101
|
Davies R, Minhas S, Jayasena CN. Next-Generation Sequencing to Elucidate the Semen Microbiome in Male Reproductive Disorders. MEDICINA (KAUNAS, LITHUANIA) 2023; 60:25. [PMID: 38256286 PMCID: PMC10819355 DOI: 10.3390/medicina60010025] [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: 11/12/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024]
Abstract
Mean sperm counts are declining at an accelerated rate and infertility is increasingly becoming a public health concern. It is now understood that human semen, previously considered to be sterile, harbours its own specific microbiome. Via activated leucocytes and the generation of reactive oxygen species, bacteria have the capability of evoking an immune response which may lead to sperm damage. Men with infertility have higher rates of both reactive oxygen species and sperm DNA damage. Due to the lack of sensitivity of routine culture and PCR-based methods, next-generation sequencing technology is being employed to characterise the seminal microbiome. There is a mounting body of studies that share a number of similarities but also a great range of conflicting findings. A lack of stringent decontamination procedures, small sample sizes and heterogeneity in other aspects of methodology makes it difficult to draw firm conclusions from these studies. However, various themes have emerged and evidence of highly conserved clusters of common bacteria can be seen. Depletion or over-representation of specific bacteria may be associated with aberrations in traditional and functional seminal parameters. Currently, the evidence is too limited to inform clinical practice and larger studies are needed.
Collapse
Affiliation(s)
- Rhianna Davies
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0HS, UK;
| | - Suks Minhas
- Department of Urology, Charing Cross Hospital, Imperial College NHS Trust, London W6 8RF, UK;
| | - Channa N. Jayasena
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0HS, UK;
| |
Collapse
|
102
|
Xu H, Yi X, Cui Z, Li H, Zhu L, Zhang L, Chen J, Fan X, Zhou P, Li MJ, Yu Y, Liu Q, Huang D, Yao Z, Zhou J. Maternal antibiotic exposure enhances ILC2 activation in neonates via downregulation of IFN1 signaling. Nat Commun 2023; 14:8332. [PMID: 38097561 PMCID: PMC10721923 DOI: 10.1038/s41467-023-43903-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 11/23/2023] [Indexed: 12/17/2023] Open
Abstract
Microbiota have an important function in shaping and priming neonatal immunity, although the cellular and molecular mechanisms underlying these effects remain obscure. Here we report that prenatal antibiotic exposure causes significant elevation of group 2 innate lymphoid cells (ILC2s) in neonatal lungs, in both cell numbers and functionality. Downregulation of type 1 interferon signaling in ILC2s due to diminished production of microbiota-derived butyrate represents the underlying mechanism. Mice lacking butyrate receptor GPR41 (Gpr41-/-) or type 1 interferon receptor IFNAR1 (Ifnar1-/-) recapitulate the phenotype of neonatal ILC2s upon maternal antibiotic exposure. Furthermore, prenatal antibiotic exposure induces epigenetic changes in ILC2s and has a long-lasting deteriorative effect on allergic airway inflammation in adult offspring. Prenatal supplementation of butyrate ameliorates airway inflammation in adult mice born to antibiotic-exposed dams. These observations demonstrate an essential role for the microbiota in the control of type 2 innate immunity at the neonatal stage, which suggests a therapeutic window for treating asthma in early life.
Collapse
Affiliation(s)
- Haixu Xu
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xianfu Yi
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Zhaohai Cui
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Hui Li
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Lin Zhu
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Lijuan Zhang
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - JiaLe Chen
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xutong Fan
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Pan Zhou
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Mulin Jun Li
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ying Yu
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Qiang Liu
- Department of Neurology, Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Dandan Huang
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| | - Zhi Yao
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| | - Jie Zhou
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
103
|
Chalifour B, Holzhausen EA, Lim JJ, Yeo EN, Shen N, Jones DP, Peterson BS, Goran MI, Liang D, Alderete TL. The potential role of early life feeding patterns in shaping the infant fecal metabolome: implications for neurodevelopmental outcomes. NPJ METABOLIC HEALTH AND DISEASE 2023; 1:2. [PMID: 38299034 PMCID: PMC10828959 DOI: 10.1038/s44324-023-00001-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/24/2023] [Indexed: 02/02/2024]
Abstract
Infant fecal metabolomics can provide valuable insights into the associations of nutrition, dietary patterns, and health outcomes in early life. Breastmilk is typically classified as the best source of nutrition for nearly all infants. However, exclusive breastfeeding may not always be possible for all infants. This study aimed to characterize associations between levels of mixed breastfeeding and formula feeding, along with solid food consumption and the infant fecal metabolome at 1- and 6-months of age. As a secondary aim, we examined how feeding-associated metabolites may be associated with early life neurodevelopmental outcomes. Fecal samples were collected at 1- and 6-months, and metabolic features were assessed via untargeted liquid chromatography/high-resolution mass spectrometry. Feeding groups were defined at 1-month as 1) exclusively breastfed, 2) breastfed >50% of feedings, or 3) formula fed ≥50% of feedings. Six-month groups were defined as majority breastmilk (>50%) or majority formula fed (≥50%) complemented by solid foods. Neurodevelopmental outcomes were assessed using the Bayley Scales of Infant Development at 2 years. Changes in the infant fecal metabolome were associated with feeding patterns at 1- and 6-months. Feeding patterns were associated with the intensities of a total of 57 fecal metabolites at 1-month and 25 metabolites at 6-months, which were either associated with increased breastmilk or increased formula feeding. Most breastmilk-associated metabolites, which are involved in lipid metabolism and cellular processes like cell signaling, were associated with higher neurodevelopmental scores, while formula-associated metabolites were associated with lower neurodevelopmental scores. These findings offer preliminary evidence that feeding patterns are associated with altered infant fecal metabolomes, which may be associated with cognitive development later in life.
Collapse
Affiliation(s)
- Bridget Chalifour
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO USA
| | | | - Joseph J. Lim
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO USA
| | - Emily N. Yeo
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO USA
| | - Natalie Shen
- Rollins School of Public Health, Emory University, Atlanta, GA USA
| | - Dean P. Jones
- School of Medicine, Emory University, Atlanta, GA USA
| | | | | | - Donghai Liang
- Rollins School of Public Health, Emory University, Atlanta, GA USA
| | - Tanya L. Alderete
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO USA
| |
Collapse
|
104
|
Morella I, Negro M, Dossena M, Brambilla R, D'Antona G. Gut-muscle-brain axis: Molecular mechanisms in neurodegenerative disorders and potential therapeutic efficacy of probiotic supplementation coupled with exercise. Neuropharmacology 2023; 240:109718. [PMID: 37774944 DOI: 10.1016/j.neuropharm.2023.109718] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 09/13/2023] [Accepted: 09/16/2023] [Indexed: 10/01/2023]
Abstract
Increased longevity is often associated with age-related conditions. The most common neurodegenerative disorders in the older population are Alzheimer's disease (AD) and Parkinson's disease (PD), associated with progressive neuronal loss leading to functional and cognitive impairments. Although symptomatic treatments are available, there is currently no cure for these conditions. Gut dysbiosis has been involved in the pathogenesis of AD and PD, thus interventions targeting the "gut-brain axis" could potentially prevent or delay these pathologies. Recent evidence suggests that the skeletal muscle and the gut microbiota can affect each other via the "gut-muscle axis". Importantly, cognitive functions in AD and PD patients significantly benefit from physical activity. In this review, we aim to provide a comprehensive picture of the crosstalk between the brain, the skeletal muscle and the gut microbiota, introducing the concept of "gut-muscle-brain axis". Moreover, we discuss human and animal studies exploring the modulatory role of exercise and probiotics on cognition in AD and PD. Collectively, the findings presented here support the potential benefits of physical activity and probiotic supplementation in AD and PD. Further studies will be needed to develop targeted and multimodal strategies, including lifestyle changes, to prevent or delay the course of these pathologies.
Collapse
Affiliation(s)
- Ilaria Morella
- Neuroscience and Mental Health Innovation Institute, School of Biosciences, Cardiff University, Cardiff, UK
| | - Massimo Negro
- Centro di Ricerca Interdipartimentale Nelle Attività Motorie e Sportive (CRIAMS)-Sport Medicine Centre, University of Pavia, Voghera, Italy
| | - Maurizia Dossena
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Riccardo Brambilla
- Neuroscience and Mental Health Innovation Institute, School of Biosciences, Cardiff University, Cardiff, UK; Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Giuseppe D'Antona
- Centro di Ricerca Interdipartimentale Nelle Attività Motorie e Sportive (CRIAMS)-Sport Medicine Centre, University of Pavia, Voghera, Italy; Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy.
| |
Collapse
|
105
|
Beretta S, Apparicio M, Toniollo GH, Cardozo MV. The importance of the intestinal microbiota in humans and dogs in the neonatal period. Anim Reprod 2023; 20:e20230082. [PMID: 38026003 PMCID: PMC10681130 DOI: 10.1590/1984-3143-ar2023-0082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/01/2023] [Indexed: 12/01/2023] Open
Abstract
The neonatal period represents a critical stage for the establishment and development of the gut microbiota, which profoundly influences the future health trajectory of individuals. This review examines the importance of intestinal microbiota in humans and dogs, aiming to elucidate the distinct characteristics and variations in the composition between these two species. In humans, the intestinal microbiota contributes to several crucial physiological processes, including digestion, nutrient absorption, immune system development, and modulation of host metabolism. Dysbiosis, an imbalance or disruption of the gut microbial community, has been linked to various disorders, such as inflammatory bowel disease, obesity, and even neurological conditions. Furthermore, recent research has unveiled the profound influence of the gut-brain axis, emphasizing the bidirectional communication between the gut microbiota and the central nervous system, impacting cognitive function and mental health. Similarly, alterations in the canine intestinal microbiota have been associated with gastrointestinal disorders, including chronic enteropathy, such as inflammatory bowel disease, food allergies, and ulcerative histiocytic colitis. However, our understanding of the intricacies and functional significance of the intestinal microbiota in dogs remains limited. Understanding the complex dynamics of the intestinal microbiota in both humans and dogs is crucial for devising effective strategies to promote health and manage disease. Moreover, exploring the similarities and differences in the gut microbial composition between these two species can facilitate translational research, potentially leading to innovative therapeutic interventions and strategies to enhance the well-being of both humans and dogs.
Collapse
Affiliation(s)
- Samara Beretta
- Departamento de Patologia, Reprodução e Saúde Única, Faculdade de Ciências Agrárias e Veterinárias (FCAV), Universidade Estadual Paulista (UNESP), Jaboticabal, SP, Brasil
| | - Maricy Apparicio
- Departamento de Cirurgia Veterinária e Reprodução Animal, Faculdade de Medicina Veterinária e Zootecnia (FMVZ), Universidade Estadual Paulista (UNESP), Botucatu, SP, Brasil
| | - Gilson Hélio Toniollo
- Departamento de Patologia, Reprodução e Saúde Única, Faculdade de Ciências Agrárias e Veterinárias (FCAV), Universidade Estadual Paulista (UNESP), Jaboticabal, SP, Brasil
| | - Marita Vedovelli Cardozo
- Laboratório de Fisiologia de Microorganismos, Departamento de Ciências Biomédicas e Saúde, Universidade do Estado de Minas Gerais (UEMG), Passos, MG, Brasil
| |
Collapse
|
106
|
Switzer AD, Callahan BJ, Costello EK, Bik EM, Fontaine C, Gulland FM, Relman DA. Rookery through rehabilitation: Microbial community assembly in newborn harbour seals after maternal separation. Environ Microbiol 2023; 25:2182-2202. [PMID: 37329141 PMCID: PMC11180496 DOI: 10.1111/1462-2920.16444] [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: 09/05/2022] [Accepted: 05/22/2023] [Indexed: 06/18/2023]
Abstract
Microbial community assembly remains largely unexplored in marine mammals, despite its potential importance for conservation and management. Here, neonatal microbiota assembly was studied in harbour seals (Phoca vitulina richardii) at a rehabilitation facility soon after maternal separation, through weaning, to the time of release back to their native environment. We found that the gingival and rectal communities of rehabilitated harbour seals were distinct from the microbiotas of formula and pool water, and became increasingly diverse and dissimilar over time, ultimately resembling the gingival and rectal communities of local wild harbour seals. Harbour seal microbiota assembly was compared to that of human infants, revealing the rapid emergence of host specificity and evidence of phylosymbiosis even though these harbour seals had been raised by humans. Early life prophylactic antibiotics were associated with changes in the composition of the harbour seal gingival and rectal communities and surprisingly, with transient increases in alpha diversity, perhaps because of microbiota sharing during close cohabitation with other harbour seals. Antibiotic-associated effects dissipated over time. These results suggest that while early life maternal contact may provide seeding for microbial assembly, co-housing of conspecifics during rehabilitation may help neonatal mammals achieve a healthy host-specific microbiota with features of resilience.
Collapse
Affiliation(s)
- Alexandra D. Switzer
- Department of Medicine, School of Medicine, Stanford University, Stanford, CA, United States
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Benjamin J. Callahan
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Department of Statistics, Stanford University, Stanford, CA, United States
| | - Elizabeth K. Costello
- Department of Medicine, School of Medicine, Stanford University, Stanford, CA, United States
| | | | | | - Frances M.D. Gulland
- The Marine Mammal Center, Sausalito, CA, United States
- Wildlife Health Center, School of Veterinary Medicine, University of California at Davis, Davis, CA, United States
| | - David A. Relman
- Department of Medicine, School of Medicine, Stanford University, Stanford, CA, United States
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA, United States
- Infectious Diseases Section, VA Palo Alto Health Care System, Palo Alto, CA, United States
| |
Collapse
|
107
|
Ross SM. Microbiota and the Immune System, Part 1. Holist Nurs Pract 2023; 37:363-365. [PMID: 37851352 DOI: 10.1097/hnp.0000000000000620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Affiliation(s)
- Stephanie Maxine Ross
- College of Nursing and Health Professions, Drexel University, Philadelphia, Pennsylvania
| |
Collapse
|
108
|
Pichichero ME. Variability of vaccine responsiveness in early life. Cell Immunol 2023; 393-394:104777. [PMID: 37866234 DOI: 10.1016/j.cellimm.2023.104777] [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: 06/15/2023] [Revised: 09/18/2023] [Accepted: 10/14/2023] [Indexed: 10/24/2023]
Abstract
Vaccinations in early life elicit variable antibody and cellular immune responses, sometimes leaving fully vaccinated children unprotected against life-threatening infectious diseases. Specific immune cell populations and immune networks may have a critical period of development and calibration in a window of opportunity occurring during the first 100 days of early life. Among the early life determinants of vaccine responses, this review will focus on modifiable factors involving development of the infant microbiota and metabolome: antibiotic exposure, breast versus formula feeding, and Caesarian section versus vaginal delivery of newborns. How microbiota may serve as natural adjuvants for vaccine responses and how microbiota-derived metabolites influence vaccine responses are also reviewed. Early life poor vaccine responsiveness can be linked to increased infection susceptibility because both phenotypes share similar immunity dysregulation profiles. An early life pre-vaccination endotype, when interventions have the highest potential for success, should be sought that predicts vaccine response trajectories.
Collapse
Affiliation(s)
- Michael E Pichichero
- Center for Infectious Diseases and Immunology, Research Institute, Rochester General Hospital, 1425 Portland Ave, Rochester, NY 14621, USA.
| |
Collapse
|
109
|
Zhu R, Gao Y, Dong J, Li Z, Ren Z. The changes of gut microbiota and metabolites in different drug-induced liver injuries. J Med Microbiol 2023; 72. [PMID: 38015063 DOI: 10.1099/jmm.0.001778] [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] [Indexed: 11/29/2023] Open
Abstract
The increasing incidence of drug-induced liver injury (DILI) has become a major concern. Gut microbiota, as another organ of the human body, has been studied in various tumors, cardiovascular metabolic diseases, inflammatory bowel disease and human immunity. The studies mentioned above have confirmed its important impact on the occurrence and development of DILI. The gut-liver axis explains the close relationship between the gut and the liver, and it may be a pathway by which gut microbes contribute to DILI. In addition, the interaction between drugs and gut microbes affects both separately, which in turn may have positive or negative effects on the body, including DILI. There are both common and specific changes in liver injury caused by different drugs. The alteration of metabolites in DILI is also a new direction of therapeutic exploration. The application of microbiomics, metabolomics and other multi-omics to DILI has also explored new ideas for DILI. In this review, we conclude the alterations of gut microbes and metabolites under different DILI, and the significance of applying gut microbiome-metabolomics to DILI, so as to explore the metabolic characteristics of DILI and possible novel metabolic biomarkers.
Collapse
Affiliation(s)
- Ruirui Zhu
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250000, PR China
- Gene Hospital of Henan Province; Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Yinghui Gao
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
- Gene Hospital of Henan Province; Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Jianxia Dong
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
- Gene Hospital of Henan Province; Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Zhiqin Li
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
- Gene Hospital of Henan Province; Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Zhigang Ren
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250000, PR China
- Gene Hospital of Henan Province; Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| |
Collapse
|
110
|
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
|
111
|
Zaongo SD, Harypursat V, Rashid F, Dahourou DL, Ouedraogo AS, Chen Y. Influence of HIV infection on cognition and overall intelligence in HIV-infected individuals: advances and perspectives. Front Behav Neurosci 2023; 17:1261784. [PMID: 37953826 PMCID: PMC10637382 DOI: 10.3389/fnbeh.2023.1261784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/10/2023] [Indexed: 11/14/2023] Open
Abstract
It is now well understood that HIV-positive individuals, even those under effective ART, tend to develop a spectrum of cognitive, motor, and/or mood conditions which are contemporarily referred to as HIV-associated neurocognitive disorder (HAND), and which is directly related to HIV-1 infection and HIV-1 replication in the central nervous system (CNS). As HAND is known to induce difficulties associated with attention, concentration, and memory, it is thus legitimate and pertinent to speculate upon the possibility that HIV infection may well influence human cognition and intelligence. We therefore propose herein to review the concept of intelligence, the concept of cells of intelligence, the influence of HIV on these particular cells, and the evidence pointing to differences in observed intelligence quotient (IQ) scores between HIV-positive and HIV-negative individuals. Additionally, cumulative research evidence continues to draw attention to the influence of the gut on human intelligence. Up to now, although it is known that HIV infection profoundly alters both the composition and diversity of the gut microbiota and the structural integrity of the gut, the influence of the gut on intelligence in the context of HIV infection remains poorly described. As such, we also provide herein a review of the different ways in which HIV may influence human intelligence via the gut-brain axis. Finally, we provide a discourse on perspectives related to HIV and human intelligence which may assist in generating more robust evidence with respect to this issue in future studies. Our aim is to provide insightful knowledge for the identification of novel areas of investigation, in order to reveal and explain some of the enigmas related to HIV infection.
Collapse
Affiliation(s)
- Silvere D. Zaongo
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Vijay Harypursat
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Farooq Rashid
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Désiré Lucien Dahourou
- Département Biomédical/Santé Publique, Institut de Recherche en Sciences de la Santé/CNRST, Ouagadougou, Burkina Faso
| | - Abdoul-Salam Ouedraogo
- Centre Muraz, Bobo-Dioulasso, Burkina Faso
- Department of Bacteriology and Virology, Souro Sanou University Hospital, Bobo-Dioulasso, Burkina Faso
| | - Yaokai Chen
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| |
Collapse
|
112
|
Raman V, Deshpande CP, Khanduja S, Howell LM, Van Dessel N, Forbes NS. Build-a-bug workshop: Using microbial-host interactions and synthetic biology tools to create cancer therapies. Cell Host Microbe 2023; 31:1574-1592. [PMID: 37827116 DOI: 10.1016/j.chom.2023.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/16/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023]
Abstract
Many systemically administered cancer therapies exhibit dose-limiting toxicities that reduce their effectiveness. To increase efficacy, bacterial delivery platforms have been developed that improve safety and prolong treatment. Bacteria are a unique class of therapy that selectively colonizes most solid tumors. As delivery vehicles, bacteria have been genetically modified to express a range of therapies that match multiple cancer indications. In this review, we describe a modular "build-a-bug" method that focuses on five design characteristics: bacterial strain (chassis), therapeutic compound, delivery method, immune-modulating features, and genetic control circuits. We emphasize how fundamental research into gut microbe pathogenesis has created safe bacterial therapies, some of which have entered clinical trials. The genomes of gut microbes are fertile grounds for discovery of components to improve delivery and modulate host immune responses. Future work coupling these delivery vehicles with insights from gut microbes could lead to the next generation of microbial cancer therapy.
Collapse
Affiliation(s)
- Vishnu Raman
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA; Ernest Pharmaceuticals, LLC, Hadley, MA, USA
| | - Chinmay P Deshpande
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
| | - Shradha Khanduja
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
| | - Lars M Howell
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
| | | | - Neil S Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA; Molecular and Cell Biology Program, University of Massachusetts, Amherst, Amherst, MA, USA; Institute for Applied Life Science, University of Massachusetts, Amherst, Amherst, MA, USA.
| |
Collapse
|
113
|
Beharry KD, Latkowska M, Valencia AM, Allana A, Soto J, Cai CL, Golombek S, Hand I, Aranda JV. Factors Influencing Neonatal Gut Microbiome and Health with a Focus on Necrotizing Enterocolitis. Microorganisms 2023; 11:2528. [PMID: 37894186 PMCID: PMC10608807 DOI: 10.3390/microorganisms11102528] [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: 06/30/2023] [Revised: 09/21/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Maturational changes in the gut start in utero and rapidly progress after birth, with some functions becoming fully developed several months or years post birth including the acquisition of a full gut microbiome, which is made up of trillions of bacteria of thousands of species. Many factors influence the normal development of the neonatal and infantile microbiome, resulting in dysbiosis, which is associated with various interventions used for neonatal morbidities and survival. Extremely low gestational age neonates (<28 weeks' gestation) frequently experience recurring arterial oxygen desaturations, or apneas, during the first few weeks of life. Apnea, or the cessation of breathing lasting 15-20 s or more, occurs due to immature respiratory control and is commonly associated with intermittent hypoxia (IH). Chronic IH induces oxygen radical diseases of the neonate, including necrotizing enterocolitis (NEC), the most common and devastating gastrointestinal disease in preterm infants. NEC is associated with an immature intestinal structure and function and involves dysbiosis of the gut microbiome, inflammation, and necrosis of the intestinal mucosal layer. This review describes the factors that influence the neonatal gut microbiome and dysbiosis, which predispose preterm infants to NEC. Current and future management and therapies, including the avoidance of dysbiosis, the use of a human milk diet, probiotics, prebiotics, synbiotics, restricted antibiotics, and fecal transplantation, for the prevention of NEC and the promotion of a healthy gut microbiome are also reviewed. Interventions directed at boosting endogenous and/or exogenous antioxidant supplementation may not only help with prevention, but may also lessen the severity or shorten the course of the disease.
Collapse
Affiliation(s)
- Kay D. Beharry
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (M.L.); (C.L.C.); (S.G.); (J.V.A.)
| | - Magdalena Latkowska
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (M.L.); (C.L.C.); (S.G.); (J.V.A.)
| | - Arwin M. Valencia
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Saddleback Memorial Medical Center, Laguna Hills, CA 92653, USA;
| | - Ahreen Allana
- Department of Pediatrics, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (A.A.); (J.S.)
| | - Jatnna Soto
- Department of Pediatrics, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (A.A.); (J.S.)
| | - Charles L. Cai
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (M.L.); (C.L.C.); (S.G.); (J.V.A.)
| | - Sergio Golombek
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (M.L.); (C.L.C.); (S.G.); (J.V.A.)
| | - Ivan Hand
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Kings County Hospital Center, Brooklyn, NY 11203, USA;
| | - Jacob V. Aranda
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (M.L.); (C.L.C.); (S.G.); (J.V.A.)
| |
Collapse
|
114
|
Sucu S, Basarir KE, Mihaylov P, Balik E, Lee JTC, Fridell JA, Emamaullee JA, Ekser B. Impact of gut microbiota on liver transplantation. Am J Transplant 2023; 23:1485-1495. [PMID: 37277064 DOI: 10.1016/j.ajt.2023.05.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/26/2023] [Accepted: 05/24/2023] [Indexed: 06/07/2023]
Abstract
The gut microbiota has been gaining attention due to its interactions with the human body and its role in pathophysiological processes. One of the main interactions is the "gut-liver axis," in which disruption of the gut mucosal barrier seen in portal hypertension and liver disease can influence liver allograft function over time. For example, in patients who are undergoing liver transplantation, preexisting dysbiosis, perioperative antibiotic use, surgical stress, and immunosuppressive use have each been associated with alterations in gut microbiota, potentially impacting overall morbidity and mortality. In this review, studies exploring gut microbiota changes in patients undergoing liver transplantation are reviewed, including both human and experimental animal studies. Common themes include an increase in Enterobacteriaceae and Enterococcaceae species and a decrease in Faecalibacterium prausnitzii and Bacteriodes, while a decrease in the overall diversity of gut microbiota after liver transplantation.
Collapse
Affiliation(s)
- Serkan Sucu
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA; Department of Surgery, Koc University School of Medicine, Istanbul, Turkey
| | - Kerem E Basarir
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Plamen Mihaylov
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Emre Balik
- Department of Surgery, Koc University School of Medicine, Istanbul, Turkey
| | - Jason T C Lee
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA; Division of Abdominal Organ Transplantation and Hepatobiliary Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jonathan A Fridell
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Juliet A Emamaullee
- Division of Abdominal Organ Transplantation and Hepatobiliary Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Burcin Ekser
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|
115
|
Algrafi AS, Jamal AA, Ismaeel DM. Microbiota as a New Target in Cancer Pathogenesis and Treatment. Cureus 2023; 15:e47072. [PMID: 38021696 PMCID: PMC10645418 DOI: 10.7759/cureus.47072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2023] [Indexed: 12/01/2023] Open
Abstract
The microbial ecosystem of humans is an integral part of human health and disease. A significant percentage of tumors worldwide are thought to be microbially induced. The relationship between cancer and microbes is complex. In this article review, we aim to give an overview of human microbiota and its role in carcinogenesis, emphasize the relation between microbiota and cancer immunity, and highlight its role in the future of cancer therapy. The term microbiota refers to the collection of microorganisms that are located in an individual, whereas the total genome of these microorganisms is referred to as the microbiome. The microbiota in humans has many physiological functions. The microbiota within the gut lumen has a profound effect on the local and systemic immune system. The immune system can change the gut microbiota. Microbiota may induce carcinogenesis by several mechanisms. It also affects tumor progression. Thus, microbiota modulation may aid in the prevention and treatment of cancer. Intentionally introducing microorganisms into the oncological patient is assumed to mobilize the immune system to become able to, at least, limit the development of cancer. Microbes are used as vectors which are carriers of particular antineoplastic agents that reduce the side effects of chemotherapy. Inflammation and tumor microenvironment play an essential role in promoting chemo-resistance. There is now considerable evidence, both in humans as well as in laboratory animals, that the commensal microbiota has important effects on carcinogenesis, tumor growth, and therapy response.
Collapse
Affiliation(s)
- Abeer S Algrafi
- Internal Medicine, College of Medicine, Taibah University, Madinah, SAU
| | - Aisha A Jamal
- General Practice, College of Medicine, Taibah University, Madinah, SAU
| | - Dana M Ismaeel
- General Practice, College of Medicine, Taibah University, Madinah, SAU
| |
Collapse
|
116
|
Ameer A, Cheng Y, Saleem F, Uzma, McKenna A, Richmond A, Gundogdu O, Sloan WT, Javed S, Ijaz UZ. Temporal stability and community assembly mechanisms in healthy broiler cecum. Front Microbiol 2023; 14:1197838. [PMID: 37779716 PMCID: PMC10534011 DOI: 10.3389/fmicb.2023.1197838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/21/2023] [Indexed: 10/03/2023] Open
Abstract
In recent years, there has been an unprecedented advancement in in situ analytical approaches that contribute to the mechanistic understanding of microbial communities by explicitly incorporating ecology and studying their assembly. In this study, we have analyzed the temporal profiles of the healthy broiler cecal microbiome from day 3 to day 35 to recover the stable and varying components of microbial communities. During this period, the broilers were fed three different diets chronologically, and therefore, we have recovered signature microbial species that dominate during each dietary regime. Since broilers were raised in multiple pens, we have also parameterized these as an environmental condition to explore microbial niches and their overlap. All of these analyses were performed in view of different parameters such as body weight (BW-mean), feed intake (FI), feed conversion ratio (FCR), and age (days) to link them to a subset of microbes that these parameters have a bearing upon. We found that gut microbial communities exhibited strong and statistically significant specificity for several environmental variables. Through regression models, genera that positively/negatively correlate with the bird's age were identified. Some short-chain fatty acids (SCFAs)-producing bacteria, including Izemoplasmatales, Gastranaerophilales, and Roseburia, have a positive correlation with age. Certain pathogens, such as Escherichia-Shigella, Sporomusa, Campylobacter, and Enterococcus, negatively correlated with the bird's age, which indicated a high disease risk in the initial days. Moreover, the majority of pathways involved in amino acid biosynthesis were also positively correlated with the bird's age. Some probiotic genera associated with improved performance included Oscillospirales; UCG-010, Shuttleworthia, Bifidobacterium, and Butyricicoccaceae; UCG-009. In general, predicted antimicrobial resistance genes (piARGs) contributed at a stable level, but there was a slight increase in abundance when the diet was changed. To the best of the authors' knowledge, this is one of the first studies looking at the stability, complexity, and ecology of natural broiler microbiota development in a temporal setting.
Collapse
Affiliation(s)
- Aqsa Ameer
- Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Youqi Cheng
- Water and Environment Research Group, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Farrukh Saleem
- Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Uzma
- Water and Environment Research Group, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, United Kingdom
| | | | | | - Ozan Gundogdu
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - William T. Sloan
- Water and Environment Research Group, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Sundus Javed
- Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Umer Zeeshan Ijaz
- Water and Environment Research Group, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, United Kingdom
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
- College of Science and Engineering, University of Galway, Galway, Ireland
| |
Collapse
|
117
|
Park H, Park NY, Koh A. Scarring the early-life microbiome: its potential life-long effects on human health and diseases. BMB Rep 2023; 56:469-481. [PMID: 37605613 PMCID: PMC10547969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/30/2023] [Accepted: 08/14/2023] [Indexed: 08/23/2023] Open
Abstract
The gut microbiome is widely recognized as a dynamic organ with a profound influence on human physiology and pathology. Extensive epidemiological and longitudinal cohort studies have provided compelling evidence that disruptions in the early-life microbiome can have long-lasting health implications. Various factors before, during, and after birth contribute to shaping the composition and function of the neonatal and infant microbiome. While these alterations can be partially restored over time, metabolic phenotypes may persist, necessitating research to identify the critical period for early intervention to achieve phenotypic recovery beyond microbiome composition. In this review, we provide current understanding of changes in the gut microbiota throughout life and the various factors affecting these changes. Specifically, we highlight the profound impact of early-life gut microbiota disruption on the development of diseases later in life and discuss perspectives on efforts to recover from such disruptions. [BMB Reports 2023; 56(9): 469-481].
Collapse
Affiliation(s)
- Hyunji Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Na-Young Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Ara Koh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
- Institute of Convergence Science, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
118
|
Pantazi AC, Balasa AL, Mihai CM, Chisnoiu T, Lupu VV, Kassim MAK, Mihai L, Frecus CE, Chirila SI, Lupu A, Andrusca A, Ionescu C, Cuzic V, Cambrea SC. Development of Gut Microbiota in the First 1000 Days after Birth and Potential Interventions. Nutrients 2023; 15:3647. [PMID: 37630837 PMCID: PMC10457741 DOI: 10.3390/nu15163647] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
The first 1000 days after birth represent a critical window for gut microbiome development, which is essential for immune system maturation and overall health. The gut microbiome undergoes major changes during this period due to shifts in diet and environment. Disruptions to the microbiota early in life can have lasting health effects, including increased risks of inflammatory disorders, autoimmune diseases, neurological disorders, and obesity. Maternal and environmental factors during pregnancy and infancy shape the infant gut microbiota. In this article, we will review how maintaining a healthy gut microbiome in pregnancy and infancy is important for long-term infant health. Furthermore, we briefly include fungal colonization and its effects on the host immune function, which are discussed as part of gut microbiome ecosystem. Additionally, we will describe how potential approaches such as hydrogels enriched with prebiotics and probiotics, gut microbiota transplantation (GMT) during pregnancy, age-specific microbial ecosystem therapeutics, and CRISPR therapies targeting the gut microbiota hold potential for advancing research and development. Nevertheless, thorough evaluation of their safety, effectiveness, and lasting impacts is crucial prior to their application in clinical approach. The article emphasizes the need for continued research to optimize gut microbiota and immune system development through targeted early-life interventions.
Collapse
Affiliation(s)
- Alexandru Cosmin Pantazi
- Pediatrics Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (A.L.B.)
- Pediatrics Department, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Adriana Luminita Balasa
- Pediatrics Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (A.L.B.)
- Pediatrics Department, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Cristina Maria Mihai
- Pediatrics Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (A.L.B.)
- Pediatrics Department, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Tatiana Chisnoiu
- Pediatrics Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (A.L.B.)
- Pediatrics Department, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Vasile Valeriu Lupu
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | | | - Larisia Mihai
- Pediatrics Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (A.L.B.)
- Pediatrics Department, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Corina Elena Frecus
- Pediatrics Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (A.L.B.)
- Pediatrics Department, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | | | - Ancuta Lupu
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Antonio Andrusca
- Pediatrics Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (A.L.B.)
- Pediatrics Department, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Constantin Ionescu
- Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (S.I.C.)
| | - Viviana Cuzic
- Pediatrics Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (A.L.B.)
- Pediatrics Department, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Simona Claudia Cambrea
- Infectious Diseases Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania
| |
Collapse
|
119
|
Seradj DS, Beeck R, Haase A, Krause J, Schick P, Weitschies W. Influence of Different Diets on the Degradation of Sulfasalazine by Colon Bacteria Determined Using MimiCol 3. Pharmaceuticals (Basel) 2023; 16:1128. [PMID: 37631043 PMCID: PMC10459966 DOI: 10.3390/ph16081128] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/04/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
The microbiome of the colon is characterized by its great diversity. This varies not only intra- but also interindividually and is influenced by endogenous and exogenous factors, such as dietary and lifestyle factors. The aim of this work was to investigate the extent to which the degradation of the drug sulfasalazine is influenced by different microbiota. Therefore, the in vitro model MimiCol3 was used, which represents the physiological conditions of the ascending colon. In addition to a representative physiological volume, the pH value, redox potential and an anaerobic atmosphere are important to provide the bacteria with the best possible growth conditions. Stool samples were taken from three healthy subjects, comparing omnivorous, vegetarian and meat-rich diets, and cultured for 24 h. However, the nutrient medium used for cultivation led to the alignment of the bacterial composition of the microbiota. The previously observed differences between the diets could not be maintained. Nevertheless, the similar degradation of sulfasalazine was observed in all microbiota studied in MimiCol3. This makes MimiCol3 a suitable in vitro model for metabolism studies in the gut microbiome.
Collapse
Affiliation(s)
| | | | | | | | | | - Werner Weitschies
- Center of Drug Absorption and Transport, Department of Biopharmaceutics and Pharmaceutical Technology, University of Greifswald, Felix-Hausdorff-Str. 3, D-17489 Greifswald, Germany; (D.-S.S.); (R.B.); (A.H.); (J.K.); (P.S.)
| |
Collapse
|
120
|
Rodriguez MT, McLaurin KA, Shtutman M, Kubinak JL, Mactutus CF, Booze RM. Therapeutically targeting the consequences of HIV-1-associated gastrointestinal dysbiosis: Implications for neurocognitive and affective alterations. Pharmacol Biochem Behav 2023; 229:173592. [PMID: 37390973 PMCID: PMC10494709 DOI: 10.1016/j.pbb.2023.173592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
Approximately 50 % of the individuals living with human immunodeficiency virus type 1 (HIV-1) are plagued by debilitating neurocognitive impairments (NCI) and/or affective alterations. Sizeable alterations in the composition of the gut microbiome, or gastrointestinal dysbiosis, may underlie, at least in part, the NCI, apathy, and/or depression observed in this population. Herein, two interrelated aims will be critically addressed, including: 1) the evidence for, and functional implications of, gastrointestinal microbiome dysbiosis in HIV-1 seropositive individuals; and 2) the potential for therapeutically targeting the consequences of this dysbiosis for the treatment of HIV-1-associated NCI and affective alterations. First, gastrointestinal microbiome dysbiosis in HIV-1 seropositive individuals is characterized by decreased alpha (α) diversity, a decreased relative abundance of bacterial species belonging to the Bacteroidetes phylum, and geographic-specific alterations in Bacillota (formerly Firmicutes) spp. Fundamentally, changes in the relative abundance of Bacteroidetes and Bacillota spp. may underlie, at least in part, the deficits in γ-aminobutyric acid and serotonin neurotransmission, as well as prominent synaptodendritic dysfunction, observed in this population. Second, there is compelling evidence for the therapeutic utility of targeting synaptodendritic dysfunction as a method to enhance neurocognitive function and improve motivational dysregulation in HIV-1. Further research is needed to determine whether the therapeutics enhancing synaptic efficacy exert their effects by altering the gut microbiome. Taken together, understanding gastrointestinal microbiome dysbiosis resulting from chronic HIV-1 viral protein exposure may afford insight into the mechanisms underlying HIV-1-associated neurocognitive and/or affective alterations; mechanisms which can be subsequently targeted via novel therapeutics.
Collapse
Affiliation(s)
- Mason T Rodriguez
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, 1512 Pendleton Street, University of South Carolina, Columbia, SC 29208, United States of America
| | - Kristen A McLaurin
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, 1512 Pendleton Street, University of South Carolina, Columbia, SC 29208, United States of America
| | - Michael Shtutman
- Drug Discovery and Biomedical Sciences, College of Pharmacy, 715 Sumter Street, University of South Carolina, Columbia, SC 29208, United States of America
| | - Jason L Kubinak
- Pathology, Microbiology & Immunology, School of Medicine Columbia, 6311 Garners Ferry Road, Building 2, Columbia, SC 29209, United States of America
| | - Charles F Mactutus
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, 1512 Pendleton Street, University of South Carolina, Columbia, SC 29208, United States of America
| | - Rosemarie M Booze
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, 1512 Pendleton Street, University of South Carolina, Columbia, SC 29208, United States of America.
| |
Collapse
|
121
|
Enya Y, Hiramatsu H, Ihira M, Suzuki R, Higashimoto Y, Funato Y, Kozawa K, Miura H, Miyata M, Kawamura Y, Ishihara T, Taniguchi K, Komoto S, Yoshikawa T. Similarities in rotavirus vaccine viral shedding and immune responses in pairs of twins. FUJITA MEDICAL JOURNAL 2023; 9:253-258. [PMID: 37554946 PMCID: PMC10405898 DOI: 10.20407/fmj.2022-039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 12/14/2022] [Indexed: 08/10/2023]
Abstract
OBJECTIVES Intestinal rotavirus (RV) vaccine replication and host immune response are suggested to be affected by several factors, including maternal antibodies, breastfeeding history, and gut microbiome, which are thought to be similar in pairs of twins. The aim of this study was to determine whether viral shedding from the fecal RV vaccine strain Rotarix® (RV1) and IgG and IgA responses to RV show similarity in pairs of twins. METHODS Quantitative reverse transcription polymerase chain reaction specific to RV vaccine strain RV1 was used to monitor fecal RV1 viral shedding. RV IgG and IgA titers were measured using an in-house enzyme-linked immunosorbent assay. Fecal RV1 viral shedding and immune responses were compared between twins and singletons with mixed effects and fixed effects models. RESULTS A total of 347 stool and 54 blood samples were collected from four pairs of twins and twelve singletons during the observation period. Although the kinetics of fecal RV1 viral shedding and immune responses differed among vaccinated individuals, they appeared to be similar within twin pairs. RV shedding after the first dose (P=0.049) and RV IgG titers during the entire observation period (P=0.015) had a significantly better fit in the fixed effect model that assumed that twins have the same response versus the model that assumed that twins have a different response. CONCLUSIONS The similarity of RV vaccine viral replication in intestine and host immune responses in twin pairs was demonstrated using statistical analysis.
Collapse
Affiliation(s)
- Yasuko Enya
- Faculty of Clinical Engineering, Fujita Health University, School of Medical Sciences, Toyoake, Aichi, Japan
| | - Hiroyuki Hiramatsu
- Department of Clinical Pharmacy, Fujita Health University Hospital, Toyoake, Aichi, Japan
| | - Masaru Ihira
- Faculty of Clinical Science for Biological Monitoring, Fujita Health University, School of Medical Sciences, Toyoake, Aichi, Japan
| | - Ryota Suzuki
- Department of Clinical Pharmacy, Fujita Health University Hospital, Toyoake, Aichi, Japan
| | - Yuki Higashimoto
- Faculty of Cellular and Molecular Biology, Fujita Health University, School of Medical Sciences, Toyoake, Aichi, Japan
| | - Yusuke Funato
- Department of Pediatrics, Fujita Health University, School of Medicine, Toyoake, Aichi, Japan
| | - Kei Kozawa
- Department of Pediatrics, Fujita Health University, School of Medicine, Toyoake, Aichi, Japan
| | - Hiroki Miura
- Department of Pediatrics, Fujita Health University, School of Medicine, Toyoake, Aichi, Japan
| | - Masafumi Miyata
- Department of Pediatrics, Fujita Health University, School of Medicine, Toyoake, Aichi, Japan
| | - Yoshiki Kawamura
- Department of Pediatrics, Fujita Health University, School of Medicine, Toyoake, Aichi, Japan
| | - Takuma Ishihara
- Innovative and Clinical Research Promotion Center, Gifu University Hospital, Gifu, Gifu, Japan
| | - Koki Taniguchi
- Department of Virology and Parasitology, Fujita Health University, School of Medicine, Toyoake, Aichi, Japan
| | - Satoshi Komoto
- Department of Virology and Parasitology, Fujita Health University, School of Medicine, Toyoake, Aichi, Japan
| | - Tetsushi Yoshikawa
- Department of Pediatrics, Fujita Health University, School of Medicine, Toyoake, Aichi, Japan
| |
Collapse
|
122
|
McKenna BG, Dunlop AL, Corwin E, Smith AK, Venkateswaran S, Brennan PA. Intergenerational and early life associations of the gut microbiome and stress-related symptomatology among Black American mothers and children. Brain Behav Immun Health 2023; 31:100651. [PMID: 37449285 PMCID: PMC10336162 DOI: 10.1016/j.bbih.2023.100651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/02/2023] [Accepted: 06/03/2023] [Indexed: 07/18/2023] Open
Abstract
Recent evidence suggests that maternal childhood adversity may have an intergenerational impact, with children of adversity-exposed mothers exhibiting elevated symptoms of psychopathology. At the same time, many children demonstrate resilience to these intergenerational effects. Among the variety of factors that likely contribute to resilience, the composition of the gut microbiome may play a role in buffering the negative impacts of trauma and stress. The current prospective cohort study tested the novel hypothesis that offspring gut microbiome composition is a potential moderator in the relationship between maternal exposure to childhood adversity and offspring symptomatology (i.e., internalizing, externalizing, and posttraumatic stress symptoms). Maternal childhood adversity was self-reported during pregnancy via the Childhood Trauma Questionnaire and Adverse Childhood Experiences (ACEs) survey, and offspring symptomatology was assessed with the Child Behavior Checklist/1.5-5 when offspring were 2-4 years old. Offspring fecal samples were collected between these timepoints (i.e., during 6- to 24-month follow-up visits) for microbiome sequencing. Results indicated that maternal ACEs and the relative abundances of Bifidobacterium, Lactobacillus, and Prevotella were associated with offspring symptomatology. However, there was little evidence that microbial abundance moderated the association between maternal adversity and offspring symptoms. Overall, these findings further our understanding of how the gut microbiome associates with psychopathology, and informs future studies aimed at targeting modifiable factors that may buffer the intergenerational effects of childhood adversity.
Collapse
Affiliation(s)
| | - Anne L. Dunlop
- School of Nursing, Emory University, Atlanta, GA, 30322, USA
| | - Elizabeth Corwin
- School of Nursing, Columbia University, New York, NY, 10032, USA
| | - Alicia K. Smith
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, 30322, USA
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, 30322, USA
| | | | | |
Collapse
|
123
|
Kiouptsi K, Pontarollo G, Reinhardt C. Gut Microbiota and the Microvasculature. Cold Spring Harb Perspect Med 2023; 13:a041179. [PMID: 37460157 PMCID: PMC10411863 DOI: 10.1101/cshperspect.a041179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
The gut microbiota is increasingly recognized as an actuating variable shaping vascular development and endothelial cell function in the intestinal mucosa but also affecting the microvasculature of remote organs. In the small intestine, colonization with gut microbiota and subsequent activation of innate immune pathways promotes the development of intricate capillary networks and lacteals, influencing the integrity of the gut-vascular barrier as well as nutrient uptake. Since the liver yields most of its blood supply via the portal circulation, the hepatic microcirculation steadily encounters microbiota-derived patterns and active signaling metabolites that induce changes in the organization of the liver sinusoidal endothelium, influencing immune zonation of sinusoids and impacting on metabolic processes. In addition, microbiota-derived signals may affect the vasculature of distant organ systems such as the brain and the eye microvasculature. In recent years, this gut-resident microbial ecosystem was revealed to contribute to the development of several vascular disease phenotypes.
Collapse
Affiliation(s)
- Klytaimnistra Kiouptsi
- Center for Thrombosis and Hemostasis (CTH), Partner Site Rhine Main, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Giulia Pontarollo
- Center for Thrombosis and Hemostasis (CTH), Partner Site Rhine Main, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis (CTH), Partner Site Rhine Main, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
124
|
Andres SF, Zhang Y, Kuhn M, Scottoline B. Building better barriers: how nutrition and undernutrition impact pediatric intestinal health. Front Immunol 2023; 14:1192936. [PMID: 37545496 PMCID: PMC10401430 DOI: 10.3389/fimmu.2023.1192936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/26/2023] [Indexed: 08/08/2023] Open
Abstract
Chronic undernutrition is a major cause of death for children under five, leaving survivors at risk for adverse long-term consequences. This review focuses on the role of nutrients in normal intestinal development and function, from the intestinal epithelium, to the closely-associated mucosal immune system and intestinal microbiota. We examine what is known about the impacts of undernutrition on intestinal physiology, with focus again on the same systems. We provide a discussion of existing animal models of undernutrition, and review the evidence demonstrating that correcting undernutrition alone does not fully ameliorate effects on intestinal function, the microbiome, or growth. We review efforts to treat undernutrition that incorporate data indicating that improved recovery is possible with interventions focused not only on delivery of sufficient energy, macronutrients, and micronutrients, but also on efforts to correct the abnormal intestinal microbiome that is a consequence of undernutrition. Understanding of the role of the intestinal microbiome in the undernourished state and correction of the phenotype is both complex and a subject that holds great potential to improve recovery. We conclude with critical unanswered questions in the field, including the need for greater mechanistic research, improved models for the impacts of undernourishment, and new interventions that incorporate recent research gains. This review highlights the importance of understanding the mechanistic effects of undernutrition on the intestinal ecosystem to better treat and improve long-term outcomes for survivors.
Collapse
Affiliation(s)
- Sarah F. Andres
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Yang Zhang
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Madeline Kuhn
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Brian Scottoline
- Division of Neonatology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
125
|
Kazura W, Michalczyk K, Stygar D. The Relationship between the Source of Dietary Animal Fats and Proteins and the Gut Microbiota Condition and Obesity in Humans. Nutrients 2023; 15:3082. [PMID: 37513500 PMCID: PMC10385089 DOI: 10.3390/nu15143082] [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: 06/09/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
The relationship between gut microbiota and obesity is well documented in humans and animal models. Dietary factors can change the intestinal microbiota composition and influence obesity development. However, knowledge of how diet, metabolism, and intestinal microbiota interact and modulate energy metabolism and obesity development is still limited. Epidemiological studies show a link between consuming dietary proteins and fats from specific sources and obesity. Animal studies confirm that proteins and fats of different origins differ in their ability to prevent or induce obesity. Protein sources, such as meat, dairy products, vegetables, pulses, and seafood, vary in their amino acid composition. In addition, the type and level of other factors, such as fatty acids or persistent organic pollutants, vary depending on the source of dietary protein. All these factors can modulate the intestinal microbiota composition and, thus, may influence obesity development. This review summarizes selected evidence of how proteins and fats of different origins affect energy efficiency, obesity development, and intestinal microbiota, linking protein and fat-dependent changes in the intestinal microbiota with obesity.
Collapse
Affiliation(s)
- Wojciech Kazura
- Department of Physiology, Faculty of Medical Sciences, Medical University of Silesia, Jordana Street 19, 41-808 Zabrze, Poland
| | - Katarzyna Michalczyk
- Department of Physiology, Faculty of Medical Sciences, Medical University of Silesia, Jordana Street 19, 41-808 Zabrze, Poland
| | - Dominika Stygar
- Department of Physiology, Faculty of Medical Sciences, Medical University of Silesia, Jordana Street 19, 41-808 Zabrze, Poland
- SLU University Animal Hospital, Swedish University of Agricultural Sciences, SE-750 07 Uppsala, Sweden
| |
Collapse
|
126
|
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
|
127
|
Barone M, Ramayo-Caldas Y, Estellé J, Tambosco K, Chadi S, Maillard F, Gallopin M, Planchais J, Chain F, Kropp C, Rios-Covian D, Sokol H, Brigidi P, Langella P, Martín R. Gut barrier-microbiota imbalances in early life lead to higher sensitivity to inflammation in a murine model of C-section delivery. MICROBIOME 2023; 11:140. [PMID: 37394428 PMCID: PMC10316582 DOI: 10.1186/s40168-023-01584-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 05/25/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND Most interactions between the host and its microbiota occur at the gut barrier, and primary colonizers are essential in the gut barrier maturation in the early life. The mother-offspring transmission of microorganisms is the most important factor influencing microbial colonization in mammals, and C-section delivery (CSD) is an important disruptive factor of this transfer. Recently, the deregulation of symbiotic host-microbe interactions in early life has been shown to alter the maturation of the immune system, predisposing the host to gut barrier dysfunction and inflammation. The main goal of this study is to decipher the role of the early-life gut microbiota-barrier alterations and its links with later-life risks of intestinal inflammation in a murine model of CSD. RESULTS The higher sensitivity to chemically induced inflammation in CSD mice is related to excessive exposure to a too diverse microbiota too early in life. This early microbial stimulus has short-term consequences on the host homeostasis. It switches the pup's immune response to an inflammatory context and alters the epithelium structure and the mucus-producing cells, disrupting gut homeostasis. This presence of a too diverse microbiota in the very early life involves a disproportionate short-chain fatty acids ratio and an excessive antigen exposure across the vulnerable gut barrier in the first days of life, before the gut closure. Besides, as shown by microbiota transfer experiments, the microbiota is causal in the high sensitivity of CSD mice to chemical-induced colitis and in most of the phenotypical parameters found altered in early life. Finally, supplementation with lactobacilli, the main bacterial group impacted by CSD in mice, reverts the higher sensitivity to inflammation in ex-germ-free mice colonized by CSD pups' microbiota. CONCLUSIONS Early-life gut microbiota-host crosstalk alterations related to CSD could be the linchpin behind the phenotypic effects that lead to increased susceptibility to an induced inflammation later in life in mice. Video Abstract.
Collapse
Affiliation(s)
- M. Barone
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Y. Ramayo-Caldas
- INRAE, AgroParisTech, GABI, Paris-Saclay University, 78350 Jouy-en-Josas, France
- Animal Breeding and Genetics Program, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, 08140 Caldes de Montbui, Spain
| | - J. Estellé
- INRAE, AgroParisTech, GABI, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - K. Tambosco
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - S. Chadi
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - F. Maillard
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - M. Gallopin
- CNRS, CEA, l’Institut de Biologie Intégrative de La Cellule (I2BC), Paris-Saclay University, 91405 Orsay, France
| | - J. Planchais
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - F. Chain
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - C. Kropp
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - D. Rios-Covian
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - H. Sokol
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
- Gastroenterology Department, Centre de Recherche Saint-Antoine, Centre de Recherche Saint-Antoine, CRSA, AP-HP, INSERM, Saint Antoine Hospital, Sorbonne Université, 75012 Paris, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - P. Brigidi
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - P. Langella
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - R. Martín
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| |
Collapse
|
128
|
Jiang J. Composition, Diversity and Sex-Related Differences in Intestinal Microbiota in Captive African Penguins ( Spheniscus demersus). Animals (Basel) 2023; 13:2106. [PMID: 37443905 DOI: 10.3390/ani13132106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/16/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
An understanding of the microbial communities in African penguins (Spheniscus demersus) could provide valuable information for saving this endangered species. The objective of this study was to investigate the composition, diversity and sex-related differences in the intestinal microbiota of captive African penguins. Fecal samples were collected from 21 captive adult African penguins reared in the same conditions at Shanghai Zoo. The results show that Proteobacteria, Actinobacteria and Firmicutes were the predominant bacteria in the intestinal microbiota of the captive African penguins. No difference was found in microbial diversity between female and male African penguins, as shown by their similar alpha and beta diversities. However, a notable sex-related difference was found between their microbial compositions. Female African penguins have a higher abundance of Pseudomonas and a lower abundance of Kocuria than males. A functional prediction indicates that the "mRNA surveillance pathway", "Polyketide sugar unit biosynthesis", "Wnt signaling pathway", "Lysosome" and "Cell cycle" pathways were significantly enriched in the microbiota of female African penguins. In conclusion, the present study indicates that the compositions and predicted functions of the intestinal microbiota are significantly different between the sexes. Our data suggest that the intestinal microbiota of female African penguins are more unstable than the intestinal microbiota of males in captivity.
Collapse
Affiliation(s)
- Jingle Jiang
- Shanghai Endangered Species Conservation and Research Centre, Shanghai Zoo, Shanghai 200335, China
| |
Collapse
|
129
|
Kim CS, Shin DM. Gut microbiota and cognitive development in infant mice: Quantity and source of potable water. PLoS One 2023; 18:e0286951. [PMID: 37315057 DOI: 10.1371/journal.pone.0286951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/29/2023] [Indexed: 06/16/2023] Open
Abstract
Not only the water quantity consumed but also the source of drinking water has been considered for their health benefits, but there is limited evidence. We aimed to determine whether the amount and type of drinking water affect physiological and biological functions, including brain function, by confirming how it affects gut microbiota which has an important regulatory role in host physiology. Three-week-old infant mice were subjected to 1) a water restriction experiment (control group, ad libitum consumption of distilled water; dehydration group, time-limited access to distilled water [15 min/day]) and 2) different water source experiment (distilled water, purified water, spring water, and tap water groups). The gut microbiota and cognitive development were analyzed using the 16S ribosomal ribonucleic acid sequencing method and the Barnes maze, respectively. The relative abundance of Firmicutes and Bacteroidetes and the Firmicutes-to-Bacteroidetes ratio (F/B ratio) changed depending on age (juveniles vs. infants). Insufficient water intake reversed these developmental changes, showing that the relative abundances of Bacteroidetes and Firmicutes and the F/B ratio in dehydrated juvenile mice were similar to those in normal infant mice. Additionally, clustering analysis revealed no significant differences in the intestinal flora in the mice from the different drinking water sources; however, dehydration significantly altered the composition of the genera compared to the other water source groups wherein water was provided ad libitum. Moreover, cognitive development was significantly disrupted by insufficient water intake, although the type of drinking water had no significant influence. Cognitive decline, measured by relative latency, was positively associated with the relative abundance of unclassified Erysipelotrichaceae that were in significantly high relative abundance in the dehydration group. These results suggest that the water quantity consumed, rather than the mineral content of drinking water, is imperative for shaping the early gut microbiota associated with cognitive development during infancy.
Collapse
Affiliation(s)
- Chong-Su Kim
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul, Republic of Korea
- Research Institute of Human Ecology, Seoul National University, Seoul, Republic of Korea
| | - Dong-Mi Shin
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul, Republic of Korea
- Research Institute of Human Ecology, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
130
|
Shin H, Martinez KA, Henderson N, Jay M, Schweizer W, Bogaert D, Park G, Bokulich NA, Blaser MJ, Dominguez-Bello MG. Partial convergence of the human vaginal and rectal maternal microbiota in late gestation and early post-partum. NPJ Biofilms Microbiomes 2023; 9:37. [PMID: 37311781 PMCID: PMC10264455 DOI: 10.1038/s41522-023-00404-5] [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] [Received: 05/02/2021] [Accepted: 06/02/2023] [Indexed: 06/15/2023] Open
Abstract
The human vaginal and fecal microbiota change during pregnancy. Because of the proximity of these perineal sites and the evolutionarily conserved maternal-to-neonatal transmission of the microbiota, we hypothesized that the microbiota of these two sites (rectal and vaginal) converge during the last gestational trimester as part of the preparation for parturition. To test this hypothesis, we analyzed 16S rRNA sequences from vaginal introitus and rectal samples in 41 women at gestational ages 6 and 8 months, and at 2 months post-partum. The results show that the human vaginal and rectal bacterial microbiota converged during the last gestational trimester and into the 2nd month after birth, with a significant decrease in Lactobacillus species in both sites, as alpha diversity progressively increased in the vagina and decreased in the rectum. The microbiota convergence of the maternal vaginal-anal sites perinatally might hold significance for the inter-generational transmission of the maternal microbiota.
Collapse
Affiliation(s)
- Hakdong Shin
- Department of Food Science & Biotechnology, and Carbohydrate Bioproduct Research Center, College of Life Science, Sejong University, Seoul, South Korea
| | - Keith A Martinez
- Department of Medicine, New York University Langone Medical Center, New York, NY, USA
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ, USA
| | - Nora Henderson
- Department of Medicine, New York University Langone Medical Center, New York, NY, USA
| | - Melanie Jay
- Department of Medicine, New York University Langone Medical Center, New York, NY, USA
- Department of Population Health, New York University Langone Medical Center, New York, NY, USA
| | - William Schweizer
- Department of Obstetrics and Gynecology, New York University Langone Medical Center, New York, NY, USA
| | - Debby Bogaert
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, Scotland
| | - Gwoncheol Park
- Department of Food Science & Biotechnology, and Carbohydrate Bioproduct Research Center, College of Life Science, Sejong University, Seoul, South Korea
| | - Nicholas A Bokulich
- Laboratory of Food Systems Biotechnology, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Martin J Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, NJ, USA
| | - Maria Gloria Dominguez-Bello
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ, USA.
- Department of Anthropology, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
131
|
Logoń K, Świrkosz G, Nowak M, Wrześniewska M, Szczygieł A, Gomułka K. The Role of the Microbiome in the Pathogenesis and Treatment of Asthma. Biomedicines 2023; 11:1618. [PMID: 37371713 DOI: 10.3390/biomedicines11061618] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 05/27/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
The role of the microbiome in the pathogenesis and treatment of asthma is significant. The purpose of this article is to show the interplay between asthma and the microbiome, and main areas that require further research are also highlighted. The literature search was conducted using the PubMed database. After a screening process of studies published before May 2023, a total of 128 articles were selected in our paper. The pre-treatment bronchial microbiome in asthmatic patients plays a role in their responsiveness to treatment. Gut microbiota and its dysbiosis can contribute to immune system modulation and the development of asthma. The association between the microbiome and asthma is complex. Further research is necessary to clarify which factors might moderate that relationship. An appropriate gut microbiome and its intestinal metabolites are a protective factor for asthma development. Prebiotics and certain dietary strategies may have a prophylactic or therapeutic effect, but more research is needed to establish final conclusions. Although the evidence regarding probiotics is ambiguous, and most meta-analyses do not support the use of probiotic intake to reduce asthma, several of the most recent studies have provided promising effects. Further studies should focus on the investigation of specific strains and the examination of their mechanistic and genetic aspects.
Collapse
Affiliation(s)
- Katarzyna Logoń
- Student Scientific Group of Adult Allergology, Wroclaw Medical University, 50-369 Wrocław, Poland
| | - Gabriela Świrkosz
- Student Scientific Group of Adult Allergology, Wroclaw Medical University, 50-369 Wrocław, Poland
| | - Monika Nowak
- Student Scientific Group of Adult Allergology, Wroclaw Medical University, 50-369 Wrocław, Poland
| | - Martyna Wrześniewska
- Student Scientific Group of Adult Allergology, Wroclaw Medical University, 50-369 Wrocław, Poland
| | - Aleksandra Szczygieł
- Student Scientific Group of Adult Allergology, Wroclaw Medical University, 50-369 Wrocław, Poland
| | - Krzysztof Gomułka
- Clinical Department of Internal Medicine, Pneumology and Allergology, Wroclaw Medical University, 50-369 Wrocław, Poland
| |
Collapse
|
132
|
Street ME, Shulhai AM, Rotondo R, Giannì G, Caffarelli C. Current knowledge on the effects of environmental contaminants in early life nutrition. Front Nutr 2023; 10:1120293. [PMID: 37324741 PMCID: PMC10267348 DOI: 10.3389/fnut.2023.1120293] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/10/2023] [Indexed: 06/17/2023] Open
Abstract
Breast milk represents the optimal source of feeding for newborns, in terms of nutritional compounds and as it provides immunological, metabolic, organic, and neurological well-being. As a complex biological fluid, it consists not only of nutritional compounds but also contains environmental contaminants. Formulas through production, contact with bottles and cups, and complementary feeding can also be contaminated. The current review focuses on endocrine-disrupting chemicals, and made-man xenoestrogens present in the environment and both commonly present in food sources, agricultural practices, packaging, consumer products, industry, and medical care. These contaminants are transferred by passive diffusion to breast milk and are delivered during breastfeeding. They mainly act by activating or antagonizing hormonal receptors. We summarize the effects on the immune system, gut microbiota, and metabolism. Exposure to endocrine-disrupting chemicals and indirect food additives may induce tissue inflammation and polarize lymphocytes, increase proinflammatory cytokines, promote allergic sensitization, and microbial dysbiosis, activate nuclear receptors and increase the incidence of allergic, autoimmune, and metabolic diseases. Breast milk is the most important optimal source in early life. This mini-review summarizes current knowledge on environmental contaminants and paves the way for strategies to prevent milk contamination and limit maternal and infant exposure during pregnancy and the first months of life.
Collapse
Affiliation(s)
- Maria E. Street
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Unit of Pediatrics, University Hospital of Parma, Parma, Italy
| | - Anna-Mariia Shulhai
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Unit of Pediatrics, University Hospital of Parma, Parma, Italy
| | - Roberta Rotondo
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Unit of Pediatrics, University Hospital of Parma, Parma, Italy
| | - Giuliana Giannì
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Unit of Pediatrics, University Hospital of Parma, Parma, Italy
| | - Carlo Caffarelli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Unit of Pediatrics, University Hospital of Parma, Parma, Italy
| |
Collapse
|
133
|
Snyder KB, Hunter CJ. Bugs and the barrier: A review of the gut microbiome and intestinal barrier in necrotizing enterocolitis. Semin Pediatr Surg 2023; 32:151310. [PMID: 37290337 DOI: 10.1016/j.sempedsurg.2023.151310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease that affects premature neonates. It frequently results in significant morbidity and mortality for those affected. Years of research into the pathophysiology of NEC have revealed it to be a variable and multifactorial disease. However, there are risk factors associated with NEC including low birth weight, prematurity, intestinal immaturity, alterations in microbial colonization, and history of rapid or formula based enteral feeds (Fig. 1).1-3 An accepted generalization of the pathogenesis of NEC includes a hyperresponsive immune reaction to insults such as ischemia, starting formula feeds, or alterations in the microbiome with pathologic bacterial colonization and translocation. This reaction causes a hyperinflammatory response disrupting the normal intestinal barrier, allowing abnormal bacterial translocation and ultimately sepsis.1,2,4 This review will focus specifically on the interactions with the microbiome and intestinal barrier function in NEC.
Collapse
Affiliation(s)
- K Brooke Snyder
- Division of Pediatric Surgery, Oklahoma Children's Hospital, 1200 Everett Drive, ET NP 2320 Oklahoma City, OK 73104, United States; The University of Oklahoma Health Sciences Center, Department of Surgery, 800 Research Parkway, Suite 449, Oklahoma City, OK 73104, United States
| | - Catherine J Hunter
- Division of Pediatric Surgery, Oklahoma Children's Hospital, 1200 Everett Drive, ET NP 2320 Oklahoma City, OK 73104, United States; The University of Oklahoma Health Sciences Center, Department of Surgery, 800 Research Parkway, Suite 449, Oklahoma City, OK 73104, United States.
| |
Collapse
|
134
|
Ji H, Tan D, Chen Y, Cheng Z, Zhao J, Lin M. Effects of different manganese sources on nutrient digestibility, fecal bacterial community, and mineral excretion of weaning dairy calves. Front Microbiol 2023; 14:1163468. [PMID: 37275150 PMCID: PMC10232960 DOI: 10.3389/fmicb.2023.1163468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/20/2023] [Indexed: 06/07/2023] Open
Abstract
Introduction Mn, which is an essential trace mineral for all animals, has functions in skeletal system development, carbohydrate and lipid metabolism. The aim of this study was to clarify the effects of different manganese (Mn) sources in basal diets on nutrient apparent digestibility, fecal microbes, and mineral elements excretion before and after weaning. Methods A total of 15 Holstein heifer calves (6-week-old, 82.71 ± 1.35, mean ± standard error) were randomly designed into three groups (five each): no extra Mn supplemented (CON), 20 mg Mn/kg (dry matter basis) in the form of chelates of lysine and glutamic acid in a mixture of 1:1 (LGM), and 20 mg Mn/kg (dry matter basis) in the form of MnSO4. All calves were weaned at 8 weeks of age. The experiment lasted for 28 days (14 days before weaning and 14 days after weaning). Dry matter intake (DMI) was recorded daily. The animals were weighed by electronic walk-over, and body size indices were collected using tape on days -14, -1, and 14 of weaning. The feces of calves was collected to measure the apparent digestibility of nutrients (acid insoluble ash was an internal marker) and bacterial community on days -1, 1, 3, 7, and 14 of weaning. Fecal mineral concentration was determined by inductively coupled plasma emission spectroscopy on days -1, 1, 7, and 14 of weaning. Results The results showed that, compared with the CON group, adding LGM to diets containing 158.82 mg/kg Mn increased the apparent digestibility (P < 0.05). The Chao 1 and Shannon index of fecal bacteria decreased at day 1 in the LGM and MnSO4 groups and increased after weaning. The PCoA results indicated that the LGM group was distinctly separate from the CON and MnSO4 groups during the whole experimental period. Significant differences (P < 0.05) were observed in the relative abundance of two phyla (Proteobacteria and Spirochaetota) and eight genera (Alloprevotella, Prevotellaceae_UCG-001, Clostridia UCG 014, RF39, UCG-010, Pseudomonas, Ralstonia, and Treponema) in three groups. Moreover, the LGM group showed less excretion of Fe, P, and Mn than the MnSO4 group. Discussion In summary, 20 mg Mn/kg diet supplementation improved nutrient digestibility, changed the fecal microbial community, and reduced mineral excretion. Organic Mn supplementation in the diet had more advantages over the sulfate forms in weaning calves.
Collapse
Affiliation(s)
- Huimin Ji
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Dejin Tan
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yuhua Chen
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Zhiqiang Cheng
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Jingwen Zhao
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Miao Lin
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| |
Collapse
|
135
|
Cha T, Kim HH, Keum J, Kwak MJ, Park JY, Hoh JK, Kim CR, Jeon BH, Park HK. Gut microbiome profiling of neonates using Nanopore MinION and Illumina MiSeq sequencing. Front Microbiol 2023; 14:1148466. [PMID: 37256051 PMCID: PMC10225602 DOI: 10.3389/fmicb.2023.1148466] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/17/2023] [Indexed: 06/01/2023] Open
Abstract
This study aimed to evaluate the difference in gut microbiomes between preterm and term infants using third-generation long-read sequencing (Oxford Nanopore Technologies, ONT) compared with an established gold standard, Illumina (second-generation short-read sequencing). A total of 69 fecal samples from 51 term (T) and preterm (P) infants were collected at 7 and 28 days of life. Gut colonization profiling was performed by 16S rRNA gene sequencing using ONT. We used Illumina to validate and compare the patterns in 13 neonates. Using bioinformatic analysis, we identified features that differed between P and T. Both T1 and P1 microbiomes were dominated by Firmicutes (Staphylococcus and Enterococcus), whereas sequentially showed dominant transitions to Lactobacillus (p < 0.001) and Streptococcus in T2 (p = 0.001), and pathogenic bacteria (Klebsiella) in P2 (p = 0.001). The abundance of beneficial bacteria (Bifidobacterium and Lactobacillus) increased in T2 (p = 0.026 and p < 0.001, respectively). These assignments were correlated with the abundance at the species-level. Bacterial α-diversity increased in T (p = 0.005) but not in P (p = 0.156), and P2 showed distinct β-diversity clustering than T2 (p = 0.001). The ONT reliably identified pathogenic bacteria at the genus level, and taxonomic profiles were comparable to those identified by Illumina at the genus level. This study shows that ONT and Illumina are highly correlated. P and T had different microbiome profiles, and the α- and β-diversity varied. ONT sequencing has potential for pathogen detection in neonates in clinical settings.
Collapse
Affiliation(s)
- Teahyen Cha
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Hoo Hugo Kim
- Department of Earth Resources and Environmental Engineering, Hanyang University, Seoul, Republic of Korea
| | - Jihyun Keum
- Department of Obstetrics and Gynecology, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Min-Jin Kwak
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jae Yong Park
- Division of Microbiome, Int-Gen Company, Seoul, Republic of Korea
| | - Jeong Kyu Hoh
- Department of Obstetrics and Gynecology, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Chang-Ryul Kim
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Byong-Hun Jeon
- Department of Earth Resources and Environmental Engineering, Hanyang University, Seoul, Republic of Korea
| | - Hyun-Kyung Park
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
136
|
Wang L, Wang F, Xiong L, Song H, Ren B, Shen X. A nexus of dietary restriction and gut microbiota: Recent insights into metabolic health. Crit Rev Food Sci Nutr 2023; 64:8649-8671. [PMID: 37154021 DOI: 10.1080/10408398.2023.2202750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
In recent times, dietary restriction (DR) has received considerable attention for its promising effects on metabolism and longevity. Previous studies on DR have mainly focused on the health benefits produced by different restriction patterns, whereas comprehensive reviews of the role of gut microbiota during DR are limited. In this review, we discuss the effects of caloric restriction, fasting, protein restriction, and amino acid restriction from a microbiome perspective. Furthermore, the underlying mechanisms by which DR affects metabolic health by regulating intestinal homeostasis are summarized. Specifically, we reviewed the impacts of different DRs on specific gut microbiota. Additionally, we put forward the limitations of the current research and suggest the development of personalized microbes-directed DR for different populations and corresponding next-generation sequencing technologies for accurate microbiological analysis. DR effectively modulates the composition of the gut microbiota and microbial metabolites. In particular, DR markedly affects the rhythmic oscillation of microbes which may be related to the circadian clock system. Moreover, increasing evidence supports that DR profoundly improves metabolic syndrome, inflammatory bowel disease, and cognitive impairment. To summarize, DR may be an effective and executable dietary manipulation strategy for maintaining metabolic health, however, further investigation is needed to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Luanfeng Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Fang Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Ling Xiong
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Haizhao Song
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Bo Ren
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Xinchun Shen
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| |
Collapse
|
137
|
Kedia S, Ahuja V. Human gut microbiome: A primer for the clinician. JGH Open 2023; 7:337-350. [PMID: 37265934 PMCID: PMC10230107 DOI: 10.1002/jgh3.12902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 12/14/2022] [Accepted: 04/01/2023] [Indexed: 06/03/2023]
Abstract
The human host gets tremendously influenced by a genetically and phenotypically distinct and heterogeneous constellation of microbial species-the human microbiome-the gut being one of the most densely populated and characterized site for these organisms. Microbiome science has advanced rapidly, technically with respect to the analytical methods and biologically with respect to its mechanistic influence in health and disease states. A clinician conducting a microbiome study should be aware of the nuances related to microbiome research, especially with respect to the technical and biological factors that can influence the interpretation of research outcomes. Hence, this review is an attempt to detail these aspects of the human gut microbiome, with emphasis on its determinants in a healthy state.
Collapse
Affiliation(s)
- Saurabh Kedia
- Department of GastroenterologyAll India Institute of Medical SciencesNew DelhiIndia
| | - Vineet Ahuja
- Department of GastroenterologyAll India Institute of Medical SciencesNew DelhiIndia
| |
Collapse
|
138
|
Lubin JB, Green J, Maddux S, Denu L, Duranova T, Lanza M, Wynosky-Dolfi M, Flores JN, Grimes LP, Brodsky IE, Planet PJ, Silverman MA. Arresting microbiome development limits immune system maturation and resistance to infection in mice. Cell Host Microbe 2023; 31:554-570.e7. [PMID: 36996818 PMCID: PMC10935632 DOI: 10.1016/j.chom.2023.03.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 01/09/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023]
Abstract
Disruptions to the intestinal microbiome during weaning lead to negative effects on host immune function. However, the critical host-microbe interactions during weaning that are required for immune system development remain poorly understood. We find that restricting microbiome maturation during weaning stunts immune system development and increases susceptibility to enteric infection. We developed a gnotobiotic mouse model of the early-life microbiome Pediatric Community (PedsCom). These mice develop fewer peripheral regulatory T cells and less IgA, hallmarks of microbiota-driven immune system development. Furthermore, adult PedsCom mice retain high susceptibility to Salmonella infection, which is characteristic of young mice and children. Altogether, our work illustrates how the post-weaning transition in microbiome composition contributes to normal immune maturation and protection from infection. Accurate modeling of the pre-weaning microbiome provides a window into the microbial requirements for healthy development and suggests an opportunity to design microbial interventions at weaning to improve immune development in human infants.
Collapse
Affiliation(s)
- Jean-Bernard Lubin
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jamal Green
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah Maddux
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lidiya Denu
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Tereza Duranova
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Matthew Lanza
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | | | - Julia N Flores
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Logan P Grimes
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Igor E Brodsky
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA; Institute for Immunology, IFI, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul J Planet
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Michael A Silverman
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Immunology Research Unit, GlaxoSmithKline, Collegeville, PA, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
139
|
Veeraraghavan B, Kesavelu D, Yadav B. Gut Microbiota Composition in Indian and Western Infants (0–24 Months): A Systematic Review. NUTRITION AND DIETARY SUPPLEMENTS 2023. [DOI: 10.2147/nds.s402256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
|
140
|
Loy MH, Usseglio J, Lasalandra D, Gold MA. Probiotic Use in Children and Adolescents with Overweight or Obesity: A Scoping Review. Child Obes 2023; 19:145-159. [PMID: 35723657 DOI: 10.1089/chi.2022.0059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Context: Probiotics have been proposed as a prevention or treatment for pediatric overweight and obesity. Objective: Conduct a scoping review on probiotic use in children and adolescents with overweight or obesity and those with weight-related conditions and to identify knowledge gaps and research priorities. Data Sources: Seven databases using keywords and medical subject heading terms for articles reporting probiotic use in children or adolescents with overweight or obesity published from database conception until initiation of the study. Study Selection: Articles reporting primary data on probiotics use in children or adolescents with overweight or obesity. Data Extraction: We utilized the Arksey and O'Malley framework, PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analysis) guidelines, followed a predetermined study protocol for level-one abstract and level-two full-text screenings, synthesized information into subject-area domains, and identified research gaps. Limitations: Heterogeneity of probiotic interventions, host factors, and genomics. Results: Database search yielded 1356 unique articles with 19 randomized placebo-controlled studies, 945 participants, duration of interventions from 8 weeks to 9 months. Disease indications included Nonalcoholic Fatty Liver Disease, insulin resistance, hypercholesterolemia, Prader-Willi Syndrome, metabolic syndrome, and obesity. Limited and heterogeneous evidence for probiotic use in children and adolescents with weight-related conditions noted. Heterogeneity among published articles in probiotic strains, doses, design, biomarkers, confirmation, and outcomes observed. Conclusions: Despite complex existing and limited data, studies to date of children and adolescents with overweight and obesity demonstrate potential beneficial treatment effects of probiotics on BMI, adiposity, metabolic parameters, inflammatory markers, fatty liver, transaminase levels, and glucose metabolism. Clinical trials to address heterogeneous results are needed.
Collapse
Affiliation(s)
- Michelle H Loy
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Integrative Health and Well-Being, Weill Cornell Medicine/New York Presbyterian Hospital, New York, NY, USA
| | - John Usseglio
- Health Sciences Library, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Melanie A Gold
- Heilbrunn Department of Population and Family Health, Columbia University Mailman School of Public Health, New York, NY, USA
- Section of Adolescent Medicine, Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
- Center for Community Health and Education, School-Based Health Centers, NewYork Presbyterian, New York, NY, USA
| |
Collapse
|
141
|
Sun Z, Lee-Sarwar K, Kelly RS, Lasky-Su JA, Litonjua AA, Weiss ST, Liu YY. Revealing the importance of prenatal gut microbiome in offspring neurodevelopment in humans. EBioMedicine 2023; 90:104491. [PMID: 36868051 PMCID: PMC9996363 DOI: 10.1016/j.ebiom.2023.104491] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/01/2023] [Accepted: 02/09/2023] [Indexed: 03/05/2023] Open
Abstract
BACKGROUND It has been widely recognized that a critical time window for neurodevelopment occurs in early life and the host's gut microbiome plays an important role in neurodevelopment. Following recent demonstrations that the maternal prenatal gut microbiome influences offspring brain development in murine models, we aim to explore whether the critical time window for the association between the gut microbiome and neurodevelopment is prenatal or postnatal for human. METHODS Here we leverage a large-scale human study and compare the associations between the gut microbiota and metabolites from mothers during pregnancy and their children with the children's neurodevelopment. Specifically, using multinomial regression integrated in Songbird, we assessed the discriminating power of the maternal prenatal and child gut microbiome for children's neurodevelopment at early life as measured by the Ages & Stages Questionnaires (ASQ). FINDINGS We show that the maternal prenatal gut microbiome is more relevant than the children's gut microbiome to the children's neurodevelopment in the first year of life (maximum Q2 = 0.212 and 0.096 separately using the taxa at the class level). Moreover, we found that Fusobacteriia is more associated with high fine motor skills in ASQ in the maternal prenatal gut microbiota but become more associated with low fine motor skills in the infant gut microbiota (rank = 0.084 and -0.047 separately), suggesting the roles of the same taxa with respect to neurodevelopment can be opposite at the two stages of fetal neurodevelopment. INTERPRETATION These findings shed light, especially in terms of timing, on potential therapeutic interventions to prevent neurodevelopmental disorders. FUNDING This work was supported by the National Institutes of Health (grant numbers: R01AI141529, R01HD093761, RF1AG067744, UH3OD023268, U19AI095219, U01HL089856, R01HL141826, K08HL148178, K01HL146980), and the Charles A. King Trust Postdoctoral Fellowship.
Collapse
Affiliation(s)
- Zheng Sun
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Kathleen Lee-Sarwar
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Rachel S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Jessica A Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Augusto A Litonjua
- Division of Pediatric Pulmonary Medicine, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Yang-Yu Liu
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA; Center for Artificial Intelligence and Modeling, The Carl R. Woese Institute of Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, 61801, USA.
| |
Collapse
|
142
|
Wilson A, Bogie B, Chaaban H, Burge K. The Nonbacterial Microbiome: Fungal and Viral Contributions to the Preterm Infant Gut in Health and Disease. Microorganisms 2023; 11:909. [PMID: 37110332 PMCID: PMC10144239 DOI: 10.3390/microorganisms11040909] [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: 03/03/2023] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023] Open
Abstract
The intestinal microbiome is frequently implicated in necrotizing enterocolitis (NEC) pathogenesis. While no particular organism has been associated with NEC development, a general reduction in bacterial diversity and increase in pathobiont abundance has been noted preceding disease onset. However, nearly all evaluations of the preterm infant microbiome focus exclusively on the bacterial constituents, completely ignoring any fungi, protozoa, archaea, and viruses present. The abundance, diversity, and function of these nonbacterial microbes within the preterm intestinal ecosystem are largely unknown. Here, we review findings on the role of fungi and viruses, including bacteriophages, in preterm intestinal development and neonatal intestinal inflammation, with potential roles in NEC pathogenesis yet to be determined. In addition, we highlight the importance of host and environmental influences, interkingdom interactions, and the role of human milk in shaping fungal and viral abundance, diversity, and function within the preterm intestinal ecosystem.
Collapse
Affiliation(s)
| | | | - Hala Chaaban
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Kathryn Burge
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
143
|
Liukkonen M, Hukkanen M, Cossin-Sevrin N, Stier A, Vesterinen E, Grond K, Ruuskanen S. No evidence for associations between brood size, gut microbiome diversity and survival in great tit (Parus major) nestlings. Anim Microbiome 2023; 5:19. [PMID: 36949549 PMCID: PMC10031902 DOI: 10.1186/s42523-023-00241-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 03/13/2023] [Indexed: 03/24/2023] Open
Abstract
BACKGROUND The gut microbiome forms at an early stage, yet data on the environmental factors influencing the development of wild avian microbiomes is limited. As the gut microbiome is a vital part of organismal health, it is important to understand how it may connect to host performance. The early studies with wild gut microbiome have shown that the rearing environment may be of importance in gut microbiome formation, yet the results vary across taxa, and the effects of specific environmental factors have not been characterized. Here, wild great tit (Parus major) broods were manipulated to either reduce or enlarge the original brood soon after hatching. We investigated if brood size was associated with nestling bacterial gut microbiome, and whether gut microbiome diversity predicted survival. Fecal samples were collected at mid-nestling stage and sequenced with the 16S rRNA gene amplicon sequencing, and nestling growth and survival were measured. RESULTS Gut microbiome diversity showed high variation between individuals, but this variation was not significantly explained by brood size or body mass. Additionally, we did not find a significant effect of brood size on body mass or gut microbiome composition. We also demonstrated that early handling had no impact on nestling performance or gut microbiome. Furthermore, we found no significant association between gut microbiome diversity and short-term (survival to fledging) or mid-term (apparent juvenile) survival. CONCLUSIONS We found no clear association between early-life environment, offspring condition and gut microbiome. This suggests that brood size is not a significantly contributing factor to great tit nestling condition, and that other environmental and genetic factors may be more strongly linked to offspring condition and gut microbiome. Future studies should expand into other early-life environmental factors e.g., diet composition and quality, and parental influences.
Collapse
Affiliation(s)
- Martta Liukkonen
- Department of Biological and Environmental Science, University of Jyväskylä, Jyvaskyla, Finland.
| | - Mikaela Hukkanen
- Department of Biology, University of Turku, Turku, Finland
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | | | - Antoine Stier
- Department of Biology, University of Turku, Turku, Finland
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, ENTPE, UMR 5023 LEHNA, 69622, Lyon, France
- Institut Pluridisciplinaire Hubert Curien, UMR7178, Université de Strasbourg, CNRS, Strasbourg, France
| | | | - Kirsten Grond
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK, 99508, USA
| | - Suvi Ruuskanen
- Department of Biological and Environmental Science, University of Jyväskylä, Jyvaskyla, Finland
- Department of Biology, University of Turku, Turku, Finland
| |
Collapse
|
144
|
Li L, Zhao X, He JJ. HIV Tat Expression and Cocaine Exposure Lead to Sex- and Age-Specific Changes of the Microbiota Composition in the Gut. Microorganisms 2023; 11:799. [PMID: 36985373 PMCID: PMC10054272 DOI: 10.3390/microorganisms11030799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/08/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
The balance of microbial communities in the gut is extremely important for normal physiological function. Disruption of the balance is often associated with various disorders and diseases. Both HIV infection and cocaine use are known to change the gut microbiota and the epithelial barrier integrity, which contribute to inflammation and immune activation. Our recent study shows that Tat expression and cocaine exposure result in changes of genome-wide DNA methylation and gene expression and lead to worsen the learning and memory impairments. In the current study, we extended the study to determine effects of Tat and cocaine on the gut microbiota composition. We found that both Tat expression and cocaine exposure increased Alteromonadaceae in 6-month-old female/male mice. In addition, we found that Tat, cocaine, or both increased Alteromonadaceae, Bacteroidaceae, Cyanobiaceae, Erysipelotrichaceae, and Muribaculaceae but decreased Clostridiales_vadinBB60_group, Desulfovibrionaceae, Helicobacteraceae, Lachnospiraceae, and Ruminococcaceae in 12-month-old female mice. Lastly, we analyzed changes of metabolic pathways and found that Tat decreased energy metabolism and nucleotide metabolism, and increased lipid metabolism and metabolism of other amino acids while cocaine increased lipid metabolism in 12-month-old female mice. These results demonstrated that Tat expression and cocaine exposure resulted in significant changes of the gut microbiota in an age- and sex-dependent manner and provide additional evidence to support the bidirectional gut-brain axis hypothesis.
Collapse
Affiliation(s)
- Lu Li
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University, 3333 Green Bay Road, North Chicago, IL 60064, USA
- Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, IL 60064, USA
| | - Xiaojie Zhao
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University, 3333 Green Bay Road, North Chicago, IL 60064, USA
- Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, IL 60064, USA
| | - Johnny J. He
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University, 3333 Green Bay Road, North Chicago, IL 60064, USA
- Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, IL 60064, USA
| |
Collapse
|
145
|
An J, Song Y, Kim S, Kong H, Kim K. Alteration of Gut Microbes in Benign Prostatic Hyperplasia Model and Finasteride Treatment Model. Int J Mol Sci 2023; 24:ijms24065904. [PMID: 36982979 PMCID: PMC10057928 DOI: 10.3390/ijms24065904] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Gut microbes are closely associated with disease onset and improvement. However, the effects of gut microbes on the occurrence, prevention, and treatment of benign prostatic hyperplasia (BPH) are still unclear. We investigated the alteration of gut microbiota with implications for the diagnosis, prevention, and treatment of BPH and identified correlations among various indicators, including hormone indicators, apoptosis markers in BPH, and finasteride treatment models. BPH induction altered the abundance of Lactobacillus, Flavonifractor, Acetatifactor, Oscillibacter, Pseudoflavonifractor, Intestinimonas, and Butyricimonas genera, which are related to BPH indicators. Among these, the altered abundance of Lactobacillus and Acetatifactor was associated with the promotion and inhibition of prostate apoptosis, respectively. Finasteride treatment altered the abundance of Barnesiella, Acetatifactor, Butyricimonas, Desulfovibrio, Anaerobacterium, and Robinsoniella genera, which are related to BPH indicators. Among these, altered abundances of Desulfovibrio and Acetatifactor were associated with the promotion and inhibition of prostate apoptosis, respectively. In addition, the abundances of Lactobacillus and Acetatifactor were normalized after finasteride treatment. In conclusion, the association between apoptosis and altered abundances of Lactobacillus and Acetatifactor, among other gut microbes, suggests their potential utility in the diagnosis, prevention, and treatment of BPH.
Collapse
Affiliation(s)
- Jinho An
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
- PADAM Natural Material Research Institute, Sahmyook University, Seoul 01795, Republic of Korea
| | - Youngcheon Song
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
- PADAM Natural Material Research Institute, Sahmyook University, Seoul 01795, Republic of Korea
| | - Sangbum Kim
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Hyunseok Kong
- PADAM Natural Material Research Institute, Sahmyook University, Seoul 01795, Republic of Korea
- College of Animal Biotechnology and Resource, Sahmyook University, Seoul 01795, Republic of Korea
| | - Kyungjae Kim
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
- PADAM Natural Material Research Institute, Sahmyook University, Seoul 01795, Republic of Korea
| |
Collapse
|
146
|
Rahman T, Sarwar PF, Potter C, Comstock SS, Klepac-Ceraj V. Role of human milk oligosaccharide metabolizing bacteria in the development of atopic dermatitis/eczema. Front Pediatr 2023; 11:1090048. [PMID: 37020647 PMCID: PMC10069630 DOI: 10.3389/fped.2023.1090048] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 02/23/2023] [Indexed: 04/07/2023] Open
Abstract
Despite affecting up to 20% of infants in the United States, there is no cure for atopic dermatitis (AD), also known as eczema. Atopy usually manifests during the first six months of an infant's life and is one predictor of later allergic health problems. A diet of human milk may offer protection against developing atopic dermatitis. One milk component, human milk oligosaccharides (HMOs), plays an important role as a prebiotic in establishing the infant gut microbiome and has immunomodulatory effects on the infant immune system. The purpose of this review is to summarize the available information about bacterial members of the intestinal microbiota capable of metabolizing HMOs, the bacterial genes or metabolic products present in the intestinal tract during early life, and the relationship of these genes and metabolic products to the development of AD/eczema in infants. We find that specific HMO metabolism gene sets and the metabolites produced by HMO metabolizing bacteria may enable the protective role of human milk against the development of atopy because of interactions with the immune system. We also identify areas for additional research to further elucidate the relationship between the human milk metabolizing bacteria and atopy. Detailed metagenomic studies of the infant gut microbiota and its associated metabolomes are essential for characterizing the potential impact of human milk-feeding on the development of atopic dermatitis.
Collapse
Affiliation(s)
- Trisha Rahman
- Department of Biological Sciences, Wellesley College, Wellesley, MA, United States
| | - Prioty F. Sarwar
- Department of Biological Sciences, Wellesley College, Wellesley, MA, United States
| | - Cassie Potter
- Department of Biological Sciences, Wellesley College, Wellesley, MA, United States
| | - Sarah S. Comstock
- Department of Food Science & Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Vanja Klepac-Ceraj
- Department of Biological Sciences, Wellesley College, Wellesley, MA, United States
| |
Collapse
|
147
|
Torow N, Hand TW, Hornef MW. Programmed and environmental determinants driving neonatal mucosal immune development. Immunity 2023; 56:485-499. [PMID: 36921575 PMCID: PMC10079302 DOI: 10.1016/j.immuni.2023.02.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/17/2023] [Indexed: 03/15/2023]
Abstract
The mucosal immune system of neonates goes through successive, non-redundant phases that support the developmental needs of the infant and ultimately establish immune homeostasis. These phases are informed by environmental cues, including dietary and microbial stimuli, but also evolutionary developmental programming that functions independently of external stimuli. The immune response to exogenous stimuli is tightly regulated during early life; thresholds are set within this neonatal "window of opportunity" that govern how the immune system will respond to diet, the microbiota, and pathogenic microorganisms in the future. Thus, changes in early-life exposure, such as breastfeeding or environmental and microbial stimuli, influence immunological and metabolic homeostasis and the risk of developing diseases such as asthma/allergy and obesity.
Collapse
Affiliation(s)
- Natalia Torow
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Timothy W Hand
- Pediatrics Department, Infectious Disease Section, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA.
| | - Mathias W Hornef
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany.
| |
Collapse
|
148
|
Murray E, Butcher J, May Kearns M, Lamba S, Liang J, Stintzi A, Ismail N. Effects of pair-housing pubertal and adult male and female mice on LPS-induced age-dependent immune responses: A potential role for the gut microbiota. Brain Behav Immun 2023; 110:297-309. [PMID: 36914014 DOI: 10.1016/j.bbi.2023.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 03/06/2023] [Accepted: 03/08/2023] [Indexed: 03/15/2023] Open
Abstract
Puberty is a critical period of development that is marked by the maturation of the stress and immune systems. There are marked age and sex differences in peripheral and central inflammatory responses to an immune challenge between pubertal and adult mice. Given the strong link between the gut microbiome and immune system, it is possible that the age and sex differences in immune responses are mediated by age and sex differences in gut microbial composition. The current study investigated whether cohousing adult and pubertal CD1 mice through three weeks of pair-housing, with the potential for microbiome exchange via coprophagy and other close contact, could mitigate age-dependent immune responses. Cytokine concentrations in the blood and cytokine mRNA expression in the brain were assessed following exposure to the immune challenge lipopolysaccharide (LPS). The results show that all mice displayed increased cytokine concentrations in serum and central cytokine mRNA expression in the hippocampus, hypothalamus and prefrontal cortex (PFC) at eight hours following LPS treatment. Pubertal male and female mice, that were pair-housed with a pubertal counterpart, displayed lower cytokine concentrations in serum and lower cytokine mRNA expression in the brain compared to adult mice that were pair-housed with an adult counterpart. However, when adult and pubertal mice were pair-housed, the age differences in both peripheral cytokine concentrations and central cytokine mRNA expression were mitigated. We also found that pair-housing adult and pubertal mice eliminated the age difference in gut bacterial diversity. These results suggest that microbial composition could be involved in modulating these age-associated immune responses and thus may represent a potential therapeutic target.
Collapse
Affiliation(s)
- Emma Murray
- Neuroimmunology, Stress and Endocrinology (NISE) Lab, School of Psychology, Faculty of Social Science, University of Ottawa, Canada
| | - James Butcher
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada
| | - Madeleine May Kearns
- Neuroimmunology, Stress and Endocrinology (NISE) Lab, School of Psychology, Faculty of Social Science, University of Ottawa, Canada
| | - Sanjeevani Lamba
- School of Biosciences, Cardiff University, Wales, United Kingdom
| | - Jacky Liang
- Neuroimmunology, Stress and Endocrinology (NISE) Lab, School of Psychology, Faculty of Social Science, University of Ottawa, Canada
| | - Alain Stintzi
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada
| | - Nafissa Ismail
- Neuroimmunology, Stress and Endocrinology (NISE) Lab, School of Psychology, Faculty of Social Science, University of Ottawa, Canada; University of Ottawa Brain and Mind Research Institute, Canada.
| |
Collapse
|
149
|
Zhao H, Ren Q, Wang HY, Zong Y, Zhao W, Wang Y, Qu M, Wang J. Alterations in gut microbiota and urine metabolomics in infants with yin-deficiency constitution aged 0–2 years. Heliyon 2023; 9:e14684. [PMID: 37064462 PMCID: PMC10102239 DOI: 10.1016/j.heliyon.2023.e14684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/15/2023] [Accepted: 03/15/2023] [Indexed: 03/31/2023] Open
Abstract
Background Based on the constitution theroy, infants are classified into balanced constitution (BC) and unbalanced constitution. Yin-deficiency constitution (YINDC) is a common type of unbalanced constitutions in Chinese infants. An infant's gut microbiota directly affects the child's health and has long-term effects on the maturation of the immune and endocrine systems throughout life. However, the gut microbiota of infants with YINDC remains unknown. Herein, we aimed to evaluate the intestinal flora profiles and urinary metabolites in infant with YINDC, find biomarkers to identify YINDC, and promote our understanding of infant constitution classification. Methods Constitutional Medicine Questionnaires were used to assess the infants' constitution types. 47 infants with 21 cases of YINDC and 26 cases of BC were included, and a cross-sectional sampling of stool and urine was conducted. Fecal microbiota was characterized using 16S rRNA sequencing, and urinary metabolomics was profiled using UPLC-Q-TOF/MS method. YINDC markers with high accuracy were identified using receiver operating characteristic (ROC) analysis. Results The diversity and composition of intestinal flora and urinary metabolites differed significantly between the YINDC and BC groups. A total of 13 obviously different genera and 55 altered metabolites were identified. Stool microbiome shifts were associated with urine metabolite changes. A combined marker comprising two genera may have a high potential to identify YINDC with an AUC of 0.845. Conclusions Infants with YINDC had a unique gut microbiota and metabolomic profile resulting in a constitutional microclassification. The altered gut microbiome in YINDC may account for the higher risk of cardiovascular diseases. Metabolomic analysis of urine showed that metabolic pathways, including histidine metabolism, proximal tubule bicarbonate reclamation, arginine biosynthesis, and steroid hormone biosynthesis, were altered in infants with YINDC. Additionally, the combined bacterial biomarker had the ability to identify YINDC. Identifying YINDC in infancy and intervening at an early stage is crucial for preventing cardiovascular diseases.
Collapse
|
150
|
Awan I, Schultz E, Sterrett JD, Dawud LM, Kessler LR, Schoch D, Lowry CA, Feldman-Winter L, Phadtare S. A Pilot Study Exploring Temporal Development of Gut Microbiome/Metabolome in Breastfed Neonates during the First Week of Life. Pediatr Gastroenterol Hepatol Nutr 2023; 26:99-115. [PMID: 36950061 PMCID: PMC10025571 DOI: 10.5223/pghn.2023.26.2.99] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/08/2022] [Accepted: 01/07/2023] [Indexed: 03/24/2023] Open
Abstract
PURPOSE Exclusive breastfeeding promotes gut microbial compositions associated with lower rates of metabolic and autoimmune diseases. Its cessation is implicated in increased microbiome-metabolome discordance, suggesting a vulnerability to dietary changes. Formula supplementation is common within our low-income, ethnic-minority community. We studied exclusively breastfed (EBF) neonates' early microbiome-metabolome coupling in efforts to build foundational knowledge needed to target this inequality. METHODS Maternal surveys and stool samples from seven EBF neonates at first transitional stool (0-24 hours), discharge (30-48 hours), and at first appointment (days 3-5) were collected. Survey included demographics, feeding method, medications, medical history and tobacco and alcohol use. Stool samples were processed for 16S rRNA gene sequencing and lipid analysis by gas chromatography-mass spectrometry. Alpha and beta diversity analyses and Procrustes randomization for associations were carried out. RESULTS Firmicutes, Proteobacteria, Bacteroidetes and Actinobacteria were the most abundant taxa. Variation in microbiome composition was greater between individuals than within (p=0.001). Palmitic, oleic, stearic, and linoleic acids were the most abundant lipids. Variation in lipid composition was greater between individuals than within (p=0.040). Multivariate composition of the metabolome, but not microbiome, correlated with time (p=0.030). Total lipids, saturated lipids, and unsaturated lipids concentrations increased over time (p=0.012, p=0.008, p=0.023). Alpha diversity did not correlate with time (p=0.403). Microbiome composition was not associated with each samples' metabolome (p=0.450). CONCLUSION Neonate gut microbiomes were unique to each neonate; respective metabolome profiles demonstrated generalizable temporal developments. The overall variability suggests potential interplay between influences including maternal breastmilk composition, amount consumed and living environment.
Collapse
Affiliation(s)
- Imad Awan
- Department of Medicine, Loma Linda University Medical Center, Loma Linda, CA, USA
| | - Emily Schultz
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - John D. Sterrett
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Lamya’a M. Dawud
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Lyanna R. Kessler
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Deborah Schoch
- Cooper Medical School of Rowan University and Cooper University Hospital, Camden, NJ, USA
| | - Christopher A. Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Lori Feldman-Winter
- Cooper Medical School of Rowan University and Cooper University Hospital, Camden, NJ, USA
| | - Sangita Phadtare
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| |
Collapse
|