151
|
Medeleanu MV, Qian YC, Moraes TJ, Subbarao P. Early-immune development in asthma: A review of the literature. Cell Immunol 2023; 393-394:104770. [PMID: 37837916 DOI: 10.1016/j.cellimm.2023.104770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 10/16/2023]
Abstract
This review presents a comprehensive examination of the various factors contributing to the immunopathogenesis of asthma from the prenatal to preschool period. We focus on the contributions of genetic and environmental components as well as the role of the nasal and gut microbiome on immune development. Predisposing genetic factors, including inherited genes associated with increased susceptibility to asthma, are discussed alongside environmental factors such as respiratory viruses and pollutant exposure, which can trigger or exacerbate asthma symptoms. Furthermore, the intricate interplay between the nasal and gut microbiome and the immune system is explored, emphasizing their influence on allergic immune development and response to environmental stimuli. This body of literature underscores the necessity of a comprehensive approach to comprehend and manage asthma, as it emphasizes the interactions of multiple factors in immune development and disease progression.
Collapse
Affiliation(s)
- Maria V Medeleanu
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Canada; Translational Medicine, SickKids Research Institute, Hospital for Sick Children, Canada
| | - Yu Chen Qian
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Canada; Translational Medicine, SickKids Research Institute, Hospital for Sick Children, Canada
| | - Theo J Moraes
- Translational Medicine, SickKids Research Institute, Hospital for Sick Children, Canada; Laboratory Medicine and Pathology, Temerty Faculty of Medicine, University of Toronto, Canada; Department of Paediatrics, Temerty Faculty of Medicine, University of Toronto, Canada; Division of Respiratory Medicine, Hospital for Sick Children, Canada
| | - Padmaja Subbarao
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Canada; Translational Medicine, SickKids Research Institute, Hospital for Sick Children, Canada; Department of Paediatrics, Temerty Faculty of Medicine, University of Toronto, Canada; Division of Respiratory Medicine, Hospital for Sick Children, Canada; Epidemiology Division, Dalla Lana School of Public Health, University of Toronto, Canada.
| |
Collapse
|
152
|
Zhao Q, Chen Y, Huang W, Zhou H, Zhang W. Drug-microbiota interactions: an emerging priority for precision medicine. Signal Transduct Target Ther 2023; 8:386. [PMID: 37806986 PMCID: PMC10560686 DOI: 10.1038/s41392-023-01619-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 07/20/2023] [Accepted: 08/24/2023] [Indexed: 10/10/2023] Open
Abstract
Individual variability in drug response (IVDR) can be a major cause of adverse drug reactions (ADRs) and prolonged therapy, resulting in a substantial health and economic burden. Despite extensive research in pharmacogenomics regarding the impact of individual genetic background on pharmacokinetics (PK) and pharmacodynamics (PD), genetic diversity explains only a limited proportion of IVDR. The role of gut microbiota, also known as the second genome, and its metabolites in modulating therapeutic outcomes in human diseases have been highlighted by recent studies. Consequently, the burgeoning field of pharmacomicrobiomics aims to explore the correlation between microbiota variation and IVDR or ADRs. This review presents an up-to-date overview of the intricate interactions between gut microbiota and classical therapeutic agents for human systemic diseases, including cancer, cardiovascular diseases (CVDs), endocrine diseases, and others. We summarise how microbiota, directly and indirectly, modify the absorption, distribution, metabolism, and excretion (ADME) of drugs. Conversely, drugs can also modulate the composition and function of gut microbiota, leading to changes in microbial metabolism and immune response. We also discuss the practical challenges, strategies, and opportunities in this field, emphasizing the critical need to develop an innovative approach to multi-omics, integrate various data types, including human and microbiota genomic data, as well as translate lab data into clinical practice. To sum up, pharmacomicrobiomics represents a promising avenue to address IVDR and improve patient outcomes, and further research in this field is imperative to unlock its full potential for precision medicine.
Collapse
Affiliation(s)
- Qing Zhao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, PR China
| | - Yao Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, PR China
| | - Weihua Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, PR China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, PR China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China.
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, PR China.
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, PR China.
- Central Laboratory of Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, Changsha, 410013, PR China.
| |
Collapse
|
153
|
Li Y, Wei B, Xue X, Li H, Li J. Microbiome changes in esophageal cancer: implications for pathogenesis and prognosis. Cancer Biol Med 2023; 21:j.issn.2095-3941.2023.0177. [PMID: 37817487 PMCID: PMC10884538 DOI: 10.20892/j.issn.2095-3941.2023.0177] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 09/06/2023] [Indexed: 10/12/2023] Open
Abstract
Esophageal cancer (EC) is an aggressive malignancy with a poor prognosis. Various factors, including dietary habits, and antacid and antibiotic use, have been shown to influence the esophageal microbiome. Conversely, enrichment and diversity of the esophageal microbiome can also impact its function. Recent studies have revealed prevalent changes in the esophageal microbiome among patients with EC, thus suggesting the potential contribution of the esophageal microbiome to EC development. Additionally, distinct microbiome compositions have been observed in patients with different responses to radiotherapy and chemotherapy, indicating the role of the esophageal microbiome in modulating treatment outcomes. In this review, we have examined previous studies on the esophageal microbiome in healthy individuals and patients with EC or other esophageal diseases, with a focus on identifying microbial communities associated with EC pathogenesis and prognosis. Understanding the role of the microbiome in EC may aid in early detection and optimized treatment strategies, ultimately leading to better outcomes for patients.
Collapse
Affiliation(s)
- Yi Li
- Department of Molecular Pathology, Clinical Pathology Center, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou 450003, China
| | - Bing Wei
- Department of Molecular Pathology, Clinical Pathology Center, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou 450003, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou 450003, China
| | - Xia Xue
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hongle Li
- Department of Molecular Pathology, Clinical Pathology Center, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou 450003, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou 450003, China
| | - Jun Li
- Department of Molecular Pathology, Clinical Pathology Center, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou 450003, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou 450003, China
| |
Collapse
|
154
|
Shao T, Hsu R, Hacein-Bey C, Zhang W, Gao L, Kurth MJ, Zhao H, Shuai Z, Leung PSC. The Evolving Landscape of Fecal Microbial Transplantation. Clin Rev Allergy Immunol 2023; 65:101-120. [PMID: 36757537 PMCID: PMC9909675 DOI: 10.1007/s12016-023-08958-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2023] [Indexed: 02/10/2023]
Abstract
The human gastrointestinal tract houses an enormous microbial ecosystem. Recent studies have shown that the gut microbiota plays significant physiological roles and maintains immune homeostasis in the human body. Dysbiosis, an imbalanced gut microbiome, can be associated with various disease states, as observed in infectious diseases, inflammatory diseases, autoimmune diseases, and cancer. Modulation of the gut microbiome has become a therapeutic target in treating these disorders. Fecal microbiota transplantation (FMT) from a healthy donor restores the normal gut microbiota homeostasis in the diseased host. Ample evidence has demonstrated the efficacy of FMT in recurrent Clostridioides difficile infection (rCDI). The application of FMT in other human diseases is gaining attention. This review aims to increase our understanding of the mechanisms of FMT and its efficacies in human diseases. We discuss the application, route of administration, limitations, safety, efficacies, and suggested mechanisms of FMT in rCDI, autoimmune diseases, and cancer. Finally, we address the future perspectives of FMT in human medicine.
Collapse
Affiliation(s)
- Tihong Shao
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis School of Medicine, Davis, CA, 95616, USA
| | - Ronald Hsu
- Division of Gastroenterology, University of California Davis School of Medicine, Davis, CA, 95616, USA
| | - Camelia Hacein-Bey
- Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis School of Medicine, Davis, CA, 95616, USA
| | - Weici Zhang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Lixia Gao
- Department of Rheumatology and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Mark J Kurth
- Department of Chemistry, University of California Davis, Davis, CA, 95616, USA
| | - Huanhuan Zhao
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Zongwen Shuai
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Patrick S C Leung
- Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis School of Medicine, Davis, CA, 95616, USA.
| |
Collapse
|
155
|
Theodosiou AA, Jones CE, Read RC, Bogaert D. Microbiotoxicity: antibiotic usage and its unintended harm to the microbiome. Curr Opin Infect Dis 2023; 36:371-378. [PMID: 37466039 PMCID: PMC10487351 DOI: 10.1097/qco.0000000000000945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
PURPOSE OF REVIEW Antibiotic use is associated with development of antimicrobial resistance and dysregulation of the microbiome (the overall host microbial community). These changes have in turn been associated with downstream adverse health outcomes. This review analyses recent important publications in a rapidly evolving field, contextualizing the available evidence to assist clinicians weighing the potential risks of antibiotics on a patient's microbiome. RECENT FINDING Although the majority of microbiome research is observational, we highlight recent interventional studies probing the associations between antibiotic use, microbiome disruption, and ill-health. These studies include germ-free mouse models, antibiotic challenge in healthy human volunteers, and a phase III study of the world's first approved microbiome-based medicine. SUMMARY The growing body of relevant clinical and experimental evidence for antibiotic-mediated microbiome perturbation is concerning, although further causal evidence is required. Within the limits of this evidence, we propose the novel term 'microbiotoxicity' to describe the unintended harms of antibiotics on a patient's microbiome. We suggest a framework for prescribers to weigh microbiotoxic effects against the intended benefits of antibiotic use.
Collapse
Affiliation(s)
- Anastasia A. Theodosiou
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton
| | - Christine E. Jones
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton
| | - Robert C. Read
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton
| | - Debby Bogaert
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
156
|
Wang J, Liu X, Sun R, Mao H, Liu M, Jin X. Akkermansia muciniphila participates in the host protection against helminth-induced cardiac fibrosis via TLR2. PLoS Pathog 2023; 19:e1011683. [PMID: 37788279 PMCID: PMC10547169 DOI: 10.1371/journal.ppat.1011683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 09/12/2023] [Indexed: 10/05/2023] Open
Abstract
Helminth Trichinella spiralis (Ts) is one of the major pathogens of human infective myocarditis that can lead to cardiac fibrosis (CF). The gut microbiota involved in this pathology are of interest. Here, we use mice infected with Ts as a model to examine the interactions between gut microbes and host protection to CF. Infected mice show enhanced CF severity. We find that antibiotics treatment to deplete the microbiota aggravates the disease phenotype. Attempts to restore microbiota using fecal microbiota transplantation ameliorates helminth-induced CF. 16S rRNA gene sequencing and metagenomics sequencing reveal a higher abundance of Akkermansia muciniphila in gut microbiomes of Ts-infected mice. Oral supplementation with alive or pasteurized A. muciniphila improves CF via TLR2. This work represents a substantial advance toward our understanding of causative rather than correlative relationships between the gut microbiota and CF.
Collapse
Affiliation(s)
- Jiaqi Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Animal Sciences, Jilin University, Changchun, China
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaolei Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ruohang Sun
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hanhai Mao
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Mingyuan Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Xuemin Jin
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
157
|
Erhardt R, Harnett JE, Steels E, Steadman KJ. Functional constipation and the effect of prebiotics on the gut microbiota: a review. Br J Nutr 2023; 130:1015-1023. [PMID: 36458339 PMCID: PMC10442792 DOI: 10.1017/s0007114522003853] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/23/2022] [Accepted: 11/29/2022] [Indexed: 12/04/2022]
Abstract
Functional constipation is a significant health issue impacting the lives of an estimated 14 % of the global population. Non-pharmaceutical treatment advice for cases with no underlying medical conditions focuses on exercise, hydration and an increase in dietary fibre intake. An alteration in the composition of the gut microbiota is thought to play a role in constipation. Prebiotics are non-digestible food ingredients that selectively stimulate the growth of a limited number of bacteria in the colon with a benefit for host health. Various types of dietary fibre, though not all, can act as a prebiotic. Short-chain fatty acids produced by these microbes play a critical role as signalling molecules in a range of metabolic and physiological processes including laxation, although details are unclear. Prebiotics have a history of safe use in the food industry spanning several decades and are increasingly used as supplements to alleviate constipation. Most scientific research on the effects of prebiotics and gut microbiota has focussed on inflammatory bowel disease rather than functional constipation. Very few clinical studies evaluated the efficacy of prebiotics in the management of constipation and their effect on the microbiota, with highly variable designs and conflicting results. Despite this, broad health claims are made by manufacturers of prebiotic supplements. This narrative review provides an overview of the literature on the interaction of prebiotics with the gut microbiota and their potential clinical role in the alleviation of functional constipation.
Collapse
Affiliation(s)
- Rene Erhardt
- School of Pharmacy, The University of Queensland, Brisbane, QLD4102, Australia
| | - Joanna E Harnett
- School of Pharmacy, The University of Sydney, Camperdown, NSW2006, Australia
| | - Elizabeth Steels
- School of Pharmacy, The University of Queensland, Brisbane, QLD4102, Australia
- Evidence Sciences, 3/884 Brunswick St, New Farm, QLD4005, Australia
| | - Kathryn J Steadman
- School of Pharmacy, The University of Queensland, Brisbane, QLD4102, Australia
| |
Collapse
|
158
|
Nagakubo D, Kaibori Y. Oral Microbiota: The Influences and Interactions of Saliva, IgA, and Dietary Factors in Health and Disease. Microorganisms 2023; 11:2307. [PMID: 37764151 PMCID: PMC10535076 DOI: 10.3390/microorganisms11092307] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/31/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Recent advances in metagenomic analyses have made it easier to analyze microbiota. The microbiota, a symbiotic community of microorganisms including bacteria, archaea, fungi, and viruses within a specific environment in tissues such as the digestive tract and skin, has a complex relationship with the host. Recent studies have revealed that microbiota composition and balance particularly affect the health of the host and the onset of disease. Influences such as diet, food preferences, and sanitation play crucial roles in microbiota composition. The oral cavity is where the digestive tract directly communicates with the outside. Stable temperature and humidity provide optimal growth environments for many bacteria. However, the oral cavity is a unique environment that is susceptible to pH changes, salinity, food nutrients, and external pathogens. Recent studies have emphasized the importance of the oral microbiota, as changes in bacterial composition and balance could contribute to the development of systemic diseases. This review focuses on saliva, IgA, and fermented foods because they play critical roles in maintaining the oral bacterial environment by regulating its composition and balance. More attention should be paid to the oral microbiota and its regulatory factors in oral and systemic health.
Collapse
Affiliation(s)
- Daisuke Nagakubo
- Division of Health and Hygienic Sciences, Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 7-2-1 Kamiohno, Himeji 670-8524, Hyogo, Japan
| | - Yuichiro Kaibori
- Division of Health and Hygienic Sciences, Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 7-2-1 Kamiohno, Himeji 670-8524, Hyogo, Japan
- Laboratory of Analytics for Biomolecules, Faculty of Pharmaceutical Science, Setsunan University, 45-1 Nagaotoge-cho, Hirakata-shi 573-0101, Osaka, Japan;
| |
Collapse
|
159
|
Strout N, Pasic L, Hicks C, Chua XY, Tashvighi N, Butler P, Liu Z, El-Assaad F, Holmes E, Susic D, Samaras K, Craig ME, Davis GK, Henry A, Ledger WL, El-Omar EM. The MothersBabies Study, an Australian Prospective Cohort Study Analyzing the Microbiome in the Preconception and Perinatal Period to Determine Risk of Adverse Pregnancy, Postpartum, and Child-Related Health Outcomes: Study Protocol. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:6736. [PMID: 37754596 PMCID: PMC10531411 DOI: 10.3390/ijerph20186736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/03/2023] [Accepted: 09/06/2023] [Indexed: 09/28/2023]
Abstract
The microbiome has emerged as a key determinant of human health and reproduction, with recent evidence suggesting a dysbiotic microbiome is implicated in adverse perinatal health outcomes. The existing research has been limited by the sample collection and timing, cohort design, sample design, and lack of data on the preconception microbiome. This prospective, longitudinal cohort study will recruit 2000 Australian women, in order to fully explore the role of the microbiome in the development of adverse perinatal outcomes. Participants are enrolled for a maximum of 7 years, from 1 year preconception, through to 5 years postpartum. Assessment occurs every three months until pregnancy occurs, then during Trimester 1 (5 + 0-12 + 6 weeks gestation), Trimester 2 (20 + 0-24 + 6 weeks gestation), Trimester 3 (32 + 0-36 + 6 weeks gestation), and postpartum at 1 week, 2 months, 6 months, and then annually from 1 to 5 years. At each assessment, maternal participants self-collect oral, skin, vaginal, urine, and stool samples. Oral, skin, urine, and stool samples will be collected from children. Blood samples will be obtained from maternal participants who can access a study collection center. The measurements taken will include anthropometric, blood pressure, heart rate, and serum hormonal and metabolic parameters. Validated self-report questionnaires will be administered to assess diet, physical activity, mental health, and child developmental milestones. Medications, medical, surgical, obstetric history, the impact of COVID-19, living environments, and pregnancy and child health outcomes will be recorded. Multiomic bioinformatic and statistical analyses will assess the association between participants who developed high-risk and low-risk pregnancies, adverse postnatal conditions, and/or childhood disease, and their microbiome for the different sample types.
Collapse
Affiliation(s)
- Naomi Strout
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
| | - Lana Pasic
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
| | - Chloe Hicks
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
| | - Xin-Yi Chua
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
| | - Niki Tashvighi
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
| | - Phoebe Butler
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
| | - Zhixin Liu
- UNSW Stats Central, Biological Sciences South Building (E26), Level 2 Kensington, UNSW Sydney, Sydney, NSW 2052, Australia
- Healthdirect Australia, Level 4, 477 Pitt Street, Sydney, NSW 2000, Australia
| | - Fatima El-Assaad
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
| | - Elaine Holmes
- The Australian National Phenome Centre, Harry Perkins Institute, Murdoch University, Perth, WA 6150, Australia;
| | - Daniella Susic
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
- Department of Women’s and Children’s Health, St George Hospital, Kogarah, NSW 2217, Australia; (G.K.D.); (A.H.)
- Discipline of Women’s Health, School of Clinical Medicine, UNSW Sydney, Sydney, NSW 2052, Australia; (M.E.C.); (W.L.L.)
| | - Katherine Samaras
- Complex Diseases Program, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia;
- Department of Endocrinology, St Vincent’s Hospital, Darlinghurst, NSW 2010, Australia
- St Vincent’s Clinical Campus, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Maria E. Craig
- Discipline of Women’s Health, School of Clinical Medicine, UNSW Sydney, Sydney, NSW 2052, Australia; (M.E.C.); (W.L.L.)
| | - Gregory K. Davis
- Department of Women’s and Children’s Health, St George Hospital, Kogarah, NSW 2217, Australia; (G.K.D.); (A.H.)
- Discipline of Women’s Health, School of Clinical Medicine, UNSW Sydney, Sydney, NSW 2052, Australia; (M.E.C.); (W.L.L.)
| | - Amanda Henry
- Department of Women’s and Children’s Health, St George Hospital, Kogarah, NSW 2217, Australia; (G.K.D.); (A.H.)
- Discipline of Women’s Health, School of Clinical Medicine, UNSW Sydney, Sydney, NSW 2052, Australia; (M.E.C.); (W.L.L.)
| | - William L. Ledger
- Discipline of Women’s Health, School of Clinical Medicine, UNSW Sydney, Sydney, NSW 2052, Australia; (M.E.C.); (W.L.L.)
| | - Emad M. El-Omar
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
| |
Collapse
|
160
|
Douglas P. Does the Academy of Breastfeeding Medicine's Clinical Protocol #36 'The Mastitis Spectrum' promote overtreatment and risk worsened outcomes for breastfeeding families? Commentary. Int Breastfeed J 2023; 18:51. [PMID: 37670315 PMCID: PMC10481477 DOI: 10.1186/s13006-023-00588-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 08/25/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND In 2022 the Academy of Breastfeeding Medicine (ABM) published Clinical Protocol #36: The Mastitis Spectrum, which aims to update clinical approaches to management of benign lactation-related breast inflammation. The protocol has been timely because of the exponential increase in knowledge about the human milk microbiome over the past decade. This Commentary aims to continue respectful debate amongst clinicians and researchers within the Academy of Breastfeeding Medicine and more broadly, confident that we share a fundamental commitment to promote breastfeeding and support the well-being of lactating women, their infants and their families. ANALYSIS Although Clinical Protocol #36 offers advances, it does not fulfil the principles of best practice implementation science for translation of evidence into clinical guidelines. Clinical Protocol #36 inaccurately represents studies; misrepresents theoretical models as proven aetiologies; does not consistently attribute sources; does not reliably apply the SORT taxonomy; and relies upon single case reports. As a result, various recommendations in Clinical Protocol #36 lack an evidence-base or credible underlying theoretical model. This includes recommendations to use 'lymphatic drainage' massage, therapeutic ultrasound, and oral lecithin. Similarly, based on a contestable theoretical model which is presented as fact, Clinical Protocol #36 makes the recommendation to either reduce frequency of milk removal or to maintain current frequency of milk removal during an episode of breast inflammation. Although Clinical Protocol #36 limits this advice to cases of 'hyperlactation', the diagnosis 'hyperlactation' itself is undefinable. As a result, this recommendation may put breastfeeding women who present with breast inflammation at risk of worsened inflammation and decreased breast milk production. CONCLUSION Clinical Protocol #36 offers some advances in the management of breast inflammation. However, Clinical Protocol #36 also exposes clinicians to two international trends in healthcare which undermine health system sustainability: overdiagnosis, including by over-definition, which increases risk of overtreatment; and antibiotic over-use, which worsens the crisis of global antimicrobial resistance. Clinical Protocol #36 also recommends unnecessary or ineffective interventions which may be accessed by affluent patients within advanced economies but are difficult to access for the global majority. The Academy of Breastfeeding Medicine may benefit from a review of processes for development of Clinical Protocols.
Collapse
Affiliation(s)
- Pamela Douglas
- The School of Nursing and Midwifery, Griffith University, Brisbane, Australia.
- General Practice Clinical Unit, The University of Queensland, Brisbane, Australia.
- Medical Director, The NDC Institute, ndcinstitute.com.au, Brisbane, Australia.
| |
Collapse
|
161
|
Pérez-Cobas AE, Rodríguez-Beltrán J, Baquero F, Coque TM. Ecology of the respiratory tract microbiome. Trends Microbiol 2023; 31:972-984. [PMID: 37173205 DOI: 10.1016/j.tim.2023.04.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 05/15/2023]
Abstract
A thriving multi-kingdom microbial ecosystem inhabits the respiratory tract: the respiratory tract microbiome (RTM). In recent years, the contribution of the RTM to human health has become a crucial research aspect. However, research into the key ecological processes, such as robustness, resilience, and microbial interaction networks, has only recently started. This review leans on an ecological framework to interpret the human RTM and determine how the ecosystem functions and assembles. Specifically, the review illustrates the ecological RTM models and discusses microbiome establishment, community structure, diversity stability, and critical microbial interactions. Lastly, the review outlines the RTM responses to ecological disturbances, as well as the promising approaches for restoring ecological balance.
Collapse
Affiliation(s)
- Ana Elena Pérez-Cobas
- Department of Microbiology, Ramón y Cajal Institute for Health Research (IRYCIS), Ramón y Cajal University Hospital, Madrid, Spain; CIBER in Infectious Diseases (CIBERINFEC), Madrid, Spain.
| | - Jerónimo Rodríguez-Beltrán
- Department of Microbiology, Ramón y Cajal Institute for Health Research (IRYCIS), Ramón y Cajal University Hospital, Madrid, Spain; CIBER in Infectious Diseases (CIBERINFEC), Madrid, Spain
| | - Fernando Baquero
- Department of Microbiology, Ramón y Cajal Institute for Health Research (IRYCIS), Ramón y Cajal University Hospital, Madrid, Spain; CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Teresa M Coque
- Department of Microbiology, Ramón y Cajal Institute for Health Research (IRYCIS), Ramón y Cajal University Hospital, Madrid, Spain; CIBER in Infectious Diseases (CIBERINFEC), Madrid, Spain
| |
Collapse
|
162
|
Lüll K, Org E. Uterine Microbiome: Does the Sampling Technique Matter? Semin Reprod Med 2023; 41:144-150. [PMID: 38065552 DOI: 10.1055/s-0043-1777361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Studies have proven the significance of microbial communities in various parts of the human body for health. In recent years it has been discovered that the uterine cavity is not sterile, and endometrium has its own microbiome which appears to have an impact on female fertility and gynecological pathologies. Lactobacillus has shown to dominate the microbial profile in the uterus and is considered an indicator of a healthy uterine environment. Yet, many argue that the Lactobacillus dominance is due to vaginal contamination during the sampling process. To date there is no clearly defined healthy endometrial microbial profile, which is largely due to the fact that determining the microbial community from the endometrium is complicated, and there is currently no consensus on sampling methods for the endometrial microbiome. As a result, this restricts ability to replicate discoveries made in other cohorts. Here we aim to give an overview of the sampling methods used and discuss what impedes the endometrial microbiome studies as well as how to reach a consensus on the study design. This knowledge could be incorporated into the future research and the knowledge on endometrial microbiome could be included into the diagnostics and treatment of female reproductive health.
Collapse
Affiliation(s)
- Kreete Lüll
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia
| | - Elin Org
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia
| |
Collapse
|
163
|
Ma LC, Zhao HQ, Wu LB, Cheng ZL, Liu C. Impact of the microbiome on human, animal, and environmental health from a One Health perspective. SCIENCE IN ONE HEALTH 2023; 2:100037. [PMID: 39077043 PMCID: PMC11262275 DOI: 10.1016/j.soh.2023.100037] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 08/21/2023] [Indexed: 07/31/2024]
Abstract
The microbiome encompasses the genomes of the microorganisms that inhabit specific environments. One Health is an emerging concept, recognised as a cohesive, harmonising approach aimed at sustainably improving the well-being of humans, animals, and the environment. The microbiome plays a crucial role in the One Health domain, facilitating interactions among humans, animals, and the environment, along with co-evolution, co-development, co-metabolism, and co-regulation with their associated humans and animals. In addition, the microbiome regulates environmental health through interactions with plant microbiota, which actively participate in substance cycling (particularly the carbon and nitrogen cycles) and influence the overall energy flow in the biosphere. Moreover, antibiotic resistance genes present in microbiota can lead to widespread drug resistance in both humans and animals. This review explores the impact of the microbiome on humans, animals, and the environment, highlighting the significance of focusing on this field in One Health research.
Collapse
Affiliation(s)
- Ling-chao Ma
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- One Health Centre, Shanghai Jiao Tong University and the University of Edinburgh, Shanghai, China
| | - Han-qing Zhao
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- One Health Centre, Shanghai Jiao Tong University and the University of Edinburgh, Shanghai, China
| | - Logan Blair Wu
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- One Health Centre, Shanghai Jiao Tong University and the University of Edinburgh, Shanghai, China
- Population Health & Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Zi-le Cheng
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- One Health Centre, Shanghai Jiao Tong University and the University of Edinburgh, Shanghai, China
| | - Chang Liu
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
164
|
Kaluanga Bwanga P, Tremblay-Lemoine PL, Timmermans M, Ravet S, Munaut C, Nisolle M, Henry L. The Endometrial Microbiota: Challenges and Prospects. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1540. [PMID: 37763663 PMCID: PMC10534531 DOI: 10.3390/medicina59091540] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/14/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023]
Abstract
Contrary to popular belief, we have known for many years that the endometrium is not a sterile environment and is considered to be a low-biomass milieu compared to the vagina. Numerous trials and studies have attempted to establish a valid sampling method and assess its physiological composition, but no consensus has been reached. Many factors, such as ethnicity, age and inflammation, can influence the microbiome. Moreover, it possesses a higher alpha-diversity and, therefore, contains more diverse bacteria than the vagina. For instance, Lactobacillus has been shown to be a predominant genus in the vaginal microbiome of healthy women. Consequently, even if a majority of scientists postulate that a predominance of Lactobacillus inside the uterus improves reproductive outcomes, vaginal contamination by these bacteria during sampling cannot be ruled out. Certain pathologies, such as chronic endometritis, have been identified as inflammation perpetrators that hinder the embryo implantation process. This pro-inflammatory climate created by dysbiosis of the endometrial microbiota could induce secondary inflammatory mediators via Toll-like receptors, creating an environment conducive to the development of endometriosis and even promoting carcinogenesis. However, studies to this day have focused on small populations. In addition, there is no clearly defined healthy uterine composition yet. At most, only a few taxa have been identified as pathogenic. As sampling and analysis methods become increasingly precise, we can expect the endometrial microbiota to be incorporated into future diagnostic tools and treatments for women's health.
Collapse
Affiliation(s)
| | - Pierre-Luc Tremblay-Lemoine
- Department of Obstetrics and Gynecology, CHU of Liege-Citadelle Site, University of Liège, 4000 Liège, Belgium
| | - Marie Timmermans
- Department of Obstetrics and Gynecology, CHU of Liege-Citadelle Site, University of Liège, 4000 Liège, Belgium
| | - Stéphanie Ravet
- Center for Reproductive Medicine, University of Liège-Citadelle Site, 4000 Liege, Belgium
| | - Carine Munaut
- Laboratory of Tumor and Development Biology, Giga-Cancer, University of Liège, 4000 Liège, Belgium
| | - Michelle Nisolle
- Department of Obstetrics and Gynecology, CHU of Liege-Citadelle Site, University of Liège, 4000 Liège, Belgium
| | - Laurie Henry
- Department of Obstetrics and Gynecology, CHU of Liege-Citadelle Site, University of Liège, 4000 Liège, Belgium
- Center for Reproductive Medicine, University of Liège-Citadelle Site, 4000 Liege, Belgium
| |
Collapse
|
165
|
Deng X, Chen X, Luo Y, Que J, Chen L. Intratumor microbiome derived glycolysis-lactate signatures depicts immune heterogeneity in lung adenocarcinoma by integration of microbiomic, transcriptomic, proteomic and single-cell data. Front Microbiol 2023; 14:1202454. [PMID: 37664112 PMCID: PMC10469687 DOI: 10.3389/fmicb.2023.1202454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
Introduction Microbiome plays roles in lung adenocarcinoma (LUAD) development and anti-tumor treatment efficacy. Aberrant glycolysis in tumor might promote lactate production that alter tumor microenvironment, affecting microbiome, cancer cells and immune cells. We aimed to construct intratumor microbiome score to predict prognosis of LUAD patients and thoroughly investigate glycolysis and lactate signature's association with LUAD immune cell infiltration. Methods The Cancer Genome Atlas-LUAD (TCGA-LUAD) microbiome data was downloaded from cBioPortal and analyzed to examine its association with overall survival to create a prognostic scoring model. Gene Set Enrichment Analysis (GSEA) was used to find each group's major mechanisms involved. Our study then investigated the glycolysis and lactate pattern in LUAD patients based on 19 genes, which were correlated with the tumor microenvironment (TME) phenotypes and immunotherapy outcomes. We developed a glycolysis-lactate risk score and signature to accurately predict TME phenotypes, prognosis, and response to immunotherapy. Results Using the univariate Cox regression analysis, the abundance of 38 genera were identified with prognostic values and a lung-resident microbial score (LMS) was then developed from the TCGA-LUAD-microbiome dataset. Glycolysis hallmark pathway was significantly enriched in high-LMS group and three distinct glycolysis-lactate patterns were generated. Patients in Cluster1 exhibited unfavorable outcomes and might be insensitive to immunotherapy. Glycolysis-lactate score was constructed for predicting prognosis with high accuracy and validated in external cohorts. Gene signature was developed and this signature was elevated in epithelial cells especially in tumor mass on single-cell level. Finally, we found that the glycolysis-lactate signature levels were consistent with the malignancy of histological subtypes. Discussion Our study demonstrated that an 18-microbe prognostic score and a 19-gene glycolysis-lactate signature for predicting prognosis of LUAD patients. Our LMS, glycolysis-lactate score and glycolysis-lactate signature have potential roles in precision therapy of LUAD patients.
Collapse
Affiliation(s)
| | | | | | - Jun Que
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liang Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
166
|
Salmeri N, Sinagra E, Dolci C, Buzzaccarini G, Sozzi G, Sutera M, Candiani M, Ungaro F, Massimino L, Danese S, Mandarino FV. Microbiota in Irritable Bowel Syndrome and Endometriosis: Birds of a Feather Flock Together-A Review. Microorganisms 2023; 11:2089. [PMID: 37630649 PMCID: PMC10458414 DOI: 10.3390/microorganisms11082089] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/09/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Endometriosis and irritable bowel syndrome (IBS) are chronic conditions affecting up to 10% of the global population, imposing significant burdens on healthcare systems and patient quality of life. Interestingly, around 20% of endometriosis patients also present with symptoms indicative of IBS. The pathogenesis of both these multifactorial conditions remains to be fully elucidated, but connections to gut microbiota are becoming more apparent. Emerging research underscores significant differences in the gut microbiota composition between healthy individuals and those suffering from either endometriosis or IBS. Intestinal dysbiosis appears pivotal in both conditions, exerting an influence via similar mechanisms. It impacts intestinal permeability, triggers inflammatory reactions, and initiates immune responses. Furthermore, it is entwined in a bidirectional relationship with the brain, as part of the gut-brain axis, whereby dysbiosis influences and is influenced by mental health and pain perception. Recent years have witnessed the development of microbiota-focused therapies, such as low FODMAP diets, prebiotics, probiotics, antibiotics, and fecal microbiota transplantation, designed to tackle dysbiosis and relieve symptoms. While promising, these treatments present inconsistent data, highlighting the need for further research. This review explores the evidence of gut dysbiosis in IBS and endometriosis, underscoring the similar role of microbiota in both conditions. A deeper understanding of this common mechanism may enable enhanced diagnostics and therapeutic advancements.
Collapse
Affiliation(s)
- Noemi Salmeri
- Gynecology/Obstetrics Unit, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (C.D.); (G.B.); (M.C.)
| | - Emanuele Sinagra
- Gastroenterology & Endoscopy Unit, Fondazione Istituto G. Giglio, Contrada Pietra Pollastra Pisciotto, 90015 Cefalù, Italy;
| | - Carolina Dolci
- Gynecology/Obstetrics Unit, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (C.D.); (G.B.); (M.C.)
| | - Giovanni Buzzaccarini
- Gynecology/Obstetrics Unit, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (C.D.); (G.B.); (M.C.)
| | - Giulio Sozzi
- Gynecology/Obstetrics Unit, Fondazione Istituto G. Giglio, Contrada Pietra Pollastra Pisciotto, 90015 Cefalù, Italy; (G.S.); (M.S.)
| | - Miriam Sutera
- Gynecology/Obstetrics Unit, Fondazione Istituto G. Giglio, Contrada Pietra Pollastra Pisciotto, 90015 Cefalù, Italy; (G.S.); (M.S.)
| | - Massimo Candiani
- Gynecology/Obstetrics Unit, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (C.D.); (G.B.); (M.C.)
| | - Federica Ungaro
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (F.U.); (L.M.); (S.D.); (F.V.M.)
| | - Luca Massimino
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (F.U.); (L.M.); (S.D.); (F.V.M.)
| | - Silvio Danese
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (F.U.); (L.M.); (S.D.); (F.V.M.)
| | - Francesco Vito Mandarino
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (F.U.); (L.M.); (S.D.); (F.V.M.)
| |
Collapse
|
167
|
Ma H, Zhao W, Song T, Baijiu Z, Zhang Z. Comparative Analysis of the Pre-Parturition and Post-Parturition Genital Tract Microbiota in Plateau Bangor Sewa Sheep. Vet Sci 2023; 10:523. [PMID: 37624310 PMCID: PMC10459245 DOI: 10.3390/vetsci10080523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/09/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023] Open
Abstract
(1) Background: Bangor Sewa sheep are an economically significant livestock species on the plateau. The roles of microbiota in reproduction are complex and critical for animal health. But little is known currently about the microbiome of plateau Bangor Sewa sheep. The purpose of this study was to discover the changes in the genital tract microbiota of pre- and post-partum Bangor Sewa sheep. (2) Methods: Samples from the birth canal were obtained for 16S rRNA sequencing, three days before and after delivery, respectively. (3) Results: The results showed that there was a noticeable difference in three phyla and 74 genera between the pre- and post-parturition groups in the microbiota of Bangor Sewa sheep. The changes included a decrease in the abundance of genera related to health (unclassified_Cellulomonadaceae, Cellulomonas, Fibrobacti, Flavobacterium, Eubacterium_ventriosum_group, Acetitomaculum, Aeromicrobium, Dietzia, Romboutsia, Ruminococcus, etc.) and an increased abundance of negatively related genera (Nocardioides, unclassified_Clostridia, Sphingobacteriaceae, unclassified_Ruminococcaceae, Prevotellaceae_UCG_004, Micromonospora, Streptococcus, Facklamia, Bosea, etc.) spp. (4) Conclusions: Microbes can serve as indicators of the physical state of Bangor Sewa sheep. These findings laid the foundation for deciphering the effects of microbial changes during birth on the reproductive health of plateau Bangor Sewa sheep.
Collapse
Affiliation(s)
- Hongcai Ma
- Institute of Animal Husbandry and Veterinary Medicine, Tibet Autonomous Region Academy of Agriculture and Animal Science, Lhasa 850009, China; (H.M.); (T.S.)
| | - Wangsheng Zhao
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China;
| | - Tianzeng Song
- Institute of Animal Husbandry and Veterinary Medicine, Tibet Autonomous Region Academy of Agriculture and Animal Science, Lhasa 850009, China; (H.M.); (T.S.)
| | - Zhaxi Baijiu
- Cultural Service Center of Maqian Township, Nagqu 852599, China;
| | - Zhenzhen Zhang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China;
| |
Collapse
|
168
|
Zhang X, Wang M, Liu Z, Wang Y, Chen L, Guo J, Zhang W, Zhang Y, Yu C, Bie T, Yu Y, Guan B. Transnasal-brain delivery of nanomedicines for neurodegenerative diseases. FRONTIERS IN DRUG DELIVERY 2023; 3. [DOI: 10.3389/fddev.2023.1247162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2024]
Abstract
Neurodegenerative diseases (NDs) have become a serious global health problem as the population ages. Traditionally, treatment strategies for NDs have included oral and intravenous administration; however, the blood–brain barrier (BBB) can prevent drugs from reaching the brain, rendering the treatment incomplete and the effect unsatisfactory. Additionally, the prolonged or excessive use of drugs that can cross the BBB can damage liver and kidney function. Recent studies have shown that nose-to-brain drug delivery can noninvasively bypass the BBB, allowing drugs to enter the brain through the olfactory or trigeminal nerve pathways; additionally, nanoparticle carriers can enhance drug delivery. This review introduces drug carrier nanoparticles for nose-to-brain delivery systems, compares the advantages and disadvantages of different nanoparticles, and discusses the factors influencing nose-to-brain nanomedicine delivery and enhancement strategies. We also summarize nose-to-brain delivery and nanomedicines for treating NDs, the current challenges of this approach, and the future promise of nanomedicine-based ND treatment.
Collapse
|
169
|
Seyedsayamdost MR, Clardy J. Discovering functional small molecules in the gut microbiome. Curr Opin Chem Biol 2023; 75:102309. [PMID: 37163788 PMCID: PMC10524162 DOI: 10.1016/j.cbpa.2023.102309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/25/2023] [Indexed: 05/12/2023]
Abstract
The human microbiome has emerged as a source of bacterially produced, functional small molecules that help regulate health and disease, and their discovery and annotation has become a popular research topic. Identifying these molecules provides an essential step in unraveling the molecular mechanisms underlying biological outcomes. The relevance of specific bacterial members of the microbiome has been demonstrated in a variety of correlative studies, and there are many possible paths from these correlations to the responsible metabolites. Herein, we summarize two studies that have recently identified gut microbiome metabolites that modulate immune responses or promote physical activity. Aside from the deep insights gained, these studies provide blueprints for successfully uncovering the molecules and mechanisms that control important physiological pathways.
Collapse
Affiliation(s)
| | - Jon Clardy
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Blavatnik Institute, Boston, MA 02115, USA.
| |
Collapse
|
170
|
Kilgore A, Belkind-Gerson J. The Bidirectional Brain-Gut-Microbiome Axis in Pediatrics: What We Know and What Lies Ahead. J Pediatr Gastroenterol Nutr 2023; 77:147-149. [PMID: 37115954 DOI: 10.1097/mpg.0000000000003817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Affiliation(s)
- Alexandra Kilgore
- From Digestive Health Institute, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO
| | | |
Collapse
|
171
|
Lupu VV, Butnariu LI, Fotea S, Morariu ID, Badescu MC, Starcea IM, Salaru DL, Popp A, Dragan F, Lupu A, Mocanu A, Chisnoiu T, Pantazi AC, Jechel E. The Disease with a Thousand Faces and the Human Microbiome-A Physiopathogenic Intercorrelation in Pediatric Practice. Nutrients 2023; 15:3359. [PMID: 37571295 PMCID: PMC10420997 DOI: 10.3390/nu15153359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/18/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Numerous interrelationships are known in the literature that have the final effect of unmasking or influencing various pathologies. Among these, the present article aims to discuss the connection between systemic lupus erythematosus (SLE) and the human microbiome. The main purpose of this work is to popularize information about the impact of dysbiosis on the pathogenesis and evolutionary course of pediatric patients with SLE. Added to this is the interest in knowledge and awareness of adjunctive therapeutic means that has the ultimate goal of increasing the quality of life. The means by which this can be achieved can be briefly divided into prophylactic or curative, depending on the phase of the condition in which the patient is. We thus reiterate the importance of the clinician acquiring an overview of SLE and the human microbiome, doubled by in-depth knowledge of the physio-pathogenic interactions between the two (in part achieved through the much-studied gut-target organ axes-brain, heart, lung, skin), with the target objective being that of obtaining individualized, multimodal and efficient management for each individual patient.
Collapse
Affiliation(s)
- Vasile Valeriu Lupu
- Pediatrics Department, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | | | - Silvia Fotea
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania
| | - Ionela Daniela Morariu
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Minerva Codruta Badescu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Iuliana Magdalena Starcea
- Pediatrics Department, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Delia Lidia Salaru
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Alina Popp
- Pediatrics Department, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Felicia Dragan
- Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Ancuta Lupu
- Pediatrics Department, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Adriana Mocanu
- Pediatrics Department, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Tatiana Chisnoiu
- Pediatrics Department, Faculty of Medicine, Ovidius University, 900470 Constanta, Romania
| | | | - Elena Jechel
- Pediatrics Department, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
172
|
da Silva Soares NF, Quagliariello A, Yigitturk S, Martino ME. Gut microbes predominantly act as living beneficial partners rather than raw nutrients. Sci Rep 2023; 13:11981. [PMID: 37488173 PMCID: PMC10366161 DOI: 10.1038/s41598-023-38669-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 07/12/2023] [Indexed: 07/26/2023] Open
Abstract
Animals and their gut microbes mutually benefit their health. Nutrition plays a central role in this, directly influencing both host and microbial fitness and the nature of their interactions. This makes nutritional symbioses a complex and dynamic tri-system of diet-microbiota-host. Despite recent discoveries on this field, full control over the interplay among these partners is challenging and hinders the resolution of fundamental questions, such as how to parse the gut microbes' effect as raw nutrition or as symbiotic partners? To tackle this, we made use of the well-characterized Drosophila melanogaster/Lactiplantibacillus plantarum experimental model of nutritional symbiosis to generate a quantitative framework of gut microbes' effect on the host. By coupling experimental assays and Random Forest analysis, we show that the beneficial effect of L. plantarum strains primarily results from the active relationship as symbionts rather than raw nutrients, regardless of the bacterial strain. Metabolomic analysis of both active and inactive bacterial cells further demonstrated the crucial role of the production of beneficial bacterial metabolites, such as N-acetylated-amino-acids, as result of active bacterial growth and function. Altogether, our results provide a ranking and quantification of the main bacterial features contributing to sustain animal growth. We demonstrate that bacterial activity is the predominant and necessary variable involved in bacteria-mediated benefit, followed by strain-specific properties and the nutritional potential of the bacterial cells. This contributes to elucidate the role of beneficial bacteria and probiotics, creating a broad quantitative framework for host-gut microbiome that can be expanded to other model systems.
Collapse
Affiliation(s)
| | - Andrea Quagliariello
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
| | - Seren Yigitturk
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
- Food Quality and Design Group, Wageningen University and Research, Wageningen, The Netherlands
| | - Maria Elena Martino
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy.
| |
Collapse
|
173
|
Vonaesch P, Billy V, Mann AE, Morien E, Habib A, Collard JM, Dédé M, Kapel N, Sansonetti PJ, Parfrey LW. The eukaryome of African children is influenced by geographic location, gut biogeography, and nutritional status. MICROLIFE 2023; 4:uqad033. [PMID: 37680753 PMCID: PMC10481997 DOI: 10.1093/femsml/uqad033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/17/2023] [Indexed: 09/09/2023]
Abstract
Eukaryotes have historically been studied as parasites, but recent evidence suggests they may be indicators of a healthy gut ecosystem. Here, we describe the eukaryome along the gastrointestinal tract of children aged 2-5 years and test for associations with clinical factors such as anaemia, intestinal inflammation, chronic undernutrition, and age. Children were enrolled from December 2016 to May 2018 in Bangui, Central African Republic and Antananarivo, Madagascar. We analyzed a total of 1104 samples representing 212 gastric, 187 duodenal, and 705 fecal samples using a metabarcoding approach targeting the full ITS2 region for fungi, and the V4 hypervariable region of the 18S rRNA gene for the overall eukaryome. Roughly, half of all fecal samples showed microeukaryotic reads. We find high intersubject variability, only a handful of taxa that are likely residents of the gastrointestinal tract, and frequent co-occurrence of eukaryotes within an individual. We also find that the eukaryome differs between the stomach, duodenum, and feces and is strongly influenced by country of origin. Our data show trends towards higher levels of Fusarium equiseti, a mycotoxin producing fungus, and lower levels of the protist Blastocystis in stunted children compared to nonstunted controls. Overall, the eukaryome is poorly correlated with clinical variables. Our study is of one of the largest cohorts analyzing the human intestinal eukaryome to date and the first to compare the eukaryome across different compartments of the gastrointestinal tract. Our results highlight the importance of studying populations across the world to uncover common features of the eukaryome in health.
Collapse
Affiliation(s)
- Pascale Vonaesch
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Vincent Billy
- Departments of Botany and Zoology, and Biodiversity Research Centre, University of British Columbia, 3200-6270 University Boulevard, V6T1Z4 Vancouver, Canada
| | - Allison E Mann
- Departments of Botany and Zoology, and Biodiversity Research Centre, University of British Columbia, 3200-6270 University Boulevard, V6T1Z4 Vancouver, Canada
| | - Evan Morien
- Departments of Botany and Zoology, and Biodiversity Research Centre, University of British Columbia, 3200-6270 University Boulevard, V6T1Z4 Vancouver, Canada
| | - Azimdine Habib
- Unité de Bactériologie Expérimentale, Institut Pasteur de Madagascar, BP1274 Ambatofotsikely Avaradoha 101 Antananarivo, Madagascar
| | - Jean-Marc Collard
- Unité de Bactériologie Expérimentale, Institut Pasteur de Madagascar, BP1274 Ambatofotsikely Avaradoha 101 Antananarivo, Madagascar
| | - Michel Dédé
- Laboratoire d’Analyse médicale, Institut Pasteur de Bangui, Avenue De Independence Bangui, 923 Central African Republic
| | - Nathalie Kapel
- Laboratoire de Coprologie Fonctionnelle, Assistance Publique- Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, 47-83 Bd de l’Hôpital, 75013 Paris, France
| | - Philippe J Sansonetti
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Laura Wegener Parfrey
- Departments of Botany and Zoology, and Biodiversity Research Centre, University of British Columbia, 3200-6270 University Boulevard, V6T1Z4 Vancouver, Canada
| |
Collapse
|
174
|
Chen X, Shi Y. Determinants of microbial colonization in the premature gut. Mol Med 2023; 29:90. [PMID: 37407941 DOI: 10.1186/s10020-023-00689-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 06/20/2023] [Indexed: 07/07/2023] Open
Abstract
Abnormal microbial colonization in the gut at an early stage of life affects growth, development, and health, resulting in short- and long-term adverse effects. Microbial colonization patterns of preterm infants differ from those of full-term infants in that preterm babies and their mothers have more complicated prenatal and postnatal medical conditions. Maternal complications, antibiotic exposure, delivery mode, feeding type, and the use of probiotics may significantly shape the gut microbiota of preterm infants at an early stage of life; however, these influences subside with age. Although some factors and processes are difficult to intervene in or avoid, understanding the potential factors and determinants will help in developing timely strategies for a healthy gut microbiota in preterm infants. This review discusses potential determinants of gut microbial colonization in preterm infants and their underlying mechanisms.
Collapse
Affiliation(s)
- Xiaoyu Chen
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Yongyan Shi
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110000, China.
| |
Collapse
|
175
|
Teixeira RA, Silva C, Ferreira AC, Martins D, Leite-Moreira A, Miranda IM, Barros AS. The Association between Gestational Diabetes and the Microbiome: A Systematic Review and Meta-Analysis. Microorganisms 2023; 11:1749. [PMID: 37512921 PMCID: PMC10385443 DOI: 10.3390/microorganisms11071749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Gestational diabetes, affecting about 10% of pregnancies, is characterized by impaired glucose regulation and can lead to complications for health of pregnant women and their offspring. The microbiota, the resident microbes within the body, have been linked to the development of several metabolic conditions. This systematic review with meta-analysis aims to summarize the evidence on the differences in microbiota composition in pregnant women with gestational diabetes and their offspring compared to healthy pregnancies. A thorough search was conducted in the PubMed, Scopus, and Web of Science databases, and data from 21 studies were analyzed utilizing 41 meta-analyses. In the gut microbiota, Bifidobacterium and Alistipes were found to be more abundant in healthy pregnancies, while Roseburia appears to be more abundant in gestational diabetes. The heterogeneity among study findings regarding the microbiota in the meconium is considerable. The placental microbiota exhibited almost no heterogeneity, with an increased abundance of Firmicutes in the gestational diabetes group and a higher abundance of Proteobacteria in the control. The role of the microbiota in gestational diabetes is reinforced by these findings, which additionally point to the potential of microbiome-targeted therapies. To completely comprehend the interactions between gestational diabetes and the microbiome, standardizing methodologies and further research is necessary.
Collapse
Affiliation(s)
- Rita Almeida Teixeira
- Cardiovascular R&D Centre, UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Alameda Professor Hernani Monteiro, 4200-319 Porto, Portugal
| | - Cláudia Silva
- Cardiovascular R&D Centre, UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Alameda Professor Hernani Monteiro, 4200-319 Porto, Portugal
| | - António Carlos Ferreira
- Cardiovascular R&D Centre, UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Alameda Professor Hernani Monteiro, 4200-319 Porto, Portugal
| | - Diana Martins
- Cardiovascular R&D Centre, UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Alameda Professor Hernani Monteiro, 4200-319 Porto, Portugal
| | - Adelino Leite-Moreira
- Cardiovascular R&D Centre, UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Alameda Professor Hernani Monteiro, 4200-319 Porto, Portugal
| | - Isabel M Miranda
- Cardiovascular R&D Centre, UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Alameda Professor Hernani Monteiro, 4200-319 Porto, Portugal
| | - António S Barros
- Cardiovascular R&D Centre, UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Alameda Professor Hernani Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
176
|
Li XY, Meng L, Shen L, Ji HF. Regulation of gut microbiota by vitamin C, vitamin E and β-carotene. Food Res Int 2023; 169:112749. [PMID: 37254375 DOI: 10.1016/j.foodres.2023.112749] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 01/04/2023] [Accepted: 03/19/2023] [Indexed: 06/01/2023]
Abstract
Vitamin C (VC), vitamin E (VE) and β-carotene (βC) are representative dietary antioxidants, which exist in daily diet and can increase the antioxidant capacity of body fluids, cells and tissues. The health benefits of vitamins like VC, VE and βC are widely demonstrated. Given that the strong associations between the gut microbiota and host health or a range of diseases has been extensively reported, it is important to explore the modulatory effects of known vitamins on the gut microbiota. Herein, this article reviews the effects of VC, VE and βC on the gut microbiota. Totally, 19 studies were included, of which eight were related to VC, nine to VE, and six to βC. Overall, VC, VE and βC can provide health benefits to the host by modulating the composition and metabolic activity of the gut microbiota, improving intestinal barrier function and maintaining the normal function of the immune system. Two perspectives are proposed for future studies: i) roles of known antioxidant activity of vitamins in regulating the gut microbiota and its molecular mechanism need to be further studied; ii) causal relationships between the regulatory effects of vitamins on gut microbiota and host health still remains to be further verified.
Collapse
Affiliation(s)
- Xin-Yu Li
- Institute of Biomedical Research, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, People's Republic of China
| | - Lei Meng
- Institute of Biomedical Research, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, People's Republic of China
| | - Liang Shen
- Institute of Biomedical Research, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, People's Republic of China.
| | - Hong-Fang Ji
- Institute of Biomedical Research, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, People's Republic of China; School of Life Sciences, Ludong University, Yantai, People's Republic of China.
| |
Collapse
|
177
|
Vaill M, Kawanishi K, Varki N, Gagneux P, Varki A. Comparative physiological anthropogeny: exploring molecular underpinnings of distinctly human phenotypes. Physiol Rev 2023; 103:2171-2229. [PMID: 36603157 PMCID: PMC10151058 DOI: 10.1152/physrev.00040.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
Anthropogeny is a classic term encompassing transdisciplinary investigations of the origins of the human species. Comparative anthropogeny is a systematic comparison of humans and other living nonhuman hominids (so-called "great apes"), aiming to identify distinctly human features in health and disease, with the overall goal of explaining human origins. We begin with a historical perspective, briefly describing how the field progressed from the earliest evolutionary insights to the current emphasis on in-depth molecular and genomic investigations of "human-specific" biology and an increased appreciation for cultural impacts on human biology. While many such genetic differences between humans and other hominids have been revealed over the last two decades, this information remains insufficient to explain the most distinctive phenotypic traits distinguishing humans from other living hominids. Here we undertake a complementary approach of "comparative physiological anthropogeny," along the lines of the preclinical medical curriculum, i.e., beginning with anatomy and considering each physiological system and in each case considering genetic and molecular components that are relevant. What is ultimately needed is a systematic comparative approach at all levels from molecular to physiological to sociocultural, building networks of related information, drawing inferences, and generating testable hypotheses. The concluding section will touch on distinctive considerations in the study of human evolution, including the importance of gene-culture interactions.
Collapse
Affiliation(s)
- Michael Vaill
- Center for Academic Research and Training in Anthropogeny, University of California, San Diego, La Jolla, California
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California
| | - Kunio Kawanishi
- Center for Academic Research and Training in Anthropogeny, University of California, San Diego, La Jolla, California
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California
- Department of Experimental Pathology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Nissi Varki
- Center for Academic Research and Training in Anthropogeny, University of California, San Diego, La Jolla, California
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California
- Department of Pathology, University of California, San Diego, La Jolla, California
| | - Pascal Gagneux
- Center for Academic Research and Training in Anthropogeny, University of California, San Diego, La Jolla, California
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California
- Department of Pathology, University of California, San Diego, La Jolla, California
| | - Ajit Varki
- Center for Academic Research and Training in Anthropogeny, University of California, San Diego, La Jolla, California
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California
| |
Collapse
|
178
|
Shatova OP, Zabolotneva AA, Shestopalov AV. Molecular Ensembles of Microbiotic Metabolites in Carcinogenesis. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:867-879. [PMID: 37751860 DOI: 10.1134/s0006297923070027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 09/28/2023]
Abstract
The mechanisms of carcinogenesis are extremely complex and involve multiple components that contribute to the malignant cell transformation, tumor growth, and metastasis. In recent decades, there has been a growing interest in the role of symbiotic human microbiota in the regulation of metabolism and functioning of host immune system. The symbiosis between a macroorganism and its microbiota has given rise to the concept of a holoorganism. Interactions between the components of a holoorganism have formed in the process of coevolution, resulting in the acquisition by microbiotic metabolites of a special role of signaling molecules and main regulators of molecular interactions in the holoorganism. As elements of signaling pathways in the host organism, bacterial metabolites have become essential participants in various physiological and pathological processes, including tumor growth. At the same time, signaling metabolites often exhibit multiple effects and impact both the functions of the host cells and metabolic activity and composition of the microbiome. This review discusses the role of microbiotic metabolites in the induction and prevention of malignant transformation of cells in the host organism and their impact on the efficacy of anticancer therapy, with special emphasis on the involvement of some components of the microbial metabolite molecular ensemble in the initiation and progression of tumor growth.
Collapse
Affiliation(s)
- Olga P Shatova
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia.
- Peoples's Friendship University of Russia (RUDN University), Moscow, 117198, Russia
| | - Anastasiya A Zabolotneva
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| | - Aleksandr V Shestopalov
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
- Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Health of the Russian Federation, Moscow, 117997, Russia
| |
Collapse
|
179
|
Toro-Ascuy D, Cárdenas JP, Zorondo-Rodríguez F, González D, Silva-Moreno E, Puebla C, Nunez-Parra A, Reyes-Cerpa S, Fuenzalida LF. Microbiota Profile of the Nasal Cavity According to Lifestyles in Healthy Adults in Santiago, Chile. Microorganisms 2023; 11:1635. [PMID: 37512807 PMCID: PMC10384449 DOI: 10.3390/microorganisms11071635] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/18/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND The respiratory microbiome is dynamic, varying between anatomical niches, and it is affected by various host and environmental factors, one of which is lifestyle. Few studies have characterized the upper respiratory tract microbiome profile according to lifestyle. We explored the association between lifestyles and microbiota profiles in the upper respiratory tract of healthy adults. METHODS We analyzed nasal samples from 110 healthy adults who were living in Santiago, Chile, using 16S ribosomal RNA gene-sequencing methods. Volunteers completed a structured questionnaire about lifestyle. RESULTS The composition and abundance of taxonomic groups varied across lifestyle attributes. Additionally, multivariate models suggested that alpha diversity varied in the function of physical activity, nutritional status, smoking, and the interaction between nutritional status and smoking, although the significant impact of those variables varied between women and men. Although physical activity and nutritional status were significantly associated with all indexes of alpha diversity among women, the diversity of microbiota among men was associated with smoking and the interaction between nutritional status and smoking. CONCLUSIONS The alpha diversity of nasal microbiota is associated with lifestyle attributes, but these associations depend on sex and nutritional status. Our results suggest that future studies of the airway microbiome may provide a better resolution if data are stratified for differences in sex and nutritional status.
Collapse
Affiliation(s)
- Daniela Toro-Ascuy
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago 8910060, Chile
| | - Juan P Cárdenas
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago 8580745, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago 8580745, Chile
| | - Francisco Zorondo-Rodríguez
- Departamento de Gestión Agraria, Facultad Tecnológica, Universidad de Santiago de Chile, Santiago 8910060, Chile
| | - Damariz González
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago 8580745, Chile
| | - Evelyn Silva-Moreno
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago 8910060, Chile
| | - Carlos Puebla
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago 8910060, Chile
| | - Alexia Nunez-Parra
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago 8910060, Chile
| | - Sebastián Reyes-Cerpa
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago 8580745, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago 8580745, Chile
| | - Loreto F Fuenzalida
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago 8910060, Chile
| |
Collapse
|
180
|
Malcangi G, Patano A, Ciocia AM, Netti A, Viapiano F, Palumbo I, Trilli I, Guglielmo M, Inchingolo AD, Dipalma G, Inchingolo F, Minetti E, Inchingolo AM. Benefits of Natural Antioxidants on Oral Health. Antioxidants (Basel) 2023; 12:1309. [PMID: 37372039 DOI: 10.3390/antiox12061309] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 05/31/2023] [Accepted: 06/19/2023] [Indexed: 06/29/2023] Open
Abstract
In recent years, special attention has been paid to the correlation between oxidation-reduction mechanisms and human health. The free radicals produced via physiological cellular biochemical processes are major contributors to oxidation phenomena. Their instability is the major cause of cellular damage. Free radical reactive oxygen species containing oxygen are the best-known ones. The body neutralises the harmful effects of free radicals via the production of endogenous antioxidants (superoxide dismutase, catalase, glutathione, and melatonin). The field of study of nutraucetics has found antioxidant capacity in substances such as vitamins A, B, C, E, coenzyme Q-10, selenium, flavonoids, lipoic acid, carotenoids, and lycopene contained in some foods. There are several areas of investigation that aim to research the interaction between reactive oxygen species, exogenous antioxidants, and the microbiota to promote increased protection via the peroxidation of macromolecules (proteins, and lipids) by maintaining a dynamic balance among the species that make up the microbiota. In this scoping review, we aim to map the scientific literature on oxidative stress related to the oral microbiota, and the use of natural antioxidants to counteract it, to assess the volume, nature, characteristics, and type of studies available to date, and to suggest the possible gaps that will emerge from the analysis.
Collapse
Affiliation(s)
- Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Assunta Patano
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Anna Maria Ciocia
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Anna Netti
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Fabio Viapiano
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Irene Palumbo
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Irma Trilli
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | | | | | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Elio Minetti
- Department of Biomedical, Surgical and Dental Science, University of Milan, 20122 Milan, Italy
| | | |
Collapse
|
181
|
Sun C, Song R, Zhou J, Jia Y, Lu J. Fermented Bamboo Fiber Improves Productive Performance by Regulating Gut Microbiota and Inhibiting Chronic Inflammation of Sows and Piglets during Late Gestation and Lactation. Microbiol Spectr 2023; 11:e0408422. [PMID: 37042787 PMCID: PMC10269633 DOI: 10.1128/spectrum.04084-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 03/21/2023] [Indexed: 04/13/2023] Open
Abstract
Sows exhibit metabolic syndrome and significant changes in intestinal microbiota during late gestation and lactation, affecting sow performance and piglet health. Dietary fiber (DF) is widely applied to improve sow performance by modulating gut microbiota and their by-products. Here, 60 sows were randomly allocated to groups, including CON (8% wheat bran), FBF-1 (1% fermented bamboo fiber), FBF-2 (2.5% fermented bamboo fiber), and FBF-3 (4% fermented bamboo fiber) from day 80 of gestation (G80d) to the end of lactation (L21d). Compared with CON, the FBF-3 diet decreased lactation backfat loss, increased average daily feed intake (ADFI) during lactation, and the weight gain of piglets, while supplementation of FBF increased fecal water content and reduced the rate of constipation in sows. Further, the yield and quality of milk of sows in FBF groups were improved. The FBF-3 diet significantly reduced markers of intestinal permeability (diamine oxidase and endotoxin) and systemic inflammation (interleukin-6 [IL-6] and tumor necrosis factor alpha) in sow serum during lactation, while it increased the anti-inflammatory marker (IL-10). Similarly, the piglets in the FBF-2 and FBF-3 groups had lower levels of IL-6 and higher levels of IgG, IgM, and insulin-like growth factor in serum. In addition, sows fed the 4% FBF diet had higher levels of acetate, propionate, butyrate, and total short-chain fatty acids (SCFAs) in feces than CON, and total SCFAs were promoted in piglets from the FBF-3 group. Spearman correlation analysis showed that immunity, inflammation, and intestinal microbiota are closely related to sow performance, which can affect piglet growth. The potential mechanism could be that FBF promoted the enrichment of beneficial genera such as Lachnospira, Lachnospiracea_XPB1014_Group, and Roseburia and the production of SCFAs in the sow's intestine, and reduced the relative abundance of harmful bacteria such as Fusobacterium, Sutterellaceae, and Sutterella. Meanwhile, the intake of FBF by sows affected the gut microbial composition of their offspring piglets, significantly increasing the relative abundance of beneficial bacteria Alistipes and Lachnoclostridium and decreasing the relative abundance of pathogenic bacteria Trueperella among colonic microorganisms. IMPORTANCE Dietary fiber is widely applied in the nutrition of sows due to its potential value in improving performance and intestinal health. Fermented bamboo fiber, rich in dietary fiber, has not been fully evaluated to be used in sow diets. Sows mobilize body reserves during gestation and lactation due to nutrients being prioritized for lactation purposes while feeding piglets, which generally leads to metabolism and immunity undergoing drastic changes. The main manifestations are increased inflammation and intestinal permeability and disturbed intestinal flora, which ultimately reduces the ADFI and milk quality, thus affecting the growth of piglets. The study described here is the first attempt to provide FBF for sows in late gestation and lactation can reverse this process. The 4% FBF was initially explored to have the most significantly beneficial effect. It provides a potentially effective method for dietary modification to control the gut microbiota and its metabolites to improve sow and piglet health. Moreover, the sow-piglet model offers a reference for investigating the impact of dietary fiber on the intestinal health of human mothers and infants.
Collapse
Affiliation(s)
- Chuansong Sun
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, China
- The National Engineering Laboratory for Feed Safety and Pollution Prevention and Controlling, National Development and Reform Commission, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science, Ministry of Agriculture and Rural Affairs, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Rui Song
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, China
- The National Engineering Laboratory for Feed Safety and Pollution Prevention and Controlling, National Development and Reform Commission, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science, Ministry of Agriculture and Rural Affairs, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Jianyong Zhou
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, China
- The National Engineering Laboratory for Feed Safety and Pollution Prevention and Controlling, National Development and Reform Commission, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science, Ministry of Agriculture and Rural Affairs, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Yubiao Jia
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, China
- The National Engineering Laboratory for Feed Safety and Pollution Prevention and Controlling, National Development and Reform Commission, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science, Ministry of Agriculture and Rural Affairs, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Jianjun Lu
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, China
- The National Engineering Laboratory for Feed Safety and Pollution Prevention and Controlling, National Development and Reform Commission, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science, Ministry of Agriculture and Rural Affairs, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
| |
Collapse
|
182
|
Choi R, Bodkhe R, Pees B, Kim D, Berg M, Monnin D, Cho J, Narayan V, Deller E, Shapira M. An Enterobacteriaceae bloom in aging animals is restrained by the gut microbiome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.13.544815. [PMID: 37398063 PMCID: PMC10312681 DOI: 10.1101/2023.06.13.544815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
The gut microbiome plays important roles in host function and health. Core microbiomes have been described for different species, and imbalances in their composition, known as dysbiosis, are associated with pathology. Changes in the gut microbiome and dysbiosis are common in aging, possibly due to multi-tissue deterioration, which includes metabolic shifts, dysregulated immunity, and disrupted epithelial barriers. However, the characteristics of these changes, as reported in different studies, are varied and sometimes conflicting. Using clonal populations of C. elegans to highlight trends shared among individuals, and employing NextGen sequencing, CFU counts and fluorescent imaging to characterize age-dependent changes in worms raised in different microbial environments, we identified an Enterobacteriaceae bloom as a common denominator in aging animals. Experiments using Enterobacter hormachei, a representative commensal, suggested that the Enterobacteriaceae bloom was facilitated by a decline in Sma/BMP immune signaling in aging animals and demonstrated its detrimental potential for increasing susceptibility to infection. However, such detrimental effects were context-dependent, mitigated by competition with commensal communities, highlighting the latter as determinants of healthy versus unhealthy aging, depending on their ability to restrain opportunistic pathobionts.
Collapse
Affiliation(s)
- Rebecca Choi
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Rahul Bodkhe
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Barbara Pees
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Dan Kim
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Maureen Berg
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - David Monnin
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Juhyun Cho
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Vivek Narayan
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Ethan Deller
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Michael Shapira
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
183
|
Krupa-Kotara K, Grajek M, Grot M, Czarnota M, Wypych-Ślusarska A, Oleksiuk K, Głogowska-Ligus J, Słowiński J. Pre- and Postnatal Determinants Shaping the Microbiome of the Newborn in the Opinion of Pregnant Women from Silesia (Poland). Life (Basel) 2023; 13:1383. [PMID: 37374165 PMCID: PMC10305644 DOI: 10.3390/life13061383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Pre- and postnatal factors influence the formation of the newborn's microbiome as early as birth and the intrauterine period has a substantial impact on the composition of the baby's gastrointestinal microbiota and its subsequent development. This study intends to measure pregnant women's knowledge of the importance of microbiota for the health of the newborn. The sample was selected based on defined inclusion and exclusion criteria. The assessment of women's knowledge was assessed by the Kolmogorov-Smirnov and Kruskal-Wallis statistical tests. This study population comprised 291 adult pregnant women with a mean age of 28.4 ± 4.7 years. A total of 34% (n = 99), 35% (n = 101), and 31.3% (n = 91) were at the 1-3 trimester, respectively. The results showed that 36.4% of the women were aware that the intrauterine period changes the makeup of the gastrointestinal microbiota, whereas 5.8% exhibited awareness of the composition of the child's normal gut microbiota. Most of the women surveyed-(72.1%)-know that colonization of the tract occurs as early as the birth period. Women with student status (those who will pursue higher education in the future) and those who had given birth to the most children exhibited higher levels of knowledge.
Collapse
Affiliation(s)
- Karolina Krupa-Kotara
- Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (A.W.-Ś.); (K.O.); (J.G.-L.); (J.S.)
| | - Mateusz Grajek
- Department of Public Health, Department of Public Health Policy, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland;
| | - Martina Grot
- Student Scientific Society, Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (M.G.); (M.C.)
- Doctoral School, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Martina Czarnota
- Student Scientific Society, Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (M.G.); (M.C.)
| | - Agata Wypych-Ślusarska
- Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (A.W.-Ś.); (K.O.); (J.G.-L.); (J.S.)
| | - Klaudia Oleksiuk
- Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (A.W.-Ś.); (K.O.); (J.G.-L.); (J.S.)
| | - Joanna Głogowska-Ligus
- Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (A.W.-Ś.); (K.O.); (J.G.-L.); (J.S.)
| | - Jerzy Słowiński
- Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (A.W.-Ś.); (K.O.); (J.G.-L.); (J.S.)
| |
Collapse
|
184
|
Ternák G, Márovics G, Sümegi K, Bánfai Z, Büki G, Magyari L, Szabó A, Melegh B. Down-Syndrome-Related Maternal Dysbiosis Might Be Triggered by Certain Classes of Antibiotics: A New Insight into the Possible Pathomechanisms. Antibiotics (Basel) 2023; 12:1029. [PMID: 37370348 DOI: 10.3390/antibiotics12061029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/24/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Down syndrome (DS) is a leading human genomic abnormality resulting from the trisomy of chromosome 21. The genomic base of the aneuploidy behind this disease is complex, and this complexity poses formidable challenges to understanding the underlying molecular basis. In the spectrum of the classic DS risk factor associations, the role of nutrients, vitamins, and, in general, the foodborne-associated background, as part of the events ultimately leading to chromosome nondisjunction, has long been recognized as a well-established clinical association. The integrity of the microbiome is a basic condition in these events, and the dysbiosis may be associated with secondary health outcomes. The possible association of DS development with maternal gut microbiota should therefore require more attention. We have hypothesized that different classes of antibiotics might promote or inhibit the proliferation of different microbial taxa; and hence, we might find associations between the use of the different classes of antibiotics and the prevalence of DS through the modification of the microbiome. As antibiotics are considered major disruptors of the microbiome, it could be hypothesized that the consumption/exposure of certain classes of antibiotics might be associated with the prevalence of DS in European countries (N = 30). By utilizing three different statistical methods, comparisons have been made between the average yearly antibiotic consumption (1997-2020) and the estimated prevalence of people living with DS for the year 2019 as a percentage of the population in European countries. We have found strong statistical correlations between the consumption of tetracycline (J01A) and the narrow-spectrum, beta-lactamase-resistant penicillin (J01CF) and the prevalence of DS.
Collapse
Affiliation(s)
- Gábor Ternák
- Institute of Migration Health, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary
| | - Gergely Márovics
- Department of Public Health Medicine, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary
| | - Katalin Sümegi
- Department of Medical Genetics, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary
- Department of Biochemistry and Chemistry, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary
| | - Zsolt Bánfai
- Department of Medical Genetics, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary
| | - Gergely Büki
- Department of Medical Genetics, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary
| | - Lili Magyari
- Department of Medical Genetics, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary
| | - András Szabó
- Department of Medical Genetics, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary
| | - Béla Melegh
- Department of Medical Genetics, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary
| |
Collapse
|
185
|
Ye J, Yao J, He F, Sun J, Zhao Z, Wang Y. Regulation of gut microbiota: a novel pretreatment for complications in patients who have undergone kidney transplantation. Front Cell Infect Microbiol 2023; 13:1169500. [PMID: 37346031 PMCID: PMC10280007 DOI: 10.3389/fcimb.2023.1169500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023] Open
Abstract
Kidney transplantation is an effective method to improve the condition of patients with end-stage renal disease. The gut microbiota significantly affects the immune system and can be used as an influencing factor to change the prognoses of patients who have undergone kidney transplantation. Recipients after kidney transplantation showed a lower abundance of Firmicutes and Faecalibacterium prausnitzii and a higher proportion of Bacteroidetes and Proteobacteria. After using prebiotics, synbiotics, and fecal microbiota transplantation to regulate the microbial community, the prognoses of patients who underwent kidney transplantation evidently improved. We aimed to determine the relationship between gut microbiota and various postoperative complications inpatients who have undergone kidney transplantation in recent years and to explore how gut microecology affects post-transplant complications. An in-depth understanding of the specific functions of gut microbiota and identification of the actual pathogenic flora during complications in patients undergoing kidney transplantation can help physicians develop strategies to restore the normal intestinal microbiome of transplant patients to maximize their survival and improve their quality of life.
Collapse
Affiliation(s)
- Jiajia Ye
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junxia Yao
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangfang He
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Sun
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Zhao
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yumei Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
186
|
Zaidi S, Ali K, Khan AU. It's all relative: analyzing microbiome compositions, its significance, pathogenesis and microbiota derived biofilms: Challenges and opportunities for disease intervention. Arch Microbiol 2023; 205:257. [PMID: 37280443 DOI: 10.1007/s00203-023-03589-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/06/2023] [Accepted: 05/18/2023] [Indexed: 06/08/2023]
Abstract
Concept of microorganisms has largely been perceived from their pathogenic view point. Nevertheless, it is being gradually revisited in terms of its significance to human health and now appears to be the most dominant force that shapes the immune system of the human body and also determines an individual's predisposition to diseases. Human inhabits bacterial diversity (which is predominant among all microbial communities in human body) occupying 0.3% of body mass, known as microbiota. On birth, a part of microbiota that child obtains is essentially a mother's legacy. So, the review was initiated with this critical topic of microbiotal inheritance. Since, each body site has distinct physiological specifications; therefore, they contain discrete microbiome composition that has been separately discussed along with dysbiosis-induced pathologies originating in different body organs. Factors affecting microbiome composition and may cause dysbiosis like antibiotics, delivery, feeding method etc. as well as the strategies that immune system adopts to prevent dysbiosis have been highlighted. We also tried to bring into attention the topic of dysbiosis induced biofilms, that enables cohort to survive stresses, evolve, disseminate and infection resurgence that is still in dormancy. Eventually, we put spotlight on microbiome significance in medical therapeutics. We didn't merely confine article to gut microbiota, that is being studied more extensively. Numerous community forms at diverse body sites are inter-related, and being exposed to awfully variable perturbations appear to be challenging to evaluate perturbation risks holistically. All aspects have been elaborately discussed to achieve a global depiction of human microbiota in order to meet urgent necessity for protocol standardisation. Demonstrates that environmental challenges (antibiotic use, alterations in diet, stress, smoking etc.) might cause dysbiosis i.e. transition of healthy microbiome composition to the one in which pathogenic microorganisms become more abundant, and eventually results in an infected state.
Collapse
Affiliation(s)
- Sahar Zaidi
- Medical Microbiology and Molecular Biology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, 202002, India
| | - Khursheed Ali
- Medical Microbiology and Molecular Biology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, 202002, India
| | - Asad U Khan
- Medical Microbiology and Molecular Biology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, 202002, India.
| |
Collapse
|
187
|
Lei Y, Liu Q, Li Q, Zhao C, Zhao M, Lu Q. Exploring the Complex Relationship Between Microbiota and Systemic Lupus Erythematosus. Curr Rheumatol Rep 2023; 25:107-116. [PMID: 37083877 DOI: 10.1007/s11926-023-01102-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2023] [Indexed: 04/22/2023]
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by various autoantibodies and multi-organ. Microbiota dysbiosis in the gut, skin, oral, and other surfaces has a significant impact on SLE development. This article summarizes relevant research and provides new microbiome-related strategies for exploring the mechanisms and treating patients with SLE. RECENT FINDINGS SLE patients have disruptions in multiple microbiomes, with the gut microbiota (bacteria, viruses, and fungi) and their metabolites being the most thoroughly researched. This dysbiosis can promote SLE progression through mechanisms such as the leaky gut, molecular mimicry, and epigenetic regulation. Notwithstanding study constraints on the relationship between microbiota and SLE, specific interventions targeting the gut microbiota, such as probiotics, dietary management, and fecal microbiota transplantation, have emerged as promising SLE therapeutics.
Collapse
Affiliation(s)
- Yu Lei
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
- Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qianmei Liu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
| | - Qilin Li
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
- Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Cheng Zhao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
- Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming Zhao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China.
- Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China.
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China.
- Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
188
|
Bicknell B, Liebert A, Borody T, Herkes G, McLachlan C, Kiat H. Neurodegenerative and Neurodevelopmental Diseases and the Gut-Brain Axis: The Potential of Therapeutic Targeting of the Microbiome. Int J Mol Sci 2023; 24:9577. [PMID: 37298527 PMCID: PMC10253993 DOI: 10.3390/ijms24119577] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 04/28/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
The human gut microbiome contains the largest number of bacteria in the body and has the potential to greatly influence metabolism, not only locally but also systemically. There is an established link between a healthy, balanced, and diverse microbiome and overall health. When the gut microbiome becomes unbalanced (dysbiosis) through dietary changes, medication use, lifestyle choices, environmental factors, and ageing, this has a profound effect on our health and is linked to many diseases, including lifestyle diseases, metabolic diseases, inflammatory diseases, and neurological diseases. While this link in humans is largely an association of dysbiosis with disease, in animal models, a causative link can be demonstrated. The link between the gut and the brain is particularly important in maintaining brain health, with a strong association between dysbiosis in the gut and neurodegenerative and neurodevelopmental diseases. This link suggests not only that the gut microbiota composition can be used to make an early diagnosis of neurodegenerative and neurodevelopmental diseases but also that modifying the gut microbiome to influence the microbiome-gut-brain axis might present a therapeutic target for diseases that have proved intractable, with the aim of altering the trajectory of neurodegenerative and neurodevelopmental diseases such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, autism spectrum disorder, and attention-deficit hyperactivity disorder, among others. There is also a microbiome-gut-brain link to other potentially reversible neurological diseases, such as migraine, post-operative cognitive dysfunction, and long COVID, which might be considered models of therapy for neurodegenerative disease. The role of traditional methods in altering the microbiome, as well as newer, more novel treatments such as faecal microbiome transplants and photobiomodulation, are discussed.
Collapse
Affiliation(s)
- Brian Bicknell
- NICM Health Research Institute, University of Western Sydney, Westmead, NSW 2145, Australia; (A.L.); (H.K.)
| | - Ann Liebert
- NICM Health Research Institute, University of Western Sydney, Westmead, NSW 2145, Australia; (A.L.); (H.K.)
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia
- Department of Governance and Research, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia;
| | - Thomas Borody
- Centre for Digestive Diseases, Five Dock, NSW 2046, Australia;
| | - Geoffrey Herkes
- Department of Governance and Research, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia;
| | - Craig McLachlan
- Centre for Healthy Futures, Torrens University Australia, Ultimo, NSW 2007, Australia;
| | - Hosen Kiat
- NICM Health Research Institute, University of Western Sydney, Westmead, NSW 2145, Australia; (A.L.); (H.K.)
- Centre for Healthy Futures, Torrens University Australia, Ultimo, NSW 2007, Australia;
- Macquarie Medical School, Macquarie University, Macquarie Park, NSW 2109, Australia
- ANU College of Health and Medicine, Australian National University, Canberra, ACT 2601, Australia
| |
Collapse
|
189
|
Wu Y, Zhang G, Wang Y, Wei X, Liu H, Zhang L, Zhang L. A Review on Maternal and Infant Microbiota and Their Implications for the Prevention and Treatment of Allergic Diseases. Nutrients 2023; 15:nu15112483. [PMID: 37299446 DOI: 10.3390/nu15112483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023] Open
Abstract
Allergic diseases, which are closely related to the composition and metabolism of maternal and infant flora, are prevalent in infants worldwide. The mother's breast milk, intestinal, and vaginal flora directly or indirectly influence the development of the infant's immune system from pregnancy to lactation, and the compositional and functional alterations of maternal flora are associated with allergic diseases in infants. Meanwhile, the infant's own flora, represented by the intestinal flora, indicates and regulates the occurrence of allergic diseases and is altered with the intervention of allergic diseases. By searching and selecting relevant literature in PubMed from 2010 to 2023, the mechanisms of allergy development in infants and the links between maternal and infant flora and infant allergic diseases are reviewed, including the effects of flora composition and its consequences on infant metabolism. The critical role of maternal and infant flora in allergic diseases has provided a window for probiotics as a microbial therapy. Therefore, the uses and mechanisms by which probiotics, such as lactic acid bacteria, can help to improve the homeostasis of both the mother and the infant, and thereby treat allergies, are also described.
Collapse
Affiliation(s)
- Yifan Wu
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Gongsheng Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yucong Wang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Xin Wei
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Huanhuan Liu
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Lili Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Lanwei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266100, China
| |
Collapse
|
190
|
Ben-Azu B, del Re EC, VanderZwaag J, Carrier M, Keshavan M, Khakpour M, Tremblay MÈ. Emerging epigenetic dynamics in gut-microglia brain axis: experimental and clinical implications for accelerated brain aging in schizophrenia. Front Cell Neurosci 2023; 17:1139357. [PMID: 37256150 PMCID: PMC10225712 DOI: 10.3389/fncel.2023.1139357] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/27/2023] [Indexed: 06/01/2023] Open
Abstract
Brain aging, which involves a progressive loss of neuronal functions, has been reported to be premature in probands affected by schizophrenia (SCZ). Evidence shows that SCZ and accelerated aging are linked to changes in epigenetic clocks. Recent cross-sectional magnetic resonance imaging analyses have uncovered reduced brain reserves and connectivity in patients with SCZ compared to typically aging individuals. These data may indicate early abnormalities of neuronal function following cyto-architectural alterations in SCZ. The current mechanistic knowledge on brain aging, epigenetic changes, and their neuropsychiatric disease association remains incomplete. With this review, we explore and summarize evidence that the dynamics of gut-resident bacteria can modulate molecular brain function and contribute to age-related neurodegenerative disorders. It is known that environmental factors such as mode of birth, dietary habits, stress, pollution, and infections can modulate the microbiota system to regulate intrinsic neuronal activity and brain reserves through the vagus nerve and enteric nervous system. Microbiota-derived molecules can trigger continuous activation of the microglial sensome, groups of receptors and proteins that permit microglia to remodel the brain neurochemistry based on complex environmental activities. This remodeling causes aberrant brain plasticity as early as fetal developmental stages, and after the onset of first-episode psychosis. In the central nervous system, microglia, the resident immune surveillance cells, are involved in neurogenesis, phagocytosis of synapses and neurological dysfunction. Here, we review recent emerging experimental and clinical evidence regarding the gut-brain microglia axis involvement in SCZ pathology and etiology, the hypothesis of brain reserve and accelerated aging induced by dietary habits, stress, pollution, infections, and other factors. We also include in our review the possibilities and consequences of gut dysbiosis activities on microglial function and dysfunction, together with the effects of antipsychotics on the gut microbiome: therapeutic and adverse effects, role of fecal microbiota transplant and psychobiotics on microglial sensomes, brain reserves and SCZ-derived accelerated aging. We end the review with suggestions that may be applicable to the clinical setting. For example, we propose that psychobiotics might contribute to antipsychotic-induced therapeutic benefits or adverse effects, as well as reduce the aging process through the gut-brain microglia axis. Overall, we hope that this review will help increase the understanding of SCZ pathogenesis as related to chronobiology and the gut microbiome, as well as reveal new concepts that will serve as novel treatment targets for SCZ.
Collapse
Affiliation(s)
- Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Nigeria
| | - Elisabetta C. del Re
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- VA Boston Healthcare System, Brockton, MA, United States
- Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Jared VanderZwaag
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Micaël Carrier
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Matcheri Keshavan
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- Beth Israel Deaconess Medical Center, Boston, MA, United States
| | | | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada
| |
Collapse
|
191
|
Lai X, Zhang Z, Zhang Z, Liu S, Bai C, Chen Z, Qadri QR, Fang Y, Wang Z, Pan Y, Wang Q. Integrated microbiome-metabolome-genome axis data of Laiwu and Lulai pigs. Sci Data 2023; 10:280. [PMID: 37179393 PMCID: PMC10183000 DOI: 10.1038/s41597-023-02191-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Excessive fat deposition can trigger metabolic diseases, and it is crucial to identify factors that can break the link between fat deposition and metabolic diseases. Healthy obese Laiwu pigs (LW) are high in fat content but resistant to metabolic diseases. In this study, we compared the fecal microbiome, fecal and blood metabolome, and genome of LW and Lulai pigs (LU) to identify factors that can block the link between fat deposition and metabolic diseases. Our results show significant differences in Spirochetes and Treponema, which are involved in carbohydrate metabolism, between LW and LU. The fecal and blood metabolome composition was similar, and some anti-metabolic disease components of blood metabolites were different between the two breeds of pigs. The predicted differential RNA is mainly enriched in lipid metabolism and glucose metabolism, which is consistent with the functions of differential microbiota and metabolites. The down-regulated gene RGP1 is strongly negatively correlated with Treponema. Our omics data would provide valuable resources for further scientific research on healthy obesity in both human and porcine.
Collapse
Affiliation(s)
- Xueshuang Lai
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
- Department of Animal Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310030, PR China
| | - Zhenyang Zhang
- Department of Animal Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310030, PR China
| | - Zhe Zhang
- Department of Animal Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310030, PR China
| | - Shengqiang Liu
- Department of Animal Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310030, PR China
- Hainan institute, Zhejiang University, Sanya, 310014, PR China
| | - Chunyan Bai
- Department of Animal Science, College of Animal Sciences, Jilin University, Changchui, 130015, PR China
| | - Zitao Chen
- Department of Animal Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310030, PR China
| | - Qamar Raza Qadri
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Yifei Fang
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
- Department of Animal Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310030, PR China
| | - Zhen Wang
- Department of Animal Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310030, PR China
| | - Yuchun Pan
- Department of Animal Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310030, PR China.
- Hainan institute, Zhejiang University, Sanya, 310014, PR China.
| | - Qishan Wang
- Department of Animal Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310030, PR China.
- Hainan institute, Zhejiang University, Sanya, 310014, PR China.
| |
Collapse
|
192
|
Martin AJ, Serebrinsky-Duek K, Riquelme E, Saa PA, Garrido D. Microbial interactions and the homeostasis of the gut microbiome: the role of Bifidobacterium. MICROBIOME RESEARCH REPORTS 2023; 2:17. [PMID: 38046822 PMCID: PMC10688804 DOI: 10.20517/mrr.2023.10] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 12/05/2023]
Abstract
The human gut is home to trillions of microorganisms that influence several aspects of our health. This dense microbial community targets almost all dietary polysaccharides and releases multiple metabolites, some of which have physiological effects on the host. A healthy equilibrium between members of the gut microbiota, its microbial diversity, and their metabolites is required for intestinal health, promoting regulatory or anti-inflammatory immune responses. In contrast, the loss of this equilibrium due to antibiotics, low fiber intake, or other conditions results in alterations in gut microbiota composition, a term known as gut dysbiosis. This dysbiosis can be characterized by a reduction in health-associated microorganisms, such as butyrate-producing bacteria, enrichment of a small number of opportunistic pathogens, or a reduction in microbial diversity. Bifidobacterium species are key species in the gut microbiome, serving as primary degraders and contributing to a balanced gut environment in various ways. Colonization resistance is a fundamental property of gut microbiota for the prevention and control of infections. This community competes strongly with foreign microorganisms, such as gastrointestinal pathogens, antibiotic-resistant bacteria, or even probiotics. Resistance to colonization is based on microbial interactions such as metabolic cross-feeding, competition for nutrients, or antimicrobial-based inhibition. These interactions are mediated by metabolites and metabolic pathways, representing the inner workings of the gut microbiota, and play a protective role through colonization resistance. This review presents a rationale for how microbial interactions provide resistance to colonization and gut dysbiosis, highlighting the protective role of Bifidobacterium species.
Collapse
Affiliation(s)
- Alberto J.M. Martin
- Laboratorio de Redes Biológicas, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Santiago 8580702, Chile
| | - Kineret Serebrinsky-Duek
- Department of Chemical and Bioprocess Engineering, Pontificia Universidad Católica de Chile, Santiago 833115, Chile
| | - Erick Riquelme
- Department of Respiratory Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Pedro A. Saa
- Department of Chemical and Bioprocess Engineering, Pontificia Universidad Católica de Chile, Santiago 833115, Chile
- Institute for Mathematical and Computational Engineering, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Daniel Garrido
- Department of Chemical and Bioprocess Engineering, Pontificia Universidad Católica de Chile, Santiago 833115, Chile
| |
Collapse
|
193
|
Wang Z, He H, Chen M, Ni M, Yuan D, Cai H, Chen Z, Li M, Xu H. Impact of coprophagy prevention on the growth performance, serum biochemistry, and intestinal microbiome of rabbits. BMC Microbiol 2023; 23:125. [PMID: 37165350 PMCID: PMC10170819 DOI: 10.1186/s12866-023-02869-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 04/23/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Coprophagy plays a vital role in maintaining growth and development in many small herbivores. Here, we constructed a coprophagy model by dividing rabbits into three groups, namely, control group (CON), sham-coprophagy prevention group (SCP), and coprophagy prevention group (CP), to explore the effects of coprophagy prevention on growth performance and cecal microecology in rabbits. RESULTS Results showed that CP treatment decreased the feed utilization and growth performance of rabbits. Serum total cholesterol and total triglyceride in the CP group were remarkably lower than those in the other two groups. Furthermore, CP treatment destroyed cecum villi and reduced the content of short-chain fatty acids (SCFAs) in cecum contents. Gut microbiota profiling showed significant differences in the phylum and genus composition of cecal microorganisms among the three groups. At the genus level, the abundance of Oscillospira and Ruminococcus decreased significantly in the CP group. Enrichment analysis of metabolic pathways showed a significantly up-regulated differential metabolic pathway (PWY-7315, dTDP-N-acetylthomosamine biosynthesis) in the CP group compared with that in the CON group. Correlation analysis showed that the serum biochemical parameters were positively correlated with the abundance of Oscillospira, Sutterella, and Butyricimonas but negatively correlated with the abundance of Oxalobacte and Desulfovibrio. Meanwhile, the abundance of Butyricimonas and Parabacteroidesde was positively correlated with the concentration of butyric acid in the cecum. CONCLUSIONS In summary, coprophagy prevention had negative effects on serum biochemistry and gut microbiota, ultimately decreasing the growth performance of rabbits. The findings provide evidence for further revealing the biological significance of coprophagy in small herbivorous mammals.
Collapse
Affiliation(s)
- Zhitong Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Hui He
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Mengjuan Chen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Mengke Ni
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Dongdong Yuan
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Hanfang Cai
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Zhi Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225000, China
| | - Ming Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China.
| | - Huifen Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China.
| |
Collapse
|
194
|
Wang R, Yang X, Jiang Q, Chen L, Gu S, Shen G, Liu S, Xiang X. Effect of mussel polysaccharide on glucolipid metabolism and intestinal flora in type 2 diabetic mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:3353-3366. [PMID: 36750436 DOI: 10.1002/jsfa.12488] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 01/08/2023] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Type 2 diabetes (T2D) mellitus is a major metabolic disease, and its incidence and lethality have increased significantly in recent years, making it a serious threat to human health. Among numerous previous studies, polysaccharides have been shown to alleviate the adverse effects of T2D, but there are still problems such as insufficient analysis and poor understanding of the mechanisms by which polysaccharides, especially those of marine origin, regulate T2D. METHODS In this study, we used multiple allosteric approaches to further investigate the regulatory effects of mussel polysaccharides (MPs) on T2D and gut microbiota disorders in mice by identifying changes in genes, proteins, metabolites and target organs associated with glucolipid metabolism using an animal model of T2D fed with high-fat diets, and to explore the underlying molecular mechanisms. RESULTS After MP intervention, serum levels of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and high-density lipoprotein cholesterol (HDL-C) were up-regulated, and blood glucose and lipid levels were effectively reduced in T2D mice. Activation of signaling molecules related to the upstream and downstream of the insulin PI3K/Akt signaling pathway reduced hepatic insulin resistance. The relative abundance of short-chain fatty acid (SCFA)-producing bacteria (including Akkermansia, Siraeum Eubacterium and Allobaculum) increased and harmful desulfurizing Vibrio decreased. In addition, the levels of SCFAs were increased. CONCLUSION These results suggest that MP can increase SCFA levels by altering the abundance of intestinal flora, thereby activating the PI3K/Akt signaling pathway and exerting hypoglycemic effects. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Rui Wang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
- Key Laboratory of Marine Fishery Resources Exploitment and Utilization of Zhejiang Province, Hangzhou, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Xingwen Yang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
- Key Laboratory of Marine Fishery Resources Exploitment and Utilization of Zhejiang Province, Hangzhou, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Qihong Jiang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
- Key Laboratory of Marine Fishery Resources Exploitment and Utilization of Zhejiang Province, Hangzhou, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Lin Chen
- Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Saiqi Gu
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
- Key Laboratory of Marine Fishery Resources Exploitment and Utilization of Zhejiang Province, Hangzhou, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Guoxin Shen
- Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Shulai Liu
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
- Key Laboratory of Marine Fishery Resources Exploitment and Utilization of Zhejiang Province, Hangzhou, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Xingwei Xiang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
- Key Laboratory of Marine Fishery Resources Exploitment and Utilization of Zhejiang Province, Hangzhou, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| |
Collapse
|
195
|
Chen C, Liu L, Zhong Y, Wang M, Ai Y, Hou Y, Chen H, Lin X, Zhang Y, Ding M, Luo T, Li J, Li X, Xiao X. Gut microbiota-bile acids-glucagon like peptide-1 axis contributes the resistance to high fat diet-induced obesity in mice. J Nutr Biochem 2023; 117:109358. [PMID: 37085058 DOI: 10.1016/j.jnutbio.2023.109358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/03/2023] [Accepted: 04/14/2023] [Indexed: 04/23/2023]
Abstract
In human and rodents, some individuals may remain lean even when they are challenged with high calorie intake. The underlying mechanism for resistance to diet-induced obesity was poorly understood. Here, we used C57BL/6J mice to establish animal models of high-fat diet (HFD) induced obesity sensitive (DIO) mice and obesity resistant (DIR) mice. We then investigated the role of gut microbiota, bile acids (BAs) and brown adipose tissue (BAT) thermogenesis in the development of DIR. Reduced fat accumulation, increased glucose tolerance and energy expenditure through BAT activation were observed in DIR mice. The plasma BAs of DIR mice especially the unconjugated BAs were significantly decreased, while intestine tauro-conjugated bile acids (T-CA, T-β-MCA, T-ω-MCA and T-UDCA) were significantly increased in DIR mice. The composition of the gut flora also changed drastically, and negative correlation was found between metabolic profiles (plasma TG, TC, LDL and body weight) and the abundance of Ruminiclostridium in DIR mice, while genus Anaerotruncus abundance in DOR mice was found to be positively correlated. After fecal microbiota transplants, HFD fed recipient mice exhibited a trend toward reduced adiposity and improved glucose tolerance, while showing increased serum tauro-conjugated BAs levels. STC-1 cell experiments confirmed tauro-conjugated BA (T-β-MCA) activated FXR/TGR5 pathway and induced the production of GLP-1, inhibiting genes that regulate the ceramide synthesis. Our results indicated that the DIR mice exhibited higher energy expenditure by activating BAT thermogenesis, which may be related altered gut microbiota-bile acids-glucagon like peptide-1 axis.
Collapse
Affiliation(s)
- Chunxiu Chen
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Lingli Liu
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Ying Zhong
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Miaoran Wang
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Yanbiao Ai
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Yi Hou
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Experimental Teaching & Management Center, Chongqing Medical University, Chongqing 401331, China
| | - Hong Chen
- Key Laboratory of Laboratory Medical Diagnosis, Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xiaojing Lin
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yunqi Zhang
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Min Ding
- Key Laboratory of Laboratory Medical Diagnosis, Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ting Luo
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jibin Li
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing 400016, China.
| | - Xinyu Li
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Pharmacy, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Xiaoqiu Xiao
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
196
|
Yu Y, Lin X, Feng F, Wei Y, Wei S, Gong Y, Guo C, Wang Q, Shuai P, Wang T, Qin H, Li G, Yi L. Gut microbiota and ionizing radiation-induced damage: Is there a link? ENVIRONMENTAL RESEARCH 2023; 229:115947. [PMID: 37080277 DOI: 10.1016/j.envres.2023.115947] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023]
Abstract
According to observational findings, ionizing radiation (IR) triggers dysbiosis of the intestinal microbiota, affecting the structural composition, function, and species of the gut microbiome and its metabolites. These modifications can further exacerbate IR-induced damage and amplify proinflammatory immune responses. Conversely, commensal bacteria and favorable metabolites can remodel the IR-disturbed gut microbial structure, promote a balance between anti-inflammatory and proinflammatory mechanisms in the body, and mitigate IR toxicity. The discovery of effective and safe remedies to prevent and treat radiation-induced injuries is vitally needed because of the proliferation of radiation toxicity threats produced by recent radiological public health disasters and increasing medical exposures. This review examines how the gut microbiota and its metabolites are linked to the processes of IR-induced harm. We highlight protective measures based on interventions with gut microbes to optimize the distress caused by IR damage to human health. We offer prospects for research in emerging and promising areas targeting the prevention and treatment of IR-induced damage.
Collapse
Affiliation(s)
- Yueqiu Yu
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiang Lin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Feiyang Feng
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yuanyun Wei
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Shuang Wei
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yaqi Gong
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Caimao Guo
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Qingyu Wang
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Peimeng Shuai
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Tiantian Wang
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Hui Qin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Guoqing Li
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Lan Yi
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
197
|
Zuber A, Peric A, Pluchino N, Baud D, Stojanov M. Human Male Genital Tract Microbiota. Int J Mol Sci 2023; 24:ijms24086939. [PMID: 37108103 PMCID: PMC10139050 DOI: 10.3390/ijms24086939] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
The human body is vastly colonised by microorganisms, whose impact on health is increasingly recognised. The human genital tract hosts a diverse microbiota, and an increasing number of studies on the male genital tract microbiota suggest that bacteria have a role in male infertility and pathological conditions, such as prostate cancer. Nevertheless, this research field remains understudied. The study of bacterial colonisation of the male genital tract is highly impacted by the invasive nature of sampling and the low abundance of the microbiota. Therefore, most studies relied on the analysis of semen microbiota to describe the colonisation of the male genital tract (MGT), which was thought to be sterile. The aim of this narrative review is to present the results of studies that used next-generation sequencing (NGS) to profile the bacterial colonisation patterns of different male genital tract anatomical compartments and critically highlight their findings and their weaknesses. Moreover, we identified potential research axes that may be crucial for our understanding of the male genital tract microbiota and its impact on male infertility and pathophysiology.
Collapse
Affiliation(s)
- Arnaud Zuber
- Materno-fetal and Obstetrics Research Unit, Department Woman-Mother-Child, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Adriana Peric
- 360° Fertility Center Zurich, 8702 Zollikon, Switzerland
| | - Nicola Pluchino
- Fertility Medicine and Gynaecological Endocrinology Unit, Department Woman-Mother-Child, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - David Baud
- Materno-fetal and Obstetrics Research Unit, Department Woman-Mother-Child, Lausanne University Hospital, 1011 Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
| | - Milos Stojanov
- Materno-fetal and Obstetrics Research Unit, Department Woman-Mother-Child, Lausanne University Hospital, 1011 Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
| |
Collapse
|
198
|
Xia C, Su J, Liu C, Mai Z, Yin S, Yang C, Fu L. Human microbiomes in cancer development and therapy. MedComm (Beijing) 2023; 4:e221. [PMID: 36860568 PMCID: PMC9969057 DOI: 10.1002/mco2.221] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/25/2023] [Accepted: 02/01/2023] [Indexed: 03/03/2023] Open
Abstract
Colonies formed by bacteria, archaea, fungi, and viral groups and their genomes, metabolites, and expressed proteins constitute complex human microbiomes. An increasing evidences showed that carcinogenesis and disease progression were link to microbiomes. Different organ sources, their microbial species, and their metabolites are different; the mechanisms of carcinogenic or procancerous are also different. Here, we summarize how microbiomes contribute to carcinogenesis and disease progression in cancers of the skin, mouth, esophagus, lung, gastrointestinal, genital, blood, and lymph malignancy. We also insight into the molecular mechanisms of triggering, promoting, or inhibiting carcinogenesis and disease progress induced by microbiomes or/and their secretions of bioactive metabolites. And then, the strategies of application of microorganisms in cancer treatment were discussed in detail. However, the mechanisms by which human microbiomes function are still poorly understood. The bidirectional interactions between microbiotas and endocrine systems need to be clarified. Probiotics and prebiotics are believed to benefit human health via a variety of mechanisms, in particular, in tumor inhibition. It is largely unknown how microbial agents cause cancer or how cancer progresses. We expect this review may open new perspectives on possible therapeutic approaches of patients with cancer.
Collapse
Affiliation(s)
- Chenglai Xia
- Affiliated Foshan Maternity and Chlid Healthcare HospitalSouthern Medical University, Foshan, China; School of Pharmaceutical Sciences, Southern Medical UniversityGuangzhouChina
| | - Jiyan Su
- Affiliated Foshan Maternity and Chlid Healthcare HospitalSouthern Medical University, Foshan, China; School of Pharmaceutical Sciences, Southern Medical UniversityGuangzhouChina
| | - Can Liu
- Affiliated Foshan Maternity and Chlid Healthcare HospitalSouthern Medical University, Foshan, China; School of Pharmaceutical Sciences, Southern Medical UniversityGuangzhouChina
| | - Zhikai Mai
- Affiliated Foshan Maternity and Chlid Healthcare HospitalSouthern Medical University, Foshan, China; School of Pharmaceutical Sciences, Southern Medical UniversityGuangzhouChina
| | - Shuanghong Yin
- Affiliated Foshan Maternity and Chlid Healthcare HospitalSouthern Medical University, Foshan, China; School of Pharmaceutical Sciences, Southern Medical UniversityGuangzhouChina
| | - Chuansheng Yang
- Department of Head‐Neck and Breast SurgeryYuebei People's Hospital of Shantou UniversityShaoguanChina
| | - Liwu Fu
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute; Sun Yat‐sen University Cancer CenterGuangzhouPeople's Republic of China
| |
Collapse
|
199
|
Abstract
Abstract The gut has been hypothesized to be the "motor" of multiple organ dysfunction in sepsis. Although there are multiple ways in which the gut can drive systemic inflammation, increasing evidence suggests that the intestinal microbiome plays a more substantial role than previously appreciated. An English language literature review was performed to summarize the current knowledge of sepsis-induced gut microbiome dysbiosis. Conversion of a normal microbiome to a pathobiome in the setting of sepsis is associated with worsened mortality. Changes in microbiome composition and diversity signal the intestinal epithelium and immune system resulting in increased intestinal permeability and a dysregulated immune response to sepsis. Clinical approaches to return to microbiome homeostasis may be theoretically possible through a variety of methods including probiotics, prebiotics, fecal microbial transplant, and selective decontamination of the digestive tract. However, more research is required to determine the efficacy (if any) of targeting the microbiome for therapeutic gain. The gut microbiome rapidly loses diversity with emergence of virulent bacteria in sepsis. Restoring normal commensal bacterial diversity through various therapies may be an avenue to improve sepsis mortality.
Collapse
Affiliation(s)
- Nathan J. Klingensmith
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Craig M. Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
200
|
Pinto C, Aluai-Cunha C, Santos A. The human and animals' malignant melanoma: comparative tumor models and the role of microbiome in dogs and humans. Melanoma Res 2023; 33:87-103. [PMID: 36662668 DOI: 10.1097/cmr.0000000000000880] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Currently, the most progressively occurring incident cancer is melanoma. The mouse is the most popular model in human melanoma research given its various benefits as a laboratory animal. Nevertheless, unlike humans, mice do not develop melanoma spontaneously, so they need to be genetically manipulated. In opposition, there are several reports of other animals, ranging from wild to domesticated animals, that spontaneously develop melanoma and that have cancer pathways that are similar to those of humans. The influence of the gut microbiome on health and disease is being the aim of many recent studies. It has been proven that the microbiome is a determinant of the host's immune status and disease prevention. In human medicine, there is increasing evidence that changes in the microbiome influences malignant melanoma progression and response to therapy. There are several similarities between some animals and human melanoma, especially between canine and human oral malignant melanoma as well as between the gut microbiome of both species. However, microbiome studies are scarce in veterinary medicine, especially in the oncology field. Future studies need to address the relevance of gut and tissue microbiome for canine malignant melanoma development, which results will certainly benefit both species in the context of translational medicine.
Collapse
Affiliation(s)
- Catarina Pinto
- Department of Veterinary Clinics, Institute of Biomedical Sciences Abel Salazar of the University of Porto (ICBAS-UP)
| | - Catarina Aluai-Cunha
- Department of Veterinary Clinics, Institute of Biomedical Sciences Abel Salazar of the University of Porto (ICBAS-UP)
| | - Andreia Santos
- Department of Veterinary Clinics, Institute of Biomedical Sciences Abel Salazar of the University of Porto (ICBAS-UP)
- Animal Science and Study Centre (CECA), Food and Agragrian Sciences and Technologies Institute (ICETA), Apartado, Porto, Portugal
| |
Collapse
|