic, burdensome condition that is historically understudied. Consequently, there is a lack of understanding of the etiology of the disease and its associated symptoms, including infertility and chronic pelvic pain (CPP). Endometriosis development is influenced by estrogen metabolism and inflammation, which are modulated by several factors including the microbiome and the estrobolome (the collection of genes encoding estrogen-metabolizing enzymes in the gut microbiome). Therefore, there is increasing interest in understanding the role of microbiota in endometriosis etiology. OBJECTIVE AND RATIONALE To date, there is no cure for endometriosis and treatment options often are ineffective. This manuscript will review the potential relationship between the microbiome and endometriosis, infertility and CPP and highlight the available data on the microbiome in relation to endometriosis and its related symptoms. The overarching goal of this manuscript is to inform future microbiome research that will lead to a deeper understanding of the etiology of the disease and possible diagnostic modalities and treatments. The potential impact of the microbiome on estrogen regulation modulated by the estrobolome, as well as inflammation and other endometriosis-promoting mechanisms within the genital tract, will be reviewed. The methodological limitations of microbiome-related studies will be critically assessed to provide improved guidelines for future microbiome and clinical studies. SEARCH METHODS PubMed databases were searched using the following keywords: endometriosis AND microbiome, infertility AND microbiome, pelvic pain AND microbiome, IVF (in-vitro fertilization) AND microbiome, endometriosis AND infertility. Clinical and preclinical animal trials that were eligible for review, and related to microbiome and endometriosis, infertility or CPP were included. All available manuscripts were published in 2002-2021. OUTCOMES In total, 28 clinical and 6 animal studies were included in the review. In both human and animal studies, bacteria were enriched in endometriosis groups, although there was no clear consensus on specific microbiota compositions that were associated with endometriosis, and no studies included infertility or CPP with endometriosis. However, bacterial vaginosis-associated bacteria and Lactobacillus depletion in the cervicovaginal microbiome were associated with endometriosis and infertility in the majority (23/28) of studies. Interpretation of endometrial studies is limited owing to a variety of methodological factors, discussed in this review. In addition, metadata outlining antibiotic usage, age, race/ethnicity, menopausal status and timing of sample collection in relation to diagnosis of endometriosis was not consistently reported. Animal studies (6/6) support a bidirectional relationship between the gut microbiota and endometriosis onset and progression. WIDER IMPLICATIONS There is evidence that a dysbiotic gut or genital microbiota is associated with multiple gynecologic conditions, with mounting data supporting an association between the microbiome and endometriosis and infertility. These microbiomes likely play a role in the gut-brain axis, which further supports a putative association with the spectrum of symptoms associated with endometriosis, including infertility and CPP. Collectively, this review highlights the demand for more rigorous and transparent methodology and controls, consistency across the field, and inclusion of key demographic and clinical characteristics of disease and comparison participants. Rigorous study designs will allow for a better understanding of the potential role of the microbiome in endometriosis etiology and the relationship to other disorders of the female reproductive tract.
Collapse
Affiliation(s)
- Mary E Salliss
- Department of Obstetrics and Gynecology, University of Arizona-College of Medicine, Phoenix, AZ, USA.,Department of Biology and Biochemistry, Bath University, Bath, UK
| | - Leslie V Farland
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA.,Department of Obstetrics and Gynecology, University of Arizona-College of Medicine Tucson, Tucson, AZ, USA
| | - Nichole D Mahnert
- Department of Obstetrics and Gynecology, University of Arizona-College of Medicine, Phoenix, AZ, USA.,Department of Obstetrics and Gynecology, Banner-University Medical Center Phoenix, Phoenix, AZ, USA
| | - Melissa M Herbst-Kralovetz
- Department of Obstetrics and Gynecology, University of Arizona-College of Medicine, Phoenix, AZ, USA.,Department of Basic Medical Sciences, University of Arizona-College of Medicine, Phoenix, AZ, USA
| |
Collapse
|
134
|
Zeibich L, Koebele SV, Bernaud VE, Ilhan ZE, Dirks B, Northup-Smith SN, Neeley R, Maldonado J, Nirmalkar K, Files JA, Mayer AP, Bimonte-Nelson HA, Krajmalnik-Brown R. Surgical Menopause and Estrogen Therapy Modulate the Gut Microbiota, Obesity Markers, and Spatial Memory in Rats. Front Cell Infect Microbiol 2021; 11:702628. [PMID: 34660336 PMCID: PMC8515187 DOI: 10.3389/fcimb.2021.702628] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/24/2021] [Indexed: 02/06/2023] Open
Abstract
Menopause in human females and subsequent ovarian hormone deficiency, particularly concerning 17β-estradiol (E2), increase the risk for metabolic dysfunctions associated with obesity, diabetes type 2, cardiovascular diseases, and dementia. Several studies indicate that these disorders are also strongly associated with compositional changes in the intestinal microbiota; however, how E2 deficiency and hormone therapy affect the gut microbial community is not well understood. Using a rat model, we aimed to evaluate how ovariectomy (OVX) and subsequent E2 administration drive changes in metabolic health and the gut microbial community, as well as potential associations with learning and memory. Findings indicated that OVX-induced ovarian hormone deficiency and E2 treatment had significant impacts on several health-affecting parameters, including (a) the abundance of some intestinal bacterial taxa (e.g., Bifidobacteriaceae and Porphyromonadaceae), (b) the abundance of microbial short-chain fatty acids (SCFAs) (e.g., isobutyrate), (c) weight/BMI, and (d) high-demand spatial working memory following surgical menopause. Furthermore, exploratory correlations among intestinal bacteria abundance, cognition, and BMI underscored the putative influence of surgical menopause and E2 administration on gut-brain interactions. Collectively, this study showed that surgical menopause is associated with physiological and behavioral changes, and that E2-linked compositional changes in the intestinal microbiota might contribute to some of its related negative health consequences. Overall, this study provides novel insights into interactions among endocrine and gastrointestinal systems in the post-menopausal life stage that collectively alter the risk for the development and progression of cardiovascular, metabolic, and dementia-related diseases.
Collapse
Affiliation(s)
- Lydia Zeibich
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, United States
| | - Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| | - Victoria E Bernaud
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| | - Zehra Esra Ilhan
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, United States
| | - Blake Dirks
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, United States
| | - Steven N Northup-Smith
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| | - Rachel Neeley
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| | - Juan Maldonado
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, United States.,Genomics Core, Arizona State University, Tempe, AZ, United States
| | - Khemlal Nirmalkar
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, United States
| | - Julia A Files
- Division of Women's Health Internal Medicine, Mayo Clinic, Scottsdale, AZ, United States
| | - Anita P Mayer
- Division of Women's Health Internal Medicine, Mayo Clinic, Scottsdale, AZ, United States
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| | - Rosa Krajmalnik-Brown
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
135
|
Gjorgoska M, Rižner TL. Estrogens and the Schrödinger's Cat in the Ovarian Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13195011. [PMID: 34638494 PMCID: PMC8508344 DOI: 10.3390/cancers13195011] [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: 08/04/2021] [Revised: 09/29/2021] [Accepted: 10/02/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Ovarian cancer is a complex pathology for which we require effective screening and therapeutical strategies. Apart from the cancer cell portion, there exist plastic immune and non-immune cell populations, jointly constituting the context-adaptive tumor microenvironment, which is pivotal in tumorigenesis. Estrogens might be synthesized in the ovarian tumor tissue and actively contribute to the shaping of an immunosuppressive microenvironment. Current immune therapies have limited effectiveness as a multitude of factors influence the outcome. A thorough understanding of the ovarian cancer biology is crucial in the efforts to reestablish homeostasis. Abstract Ovarian cancer is a heterogeneous disease affecting the aging ovary, in concert with a complex network of cells and signals, together representing the ovarian tumor microenvironment. As in the “Schrödinger’s cat” thought experiment, the context-dependent constituents of the—by the time of diagnosis—well-established tumor microenvironment may display a tumor-protective and -destructive role. Systemic and locally synthesized estrogens contribute to the formation of a pro-tumoral microenvironment that enables the sustained tumor growth, invasion and metastasis. Here we focus on the estrogen biosynthetic and metabolic pathways in ovarian cancer and elaborate their actions on phenotypically plastic, estrogen-responsive, aging immune cells of the tumor microenvironment, altogether highlighting the multicomponent-connectedness and complexity of cancer, and contributing to a broader understanding of the ovarian cancer biology.
Collapse
|
136
|
Deng Z, Zhang Q, Zhao Z, Li Y, Chen X, Lin Z, Deng Z, Liu J, Duan L, Wang D, Li W. Crosstalk between immune cells and bone cells or chondrocytes. Int Immunopharmacol 2021; 101:108179. [PMID: 34601329 DOI: 10.1016/j.intimp.2021.108179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/10/2021] [Accepted: 09/18/2021] [Indexed: 01/12/2023]
Abstract
The term "osteoimmunology" was coined to denote the bridge between the immune system and the skeletal system. Osteoimmunology is interdisciplinary, and a full understanding and development of this "bridge" will provide an in-depth understanding of the switch between body health and disease development. B lymphocytes can promote the maturation and differentiation of osteoclasts, and osteoclasts have a negative feedback effect on B lymphocytes. Different subtypes of T lymphocytes regulate osteoclasts in different directions. T lymphocytes have a two-way regulatory effect on osteoblasts, while B lymphocytes have minimal regulatory effects on osteoblasts. In contrast, osteoblasts can promote the differentiation and maturation of T lymphocytes and B lymphocytes. Different immune cells have different effects on chondrocytes; some cooperate with each other, while some antagonize each other. In a healthy adult body, bone resorption and bone formation are in a dynamic balance under the action of multiple mechanisms. In this review, we summarize the interactions and key signaling molecular mechanisms between each type of cell in the immune system and the skeletal system.
Collapse
Affiliation(s)
- Zhiqin Deng
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital/ the First Hospital Affiliated to Shenzhen University, Shenzhen 518000, China
| | - Qian Zhang
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital/ the First Hospital Affiliated to Shenzhen University, Shenzhen 518000, China
| | - Zhe Zhao
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital/ the First Hospital Affiliated to Shenzhen University, Shenzhen 518000, China
| | - Yongshen Li
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital/ the First Hospital Affiliated to Shenzhen University, Shenzhen 518000, China
| | - Xiaoqiang Chen
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital/ the First Hospital Affiliated to Shenzhen University, Shenzhen 518000, China
| | - Zicong Lin
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital/ the First Hospital Affiliated to Shenzhen University, Shenzhen 518000, China
| | - Zhenhan Deng
- Department of Sports Medicine, Shenzhen Second People's Hospital/ the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong 518000, China
| | - Jianquan Liu
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital/ the First Hospital Affiliated to Shenzhen University, Shenzhen 518000, China
| | - Li Duan
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital/ the First Hospital Affiliated to Shenzhen University, Shenzhen 518000, China
| | - Daping Wang
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital/ the First Hospital Affiliated to Shenzhen University, Shenzhen 518000, China.
| | - Wencui Li
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital/ the First Hospital Affiliated to Shenzhen University, Shenzhen 518000, China.
| |
Collapse
|
137
|
Abstract
Anthropogenic environmental pollutants affect many physiological, biochemical, and endocrine actions as reproduction, metabolism, immunity, behavior and as such can interfere with any aspect of hormone action. Microbiota and their genes, microbiome, a large body of microorganisms, first of all bacteria and co-existing in the host´s gut, are now believed to be autonomous endocrine organ, participating at overall endocrine, neuroendocrine and immunoendocrine regulations. While an extensive literature is available on the physiological and pathological aspects of both players, information about their mutual relationships is scarce. In the review we attempted to show various examples where both, endocrine disruptors and microbiota are meeting and can act cooperatively or in opposition and to show the mechanism, if known, staying behind these actions.
Collapse
Affiliation(s)
- R Hampl
- Institute of Endocrinology, Prague, Czech Republic.
| | | |
Collapse
|
138
|
Rubinstein MM, Brown KA, Iyengar NM. Targeting obesity-related dysfunction in hormonally driven cancers. Br J Cancer 2021; 125:495-509. [PMID: 33911195 PMCID: PMC8368182 DOI: 10.1038/s41416-021-01393-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 03/05/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Obesity is a risk factor for at least 13 different types of cancer, many of which are hormonally driven, and is associated with increased cancer incidence and morbidity. Adult obesity rates are steadily increasing and a subsequent increase in cancer burden is anticipated. Obesity-related dysfunction can contribute to cancer pathogenesis and treatment resistance through various mechanisms, including those mediated by insulin, leptin, adipokine, and aromatase signalling pathways, particularly in women. Furthermore, adiposity-related changes can influence tumour vascularity and inflammation in the tumour microenvironment, which can support tumour development and growth. Trials investigating non-pharmacological approaches to target the mechanisms driving obesity-mediated cancer pathogenesis are emerging and are necessary to better appreciate the interplay between malignancy, adiposity, diet and exercise. Diet, exercise and bariatric surgery are potential strategies to reverse the cancer-promoting effects of obesity; trials of these interventions should be conducted in a scientifically rigorous manner with dose escalation and appropriate selection of tumour phenotypes and have cancer-related clinical and mechanistic endpoints. We are only beginning to understand the mechanisms by which obesity effects cell signalling and systemic factors that contribute to oncogenesis. As the rates of obesity and cancer increase, we must promote the development of non-pharmacological lifestyle trials for the treatment and prevention of malignancy.
Collapse
Affiliation(s)
- Maria M. Rubinstein
- grid.51462.340000 0001 2171 9952Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Kristy A. Brown
- grid.5386.8000000041936877XDepartment of Biochemistry in Medicine, Weill Cornell Medical College, New York, NY USA
| | - Neil M. Iyengar
- grid.51462.340000 0001 2171 9952Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY USA
| |
Collapse
|
139
|
Sui Y, Wu J, Chen J. The Role of Gut Microbial β-Glucuronidase in Estrogen Reactivation and Breast Cancer. Front Cell Dev Biol 2021; 9:631552. [PMID: 34458248 PMCID: PMC8388929 DOI: 10.3389/fcell.2021.631552] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 07/09/2021] [Indexed: 12/15/2022] Open
Abstract
Over the past decade, the gut microbiota has received considerable attention for its interactions with the host. Microbial β-glucuronidase generated by this community has hence aroused concern for its biotransformation activity to a wide range of exogenous (foreign) and endogenous compounds. Lately, the role of gut microbial β-glucuronidase in the pathogenesis of breast cancer has been proposed for its estrogen reactivation activity. This is plausible considering that estrogen glucuronides are the primary products of estrogens' hepatic phase II metabolism and are subject to β-glucuronidase-catalyzed hydrolysis in the gut via bile excretion. However, research in this field is still at its very preliminary stage. This review outlines the biology of microbial β-glucuronidase in the gastrointestinal tract and elaborates on the clues to the existence of microbial β-glucuronidase-estrogen metabolism-breast cancer axis. The research gaps in this field will be discussed and possible strategies to address these challenges are suggested.
Collapse
Affiliation(s)
- Yue Sui
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Jianming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jianping Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
| |
Collapse
|
140
|
Ruo SW, Alkayyali T, Win M, Tara A, Joseph C, Kannan A, Srivastava K, Ochuba O, Sandhu JK, Went TR, Sultan W, Kantamaneni K, Poudel S. Role of Gut Microbiota Dysbiosis in Breast Cancer and Novel Approaches in Prevention, Diagnosis, and Treatment. Cureus 2021; 13:e17472. [PMID: 34513524 PMCID: PMC8405251 DOI: 10.7759/cureus.17472] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 08/26/2021] [Indexed: 12/21/2022] Open
Abstract
Breast cancer is the most common cause of cancer-related deaths in women. Breast cancer is still a major cause of morbidity and mortality among women despite all the available diagnostic and treatment modalities. The gut microbiota has drawn keen interest as an additional environmental risk factor in breast cancer, especially in sporadic cases. This article explores factors that disrupt the normal gut microbial composition and the role of gut microbial dysbiosis in the development of breast cancer. We finalized 40 relevant articles after searching Pubmed and Google Scholar using regular keywords and the Medical Subject Headings (MeSH) strategy. Gut microbiota dysbiosis has been shown to play a role in the development of breast cancer via estrogen-dependent mechanisms and non-estrogen-dependent mechanisms involving the production of microbial-derived metabolites, immune regulation, and effects on DNA. The gut microbiota influence estrogen metabolism hence estrogen levels. The metabolites that have demonstrated anticancer properties include lithocholic acid, butyrate, and cadaverine. New approaches targeting the gut microbiota have come up and may yield new advances in the prevention, diagnosis, and treatment of breast cancer. They include the use of prebiotics, probiotics, and hormone supplements to restore normobiosis in the prevention and treatment of breast cancer.
Collapse
Affiliation(s)
- Sheila W Ruo
- General Surgery, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Tasnim Alkayyali
- Internal Medicine, Marmara University, Istanbul, TUR
- Pathology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Myat Win
- General Surgery, Nottingham University Hospitals NHS Trust, Nottingham, GBR
- General Surgery, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Anjli Tara
- General Surgery, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- General Surgery, Liaquat University of Medical and Health Sciences, Jamshoro, PAK
| | - Christine Joseph
- Urology and Obstetrics & Gynecology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Amudhan Kannan
- General Surgery, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
- General Surgery, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Kosha Srivastava
- Neurology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Olive Ochuba
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Jasmine K Sandhu
- Obstetrics & Gynecology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Terry R Went
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Waleed Sultan
- Medicine, Beni Suef University Faculty of Medicine, Beni Suef, EGY
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Surgery, Halifax Health Medical Center, Daytona Beach, USA
| | - Ketan Kantamaneni
- Surgery, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Surgery, Dr.Pinnamaneni Siddhartha Institute of Medical Sciences and Research Foundation, Gannavaram, IND
| | - Sujan Poudel
- Psychiatry and Behavioral Sciences, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Division of Research & Academic Affairs, Larkin Community Hospital, South Miami, USA
| |
Collapse
|
141
|
Graham ME, Herbert WG, Song SD, Raman HN, Zhu JE, Gonzalez PE, Walther-António MRS, Tetel MJ. Gut and vaginal microbiomes on steroids: implications for women's health. Trends Endocrinol Metab 2021; 32:554-565. [PMID: 34049772 PMCID: PMC8282721 DOI: 10.1016/j.tem.2021.04.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 04/17/2021] [Accepted: 04/26/2021] [Indexed: 12/11/2022]
Abstract
This review discusses the interactions of steroids with the gut and vaginal microbiomes within each life phase of adult women and the implications for women's health. Each phase of a woman's life is characterized by distinct hormonal states which drive overall physiology of both host and commensal microbes. These host-microbiome interactions underlie disease pathology in disorders that affect women across their lifetime, including bacterial vaginosis, gestational diabetes, polycystic ovary syndrome (PCOS), anxiety, depression, and obesity. Although many associations between host health and microbiome composition are well defined, the mechanistic role of the microbiome in women's health outcomes is largely unknown. This review addresses potential mechanisms by which the microbiota influences women's health and highlights gaps in current knowledge.
Collapse
Affiliation(s)
- Madeline E Graham
- Neuroscience Department, Wellesley College, Wellesley, MA 02481, USA
| | - William G Herbert
- Department of Surgery, Department of Obstetrics and Gynecology, and Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Stephanie D Song
- Department of Surgery, Department of Obstetrics and Gynecology, and Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Harshini N Raman
- Neuroscience Department, Wellesley College, Wellesley, MA 02481, USA
| | - Jade E Zhu
- Neuroscience Department, Wellesley College, Wellesley, MA 02481, USA
| | | | - Marina R S Walther-António
- Department of Surgery, Department of Obstetrics and Gynecology, and Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Marc J Tetel
- Neuroscience Department, Wellesley College, Wellesley, MA 02481, USA.
| |
Collapse
|
142
|
Discovery and mining of enzymes from the human gut microbiome. Trends Biotechnol 2021; 40:240-254. [PMID: 34304905 DOI: 10.1016/j.tibtech.2021.06.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 12/19/2022]
Abstract
Advances in technological and bioinformatics approaches have led to the generation of a plethora of human gut metagenomic datasets. Metabolomics has also provided substantial data regarding the small metabolites produced and modified by the microbiota. Comparatively, the microbial enzymes mediating the transformation of metabolites have not been intensively investigated. Here, we discuss the recent efforts and technologies used for discovering and mining enzymes from the human gut microbiota. The wealth of knowledge on metabolites, reactions, genome sequences, and structures of proteins, may drive the development of strategies for enzyme mining. Ongoing efforts to annotate gut microbiota enzymes will explain catalytic mechanisms that may guide the clinical applications of the gut microbiome for diagnostic and therapeutic purposes.
Collapse
|
143
|
Islam R, Yu RMK, Andrew-Priestley M, Smith N, Rahman MM, Tran TKA, Connor WAO, MacFarlane GR. Secondary treatment phase of tertiary wastewater treatment works significantly reduces estrogenic load. WATER RESEARCH 2021; 200:117257. [PMID: 34077838 DOI: 10.1016/j.watres.2021.117257] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 06/12/2023]
Abstract
Estrogenic compounds enter waterways via effluents from wastewater treatment works (WWTW), thereby indicating a potential risk to organisms inhabiting adjacent receiving waters. However, little is known about the loads or concentrations of estrogenic compounds that enter Australian WWTWs, the efficiency of removing estrogenic compounds throughout the various stages of tertiary WWTW processes (which are common in Australia), nor the concentrations released into estuarine or marine receiving waters, and the associated risk for aquatic taxa residing in these environments. Therefore, seven estrogenic compounds, comprising the natural estrogens estrone (E1), 17β-estradiol (E2) and estriol (E3), the synthetic estrogen (EE2), and the industrial chemicals bisphenol A (BPA), 4-t-octyl phenol (4-t-OP) and 4-nonyl phenol (4-NP), in wastewater samples were quantified via liquid chromatographic-mass spectrometry (LC-MS) after solid-phase extraction at different stages of wastewater treatment and associated receiving waters. The concentrations of the target compounds in wastewater ranged from < LOQ (limit of quantification) to 158 ng/L for Tanilba Bay WWTW and < LOQ to 162 ng/L for Belmont WWTW. Most target compounds significantly declined after the secondary treatment phase. Appreciable removal efficiency throughout the treatment process was observed with removal from 39.21 to 99.98% of influent values at both WWTWs. The reduction of the natural estrogens (E1, E2 and E3) and 4-t-OP were significantly greater than EE2, BPA, and 4-NP in both WWTWs. Risk quotients (RQs) were calculated to assess potential ecological risks from individual estrogenic compounds. In predicted diluted effluents, no targeted compounds showed any ecological risk (RQ ≤1.65 × 10-2) at both WWTWs. Similarly, all RQs for shore samples at both WWTWs were below 1. Finally, the hazard index (HI), which represents combined estrogenic contaminants' ecological risk, indicated no mentionable risk for predicted diluted effluents (HI = 0.0097 to 0.0218) as well as shoreline samples (HI = 0.393 to 0.522) in the receiving estuarine or marine waters.
Collapse
Affiliation(s)
- Rafiquel Islam
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia; Department of Applied Chemistry and Chemical Engineering, Islamic University, Kushtia 7003, Bangladesh
| | - Richard Man Kit Yu
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia
| | | | - Nathan Smith
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Mohammad Mahmudur Rahman
- Global Centre for Environmental Remediation (GCER), The University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Thi Kim Anh Tran
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia; School of Agriculture and Resources, Vinh University, Viet Nam
| | - Wayne A O' Connor
- New South Wales Department of Primary Industries, Port Stephens Fisheries Institute, Taylors Beach, NSW, 2316, Australia
| | - Geoff R MacFarlane
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia.
| |
Collapse
|
144
|
Glucuronides Hydrolysis by Intestinal Microbial β-Glucuronidases (GUS) Is Affected by Sampling, Enzyme Preparation, Buffer pH, and Species. Pharmaceutics 2021; 13:pharmaceutics13071043. [PMID: 34371734 PMCID: PMC8309147 DOI: 10.3390/pharmaceutics13071043] [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: 06/11/2021] [Revised: 07/03/2021] [Accepted: 07/06/2021] [Indexed: 01/02/2023] Open
Abstract
Glucuronides hydrolysis by intestinal microbial β-Glucuronidases (GUS) is an important procedure for many endogenous and exogenous compounds. The purpose of this study is to determine the impact of experimental conditions on glucuronide hydrolysis by intestinal microbial GUS. Standard probe 4-Nitrophenyl β-D-glucopyranoside (pNPG) and a natural glucuronide wogonoside were used as the model compounds. Feces collection time, buffer conditions, interindividual, and species variations were evaluated by incubating the substrates with enzymes. The relative reaction activity of pNPG, reaction rates, and reaction kinetics for wogonoside were calculated. Fresh feces showed the highest hydrolysis activities. Sonication increased total protein yield during enzyme preparation. The pH of the reaction system increased the activity in 0.69–1.32-fold, 2.9–12.9-fold, and 0.28–1.56-fold for mouse, rat, and human at three different concentrations of wogonoside, respectively. The Vmax for wogonoside hydrolysis was 2.37 ± 0.06, 4.48 ± 0.11, and 5.17 ± 0.16 μmol/min/mg and Km was 6.51 ± 0.71, 3.04 ± 0.34, and 0.34 ± 0.047 μM for mouse, rat, and human, respectively. The inter-individual difference was significant (4–6-fold) using inbred rats as the model animal. Fresh feces should be used to avoid activity loss and sonication should be utilized in enzyme preparation to increase hydrolysis activity. The buffer pH should be appropriate according to the species. Inter-individual and species variations were significant.
Collapse
|
145
|
Kudesia R, Alexander M, Gulati M, Kennard A, Tollefson M. Dietary Approaches to Women's Sexual and Reproductive Health. Am J Lifestyle Med 2021; 15:414-424. [PMID: 34366740 PMCID: PMC8299929 DOI: 10.1177/15598276211007113] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 12/26/2022] Open
Abstract
Over the course of the reproductive life span, it is common for women to experience one or more of the most common gynecologic conditions, including sexual dysfunction, polycystic ovary syndrome, fibroids, endometriosis, and infertility. Although current management guidelines often turn to the established pharmaceutical approaches for each of these diagnoses, the scientific literature also supports an evidence-based approach rooted in the paradigm of food as medicine. Achieving healthy dietary patterns is a core goal of lifestyle medicine, and a plant-forward approach akin to the Mediterranean diet holds great promise for improving many chronic gynecologic diseases. Furthermore, creating an optimal preconception environment from a nutritional standpoint may facilitate epigenetic signaling, thus improving the health of future generations. This state-of-the-art review explores the literature connecting diet with sexual and reproductive health in premenopausal women.
Collapse
Affiliation(s)
- Rashmi Kudesia
- Houston Methodist Hospital and CCRM Fertility Houston, Texas
| | | | - Mahima Gulati
- Division of Endocrinology, Diabetes, and Metabolism, Middlesex Health, Middletown, Connecticut
| | - Anne Kennard
- Marian Regional Medical Center, San Luis Obispo, California
| | | |
Collapse
|
146
|
Laborda-Illanes A, Sánchez-Alcoholado L, Boutriq S, Plaza-Andrades I, Peralta-Linero J, Alba E, González-González A, Queipo-Ortuño MI. A New Paradigm in the Relationship between Melatonin and Breast Cancer: Gut Microbiota Identified as a Potential Regulatory Agent. Cancers (Basel) 2021; 13:3141. [PMID: 34201776 PMCID: PMC8269379 DOI: 10.3390/cancers13133141] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 01/18/2023] Open
Abstract
In this review we summarize a possible connection between gut microbiota, melatonin production, and breast cancer. An imbalance in gut bacterial population composition (dysbiosis), or changes in the production of melatonin (circadian disruption) alters estrogen levels. On the one hand, this may be due to the bacterial composition of estrobolome, since bacteria with β-glucuronidase activity favour estrogens in a deconjugated state, which may ultimately lead to pathologies, including breast cancer. On the other hand, it has been shown that these changes in intestinal microbiota stimulate the kynurenine pathway, moving tryptophan away from the melatonergic pathway, thereby reducing circulating melatonin levels. Due to the fact that melatonin has antiestrogenic properties, it affects active and inactive estrogen levels. These changes increase the risk of developing breast cancer. Additionally, melatonin stimulates the differentiation of preadipocytes into adipocytes, which have low estrogen levels due to the fact that adipocytes do not express aromatase. Consequently, melatonin also reduces the risk of breast cancer. However, more studies are needed to determine the relationship between microbiota, melatonin, and breast cancer, in addition to clinical trials to confirm the sensitizing effects of melatonin to chemotherapy and radiotherapy, and its ability to ameliorate or prevent the side effects of these therapies.
Collapse
Affiliation(s)
- Aurora Laborda-Illanes
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; (A.L.-I.); (L.S.-A.); (S.B.); (I.P.-A.); (J.P.-L.); (M.I.Q.-O.)
- Facultad de Medicina, Universidad de Málaga, 29071 Málaga, Spain
| | - Lidia Sánchez-Alcoholado
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; (A.L.-I.); (L.S.-A.); (S.B.); (I.P.-A.); (J.P.-L.); (M.I.Q.-O.)
- Facultad de Medicina, Universidad de Málaga, 29071 Málaga, Spain
| | - Soukaina Boutriq
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; (A.L.-I.); (L.S.-A.); (S.B.); (I.P.-A.); (J.P.-L.); (M.I.Q.-O.)
- Facultad de Medicina, Universidad de Málaga, 29071 Málaga, Spain
| | - Isaac Plaza-Andrades
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; (A.L.-I.); (L.S.-A.); (S.B.); (I.P.-A.); (J.P.-L.); (M.I.Q.-O.)
| | - Jesús Peralta-Linero
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; (A.L.-I.); (L.S.-A.); (S.B.); (I.P.-A.); (J.P.-L.); (M.I.Q.-O.)
| | - Emilio Alba
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; (A.L.-I.); (L.S.-A.); (S.B.); (I.P.-A.); (J.P.-L.); (M.I.Q.-O.)
- Centro de Investigación Biomédica en Red de Cáncer (Ciberonc CB16/12/00481), 28029 Madrid, Spain
| | - Alicia González-González
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; (A.L.-I.); (L.S.-A.); (S.B.); (I.P.-A.); (J.P.-L.); (M.I.Q.-O.)
| | - María Isabel Queipo-Ortuño
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; (A.L.-I.); (L.S.-A.); (S.B.); (I.P.-A.); (J.P.-L.); (M.I.Q.-O.)
- Centro de Investigación Biomédica en Red de Cáncer (Ciberonc CB16/12/00481), 28029 Madrid, Spain
| |
Collapse
|
147
|
|
148
|
Średnicka P, Juszczuk-Kubiak E, Wójcicki M, Akimowicz M, Roszko MŁ. Probiotics as a biological detoxification tool of food chemical contamination: A review. Food Chem Toxicol 2021; 153:112306. [PMID: 34058235 DOI: 10.1016/j.fct.2021.112306] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/17/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022]
Abstract
Nowadays, people are exposed to diverse environmental and chemical pollutants produced by industry and agriculture. Food contaminations such as persistent organic pollutants (POPs), heavy metals, and mycotoxins are a serious concern for global food safety with economic and public health implications especially in the newly industrialized countries (NIC). Mounting evidence indicates that chronic exposure to food contaminants referred to as xenobiotics exert a negative effect on human health such as inflammation, oxidative stress, and intestinal disorders linked with perturbation of the composition and metabolic profile of the gut microflora. Although the physicochemical technologies for food decontamination are utilized in many cases but require adequate conditions which are often not feasible to be met in many industrial sectors. At present, one promising approach to reduce the risk related to the presence of xenobiotics in foodstuffs is a biological detoxification done by probiotic strains and their enzymes. Many studies confirmed that probiotics are an effective, feasible, and inexpensive tool for preventing xenobiotic-induced dysbiosis and alleviating their toxicity. This review aims to summarize the current knowledge of the direct mechanisms by which probiotics can influence the detoxification of xenobiotics. Moreover, probiotic-xenobiotic interactions with the gut microbiota and the host response were also discussed.
Collapse
Affiliation(s)
- Paulina Średnicka
- Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology, State Research Institute, Rakowiecka 36 Street, Warsaw, Poland
| | - Edyta Juszczuk-Kubiak
- Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology, State Research Institute, Rakowiecka 36 Street, Warsaw, Poland.
| | - Michał Wójcicki
- Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology, State Research Institute, Rakowiecka 36 Street, Warsaw, Poland
| | - Monika Akimowicz
- Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology, State Research Institute, Rakowiecka 36 Street, Warsaw, Poland
| | - Marek Ł Roszko
- Department of Food Safety and Chemical Analysis, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology - State Research Institute, Rakowiecka 36 Street, Warsaw, Poland.
| |
Collapse
|
149
|
Jiang I, Yong PJ, Allaire C, Bedaiwy MA. Intricate Connections between the Microbiota and Endometriosis. Int J Mol Sci 2021; 22:5644. [PMID: 34073257 PMCID: PMC8198999 DOI: 10.3390/ijms22115644] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/09/2021] [Accepted: 05/12/2021] [Indexed: 02/06/2023] Open
Abstract
Imbalances in gut and reproductive tract microbiota composition, known as dysbiosis, disrupt normal immune function, leading to the elevation of proinflammatory cytokines, compromised immunosurveillance and altered immune cell profiles, all of which may contribute to the pathogenesis of endometriosis. Over time, this immune dysregulation can progress into a chronic state of inflammation, creating an environment conducive to increased adhesion and angiogenesis, which may drive the vicious cycle of endometriosis onset and progression. Recent studies have demonstrated both the ability of endometriosis to induce microbiota changes, and the ability of antibiotics to treat endometriosis. Endometriotic microbiotas have been consistently associated with diminished Lactobacillus dominance, as well as the elevated abundance of bacterial vaginosis-related bacteria and other opportunistic pathogens. Possible explanations for the implications of dysbiosis in endometriosis include the Bacterial Contamination Theory and immune activation, cytokine-impaired gut function, altered estrogen metabolism and signaling, and aberrant progenitor and stem-cell homeostasis. Although preliminary, antibiotic and probiotic treatments have demonstrated efficacy in treating endometriosis, and female reproductive tract (FRT) microbiota sampling has successfully predicted disease risk and stage. Future research should aim to characterize the "core" upper FRT microbiota and elucidate mechanisms behind the relationship between the microbiota and endometriosis.
Collapse
Affiliation(s)
| | | | | | - Mohamed A. Bedaiwy
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of British Columbia, D415A-4500 Oak Street, Vancouver, BC V6H 3N1, Canada; (I.J.); (P.J.Y.); (C.A.)
| |
Collapse
|
150
|
R S J. The Immune Microenvironment in Human Papilloma Virus-Induced Cervical Lesions-Evidence for Estrogen as an Immunomodulator. Front Cell Infect Microbiol 2021; 11:649815. [PMID: 33996630 PMCID: PMC8120286 DOI: 10.3389/fcimb.2021.649815] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/29/2021] [Indexed: 12/24/2022] Open
Abstract
Globally, human papilloma virus (HPV) infection is a common sexually transmitted disease. However, most of the HPV infections eventually resolve aided by the body’s efficient cell-mediated immune responses. In the vast majority of the small group of patients who develop overt disease too, it is the immune response that culminates in regression of lesions. It is therefore a rarity that persistent infection by high-risk genotypes of HPV compounded by other risk factors progresses through precancer (various grades of cervical intraepithelial neoplasia—CIN) to cervical cancer (CxCa). Hence, although CxCa is a rare culmination of HPV infection, the latter is nevertheless causally linked to >90% of cancer. The three ‘Es’ of cancer immunoediting viz. elimination, equilibrium, and escape come into vogue during the gradual evolution of CIN 1 to CxCa. Both cell-intrinsic and extrinsic mechanisms operate to eliminate virally infected cells: cell-extrinsic players are anti-tumor/antiviral effectors like Th1 subset of CD4+ T cells, CD8+ cytotoxic T cells, Natural Killer cells, etc. and pro-tumorigenic/immunosuppressive cells like regulatory T cells (Tregs), Myeloid-Derived Suppressor Cells (MDSCs), type 2 macrophages, etc. And accordingly, when immunosuppressive cells overpower the effectors e.g., in high-grade lesions like CIN 2 or 3, the scale is tilted towards immune escape and the disease progresses to cancer. Estradiol has long been considered as a co-factor in cervical carcinogenesis. In addition to the gonads, the Peyer’s patches in the gut synthesize estradiol. Over and above local production of the hormone in the tissues, estradiol metabolism by the gut microbiome: estrobolome versus tryptophan non-metabolizing microbiome, regulates free estradiol levels in the intestine and extraintestinal mucosal sites. Elevated tissue levels of the hormone serve more than one purpose: besides a direct growth-promoting action on cervical epithelial cells, estradiol acting genomically via Estrogen Receptor-α also boosts the function of the stromal and infiltrating immunosuppressive cells viz. Tregs, MDSCs, and carcinoma-associated fibroblasts. Hence as a corollary, therapeutic repurposing of Selective Estrogen Receptor Disruptors or aromatase inhibitors could be useful for modulating immune function in cervical precancer/cancer. The immunomodulatory role of estradiol in HPV-mediated cervical lesions is reviewed.
Collapse
Affiliation(s)
- Jayshree R S
- Department of Microbiology, Kidwai Memorial Institute of Oncology, Bangalore, India
| |
Collapse
|