301
|
Ni Q, Zhang P, Li Q, Han Z. Oxidative Stress and Gut Microbiome in Inflammatory Skin Diseases. Front Cell Dev Biol 2022; 10:849985. [PMID: 35321240 PMCID: PMC8937033 DOI: 10.3389/fcell.2022.849985] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/18/2022] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress plays a dominant role in inflammatory skin diseases. Emerging evidence has shown that the close interaction occurred between oxidative stress and the gut microbiome. Overall, in this review, we have summarized the impact of oxidative stress and gut microbiome during the progression and treatment for inflammatory skin diseases, the interactions between gut dysbiosis and redox imbalance, and discussed the potential possible role of oxidative stress in the gut-skin axis. In addition, we have also elucidated the promising gut microbiome/redox-targeted therapeutic strategies for inflammatory skin diseases.
Collapse
Affiliation(s)
- Qingrong Ni
- Department of Dermatology, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Ping Zhang
- Department of Dermatology, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Qiang Li
- Department of Dermatology, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Zheyi Han
- Department of Gastroenterology, Air Force Medical Center, Fourth Military Medical University, Beijing, China
- *Correspondence: Zheyi Han,
| |
Collapse
|
302
|
Feng T, Liu Y. Microorganisms in the reproductive system and probiotic's regulatory effects on reproductive health. Comput Struct Biotechnol J 2022; 20:1541-1553. [PMID: 35465162 PMCID: PMC9010680 DOI: 10.1016/j.csbj.2022.03.017] [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: 10/22/2021] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 12/18/2022] Open
Abstract
The presence of microbial communities in the reproductive tract has been revealed, and this resident microbiota is involved in the maintenance of health. Intentional modulation via probiotics has been proposed as a possible strategy to enhance reproductive health and reduce the risk of diseases. The male seminal microbiota has been suggested as an important factor that influences a couple’s health, pregnancy outcomes, and offspring health. Probiotics have been reported to play a role in male fertility and to affect the health of mothers and offspring. While the female reproductive microbiota is more complicated and has been identified in both the upper and lower reproductive systems, they together contribute to health maintenance. Probiotics have shown regulatory effects on the female reproductive tract, thereby contributing to homeostasis of the tract and influencing the health of offspring. Further, through transmission of bacteria or through other indirect mechanisms, the parent’s reproductive microbiota and probiotic intervention influence infant gut colonization and immunity development, with potential health consequences. In vitro and in vivo studies have explored the mechanisms underlying the benefits of probiotic administration and intervention, and an array of positive results, such as modulation of microbiota composition, regulation of metabolism, promotion of the epithelial barrier, and improvement of immune function, have been observed. Herein, we review the state of the art in reproductive system microbiota and its role in health and reproduction, as well as the beneficial effects of probiotics on reproductive health and their contributions to the prevention of associated diseases.
Collapse
|
303
|
Liu L, Shang L, Jin D, Wu X, Long B. General anesthesia bullies the gut: a toxic relationship with dysbiosis and cognitive dysfunction. Psychopharmacology (Berl) 2022; 239:709-728. [PMID: 35187594 DOI: 10.1007/s00213-022-06096-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 02/15/2022] [Indexed: 12/12/2022]
Abstract
Perioperative neurocognitive disorder (PND) is a common surgery outcome affecting up to a third of the elderly patients, and it is associated with high morbidity and increased risk for Alzheimer's disease development. PND is characterized by cognitive impairment that can manifest acutely in the form of postoperative delirium (POD) or after hospital discharge as postoperative cognitive dysfunction (POCD). Although POD and POCD are clinically distinct, their development seems to be mediated by a systemic inflammatory reaction triggered by surgical trauma that leads to dysfunction of the blood-brain barrier and facilitates the occurrence of neuroinflammation. Recent studies have suggested that the gut microbiota composition may play a pivotal role in the PND development by modulating the risk of neuroinflammation establishment. In fact, modulation of gut microbiome composition with pre- and probiotics seems to be effective for the prevention and treatment of PND in animals. Interestingly, general anesthetics seem to have major responsibility on the gut microbiota composition changes following surgery and, consequently, can be an important element in the process of PND initiation. This concept represents an important milestone for the understanding of PND pathogenesis and may unveil new opportunities for the development of preventive or mitigatory strategies against the development of these conditions. The aim of this review is to discuss how anesthetics used in general anesthesia can interact and alter the gut microbiome composition and contribute to PND development by favoring the emergence of neuroinflammation.
Collapse
Affiliation(s)
- Lidan Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China
| | - Lihua Shang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China
| | - Dongxue Jin
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China
| | - Xiuying Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China
| | - Bo Long
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
304
|
Xie J, Tian S, Liu J, Cao R, Yue P, Cai X, Shang Q, Yang M, Han L, Zhang DK. Dual role of the nasal microbiota in neurological diseases—An unignorable risk factor or a potential therapy carrier. Pharmacol Res 2022; 179:106189. [DOI: 10.1016/j.phrs.2022.106189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/06/2022] [Accepted: 03/17/2022] [Indexed: 12/11/2022]
|
305
|
Potential for Prebiotic Stabilized Cornus mas L. Lyophilized Extract in the Prophylaxis of Diabetes Mellitus in Streptozotocin Diabetic Rats. Antioxidants (Basel) 2022; 11:antiox11020380. [PMID: 35204262 PMCID: PMC8868578 DOI: 10.3390/antiox11020380] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 12/16/2022] Open
Abstract
As a systemic disease, diabetes mellitus (DM) is characterized by the disruption of many glucose metabolic pathways. Therefore, it seems critical to study new therapies to support treatment to develop therapeutic systems that can operate across a broad metabolic spectrum. The current state of knowledge indicates an essential role of the gut microbiota in the development and course of the disease. Cornus mas fruits have demonstrated a rich biological activity profile and potential for application in the treatment of DM. As part of a preliminary analysis, the activity of four cultivars of Cornus mas fruits was analyzed. The cultivar Wydubieckij was selected as having the highest activity in in vitro conditions for further prebiotic system preparation. The study aimed to develop a unique therapeutic system based, first of all, on the mechanism of α-glucosidase inhibition and the antioxidant effect resulting from the activity of the plant extract used, combined with the prebiotic effect of inulin. The obtained system was characterized in vitro in terms of antioxidant activity and enzyme inhibition capacity, and was then tested on diabetic rats. The study was coupled with an analysis of changes in the intestinal microflora. The system of prebiotic stabilized Cornus mas L. lyophilized extract with inulin offers valuable support for the prophylaxis and treatment of DM.
Collapse
|
306
|
Therapeutic Potential of Metabolites from Lactobacillus rhamnosus and Mare’s Milk in the Treatment of Dysbiosis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3851478. [PMID: 35132375 PMCID: PMC8817857 DOI: 10.1155/2022/3851478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/08/2022] [Indexed: 12/02/2022]
Abstract
Ulcerative colitis is an inflammatory bowel disease that forms ulcerations in the mucous membrane of the colon and rectum, in which gut microbiota plays a pivotal role in its pathogenesis. Agents modulating microbial dysbiosis caused by colitis can help in the remission of this disease. The current study describes the potential therapeutic effects of active metabolites from Lactobacillus rhamnosus and mare's milk which have potential therapeutic values on the intestinal microbiota and proinflammatory cytokines. The analysis of the V1-V3 16S rDNA site revealed significant changes in the intestinal microbiome composition before and after treatment in the treated group compared to the positive control group that was treated with 5-aminosalicylic acid (5-ASA). So the effect of the study product on dextran sulfate sodium-induced dysbiosis was shown to be more potent than the positive control, 5-ASA. The level of proinflammatory cytokines also decreased under the influence of a biological product.
Collapse
|
307
|
Vatanen T, Sakwinska O, Wilson B, Combremont S, Cutfield WS, Chan SY, Godfrey KM, O'Sullivan JM. Transcription shifts in gut bacteria shared between mothers and their infants. Sci Rep 2022; 12:1276. [PMID: 35075183 PMCID: PMC8786960 DOI: 10.1038/s41598-022-04848-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/10/2021] [Indexed: 01/05/2023] Open
Abstract
The infant gut microbiome contains a portion of bacteria that originate from the maternal gut. In the infant gut these bacteria encounter a new metabolic environment that differs from the adult gut, consequently requiring adjustments in their activities. We used pilot community RNA sequencing data (metatranscriptomes) from ten mother-infant dyads participating in the NiPPeR Study to characterize bacterial gene expression shifts following mother-to-infant transmission. Maternally-derived bacterial strains exhibited large scale gene expression shifts following the transmission to the infant gut, with 12,564 activated and 14,844 deactivated gene families. The implicated genes were most numerous and the magnitude shifts greatest in Bacteroides spp. This pilot study demonstrates environment-dependent, strain-specific shifts in gut bacteria function and underscores the importance of metatranscriptomic analysis in microbiome studies.
Collapse
Affiliation(s)
- Tommi Vatanen
- Liggins Institute, The University of Auckland, Private Bag 102904, Auckland, New Zealand.
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - O Sakwinska
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., 1000, Lausanne, Switzerland
| | - B Wilson
- Liggins Institute, The University of Auckland, Private Bag 102904, Auckland, New Zealand
| | - S Combremont
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., 1000, Lausanne, Switzerland
| | - W S Cutfield
- Liggins Institute, The University of Auckland, Private Bag 102904, Auckland, New Zealand
- A Better Start - National Science Challenge, Auckland, New Zealand
| | - S Y Chan
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
| | - K M Godfrey
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, UK
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
| | - Justin M O'Sullivan
- Liggins Institute, The University of Auckland, Private Bag 102904, Auckland, New Zealand.
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK.
- The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand.
- Brain Research New Zealand, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
308
|
Benezra A. Microbial Kin: Relations of Environment and Time. Med Anthropol Q 2022; 35:511-528. [PMID: 35066930 DOI: 10.1111/maq.12680] [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: 12/09/2020] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 11/28/2022]
Abstract
Microbiome science considers human beings supraorganisms: single ecological units made up of symbiotic assemblages of human cells and microorganisms. Microbes co-evolve with humans, and microbial populations in human bodies are determined by environments/exposures including family, food and place, health care, race and gender inequities, and toxic pollution. Microbiomes are transgenerational links, disarrangements between different bodies and the outside world. This article asserts that microbes are kin-kin that are made of and making environments, across generations. Post/nonhuman theories have debated the agency, sociality, and ontologies of microbes and things like microbes, all the while appropriating and eliding Indigenous scholarship that directly address the nonhuman world. Microbial kin evokes Indigenous formulations that necessitate reciprocal, ethical accountability to more-than-human relations. This article uses fieldwork in a transnational microbiome malnutrition project in Bangladesh to explore what develops for both the biological and social sciences if we call human-microbe relations kinships, and call microbes our kin.
Collapse
Affiliation(s)
- Amber Benezra
- Science and Technology Studies, Stevens Institute of Technology
| |
Collapse
|
309
|
An Integrative Bioinformatic Analysis of Microbiome and Transcriptome for Predicting the Risk of Colon Adenocarcinoma. DISEASE MARKERS 2022; 2022:7994074. [PMID: 35096207 PMCID: PMC8794683 DOI: 10.1155/2022/7994074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/24/2021] [Accepted: 12/02/2021] [Indexed: 11/18/2022]
Abstract
The abundance of gut microbiota is significantly decreased in patients with colorectal tumors compared to healthy groups. However, few studies have been conducted to correlate the differences in gut microbiota in colon cancer patients with different prognosis. In this study, we analysed the gut microbiota among patients with colon cancer and determined the microbial characteristics of COAD and divided the overall survival of COAD data into the high- and low-risk groups. In addition, we established a microbiome-related gene map and determined the association between microbial features and immune cell infiltration in COAD. In comparison with the low-risk group, the high risk group of COAD samples exhibited a decreased proportion of activated CD4 T cells as well as an increased proportion of M2 macrophages. The current data suggested that different gut flora backgrounds lead to different gene expression profiles, which in turn affect immune cell typing and colorectal tumor prognosis.
Collapse
|
310
|
Diet leaves a genetic signature in a keystone member of the gut microbiota. Cell Host Microbe 2022; 30:183-199.e10. [PMID: 35085504 DOI: 10.1016/j.chom.2022.01.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/08/2021] [Accepted: 12/17/2021] [Indexed: 12/22/2022]
Abstract
Switching from a low-fat and high-fiber diet to a Western-style high-fat and high-sugar diet causes microbiota imbalances that underlay many pathological conditions (i.e., dysbiosis). Although the effects of dietary changes on microbiota composition and functions are well documented, their impact in gut bacterial evolution remains unexplored. We followed the emergence of mutations in Bacteroides thetaiotaomicron, a prevalent fiber-degrading microbiota member, upon colonization of the murine gut under different dietary regimens. B. thetaiotaomicron evolved rapidly in the gut and Western-style diet selected for mutations that promote degradation of mucin-derived glycans. Periodic dietary changes caused fluctuations in the frequency of such mutations and were associated with metabolic shifts, resulting in the maintenance of higher intraspecies genetic diversity compared to constant dietary regimens. These results show that dietary changes leave a genetic signature in microbiome members and suggest that B. thetaiotaomicron genetic diversity could be a biomarker for dietary differences among individuals.
Collapse
|
311
|
Gut Microbiome Alterations following Postnatal Iron Supplementation Depend on Iron Form and Persist into Adulthood. Nutrients 2022; 14:nu14030412. [PMID: 35276770 PMCID: PMC8838803 DOI: 10.3390/nu14030412] [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: 12/09/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 12/13/2022] Open
Abstract
The gut microbiota is implicated in the adverse developmental outcomes of postnatal iron supplementation. To generate hypotheses on how changes to the gut microbiota by iron adversely affect development, and to determine whether the form of iron influences microbiota outcomes, we characterized gut microbiome and metabolome changes in Sprague-Dawley rat pups given oral supplements of ferrous sulfate (FS), ferrous bis-glycinate chelate (FC), or vehicle control (CON) on postnatal day (PD) 2−14. Iron supplementation reduced microbiome alpha-diversity (p < 0.0001) and altered short-chain fatty acids (SCFAs) and trimethylamine (TMA) in a form-dependent manner. To investigate the long-term effects of iron provision in early life, an additional cohort was supplemented with FS, FC, or CON until PD 21 and then weaned onto standard chow. At ~8 weeks of age, young adult (YA) rats that received FS exhibited more diverse microbiomes compared to CON (p < 0.05), whereas FC microbiomes were less diverse (p < 0.05). Iron provision resulted in 10,000-fold reduced abundance of Lactobacilli in pre-weanling and YA animals provided iron in early life (p < 0.0001). Our results suggest that in pre-weanling rats, supplemental iron form can generate differential effects on the gut microbiota and microbial metabolism that persist into adulthood.
Collapse
|
312
|
Liu W, Sun Z, Ma C, Zhang J, Ma C, Zhao Y, Wei H, Huang S, Zhang H. Exposure to soil environments during earlier life stages is distinguishable in the gut microbiome of adult mice. Gut Microbes 2022; 13:1-13. [PMID: 33382948 PMCID: PMC7781656 DOI: 10.1080/19490976.2020.1830699] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Environmental exposure during earlier life stages can govern the assembly and development of gut microbiota, yet it is insufficiently understood. In this study, ex-germ-free mice were cohoused with distinct soil-microbiota (from desert, steppe, and forest) beddings within 60 days after birth and subsequently transferred to new soil beddings from 60 to 90th day. Using metagenomic shotgun sequencing, firstly, we found soil microbes from natural environments (birthplace) greatly influenced the gut community assembly in the housing experiment. About 27% microbial species and 12% functional components that associated with birthplaces at Day 60 were still discriminatory of birthplaces after transferring mice to new environments. Moreover, prior soil-exposure types are associated with the magnitude of temporal microbiome change due to environmental shifts. The appropriate soil-exposure (e.g., steppe) might help mice gut microbiome adapt to changing environments or host development. Our study demonstrated the continuous soil-exposure history earlier is associated with the gut microbiome individuality and development later.
Collapse
Affiliation(s)
- Wenjun Liu
- Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Zheng Sun
- Single-Cell Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, China
| | - Chen Ma
- Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Jiachao Zhang
- College of Food Science and Technology, Hainan University, Haikou, Hainan, China
| | - ChenChen Ma
- College of Food Science and Technology, Hainan University, Haikou, Hainan, China
| | - Yinqi Zhao
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Hong Wei
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, P. R. China, Heping Zhang Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Shi Huang
- Single-Cell Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, China, Heping Zhang Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China, Heping Zhang Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| |
Collapse
|
313
|
Dan W, Peng L, Yan B, Li Z, Pan F. Human Microbiota in Esophageal Adenocarcinoma: Pathogenesis, Diagnosis, Prognosis and Therapeutic Implications. Front Microbiol 2022; 12:791274. [PMID: 35126331 PMCID: PMC8815000 DOI: 10.3389/fmicb.2021.791274] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/23/2021] [Indexed: 11/29/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) is one of the main subtypes of esophageal cancer. The incidence rate of EAC increased progressively while the 5-year relative survival rates were poor in the past two decades. The mechanism of EAC has been studied extensively in relation to genetic factors, but less so with respect to human microbiota. Currently, researches about the relationship between EAC and the human microbiota is a newly emerging field of study. Herein, we present the current state of knowledge linking human microbiota to esophageal adenocarcinoma and its precursor lesion—gastroesophageal reflux disease and Barrett’s esophagus. There are specific human bacterial alternations in the process of esophageal carcinogenesis. And bacterial dysbiosis plays an important role in the process of esophageal carcinogenesis via inflammation, microbial metabolism and genotoxicity. Based on the human microbiota alternation in the EAC cascade, it provides potential microbiome-based clinical application. This review is focused on novel targets in prevention, diagnosis, prognosis, and therapy for esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Wanyue Dan
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Nankai University, Tianjin, China
| | - Lihua Peng
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Bin Yan
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhengpeng Li
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Fei Pan
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Fei Pan,
| |
Collapse
|
314
|
Amir A, Erez-Granat O, Braun T, Sosnovski K, Hadar R, BenShoshan M, Heiman S, Abbas-Egbariya H, Glick Saar E, Efroni G, Haberman Y. Gut microbiome development in early childhood is affected by day care attendance. NPJ Biofilms Microbiomes 2022; 8:2. [PMID: 35017536 PMCID: PMC8752763 DOI: 10.1038/s41522-021-00265-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/11/2021] [Indexed: 11/09/2022] Open
Abstract
The human gut microbiome develops during the first years of life, followed by a relatively stable adult microbiome. Day care attendance is a drastic change that exposes children to a large group of peers in a diverse environment for prolonged periods, at this critical time of microbial development, and therefore has the potential to affect microbial composition. We characterize the effect of day care on the gut microbial development throughout a single school year in 61 children from 4 different day care facilities, and in additional 24 age-matched home care children (n = 268 samples, median age of entering the study was 12 months). We show that day care attendance is a significant and impactful factor in shaping the microbial composition of the growing child, the specific daycare facility and class influence the gut microbiome, and each child becomes more similar to others in their day care. Furthermore, in comparison to home care children, day care children have a different gut microbial composition, with enrichment of taxa more frequently observed in older populations. Our results provide evidence that daycare may be an external factor that contributes to gut microbiome maturation and make-up in early childhood.
Collapse
Affiliation(s)
- Amnon Amir
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Ortal Erez-Granat
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Tzipi Braun
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Katya Sosnovski
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Rotem Hadar
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Marina BenShoshan
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Sophia Heiman
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Haya Abbas-Egbariya
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Efrat Glick Saar
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Gilat Efroni
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Yael Haberman
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel. .,Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
315
|
Nowicka D, Chilicka K, Dzieńdziora-Urbińska I. Host-Microbe Interaction on the Skin and Its Role in the Pathogenesis and Treatment of Atopic Dermatitis. Pathogens 2022; 11:pathogens11010071. [PMID: 35056019 PMCID: PMC8779626 DOI: 10.3390/pathogens11010071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/04/2022] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
Atopic dermatitis (AD) is a condition with a complex and unclear aetiology. Possible causes of AD encompass alterations in the structure and function of the epidermal barrier, disturbances in the skin microbiome, immune factors, allergens, bacterial and fungal infections as well as environmental and genetic factors. In patients with AD, acute skin lesions are colonized by a greater number of bacteria and fungi than chronic lesions, clinically unchanged atopic skin and the skin of healthy people. Mechanisms promoting skin colonization by pathogens include complex interplay among several factors. Apart from disturbances of the skin microbiome, increased adhesion in atopic skin, defects of innate immune response resulting in the lack of or restriction of growth of microorganisms also contribute to susceptibility to the skin colonization of and infections, especially with Staphylococcus aureus. This review of the literature attempts to identify factors that are involved in the pathogenesis of AD-related bacterial and fungal skin colonization. Studies on the microbiome, commensal microorganisms and the role of skin microorganisms in maintaining healthy skin bring additional insight into the treatment and prevention of AD. In the light of presented mechanisms, reduction in colonization may become both causative and symptomatic treatment in AD.
Collapse
Affiliation(s)
- Danuta Nowicka
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, 50-368 Wrocław, Poland
- Correspondence:
| | - Karolina Chilicka
- Department of Health Sciences, University of Opole, 45-040 Opole, Poland; (K.C.); (I.D.-U.)
| | | |
Collapse
|
316
|
Zhang J, Yu H, Zhong H, Wang Q, Tang J, Shen F, Cai H, Liu T, Feng F, Zhao M. Dietary emulsifier glycerol monodecanoate affects gut microbiota contributing to regulating lipid metabolism, insulin sensitivity and inflammation. Food Funct 2022; 13:8804-8817. [DOI: 10.1039/d2fo01689c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Glycerol monodecanoate (GMD) is a medium-chain monoacylglycerol that possesses emulsifying and antibacterial properties. Common emulsifiers carboxymethylcellulose and polysorbate-80 have been reported to cause intestinal microbiota dysbiosis and metabolic disturbances. While...
Collapse
|
317
|
Tsigalou C, Konstantinidis T, Aloizou AM, Bezirtzoglou E, Tsakris A. Future Therapeutic Prospects in Dealing with Autoimmune Diseases: Treatment Based on the Microbiome Model. ROLE OF MICROORGANISMS IN PATHOGENESIS AND MANAGEMENT OF AUTOIMMUNE DISEASES 2022:489-520. [DOI: 10.1007/978-981-19-4800-8_25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
318
|
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: 36] [Impact Index Per Article: 12.0] [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
|
319
|
Kang K, Zhou Q, McGinn L, Nguyen T, Luo Y, Djalilian A, Rosenblatt M. High fat diet induced gut dysbiosis alters corneal epithelial injury response in mice. Ocul Surf 2022; 23:49-59. [PMID: 34808360 PMCID: PMC8792274 DOI: 10.1016/j.jtos.2021.11.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/30/2021] [Accepted: 11/10/2021] [Indexed: 01/03/2023]
Abstract
PURPOSE Commensal microbiome secretes various metabolites that can exert important effects on the host immunity and inflammation and can alter cellular functions. However, little is known regarding the effect of microbiome on corneal immunity and genetic expression. The purpose of this study is to describe the effect of diet-induced gut dysbiosis on corneal immunity and corneal gene expression after wounding. METHODS This study is approved by the Animal Care and Use of the University of Illinois. Six-week-old female C57BL6 mice were fed on a normal chow diet (ND), isocaloric low-fat control diet (LFD), or a 21% milk high-fat diet (HFD) for six weeks. 2 mm corneal epithelial debridement was performed (n = 10). Fecal samples from mice were used for microbial diversity analysis (n > 3). Immunofluorescence staining of corneal wholemount tissue post-debridement was used to visualize immune cell distribution. RNA Seq was performed on tissue samples from corneas following debridement. RESULTS Mice fed differing diets had significant alterations in gut microbial diversities. After corneal debridement, HFD mice experienced delayed wound healing in comparison to LFD mice and ND mice groups. However, fecal transplantation led to normalization of wound closure rates. Increased γδTCR staining was observed in the LFD group, and decreased LY6G was observed in HFD group (p < 0.05). Gene Ontology terms of differentially expressed genes included response to external stimulus, cell proliferation, migration, adhesion, defense response and leukocyte migration. Top over-represented pathways included ECM-receptor interaction, Cytokine-cytokine receptor interaction, Focal adhesion and Leukocyte trans-endothelial migration. CONCLUSIONS Gut microbial dysbiosis alters corneal immune cell distribution, corneal response to injury, and genes related to epithelial function and corneal immunity.
Collapse
Affiliation(s)
- Kai Kang
- Illinois Eye and Ear Infirmary, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA.
| | - Qiang Zhou
- Illinois Eye and Ear Infirmary, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Lander McGinn
- Illinois Eye and Ear Infirmary, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Tara Nguyen
- Illinois Eye and Ear Infirmary, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Yuncin Luo
- Illinois Eye and Ear Infirmary, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Ali Djalilian
- Illinois Eye and Ear Infirmary, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Mark Rosenblatt
- Illinois Eye and Ear Infirmary, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
320
|
Spontaneous preterm delivery is reflected in both early neonatal and maternal gut microbiota. Pediatr Res 2022; 91:1804-1811. [PMID: 34349229 PMCID: PMC9270225 DOI: 10.1038/s41390-021-01663-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 04/22/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Aberrant gut microbiota composition in preterm neonates is linked to adverse health consequences. Little is known about the impact of perinatal factors or maternal gut microbiota on initial preterm gut colonization. METHODS Fecal samples were collected from 55 preterm neonates (<35 gestational weeks), 51 mothers, and 25 full-term neonates during the first 3-4 postpartum days. Gut microbiota composition was assessed using 16S ribosomal RNA gene sequencing. RESULTS Preterm neonates exhibited significantly lower gut microbiota alpha diversity and distinct beta diversity clustering compared to term neonates. Spontaneous preterm birth was associated with distinct initial gut microbiota beta diversity as compared to iatrogenic delivery. Gestational age or delivery mode had no impact on the preterm gut microbiota composition. The cause of preterm delivery was also reflected in the maternal gut microbiota composition. The contribution of maternal gut microbiota to initial preterm gut colonization was more pronounced after spontaneous delivery than iatrogenic delivery and not dependent on delivery mode. CONCLUSIONS The initial preterm gut microbiota is distinct from term microbiota. Spontaneous preterm birth is reflected in the early neonatal and maternal gut microbiota. Transmission of gut microbes from mother to neonate is determined by spontaneous preterm delivery, but not by mode of birth. IMPACT The initial gut microbiota in preterm neonates is distinct from those born full term. Spontaneous preterm birth is associated with changes in the gut microbiota composition of both preterm neonates and their mothers. The contribution of the maternal gut microbiota to initial neonatal gut colonization was more pronounced after spontaneous preterm delivery as compared to iatrogenic preterm delivery and not dependent on delivery mode. Our study provides new evidence regarding the early gut colonization patterns in preterm infants. Altered preterm gut microbiota has been linked to adverse health consequences and may provide a target for early intervention.
Collapse
|
321
|
Requena T, Pérez Martínez G. Probiotics, Prebiotics, Synbiotics, Postbiotics and Other Biotics. What's Next? COMPREHENSIVE GUT MICROBIOTA 2022:197-210. [DOI: 10.1016/b978-0-12-819265-8.00094-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
322
|
Solans M, Barceló MA, Morales-Suárez-Varela M, Moya A, Saez M. Prenatal exposure to antibiotics and risk of childhood overweight or obesity: A systematic review and meta-analysis. Obes Rev 2022; 23 Suppl 1:e13382. [PMID: 34859947 DOI: 10.1111/obr.13382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/08/2021] [Indexed: 10/19/2022]
Abstract
Infant antibiotic use has been modestly associated with childhood overweight, while evidence on prenatal exposures remains less clear. A systematic review and meta-analysis were conducted to examine associations between maternal antibiotic exposure and subsequent risk of childhood overweight/obesity. Publications were retrieved from PubMed and Web of Science databases up to December 2019. A random effects model was used to summarize risk estimates, overall, and by period and frequency of exposure. Ten observational studies were included in the narrative synthesis. We did not observe a clear pattern of association between prenatal antibiotic use and childhood overweight/obesity. There were suggestive associations for repeated exposures (≥3 courses) and those taking place during the second trimester of gestation, which were also pointed out in our meta-analysis (relative risk, RR2T = 1.15 (95% CI 1.04; 1.28, I2 = 18%), and RR3courses = 1.31 (95% CI 1.03; 1.67, I2 = 65%), respectively). In most studies, however, confounding by underlying infections cannot be ruled out. Overall, current data do not conclusively support the hypothesis that prenatal exposure to antibiotics is a risk factor for childhood obesity/overweight. Further studies, controlling for underlying infections and exploring the association according to frequency, period (both prenatal and intrapartum) and type of antibiotic, are needed to clarify this association.
Collapse
Affiliation(s)
- Marta Solans
- Centro de Investigación Biomédica en Red: Epidemiología y Salud Pública (CIBERESP), Girona, Spain.,Research Group on Statistics, Econometrics and Health (GRECS), University of Girona, Girona, Spain
| | - Maria A Barceló
- Centro de Investigación Biomédica en Red: Epidemiología y Salud Pública (CIBERESP), Girona, Spain.,Research Group on Statistics, Econometrics and Health (GRECS), University of Girona, Girona, Spain
| | - Maria Morales-Suárez-Varela
- Centro de Investigación Biomédica en Red: Epidemiología y Salud Pública (CIBERESP), Girona, Spain.,Department of Preventive Medicine and Public Health, Food Sciences, Toxicology, and Legal Medicine, School of Pharmacy, University of Valencia, Valencia, Spain
| | - Andrés Moya
- Centro de Investigación Biomédica en Red: Epidemiología y Salud Pública (CIBERESP), Girona, Spain.,Instituto de Biología Integrativa de Sistemas, Universitat de València and Spanish Research Council (CSIC), Valencia, Spain.,Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Valencia, Spain
| | - Marc Saez
- Centro de Investigación Biomédica en Red: Epidemiología y Salud Pública (CIBERESP), Girona, Spain.,Research Group on Statistics, Econometrics and Health (GRECS), University of Girona, Girona, Spain
| |
Collapse
|
323
|
Douglas P. Re-thinking benign inflammation of the lactating breast: A mechanobiological model. WOMEN'S HEALTH (LONDON, ENGLAND) 2022; 18:17455065221075907. [PMID: 35156466 PMCID: PMC8848036 DOI: 10.1177/17455065221075907] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 11/29/2021] [Accepted: 01/06/2022] [Indexed: 12/13/2022]
Abstract
Despite the known benefits of breastfeeding for both infant and mother, clinical support for problems such as inflammation of the lactating breast remains a research frontier. Breast pain associated with inflammation is a common reason for premature weaning. Multiple diagnoses are used for inflammatory conditions of the lactating breast, such as engorgement, blocked ducts, phlegmon, mammary candidiasis, subacute mastitis, mastitis and white spots, which lack agreed or evidence-based aetiology, definitions and treatment. This is the first in a series of three articles which review the research literature concerning benign lactation-related breast inflammation. This article investigates aetiological models. A complex systems perspective is applied to analyse heterogeneous and interdisciplinary evidence elucidating the functional anatomy and physiology of the lactating breast; the mammary immune system, including the human milk microbiome and cellular composition; the effects of mechanical forces during lactation; and the interactions between these. This analysis gives rise to a mechanobiological model of breast inflammation, in which very high intra-alveolar and intra-ductal pressures are hypothesized to strain or rupture the tight junctions between lactocytes and ductal epithelial cells, triggering inflammatory cascades and capillary dilation. Resultant elevation of stromal tension exerts pressure on lactiferous ducts, worsening intraluminal backpressure. Rising leucocyte and epithelial cell counts in the milk and alterations in the milk microbiome are signs that the mammary immune system is recruiting mechanisms to downregulate inflammatory feedback loops. From a complex systems perspective, the key mechanism for the prevention or treatment of breast inflammation is avoidance of excessively high intra-alveolar and intra-ductal pressures, which prevents a critical mass of mechanical strain and rupture of the tight junctions between lactocytes and ductal epithelial cells.
Collapse
Affiliation(s)
- Pamela Douglas
- School of Nursing and Midwifery, Griffith University, Brisbane, QLD, Australia
- General Practice Clinical Unit, The University of Queensland, Brisbane, QLD, Australia
- Possums & Co., Brisbane, QLD, Australia
| |
Collapse
|
324
|
Cao M, Peng Q, Wei ZG, Liu F, Hou YF. EdClust: A heuristic sequence clustering method with higher sensitivity. J Bioinform Comput Biol 2021; 20:2150036. [PMID: 34939905 DOI: 10.1142/s0219720021500360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The development of high-throughput technologies has produced increasing amounts of sequence data and an increasing need for efficient clustering algorithms that can process massive volumes of sequencing data for downstream analysis. Heuristic clustering methods are widely applied for sequence clustering because of their low computational complexity. Although numerous heuristic clustering methods have been developed, they suffer from two limitations: overestimation of inferred clusters and low clustering sensitivity. To address these issues, we present a new sequence clustering method (edClust) based on Edlib, a C/C[Formula: see text] library for fast, exact semi-global sequence alignment to group similar sequences. The new method edClust was tested on three large-scale sequence databases, and we compared edClust to several classic heuristic clustering methods, such as UCLUST, CD-HIT, and VSEARCH. Evaluations based on the metrics of cluster number and seed sensitivity (SS) demonstrate that edClust can produce fewer clusters than other methods and that its SS is higher than that of other methods. The source codes of edClust are available from https://github.com/zhang134/EdClust.git under the GNU GPL license.
Collapse
Affiliation(s)
- Ming Cao
- Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China.,School of Mathematics and Statistics, Shaanxi Xueqian Normal University, Xi'an, 710100, P. R. China
| | - Qinke Peng
- Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Ze-Gang Wei
- Institute of Physics and Optoelectronics Technology, Baoji University of Arts and Sciences, Baoji, 721016, P. R. China
| | - Fei Liu
- Institute of Physics and Optoelectronics Technology, Baoji University of Arts and Sciences, Baoji, 721016, P. R. China
| | - Yi-Fan Hou
- Institute of Physics and Optoelectronics Technology, Baoji University of Arts and Sciences, Baoji, 721016, P. R. China
| |
Collapse
|
325
|
Beribaka M, Jelić M, Tanasković M, Lazić C, Stamenković-Radak M. Life History Traits in Two Drosophila Species Differently Affected by Microbiota Diversity under Lead Exposure. INSECTS 2021; 12:insects12121122. [PMID: 34940211 PMCID: PMC8708062 DOI: 10.3390/insects12121122] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 01/04/2023]
Abstract
Simple Summary Microbiota have a significant functional role in the life of the host, including immunity, lifespan and reproduction. Drosophila species are attractive model organisms for investigating microbiota diversity from different aspects due to their simple gut microbiota, short generation time and high fertility. Considering such an important role of the microbiota in the life of Drosophila, we investigated the extent to which lead (Pb), as one of the most abundant heavy metals in the environment, affects the microbiota and the fitness of this insect host. The results indicate that different factors, such as population origin and sex, may affect individual traits differently and this could be species-specific. In addition, there are members of microbiota that help the host to overcome environmental stress and they could play a key role in reducing the fitness cost in such situations. Studying the influence of microbiota on the adaptive response to heavy metals and the potential implications on overall host fitness is of great pertinence. Abstract Life history traits determine the persistence and reproduction of each species. Factors that can affect life history traits are numerous and can be of different origin. We investigated the influence of population origin and heavy metal exposure on microbiota diversity and two life history traits, egg-to-adult viability and developmental time, in Drosophila melanogaster and Drosophila subobscura, grown in the laboratory on a lead (II) acetate-saturated substrate. We used 24 samples, 8 larval and 16 adult samples (two species × two substrates × two populations × two sexes). The composition of microbiota was determined by sequencing (NGS) of the V3–V4 variable regions of the 16S rRNA gene. The population origin showed a significant influence on life history traits, though each trait in the two species was affected differentially. Reduced viability in D. melanogaster could be a cost of fast development, decrease in Lactobacillus abundance and the presence of Wolbachia. The heavy metal exposure in D. subobscura caused shifts in developmental time but maintained the egg-to-adult viability at a similar level. Microbiota diversity indicated that the Komagataeibacter could be a valuable member of D. subobscura microbiota in overcoming the environmental stress. Research on the impact of microbiota on the adaptive response to heavy metals and consequently the potential tradeoffs among different life history traits is of great importance in evolutionary research.
Collapse
Affiliation(s)
- Mirjana Beribaka
- Faculty of Technology Zvornik, University of East Sarajevo, Karakaj 34A, 75400 Zvornik, Bosnia and Herzegovina;
- Correspondence:
| | - Mihailo Jelić
- Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia; (M.J.); (M.S.-R.)
| | - Marija Tanasković
- Department of Genetics of Populations and Ecogenotoxicology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia;
| | - Cvijeta Lazić
- Faculty of Technology Zvornik, University of East Sarajevo, Karakaj 34A, 75400 Zvornik, Bosnia and Herzegovina;
| | - Marina Stamenković-Radak
- Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia; (M.J.); (M.S.-R.)
| |
Collapse
|
326
|
Bianco MR, Ralli M, Modica DM, Amata M, Poma S, Mattina G, Allegra E. The Role of Probiotics in Chronic Rhinosinusitis Treatment: An Update of the Current Literature. Healthcare (Basel) 2021; 9:healthcare9121715. [PMID: 34946441 PMCID: PMC8701913 DOI: 10.3390/healthcare9121715] [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: 10/20/2021] [Revised: 11/25/2021] [Accepted: 12/10/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic rhinosinusitis (CRS) is a significant health problem. It affects 5–12% of the general population. The causes that underlie the onset of CRS are not yet well known. However, many factors may contribute to its onset, such as environmental factors and the host’s general condition. Medical treatment mainly uses local corticosteroids, nasal irrigation, and antibiotics. In recent years, a new therapeutic approach that employs the use of probiotics emerged. Probiotics have been extensively studied as a therapy for dysbiosis and inflammatory pathologies of various parts of the body. We aimed to examine the studies in vivo and in vitro and clinicals reports in the existing literature to update probiotics’ role in rhinosinusitis chronic medical treatment.
Collapse
Affiliation(s)
- Maria Rita Bianco
- Otolaryngology-Department of Health Science, University of Catanzaro, 88100 Catanzaro, Italy;
- Correspondence: ; Tel.: +39-0961-3647130; Fax: +39-0961-3647131
| | - Massimo Ralli
- Department of Sense Organs, Sapienza University of Rome, 00185 Rome, Italy;
| | - Domenico Michele Modica
- Otolaryngology Unit-“Villa Sofia”-Cervello Hospital, 90146 Palermo, Italy; (D.M.M.); (S.P.); (G.M.)
| | - Marta Amata
- Department of Biomedicine and Internal and Specialistic Medicine (DIBIMIS), University of Palermo, 90133 Palermo, Italy;
| | - Salvatore Poma
- Otolaryngology Unit-“Villa Sofia”-Cervello Hospital, 90146 Palermo, Italy; (D.M.M.); (S.P.); (G.M.)
| | - Gianfranco Mattina
- Otolaryngology Unit-“Villa Sofia”-Cervello Hospital, 90146 Palermo, Italy; (D.M.M.); (S.P.); (G.M.)
| | - Eugenia Allegra
- Otolaryngology-Department of Health Science, University of Catanzaro, 88100 Catanzaro, Italy;
| |
Collapse
|
327
|
Sansores-España LD, Melgar-Rodríguez S, Olivares-Sagredo K, Cafferata EA, Martínez-Aguilar VM, Vernal R, Paula-Lima AC, Díaz-Zúñiga J. Oral-Gut-Brain Axis in Experimental Models of Periodontitis: Associating Gut Dysbiosis With Neurodegenerative Diseases. FRONTIERS IN AGING 2021; 2:781582. [PMID: 35822001 PMCID: PMC9261337 DOI: 10.3389/fragi.2021.781582] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022]
Abstract
Periodontitis is considered a non-communicable chronic disease caused by a dysbiotic microbiota, which generates a low-grade systemic inflammation that chronically damages the organism. Several studies have associated periodontitis with other chronic non-communicable diseases, such as cardiovascular or neurodegenerative diseases. Besides, the oral bacteria considered a keystone pathogen, Porphyromonas gingivalis, has been detected in the hippocampus and brain cortex. Likewise, gut microbiota dysbiosis triggers a low-grade systemic inflammation, which also favors the risk for both cardiovascular and neurodegenerative diseases. Recently, the existence of an axis of Oral-Gut communication has been proposed, whose possible involvement in the development of neurodegenerative diseases has not been uncovered yet. The present review aims to compile evidence that the dysbiosis of the oral microbiota triggers changes in the gut microbiota, which creates a higher predisposition for the development of neuroinflammatory or neurodegenerative diseases.The Oral-Gut-Brain axis could be defined based on anatomical communications, where the mouth and the intestine are in constant communication. The oral-brain axis is mainly established from the trigeminal nerve and the gut-brain axis from the vagus nerve. The oral-gut communication is defined from an anatomical relation and the constant swallowing of oral bacteria. The gut-brain communication is more complex and due to bacteria-cells, immune and nervous system interactions. Thus, the gut-brain and oral-brain axis are in a bi-directional relationship. Through the qualitative analysis of the selected papers, we conclude that experimental periodontitis could produce both neurodegenerative pathologies and intestinal dysbiosis, and that periodontitis is likely to induce both conditions simultaneously. The severity of the neurodegenerative disease could depend, at least in part, on the effects of periodontitis in the gut microbiota, which could strengthen the immune response and create an injurious inflammatory and dysbiotic cycle. Thus, dementias would have their onset in dysbiotic phenomena that affect the oral cavity or the intestine. The selected studies allow us to speculate that oral-gut-brain communication exists, and bacteria probably get to the brain via trigeminal and vagus nerves.
Collapse
Affiliation(s)
- Luis Daniel Sansores-España
- Periodontal Biology Laboratory, Faculty of Dentistry, University of Chile, Santiago, Chile
- Faculty of Dentistry, Autonomous University of Yucatán, Mérida, México
| | | | | | - Emilio A. Cafferata
- Department of Periodontology, School of Dentistry, Universidad Científica Del Sur, Lima, Perú
| | | | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Andrea Cristina Paula-Lima
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Jaime Díaz-Zúñiga
- Periodontal Biology Laboratory, Faculty of Dentistry, University of Chile, Santiago, Chile
- Department of Medicine, Faculty of Medicine, University of Atacama, Copiapó, Chile
- *Correspondence: Jaime Díaz-Zúñiga, ,
| |
Collapse
|
328
|
Abstract
The microbiome modulates key processes in metabolism, inflammation, and immunity and plays pivotal roles in many gastrointestinal and liver diseases. Recent experimental studies have demonstrated a key role of the microbiome in hepatocarcinogenesis. Dysfunctions of the gut bacterial flora have a significant effect on liver disease. Dysbiosis is found to be associated with chronic liver diseases. Hepatocellular carcinoma (HCC) is the sixth most common cancer and the third leading cause of cancer-related mortality. The majority of HCC develops in patients with chronic liver disease, caused by chronic viral hepatitis, nonalcoholic fatty liver disease (NAFLD), and alcohol-related fatty liver disease. This review discusses molecular mechanisms of gut microbiome-related hepatocarcinogenesis and the impact of dysbiosis on chronic liver disease progression.
Collapse
Affiliation(s)
- Hikmet Akkız
- Department of Gastroenterology and Hepatology, The University of Çukurova, Adana, Turkey.
| |
Collapse
|
329
|
Larussa T, Abenavoli L, Fabiano G, Mancuso MA, Polimeni N, Dumitrascu DL, Luzza F. Gut microbiota in inflammatory bowel disease: a target for therapy not to be missed. Minerva Gastroenterol (Torino) 2021; 67:357-368. [PMID: 35040302 DOI: 10.23736/s2724-5985.21.02907-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the last years, the gut microbiota achieved great importance, since several studies demonstrated its correlation with the immune system and with the maintenance of intestinal homeostasis, as well as with the regulation of the integrity of the epithelium and the intestinal motility. An imbalance in microbial species promotes a dysbiosis, which has been associated with chronic diseases such as metabolic syndrome, inflammatory diseases, and some behavior disorders. The association with gut microbiota and dysbiosis has been demonstrated mostly in inflammatory bowel disease (IBD). Several studies investigated the application of antibiotics, prebiotics, probiotics, and fecal microbiota transplantation in the treatment strategies for IBD. In this review, we discuss the recent findings on the potential role of the gut microbiota manipulation, with particular attention to bacterial microbiota, which could be implicated for a successful IBD therapeutic approach.
Collapse
Affiliation(s)
- Tiziana Larussa
- Department of Health Sciences, Magna Græcia University, Catanzaro, Italy -
| | - Ludovico Abenavoli
- Department of Health Sciences, Magna Græcia University, Catanzaro, Italy
| | - Giulia Fabiano
- Department of Health Sciences, Magna Græcia University, Catanzaro, Italy
| | - Maria A Mancuso
- Department of Health Sciences, Magna Græcia University, Catanzaro, Italy
| | - Natale Polimeni
- Digestive Endoscopy Service, Casa di Cura Policlinico Madonna della Consolazione, Reggio Calabria, Italy
| | - Dan L Dumitrascu
- Second Medical Department, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Francesco Luzza
- Department of Health Sciences, Magna Græcia University, Catanzaro, Italy
| |
Collapse
|
330
|
Akkermansia muciniphila – obiecujący kandydat na probiotyk nowej generacji. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstrakt
W ostatnich czasach można zaobserwować duży wzrost zainteresowania relacjami między człowiekiem a mikroorganizmami zasiedlającymi jego organizm. Licznie występują w oraz na ciele człowieka, a ich liczba przekracza liczbę komórek ludzkiego organizmu. Udoskonalenie technik badawczych pozwala lepiej zrozumieć molekularne podłoże tych oddziaływań, co być może pozwoli na wykorzystanie w terapiach tych mikroorganizmów, które korzystnie wpływają na organizm człowieka. W artykule podsumowano dotychczasową wiedzę na temat fizjologii licznie występującej w przewodzie pokarmowym bakterii Akkermansia muciniphila i jej wpływu na organizm gospodarza. Opisano właściwości A. muciniphila, jej funkcjonowanie w środowisku przewodu pokarmowego oraz relacje (zarówno antagonistyczne jak i symbiotyczne) z innymi tam bytującymi mikroorganizmami. Przedstawiono także mechanizmy oddziaływania A. muciniphila na barierę jelitową, układ immunologiczny oraz metabolizm energetyczny gospodarza. Ponadto opisano jej rolę w patogenezie i terapii chorób, w tym m.in. cukrzycy typu 2, nieswoistego zapalenia jelit, zaburzeń neurologicznych, astmy, a także jej wpływ na odpowiedź pacjenta na terapie przeciwnowotworowe oparte na działaniu układu odpornościowego. Duże zainteresowanie tą bakterią, a także przytoczone wyniki badań, w tym tych sprawdzających bezpieczeństwo jej stosowania, wskazują, że A. muciniphila może być obiecującym kandydatem na probiotyk nowej generacji. Niewątpliwie jednak przed dopuszczeniem A. muciniphila do powszechnego stosowania konieczne są dalsze badania z udziałem ludzi, a także wnikliwa ocena bezpieczeństwa jej stosowania.
Collapse
|
331
|
Dey P, Chaudhuri SR, Efferth T, Pal S. The intestinal 3M (microbiota, metabolism, metabolome) zeitgeist - from fundamentals to future challenges. Free Radic Biol Med 2021; 176:265-285. [PMID: 34610364 DOI: 10.1016/j.freeradbiomed.2021.09.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/18/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022]
Abstract
The role of the intestine in human health and disease has historically been neglected and was mostly attributed to digestive and absorptive functions. In the past two decades, however, discoveries related to human nutrition and intestinal host-microbe reciprocal interaction have established the essential role of intestinal health in the pathogenesis of chronic diseases and the overall wellbeing. That transfer of gut microbiota could be a means of disease phenotype transfer has revolutionized our understanding of chronic disease pathogenesis. This narrative review highlights the major concepts related to intestinal microbiota, metabolism, and metabolome (3M) that have facilitated our fundamental understanding of the association between the intestine, and human health and disease. In line with increased interest of microbiota-dependent modulation of human health by dietary phytochemicals, we have also discussed the emerging concepts beyond the phytochemical bioactivities which emphasizes the integral role of microbial metabolites of parent phytochemicals at extraintestinal tissues. Finally, this review concludes with challenges and future prospects in defining the 3M interactions and has emphasized the fact that, it takes 'guts' to stay healthy.
Collapse
Affiliation(s)
- Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India.
| | - Saumya Ray Chaudhuri
- Council of Scientific and Industrial Research (CSIR), Institute of Microbial Technology, Chandigarh, India
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Sirshendu Pal
- Mukherjee Hospital, Mitra's Clinic and Nursing Home, Siliguri, West Bengal, India
| |
Collapse
|
332
|
Changes to human faecal microbiota after international travel. Travel Med Infect Dis 2021; 44:102199. [PMID: 34781018 DOI: 10.1016/j.tmaid.2021.102199] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND The aim was to investigate whether travelling to less-resourced destinations influences the composition of faecal microbiota in generally healthy adults. METHOD In this prospective observational study, 47 adults (median age, 24 years; 73% females) travelled from Sweden to distant destinations for 1-12 weeks. Five faecal samples, two before and three after travel, were analysed by 16S amplicon massive parallel sequencing. Subjects had taken no antibiotics within three months of each sampling. RESULTS The overall composition of faecal microbiota was not affected by travel. However, when looking at the relative abundance of individual bacterial taxa, Enterobacteriaceae demonstrated a 10-fold increase immediately after the trip as compared to the samples taken before travelling. Conversely, the relative abundance of Christensenellaceae had decreased equally much. Both these changes were reversible within nine weeks. CONCLUSIONS International travel, even to less-resourced countries, did not appear to alter the overall diversity of human faecal microbiota as studied here after travelling. However, Enterobacteriaceae bacteria, often associated with infection, inflammation, and antibiotic resistance, showed dramatically elevated levels, and Christensenellaceae, frequently associated with healthy conditions, demonstrated remarkably declined levels in relative abundance as detected immediately after travel. Both these changes returned to original pre-travel levels within nine weeks.
Collapse
|
333
|
Glinert A, Turjeman S, Elliott E, Koren O. Microbes, metabolites and (synaptic) malleability, oh my! The effect of the microbiome on synaptic plasticity. Biol Rev Camb Philos Soc 2021; 97:582-599. [PMID: 34734461 PMCID: PMC9298272 DOI: 10.1111/brv.12812] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/10/2021] [Accepted: 10/22/2021] [Indexed: 12/15/2022]
Abstract
The microbiome influences the emotional and cognitive phenotype of its host, as well as the neurodevelopment and pathophysiology of various brain processes and disorders, via the well‐established microbiome–gut–brain axis. Rapidly accumulating data link the microbiome to severe neuropsychiatric disorders in humans, including schizophrenia, Alzheimer's and Parkinson's. Moreover, preclinical work has shown that perturbation of the microbiome is closely associated with social, cognitive and behavioural deficits. The potential of the microbiome as a diagnostic and therapeutic tool is currently undercut by a lack of clear mechanistic understanding of the microbiome–gut–brain axis. This review establishes the hypothesis that the mechanism by which this influence is carried out is synaptic plasticity – long‐term changes to the physical and functional neuronal structures that enable the brain to undertake learning, memory formation, emotional regulation and more. By examining the different constituents of the microbiome–gut–brain axis through the lens of synaptic plasticity, this review explores the diverse aspects by which the microbiome shapes the behaviour and mental wellbeing of the host. Key elements of this complex bi‐directional relationship include neurotransmitters, neuronal electrophysiology, immune mediators that engage with both the central and enteric nervous systems and signalling cascades that trigger long‐term potentiation of synapses. The importance of establishing mechanistic correlations along the microbiome–gut–brain axis cannot be overstated as they hold the potential for furthering current understanding regarding the vast fields of neuroscience and neuropsychiatry. This review strives to elucidate the promising theory of microbiome‐driven synaptic plasticity in the hope of enlightening current researchers and inspiring future ones.
Collapse
Affiliation(s)
- Ayala Glinert
- Azrieli Faculty of Medicine, Bar Ilan University, 8 Henrietta Szold, Safed, 1311502, Israel
| | - Sondra Turjeman
- Azrieli Faculty of Medicine, Bar Ilan University, 8 Henrietta Szold, Safed, 1311502, Israel
| | - Evan Elliott
- Azrieli Faculty of Medicine, Bar Ilan University, 8 Henrietta Szold, Safed, 1311502, Israel
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar Ilan University, 8 Henrietta Szold, Safed, 1311502, Israel
| |
Collapse
|
334
|
Vithoulkas G. An integrated perspective on transmutation of acute inflammation into chronic and the role of the microbiome. J Med Life 2021; 14:740-747. [PMID: 35126742 PMCID: PMC8811668 DOI: 10.25122/jml-2021-0375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/30/2021] [Indexed: 11/20/2022] Open
Abstract
The Continuum theory and the Levels of Health theory were separately proposed to explain the myriad responses to treatment and understand the process of health and disease in an individual. In light of accumulating evidence on the intricate relationship between the human immune system and microbiome, an attempt is made in this article to connect these two theories to explain the transmutation of the efficiently responding immune system (through the acute inflammatory response and high fever) to one involved in a low-grade chronic inflammatory process (resulting in chronic disease). There is already enough evidence to demonstrate the role of the microbiome in all chronic inflammatory diseases. In this article, we discuss the mechanism by which subjecting a healthy person to continuous drug treatment for acute inflammatory conditions (at a certain time) leads to transmutation to chronic disease. Although this hypothesis requires further experimental evidence, it calls for a reconsideration of the manner in which we treat acute infectious diseases in the population.
Collapse
Affiliation(s)
- George Vithoulkas
- University of the Aegean, Syros, Greece
- Postgraduate Doctors’ Training Institute, Health Care Ministry of the Chuvash Republic, Cheboksary, Russian Federation
| |
Collapse
|
335
|
Fonseca W, Malinczak CA, Fujimura K, Li D, McCauley K, Li J, Best SKK, Zhu D, Rasky AJ, Johnson CC, Bermick J, Zoratti EM, Ownby D, Lynch SV, Lukacs NW, Ptaschinski C. Maternal gut microbiome regulates immunity to RSV infection in offspring. J Exp Med 2021; 218:212680. [PMID: 34613328 PMCID: PMC8500238 DOI: 10.1084/jem.20210235] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/26/2021] [Accepted: 09/07/2021] [Indexed: 12/15/2022] Open
Abstract
Development of the immune system can be influenced by diverse extrinsic and intrinsic factors that influence the risk of disease. Severe early life respiratory syncytial virus (RSV) infection is associated with persistent immune alterations. Previously, our group had shown that adult mice orally supplemented with Lactobacillus johnsonii exhibited decreased airway immunopathology following RSV infection. Here, we demonstrate that offspring of mice supplemented with L. johnsonii exhibit reduced airway mucus and Th2 cell–mediated response to RSV infection. Maternal supplementation resulted in a consistent gut microbiome in mothers and their offspring. Importantly, supplemented maternal plasma and breastmilk, and offspring plasma, exhibited decreased inflammatory metabolites. Cross-fostering studies showed that prenatal Lactobacillus exposure led to decreased Th2 cytokines and lung inflammation following RSV infection, while postnatal Lactobacillus exposure diminished goblet cell hypertrophy and mucus production in the lung in response to airway infection. These studies demonstrate that Lactobacillus modulation of the maternal microbiome and associated metabolic reprogramming enhance airway protection against RSV in neonates.
Collapse
Affiliation(s)
- Wendy Fonseca
- Department of Pathology, University of Michigan, Ann Arbor, MI
| | | | - Kei Fujimura
- Department of Medicine-Gastroenterology, University of California, San Francisco, San Francisco, CA
| | - Danny Li
- Department of Medicine-Gastroenterology, University of California, San Francisco, San Francisco, CA
| | - Kathryn McCauley
- Department of Medicine-Gastroenterology, University of California, San Francisco, San Francisco, CA
| | - Jia Li
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI
| | | | - Diana Zhu
- Department of Pathology, University of Michigan, Ann Arbor, MI
| | - Andrew J Rasky
- Department of Pathology, University of Michigan, Ann Arbor, MI
| | | | - Jennifer Bermick
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Edward M Zoratti
- Division of Allergy and Clinical Immunology, Department of Medicine, Henry Ford Health System, Detroit, MI
| | - Dennis Ownby
- Department of Pediatrics, Augusta University, Augusta, GA
| | - Susan V Lynch
- Department of Medicine-Gastroenterology, University of California, San Francisco, San Francisco, CA
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI.,Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI
| | - Catherine Ptaschinski
- Department of Pathology, University of Michigan, Ann Arbor, MI.,Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI
| |
Collapse
|
336
|
He Y, Hong Q, Zhou D, Wang S, Yang B, Yuan Y, Zhang W, Huang Y, E G. Genome-wide selective detection of Mile red-bone goat using next-generation sequencing technology. Ecol Evol 2021; 11:14805-14812. [PMID: 34765142 PMCID: PMC8571596 DOI: 10.1002/ece3.8165] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/31/2021] [Indexed: 11/23/2022] Open
Abstract
The ecotype population of goats (Capra hircus) was created by long-term artificial selection and natural adaptation. Mile red-bone goat is an indigenous breed with visible red bones, and its special bone structure has received extensive attention. This study aimed to identify genetic variants and candidate genes associated with specific bone phenotypes using next-generation sequencing technology (NGS). The results revealed that 31,828,206 single nucleotide polymorphisms (SNPs) were obtained from 72 goats (20 Mile red-bone goats and 52 common goats) by NGS. A total of 100 candidate genes were identified on the basis top 1% window interaction from nucleotide diversity (π), π ratio (π A/π B), and pairwise fixation index (F ST). Exactly 77 known signaling pathways were enriched. Specifically, three coding genes (NMNAT2, LOC102172983, and PNLIP) were annotated in the vitamin metabolism signaling pathways, and NCF2 was annotated to the osteoclast (OC) differentiation pathway. Furthermore, 5862 reliable copy number variations (CNVs) were obtained, and 14 and 24 genes were annotated with the top 1‰ CNV based on F ST (>0.490) and V ST (>0.527), respectively. Several pathways related to bone development and metabolism of exogenous substances in vivo, including calcium signaling pathway, OC differentiation, and glycerophospholipid metabolism, were annotated. Specifically, six genes from 19 candidate CNVs, which were obtained by interaction of the top 1‰ CNVs with F ST and V ST, were annotated to mucin-type O-glycan biosynthesis and metabolic pathways. Briefly, the results implied that pseudopurpurin and specific genetic variants work together to contribute to the red-bone color and specific bone structure of Mile red-bone goat. This study is helpful to understanding the genetic basis of the unique bone phenotype of Mile red-bone goats.
Collapse
Affiliation(s)
- Yong‐Meng He
- Chongqing Key Laboratory of Forage & HerbivoreCollege of Animal Science and TechnologyChongqing Engineering Research Centre for Herbivores Resource Protection and UtilizationSouthwest UniversityChongqingChina
| | - Qiong‐Hua Hong
- Yunnan Animal Science and Veterinary InstituteKunmingChina
| | - Dong‐Ke Zhou
- Chongqing Key Laboratory of Forage & HerbivoreCollege of Animal Science and TechnologyChongqing Engineering Research Centre for Herbivores Resource Protection and UtilizationSouthwest UniversityChongqingChina
| | - Shi‐Zhi Wang
- Chongqing Key Laboratory of Forage & HerbivoreCollege of Animal Science and TechnologyChongqing Engineering Research Centre for Herbivores Resource Protection and UtilizationSouthwest UniversityChongqingChina
| | - Bai‐Gao Yang
- Chongqing Key Laboratory of Forage & HerbivoreCollege of Animal Science and TechnologyChongqing Engineering Research Centre for Herbivores Resource Protection and UtilizationSouthwest UniversityChongqingChina
| | - Ying Yuan
- Chongqing Key Laboratory of Forage & HerbivoreCollege of Animal Science and TechnologyChongqing Engineering Research Centre for Herbivores Resource Protection and UtilizationSouthwest UniversityChongqingChina
| | - Wei‐Yi Zhang
- Chongqing Key Laboratory of Forage & HerbivoreCollege of Animal Science and TechnologyChongqing Engineering Research Centre for Herbivores Resource Protection and UtilizationSouthwest UniversityChongqingChina
| | - Yong‐Fu Huang
- Chongqing Key Laboratory of Forage & HerbivoreCollege of Animal Science and TechnologyChongqing Engineering Research Centre for Herbivores Resource Protection and UtilizationSouthwest UniversityChongqingChina
| | - Guang‐Xin E
- Chongqing Key Laboratory of Forage & HerbivoreCollege of Animal Science and TechnologyChongqing Engineering Research Centre for Herbivores Resource Protection and UtilizationSouthwest UniversityChongqingChina
| |
Collapse
|
337
|
Pham VT, Dold S, Rehman A, Bird JK, Steinert RE. Vitamins, the gut microbiome and gastrointestinal health in humans. Nutr Res 2021; 95:35-53. [PMID: 34798467 DOI: 10.1016/j.nutres.2021.09.001] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 02/07/2023]
Abstract
The gut microbiome plays important roles in the maintenance of host health and the pathogenesis of many diseases. Diet is a key modulator of the gut microbiome. There is increasing evidence that nutrients other than fermentable fiber affect the gut microbial composition. In this review, we discuss the effects of vitamins on the gut microbiome, and related gastrointestinal health, based on in vitro, animal and human studies. Some vitamins, when provided in large doses or when delivered to the large intestine, have been shown to beneficially modulate the gut microbiome by increasing the abundance of presumed commensals (vitamins A, B2, D, E, and beta-carotene), increasing or maintaining microbial diversity (vitamins A, B2, B3, C, K) and richness (vitamin D), increasing short chain fatty acid production (vitamin C), or increasing the abundance of short chain fatty acid producers (vitamins B2, E). Others, such as vitamins A and D, modulate the gut immune response or barrier function, thus, indirectly influencing gastrointestinal health or the microbiome. Future research is needed to explore these potential effects and to elucidate the underlying mechanisms and host health benefits.
Collapse
Affiliation(s)
- Van T Pham
- DSM Nutritional Products, Kaiseraugst, Switzerland.
| | - Susanne Dold
- DSM Nutritional Products, Kaiseraugst, Switzerland
| | | | | | - Robert E Steinert
- DSM Nutritional Products, Kaiseraugst, Switzerland; Department of Surgery, Division of Visceral and Transplantation Surgery, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
338
|
Laursen MF, Sakanaka M, von Burg N, Mörbe U, Andersen D, Moll JM, Pekmez CT, Rivollier A, Michaelsen KF, Mølgaard C, Lind MV, Dragsted LO, Katayama T, Frandsen HL, Vinggaard AM, Bahl MI, Brix S, Agace W, Licht TR, Roager HM. Bifidobacterium species associated with breastfeeding produce aromatic lactic acids in the infant gut. Nat Microbiol 2021; 6:1367-1382. [PMID: 34675385 PMCID: PMC8556157 DOI: 10.1038/s41564-021-00970-4] [Citation(s) in RCA: 243] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 08/26/2021] [Indexed: 12/12/2022]
Abstract
Breastfeeding profoundly shapes the infant gut microbiota, which is critical for early life immune development, and the gut microbiota can impact host physiology in various ways, such as through the production of metabolites. However, few breastmilk-dependent microbial metabolites mediating host-microbiota interactions are currently known. Here, we demonstrate that breastmilk-promoted Bifidobacterium species convert aromatic amino acids (tryptophan, phenylalanine and tyrosine) into their respective aromatic lactic acids (indolelactic acid, phenyllactic acid and 4-hydroxyphenyllactic acid) via a previously unrecognized aromatic lactate dehydrogenase (ALDH). The ability of Bifidobacterium species to convert aromatic amino acids to their lactic acid derivatives was confirmed using monocolonized mice. Longitudinal profiling of the faecal microbiota composition and metabolome of Danish infants (n = 25), from birth until 6 months of age, showed that faecal concentrations of aromatic lactic acids are correlated positively with the abundance of human milk oligosaccharide-degrading Bifidobacterium species containing the ALDH, including Bifidobacterium longum, B. breve and B. bifidum. We further demonstrate that faecal concentrations of Bifidobacterium-derived indolelactic acid are associated with the capacity of these samples to activate in vitro the aryl hydrocarbon receptor (AhR), a receptor important for controlling intestinal homoeostasis and immune responses. Finally, we show that indolelactic acid modulates ex vivo immune responses of human CD4+ T cells and monocytes in a dose-dependent manner by acting as an agonist of both the AhR and hydroxycarboxylic acid receptor 3 (HCA3). Our findings reveal that breastmilk-promoted Bifidobacterium species produce aromatic lactic acids in the gut of infants and suggest that these microbial metabolites may impact immune function in early life.
Collapse
Affiliation(s)
- Martin F Laursen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Mikiyasu Sakanaka
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
- Faculty of Bioresources and Environmental Sciences, Ishikawa Prefectural University, Ishikawa, Japan
| | - Nicole von Burg
- Mucosal Immunology Group, Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Urs Mörbe
- Mucosal Immunology Group, Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Daniel Andersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Janne Marie Moll
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Ceyda T Pekmez
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg C, Denmark
| | - Aymeric Rivollier
- Mucosal Immunology Group, Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Kim F Michaelsen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg C, Denmark
| | - Christian Mølgaard
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg C, Denmark
| | - Mads Vendelbo Lind
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg C, Denmark
| | - Lars O Dragsted
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg C, Denmark
| | - Takane Katayama
- Faculty of Bioresources and Environmental Sciences, Ishikawa Prefectural University, Ishikawa, Japan
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Henrik L Frandsen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | | | - Martin I Bahl
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Susanne Brix
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - William Agace
- Mucosal Immunology Group, Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
- Immunology Section, BMC D14, Department of Experimental Medicine, Lund University, Lund, Sweden
| | - Tine R Licht
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark.
| | - Henrik M Roager
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark.
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg C, Denmark.
| |
Collapse
|
339
|
Parra-Torres V, Melgar-Rodríguez S, Muñoz-Manríquez C, Sanhueza B, Cafferata EA, Paula-Lima AC, Díaz-Zúñiga J. Periodontal bacteria in the brain-Implication for Alzheimer's disease: A systematic review. Oral Dis 2021; 29:21-28. [PMID: 34698406 DOI: 10.1111/odi.14054] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 10/10/2021] [Accepted: 10/13/2021] [Indexed: 11/29/2022]
Abstract
Periodontitis is a chronic non-communicable disease caused by a dysbiotic microbiota. Pathogens can spread to the bloodstream, colonize other tissues or organs, and favor the onset of other pathologies, such as Alzheimer's disease (AD). Pathogens could permanently or transiently colonize the brain and induce an immune response. Thus, we analyzed the evidence combining oral bacteria's detection in the brain, both in animals and humans affected with AD. This systematic review was carried out following the PRISMA guideline. Studies that detected oral bacteria at the brain level were selected. The search was carried out in the Medline, Latindex, SciELO, and Cochrane Library databases. SYRCLE tool and Newcastle-Ottawa Scale were used for the risk of bias assessment. 23 studies were selected according to the eligibility criteria. Infection with oral pathogens in animals was related to developing neuropathological characteristics of AD and bacteria detection in the brain. In patients with AD, oral bacteria were detected in brain tissues, and increased levels of pro-inflammatory cytokines were also detected. There is evidence of a microbiological susceptibility to develop AD when the most dysbiosis-associated oral bacteria are present. The presence of bacteria in the brain is related to AD's pathological characteristics, suggesting an etiological oral-brain axis.
Collapse
Affiliation(s)
- Valeria Parra-Torres
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | | | | | - Benjamín Sanhueza
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Emilio A Cafferata
- Department of Periodontology, School of Dentistry, Universidad Científica del Sur, Lima, Perú
| | - Andrea C Paula-Lima
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Jaime Díaz-Zúñiga
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Department of Medicine, Faculty of Medicine, Universidad de Atacama, Copiapo, Chile
| |
Collapse
|
340
|
Caenazzo L, Tozzo P. Microbiome Forensic Biobanking: A Step toward Microbial Profiling for Forensic Human Identification. Healthcare (Basel) 2021; 9:1371. [PMID: 34683051 PMCID: PMC8544459 DOI: 10.3390/healthcare9101371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/04/2021] [Accepted: 10/11/2021] [Indexed: 11/16/2022] Open
Abstract
In recent years many studies have highlighted the great potential of microbial analysis in human identification for forensic purposes, with important differences in microbial community composition and function across different people and locations, showing a certain degree of uncertainty. Therefore, further studies are necessary to enable forensic scientists to evaluate the risk of microbial transfer and recovery from various items and to further critically evaluate the suitability of current human DNA recovery protocols for human microbial profiling for identification purposes. While the establishment and development of microbiome research biobanks for clinical applications is already very structured, the development of studies on the applicability of microbiome biobanks for forensic purposes is still in its infancy. The creation of large population microbiome biobanks, specifically dedicated to forensic human identification, could be worthwhile. This could also be useful to increase the practical applications of forensic microbiology for identification purposes, given that this type of evidence is currently absent from most real casework investigations and judicial proceedings in courts.
Collapse
Affiliation(s)
| | - Pamela Tozzo
- Laboratory of Forensic Genetics, Department of Molecular Medicine, University of Padova, 35121 Padova, Italy;
| |
Collapse
|
341
|
Nowland TL, Kirkwood RN, Pluske JR. Review: Can early-life establishment of the piglet intestinal microbiota influence production outcomes? Animal 2021; 16 Suppl 2:100368. [PMID: 34649827 DOI: 10.1016/j.animal.2021.100368] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/07/2021] [Accepted: 08/27/2021] [Indexed: 12/21/2022] Open
Abstract
The gastrointestinal tract microbiota is involved in the development and function of many body processes. Studies demonstrate that early-life microbial colonisation is the most important time for shaping intestinal and immune development, with perturbations to the microbiota during this time having long-lasting negative implications for the host. Piglets face many early-life events that shape the acquisition and development of their intestinal microbiota. The pork industry has a unique advantage in that the producer has a degree of control over what piglets are exposed to, providing conditions that allow for optimum piglet growth and development. An influx of publications within this area has occurred in recent times and with this, interest surrounding its application in pork production has increased. However, it can be difficult to distinguish which research is of most relevance to industry in terms of delivering repeatable and reliable production outcomes. In this review, we describe the literature surrounding research within pigs, predominantly during the preweaning period that has either provided solutions to industry problems or is generating information targeted at addressing relevant industry issues, with the focus being on studies demonstrating causation where possible. This review will provide a basis for the development of new studies targeted at understanding how to better support initial intestinal microbiota colonisation in order to improve piglet health and survival.
Collapse
Affiliation(s)
- T L Nowland
- Livestock Sciences, South Australian Research and Development Institute, PPPI Building, University of Adelaide, Roseworthy, SA 5371, Australia.
| | - R N Kirkwood
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA 5371, Australia
| | - J R Pluske
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
342
|
Nader-Macías MEF, De Gregorio PR, Silva JA. Probiotic lactobacilli in formulas and hygiene products for the health of the urogenital tract. Pharmacol Res Perspect 2021; 9:e00787. [PMID: 34609059 PMCID: PMC8491456 DOI: 10.1002/prp2.787] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/04/2021] [Accepted: 04/02/2021] [Indexed: 12/13/2022] Open
Abstract
Lactobacilli are the predominant microorganisms of the healthy human vagina. A novel alternative for the prevention and treatment of female urogenital tract infections (UGTI) is the inclusion of these microorganisms as active pharmaceutical ingredients in probiotic formulas, and more recently in female hygienic products. Probiotics are defined as “live microorganisms that, when administered in adequate amounts, confer a health benefit on the host.” A list of requirements must be considered during the development of probiotic product/formula for the female urogenital tract (UGT). This review aims to resume the requirements, probiotic characteristics, and clinical trial applied to determine the effect of probiotic and potentially probiotic strains on different woman’s physiological and pathological conditions, and in preterm birth prevention. A revision of female hygienic products available in the world market is included, together with novel studies applying nanotechnology for Lactobacillus incorporation in hygienic products. Further studies and well‐designed clinical trials are urgently required to complement the current knowledge and applications of probiotics in the female UGT. The use of probiotic formulas and products will improve and restore the ecological equilibrium of the UGT microbiome to prevent and treat UGTI in women under different conditions.
Collapse
Affiliation(s)
- María Elena Fátima Nader-Macías
- Centro de Referencia para Lactobacilos-Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina (CERELA-CONICET), San Miguel de Tucumán, Argentina
| | - Priscilla Romina De Gregorio
- Centro de Referencia para Lactobacilos-Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina (CERELA-CONICET), San Miguel de Tucumán, Argentina
| | - Jessica Alejandra Silva
- Centro de Referencia para Lactobacilos-Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina (CERELA-CONICET), San Miguel de Tucumán, Argentina
| |
Collapse
|
343
|
Xie M, Zhang S, Xu L, Wu Z, Yuan J, Chen X. Comparison of the Intestinal Microbiota During the Different Growth Stages of Red Swamp Crayfish ( Procambarus clarkii). Front Microbiol 2021; 12:696281. [PMID: 34589066 PMCID: PMC8473915 DOI: 10.3389/fmicb.2021.696281] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 08/12/2021] [Indexed: 12/21/2022] Open
Abstract
This study aimed to determine the effect of the growth stage of Procambarus clarkii on their intestinal microbiota. Intestinal samples of five different growth stages of P. clarkii (first instar, second instar, third instar, juvenile, and adult) from laboratory culture were analyzed through the Illumina MiSeq high-throughput sequencing platform to determine the intestinal microbiome of crayfish. The alpha diversity decreased along with the growth of the crayfish, with the relative abundance of the microbiota changing among stages; crayfish at closer development stages had a more comparable intestinal microbiota composition. A comparative analysis by principal component analysis and principal coordinate analysis showed that there were significant differences in the intestinal microbiota of crayfish among the different growth stages, except for the first two stages of larval crayfish, and the intestinal microbiota showed a consistent progression pattern from the larval stage to the juvenile stage. Some microbiota showed stage specificity, which might be the characteristic microbiota of different stages of growth. According to FAPROTAX functional clustering analysis, the three stages of larvae were clustered together, while the juvenile and adult stages were clustered separately according to the growth stage, indicating that, in the early stages of larval development, the function of the intestinal flora was similar; as the body grew and developed, the composition and function of the intestinal microbiota also changed.
Collapse
Affiliation(s)
- Mengqi Xie
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agriculture University, Wuhan, China.,Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan, China
| | - Shiyu Zhang
- Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan, China
| | - Lili Xu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agriculture University, Wuhan, China
| | - Zhixin Wu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agriculture University, Wuhan, China.,Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan, China
| | - Junfa Yuan
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agriculture University, Wuhan, China.,Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan, China
| | - Xiaoxuan Chen
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agriculture University, Wuhan, China
| |
Collapse
|
344
|
Cho NA, Sales KM, Sampsell K, Wang W, Noye Tuplin EW, Lowry DE, Reimer RA. C-section birth increases offspring obesity risk dependent on maternal diet and obesity status in rats. Obesity (Silver Spring) 2021; 29:1664-1675. [PMID: 34464518 DOI: 10.1002/oby.23258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/10/2021] [Accepted: 06/22/2021] [Indexed: 12/26/2022]
Abstract
OBJECTIVE The gut microbiota is a complex ecosystem that shapes host metabolism, especially in early life. Maternal vaginal and gut microbiota is vertically transmitted to offspring during natural birth. Offspring born by cesarean section (CS) do not receive these bacteria and exhibit higher obesity risk later in life. The objective of this study was to examine differences in obesity risk between offspring born naturally (NB) or by CS to lean/obese dams. METHODS Lean and obese rat dams gave birth to offspring naturally or by CS. Offspring obesity risk was analyzed via body weight/composition, food intake, sucrose preference, gut microbiota, and gene expression in gut and brain tissues. RESULTS Obese (O)+CS offspring showed greater weight gain and caloric intake but a reduction in hypothalamic agouti related neuropeptide, neuropeptide Y, and interleukin 1β expression compared with O+NB offspring. Lean (L)+CS offspring had higher serum corticosterone concentration and reduced liver peroxisome proliferator-activated receptor γ expression compared with L+NB. O+CS offspring had long-term alterations to gut microbiota, including increased relative abundance of Faecalibaculum and reduced Muribaculaceae. CONCLUSIONS Overall, CS alters obesity risk differentially based on maternal obesity status. Further studies looking at the risks of obesity associated with CS are needed, with special attention paid to maternal obesity status and gut microbiota.
Collapse
Affiliation(s)
- Nicole A Cho
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Kate M Sales
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Kara Sampsell
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Weilan Wang
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | | | - Dana E Lowry
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Raylene A Reimer
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
345
|
Enteric Microbiota-Mediated Serotonergic Signaling in Pathogenesis of Irritable Bowel Syndrome. Int J Mol Sci 2021; 22:ijms221910235. [PMID: 34638577 PMCID: PMC8508930 DOI: 10.3390/ijms221910235] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/18/2021] [Accepted: 09/19/2021] [Indexed: 12/12/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a chronic functional disorder that affects the gastrointestinal tract. Details regarding the pathogenesis of IBS remain largely unknown, though the dysfunction of the brain-gut-microbiome (BGM) axis is a major etiological factor, in which neurotransmitters serve as a key communication tool between enteric microbiota and the brain. One of the most important neurotransmitters in the pathology of IBS is serotonin (5-HT), as it influences gastrointestinal motility, pain sensation, mucosal inflammation, immune responses, and brain activity, all of which shape IBS features. Genome-wide association studies discovered susceptible genes for IBS in serotonergic signaling pathways. In clinical practice, treatment strategies targeting 5-HT were effective for a certain portion of IBS cases. The synthesis of 5-HT in intestinal enterochromaffin cells and host serotonergic signaling is regulated by enteric resident microbiota. Dysbiosis can trigger IBS development, potentially through aberrant 5-HT signaling in the BGM axis; thus, the manipulation of the gut microbiota may be an alternative treatment strategy. However, precise information regarding the mechanisms underlying the microbiota-mediated intestinal serotonergic pathway related to the pathogenesis of IBS remains unclear. The present review summarizes current knowledge and recent progress in understanding microbiome–serotonin interaction in IBS cases.
Collapse
|
346
|
Gut Microbiome Composition and Metabolic Status Are Differently Affected by Early Exposure to Unhealthy Diets in a Rat Model. Nutrients 2021; 13:nu13093236. [PMID: 34579113 PMCID: PMC8469890 DOI: 10.3390/nu13093236] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/11/2021] [Accepted: 09/15/2021] [Indexed: 12/11/2022] Open
Abstract
Childhood is a critical stage of development during which diet can have profound influence on the microbiota–host interactions, leading to potentially lifelong impacts. This study aimed to investigate whether the consumption of cafeteria diet (CAFD) and sugary drinks during early rat life alters the structure of the gut microbial community and the metabolic activity. Four-week-old male Wistar rats (n = 27) were fed a standard chow diet with ad libitum access to water (CD) or to sucrose solution (HSD), and a third group was fed with CAFD and a sucrose solution for 14 weeks. HSD and CAFD consumption induced alterations in Firmicutes to Bacteroidetes ratio, Proteobacteria, and Verrucomicrobia. HSD increased the abundance of Barnesiella, whereas CAFD induced a depletion of Saccharibacteria. CAFD increased total white adipose tissue (WAT) weight (p < 0.0005) compared to CD. When CAFD was compared to HSD, a significant difference was found only for retroperitoneal WAT (p < 0.0005). Unhealthy diet-fed groups presented higher glucose (p < 0.0005), total cholesterol and creatinine serum levels (p < 0.005) compared to the CD rats. Early-life consumption of HSD, and of CAFD even more so, can have long-lasting negative effects on metabolic function. The gut microbiota communities were distinctively perturbed by diet composition.
Collapse
|
347
|
Human Milk Oligosaccharide-Stimulated Bifidobacterium Species Contribute to Prevent Later Respiratory Tract Infections. Microorganisms 2021; 9:microorganisms9091939. [PMID: 34576834 PMCID: PMC8465161 DOI: 10.3390/microorganisms9091939] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/09/2021] [Accepted: 09/09/2021] [Indexed: 12/11/2022] Open
Abstract
(1) Background: Human milk oligosaccharides (HMOs) may support immune protection, partly via their action on the early-life gut microbiota. Exploratory findings of a randomized placebo-controlled trial associated 2′fucosyllactose (2′FL) and lacto-N-neotetraose (LNnT) formula feeding with reduced risk for reported bronchitis and lower respiratory tract illnesses (LRTI), as well as changes in gut microbiota composition. We sought to identify putative gut microbial mechanisms linked with these clinical observations. (2) Methods: We used stool microbiota composition, metabolites including organic acids and gut health markers in several machine-learning-based classification tools related prospectively to experiencing reported bronchitis or LRTI, as compared to no reported respiratory illness. We performed preclinical epithelial barrier function modelling to add mechanistic insight to these clinical observations. (3) Results: Among the main features discriminant for infants who did not experience any reported bronchitis (n = 80/106) or LRTI (n = 70/103) were the 2-HMO formula containing 2′FL and LNnT, higher acetate, fucosylated glycans and Bifidobacterium, as well as lower succinate, butyrate, propionate and 5-aminovalerate, along with Carnobacteriaceae members and Escherichia. Acetate correlated with several Bifidobacterium species. By univariate analysis, infants experiencing no bronchitis or LRTI, compared with those who did, showed higher acetate (p < 0.007) and B. longum subsp. infantis (p ≤ 0.03). In vitro experiments demonstrate that 2′FL, LNnT and lacto-N-tetraose (LNT) stimulated B. longum subsp. infantis (ATCC15697) metabolic activity. Metabolites in spent culture media, primarily due to acetate, supported epithelial barrier protection. (4) Conclusions: An early-life gut ecology characterized by Bifidobacterium-species-driven metabolic changes partly explains the observed clinical outcomes of reduced risk for bronchitis and LRTI in infants fed a formula with HMOs. (Trial registry number NCT01715246.).
Collapse
|
348
|
Walsh CM, Haasbroek C. The interaction between the microbiome, diet and health. SOUTH AFRICAN JOURNAL OF CLINICAL NUTRITION 2021. [DOI: 10.1080/16070658.2021.1966226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Corinna May Walsh
- Department of Nutrition and Dietetics, University of the Free State, Bloemfontein, South Africa
| | - Carina Haasbroek
- Department of Nutrition and Dietetics, University of the Free State, Bloemfontein, South Africa
| |
Collapse
|
349
|
Shen W, Qiu W, Liu Y, Liao W, Ma Y, He Y, Wang Z, Zhou H. Postnatal age is strongly correlated with the early development of the gut microbiome in preterm infants. Transl Pediatr 2021; 10:2313-2324. [PMID: 34733672 PMCID: PMC8506066 DOI: 10.21037/tp-21-367] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/27/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The gut microbiome plays a potential role in clinical events in preterm infants and may affect their lateral development. Understanding the initial colonization of microbes in the gut, their early dynamic changes, and the major factors correlated with these changes would provide crucial information about the developmental process in early life. METHODS The present study enrolled 151 preterm infants and examined the longitudinal dynamics of their fecal microbiome profiles during the period of hospitalization using 16S ribosomal RNA gene sequencing. Random forest modeling was used to predict postnatal age (Age), postmenstrual age (PMA), and gestational age (GA), using gut microbiome features. RESULTS Principal coordinate analysis revealed that the gut microbiome of the preterm infants displayed an obvious time-dependent change pattern, which showed the strongest association with Age, followed by PMA, and a much weaker association with (GA). Random forest modeling further evidenced the time-dependent change pattern, with the Pearson's correlation coefficients between the actual values and the gut microbiome-predicted values being 0.68, 0.53, and 0.38 for postnatal, postmenstrual, and gestational age, respectively. The microbiome dynamism could be further divided into four Age stages, each with its own characteristic microbial taxa. The first 1-4 days (T1 stage) represented the meconium microbiome, with colonization of a high diversity of microbes before or during delivery. During 5-15 days (T2 stage), the gut microbiome of the preterm infants underwent a rapid turnover, in which microbial diversity declined, and stabilized afterward. Enterobacteriaceae, Enterococcaceae, Streptococcaceae, Staphylococcaceae, and Clostridiaceae were the major classes in the gut microbiome in the lateral stages of development (T3-T4 stage). CONCLUSIONS Postnatal age, rather than the gestational age, is significantly correlated with the gut microbiome of preterm infants, suggesting that clinical interventions contribute more to the early dynamics of gut microbiome in preterm infants than the natural development of the gut.
Collapse
Affiliation(s)
- Wei Shen
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wen Qiu
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuting Liu
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weihua Liao
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyi Ma
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yan He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhang Wang
- Institute of Ecological Sciences, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
350
|
Zhou Y, Qi H, Yin N. Adaptations and alterations of maternal microbiota: From physiology to pathology. MEDICINE IN MICROECOLOGY 2021. [DOI: 10.1016/j.medmic.2021.100045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|