1
|
Wang Y, Ding P, Zhang K, Xu X, Li H. Correlation Between Regulation of Intestinal Flora by Danggui-Shaoyao-San and Improvement of Cognitive Impairment in Mice With Alzheimer's Disease. Brain Behav 2024; 14:e70110. [PMID: 39482855 PMCID: PMC11527834 DOI: 10.1002/brb3.70110] [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: 06/17/2024] [Revised: 09/21/2024] [Accepted: 10/06/2024] [Indexed: 11/03/2024] Open
Abstract
PURPOSE The abnormal central glucose metabolism in Alzheimer's disease (AD) is related to the brain-gut axis. This study aims to explore the target of Danggui-Shaoyao-San (DSS) in improving cognitive impairment. METHOD This study analyzed the differences in mice intestinal flora by 16S rRNA sequencing. The cognitive protective effects of DSS were observed through the Morris water maze and the new object recognition. The mitigation effects of DSS on Aβ and p-tau, regulatory effects on glucose metabolism targets, and intestinal structure effects were observed through brain and colon slices staining. The differences in neural ultrastructure were compared by transmission electron microscopy. FINDING The results showed that DSS affected the composition of intestinal dominant bacteria and bacteria genera and regulated the abundance of intestinal bacteria in AD mice. DSS improved the behavior of AD mice, alleviated the deposition of AD pathological products in the brain and colon, regulated the expression of glycometabolism-related proteins, and improved the colon barrier structure and neural ultrastructure in the brain of mice with AD. CONCLUSION Our findings suggest that DSS may affect AD central glucose metabolism and improve cognition by regulating the gut-brain axis.
Collapse
Affiliation(s)
- Ya‐Han Wang
- Department of NeurologyAffiliated Hospital of Shandong University of Traditional Chinese MedicineJinanChina
| | - Peng‐Li Ding
- The First Clinical Medical CollegeShandong University of Traditional Chinese MedicineJinanChina
| | - Kai‐Xin Zhang
- The First Clinical Medical CollegeShandong University of Traditional Chinese MedicineJinanChina
| | - Xiang‐Qing Xu
- Department of NeurologyAffiliated Hospital of Shandong University of Traditional Chinese MedicineJinanChina
| | - He Li
- The First Clinical Medical CollegeShandong University of Traditional Chinese MedicineJinanChina
| |
Collapse
|
2
|
Qiu T, Fang Q, Tian X, Cao Y, Fan X, Li Y, Tu Y, Liu L, Chen Z, Wei Y, Bai J, Huang J, Liu Y. Time-varying ambient air pollution exposure is associated with gut microbiome variation in the first 2 years of life. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 360:124705. [PMID: 39134171 DOI: 10.1016/j.envpol.2024.124705] [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: 02/27/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024]
Abstract
The infant gut microbiome matures greatly in the first year of life. Ambient air pollution (AAP) exposure is associated with the infant gut microbiome. However, whether time-varying AAP influences infant gut microbiome variation is rarely investigated. This study aimed to investigate the effects of PM2.5, PM10, and O3 on infant gut microbiome variation longitudinally. Demographic information, stool samples, and AAP exposure concentrations were collected at 6, 12, 24 months from infants. Gut microbiome was processed and analyzed using 16S rRNA V3-V4 gene regions. AAP exposure concentrations were calculated using the China High Air Pollutants (CHAP) database. Multiple pollutant models were used to assess the mixed effects of PM2.5, PM10, and O3 on infant gut microbiome variation. Infants' gut microbiomes at 6, 12, 24 months old had significant differences in alpha diversity, beta diversity, and community composition. PM2.5 and O3 respectively explained 6.3% and 5.3% of the differences in community composition for 24-month-old infants. Single pollutant exposure and multiple pollutant exposure in different periods were both associated with alpha diversity indices and specific gut microbial phyla and genera. AAP was more associated with infant gut microbial alpha diversity indices, phyla variations, and genera variations at 12-24 months than 6-12 months. Multiple pollutant exposure in 0-2 lag months showed negative correlations with 12-24 months variation in Escherichia-Shigella (β = -0.854, 95%CI: 1.398 to -0.310) and Enterococcus (β = -0.979, 95%CI: 1.429 to -0.530). This study highlighted that time-varying PM2.5, PM10, and O3 synergistically influenced the variation of alpha diversity and abundance of gut microbial taxa in infants. Further research is needed to explore the effects and mechanisms of other environmental exposures on infant gut microbiome variation.
Collapse
Affiliation(s)
- Tianlai Qiu
- Wuhan University School of Nursing, Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Qingbo Fang
- Wuhan University School of Nursing, Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Xuqi Tian
- Wuhan University School of Nursing, Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Yanan Cao
- Wuhan University School of Nursing, Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Xiaoxiao Fan
- Wuhan University School of Nursing, Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Yanting Li
- Wuhan University School of Nursing, Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Yiming Tu
- Wuhan University School of Nursing, Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Linxia Liu
- Wuhan University School of Nursing, Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Zitong Chen
- Wuhan University School of Nursing, Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Yi Wei
- Wuhan University School of Nursing, Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Jinbing Bai
- Emory University Nell Hodgson Woodruff School of Nursing, 1520 Clifton Road, Atlanta, GA, 30322, USA
| | - Jing Huang
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, 100191, China
| | - Yanqun Liu
- Wuhan University School of Nursing, Wuhan University, 169 Donghu Road, Wuhan, 430071, China; Research Center for Lifespan Health, Wuhan University, 169 Donghu Road, Wuhan, 430071, China.
| |
Collapse
|
3
|
Hasegawa-Ishii S, Komaki S, Asano H, Imai R, Osaki T. Chronic nasal inflammation early in life induces transient and long-term dysbiosis of gut microbiota in mice. Brain Behav Immun Health 2024; 41:100848. [PMID: 39280089 PMCID: PMC11402449 DOI: 10.1016/j.bbih.2024.100848] [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: 01/08/2024] [Revised: 07/30/2024] [Accepted: 08/17/2024] [Indexed: 09/18/2024] Open
Abstract
The gut microbiota begins to colonize the host body following birth, develops during the suckling period and changes to the adult type after weaning. The early gut microbiota during the suckling period is thought to have profound effects on the host physiology throughout life but it is still unclear whether early dysbiosis is retained lifelong. Our previous study indicated that chronic nasal inflammation induces dysbiosis of gut microbiota in adult mice. In the present study, we addressed the question as to whether early exposure to chronic nasal inflammation induces dysbiosis, and if so, whether the dysbiosis is retained until adulthood and the sex differences in this effect. Male and female mice received repeated intranasal administration of lipopolysaccharide (LPS) or saline twice a week from P7 to P24 and were weaned at P24. The cecal contents were obtained for 16S rRNA analysis at 2 time points: at 4 weeks (wks), just after weaning, and at maturation to adulthood at 10 wks. The body weight did not differ between saline- and LPS-treated mice till around weaning, suggesting that the mothers' milk was given similarly to all mice. At 4 wks, the beta diversity was significantly different between saline- and LPS-treated male and female mice and the composition of the gut microbiota changed in LPS-treated mice. The abundance of phylum Bacteroidota tended to decrease and that of Firmicutes increased in LPS-treated male mice, while the abundance of Deferribacterota increased in LPS-treated female mice. At 10 wks, the beta diversity was not different between saline- and LPS-treated mice, but the abundance of family Lachnospiraceae significantly decreased in LPS-treated male and female mice by LEfSe analysis. Together, chronic nasal inflammation early in life caused transient and long-term dysbiosis of gut microbiota, which may contribute to the onset and progress of metabolic and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sanae Hasegawa-Ishii
- Pathology Research Team, Faculty of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka-shi, Tokyo, 181-8612, Japan
| | - Suzuho Komaki
- Pathology Research Team, Faculty of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka-shi, Tokyo, 181-8612, Japan
| | - Hinami Asano
- Pathology Research Team, Faculty of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka-shi, Tokyo, 181-8612, Japan
| | - Ryuichi Imai
- Pathology Research Team, Faculty of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka-shi, Tokyo, 181-8612, Japan
| | - Takako Osaki
- Department of Infectious Diseases, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611, Japan
| |
Collapse
|
4
|
Gulliver EL, Di Simone SK, Chonwerawong M, Forster SC. Unlocking the potential for microbiome-based therapeutics to address the sustainable development goal of good health and wellbeing. Microb Biotechnol 2024; 17:e70041. [PMID: 39487814 DOI: 10.1111/1751-7915.70041] [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: 08/16/2024] [Accepted: 10/17/2024] [Indexed: 11/04/2024] Open
Abstract
Recent years have witnessed major advances and an ever-growing list of healthcare applications for microbiome-based therapeutics. However, these advances have disproportionately targeted diseases common in high-income countries (HICs). Within low- to middle-income countries (LMIC), opportunities for microbiome-based therapeutics include sexual health epidemics, maternal health, early life mortality, malnutrition, vaccine response and infectious diseases. In this review we detail the advances that have been achieved in microbiome-based therapeutics for these areas of healthcare and identify where further work is required. Current efforts to characterise microbiomes from LMICs will aid in targeting and optimisation of therapeutics and preventative strategies specifically suited to the unmet needs within these populations. Once achieved, opportunities from disease treatment and improved treatment efficacy through to disease prevention and vector control can be effectively addressed using probiotics and live biotherapeutics. Together these strategies have the potential to increase individual health, overcome logistical challenges and reduce overall medical, individual, societal and economic costs.
Collapse
Affiliation(s)
- Emily L Gulliver
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Sara K Di Simone
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Ritchie Centre, Hudson, Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Paediatrics, Monash University, Melbourne, Victoria, Australia
| | - Michelle Chonwerawong
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Samuel C Forster
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
5
|
Donald K, Serapio-Palacios A, Gerbec Z, Bozorgmehr T, Holani R, Cruz AR, Schnupf P, Finlay BB. Secretory IgA in breast milk protects against asthma through modulation of the gut microbiota. Cell Rep 2024; 43:114835. [PMID: 39368092 DOI: 10.1016/j.celrep.2024.114835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 07/22/2024] [Accepted: 09/20/2024] [Indexed: 10/07/2024] Open
Abstract
Asthma susceptibility is linked to dysbiosis in early-life gut microbiota, and the antibody secretory immunoglobulin (Ig)A (SIgA) is a key determinant of gut microbiota composition. SIgA is obtained through breast milk during the critical early-life window. We use a mouse model of SIgA deficiency and the house dust mite (HDM) model of asthma to elucidate the role of maternal SIgA in modulating the early-life gut microbiota and asthma protection. Mice that do not receive maternal SIgA display a transient bloom of segmented filamentous bacteria (SFB) in the small intestine during the early post-weaning period. Mice that do not receive maternal SIgA also display elevated T helper type 17 (Th17) cell activation in the intestine, which persists into adulthood and is associated with more severe inflammation in response to the HDM model of asthma. This study demonstrates a mechanism by which breast-milk-derived SIgA influences immune development and asthma susceptibility by modulating the early-life gut microbiota.
Collapse
Affiliation(s)
- Katherine Donald
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Antonio Serapio-Palacios
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Zachary Gerbec
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
| | - Tahereh Bozorgmehr
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Ravi Holani
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Ana Raquel Cruz
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Laboratory of Host-Microbiota Interaction, 75015 Paris, France
| | - Pamela Schnupf
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Laboratory of Host-Microbiota Interaction, 75015 Paris, France
| | - B Brett Finlay
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| |
Collapse
|
6
|
Donald K, Finlay BB. Mechanisms of microbe-mediated immune development in the context of antibiotics and asthma. FRONTIERS IN ALLERGY 2024; 5:1469426. [PMID: 39469482 PMCID: PMC11513386 DOI: 10.3389/falgy.2024.1469426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 08/28/2024] [Indexed: 10/30/2024] Open
Abstract
The gut houses 70%-80% of the body's immune cells and represents the main point of contact between the immune system and the outside world. Immune maturation occurs largely after birth and is guided by the gut microbiota. In addition to the many human clinical studies that have identified relationships between gut microbiota composition and disease outcomes, experimental research has demonstrated a plethora of mechanisms by which specific microbes and microbial metabolites train the developing immune system. The healthy maturation of the gut microbiota has been well-characterized and discreet stages marked by changes in abundance of specific microbes have been identified. Building on Chapter 8, which discusses experimental models used to study the relationship between the gut microbiota and asthma, the present review aims to dive deeper into the specific microbes and metabolites that drive key processes in immune development. The implications of microbiota maturation patterns in the context of asthma and allergies, as well as the effects of antibiotics on microbe-immune crosstalk, will also be discussed.
Collapse
Affiliation(s)
- Katherine Donald
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Departmentof Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - B. Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Departmentof Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
7
|
Fischer MT, Xue KS, Costello EK, Dvorak M, Raboisson G, Robaczewska A, Caty SN, Relman DA, O’Connell LA. Effects of parental care on skin microbial community composition in poison frogs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612488. [PMID: 39314287 PMCID: PMC11419107 DOI: 10.1101/2024.09.11.612488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Parent-offspring interactions constitute the first contact of many newborns with their environment, priming community assembly of microbes through priority effects. Early exposure to microbes can have lasting influences on the assembly and functionality of the host's microbiota, leaving a life-long imprint on host health and disease. Studies of the role played by parental care in microbial acquisition have primarily focused on humans and hosts with agricultural relevance. Anuran vertebrates offer the opportunity to examine microbial community composition across life stages as a function of parental investment. In this study, we investigate vertical transmission of microbiota during parental care in a poison frog (Family Dendrobatidae), where fathers transport their offspring piggyback-style from terrestrial clutches to aquatic nurseries. We found that substantial bacterial colonization of the embryo begins after hatching from the vitelline envelope, emphasizing its potential role as microbial barrier during early development. Using a laboratory cross-foster experiment, we demonstrated that poison frogs performing tadpole transport serve as a source of skin microbes for tadpoles on their back. To study how transport impacts the microbial skin communities of tadpoles in an ecologically relevant setting, we sampled frogs and tadpoles of sympatric species that do or do not exhibit tadpole transport in their natural habitat. We found more diverse microbial communities associated with tadpoles of transporting species compared to a non-transporting frog. However, we detected no difference in the degree of similarity between adult and tadpole skin microbiotas, based on whether the frog species exhibits transporting behavior or not. Using a field experiment, we confirmed that tadpole transport can result in the persistent colonization of tadpoles by isolated microbial taxa associated with the caregiver's skin, albeit often at low prevalence. This is the first study to describe vertical transmission of skin microbes in anuran amphibians, showing that offspring transport may serve as a mechanism for transmission of parental skin microbes. Overall, these findings provide a foundation for further research on how vertical transmission in this order impacts host-associated microbiota and physiology.
Collapse
Affiliation(s)
| | - Katherine S. Xue
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Elizabeth K. Costello
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mai Dvorak
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Gaëlle Raboisson
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Anna Robaczewska
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - David A. Relman
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Infectious Diseases Section, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Lauren A. O’Connell
- Department of Biology, Stanford University, Stanford, CA 94305, USA
- Wu Tsai Institute for Neuroscience, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
8
|
Louvado A, Silva DAM, Oliveira V, Castro C, Cleary DFR, Gomes NCM. Association between Turbot ( Scophthalmus maximus) Fish Phenotype and the Post-Larval Bacteriome. Microorganisms 2024; 12:2014. [PMID: 39458323 PMCID: PMC11510086 DOI: 10.3390/microorganisms12102014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/17/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Over the past decade, an increasing number of studies have emphasized the importance of the host microbiome in influencing organismal health and development. Aligned with this understanding, our study aimed to investigate the potential association between the turbot (Scophthalmus maximus) phenotypic traits and the post-larval bacteriome. Turbot post-larvae were sampled from twenty randomly selected production cycles thirty days after hatching (DAH) across multiple post-larval production batches over a three-month period (April to June). Fish were selectively sampled based on five phenotypic traits, namely, normal, large, small, malformed, and depigmented. Our results showed that small-sized post-larvae had significantly higher bacterial phylogenetic diversity in their bacterial communities than all other phenotypes. A more in-depth compositional analysis also revealed specific associations between certain bacterial taxa and fish phenotypes. For example, the genera Aliivibrio and Sulfitobacter were enriched in small-sized post-larvae, while the family Micrococcaceae were predominantly found in larger post-larvae. Furthermore, genus Exiguobacterium was linked to depigmented larvae, and genus Pantoea was more prevalent in normal post-larvae. These observations underscore the importance of further research to understand the roles of these bacterial taxa in larval growth and phenotypic differentiation. Such insights could contribute to developing microbiome modulation strategies, which may enhance turbot post-larval health and quality and improve larviculture production.
Collapse
Affiliation(s)
- Antonio Louvado
- CESAM & Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal; (A.L.); (D.A.M.S.); (V.O.); (D.F.R.C.)
| | - Davide A. M. Silva
- CESAM & Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal; (A.L.); (D.A.M.S.); (V.O.); (D.F.R.C.)
| | - Vanessa Oliveira
- CESAM & Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal; (A.L.); (D.A.M.S.); (V.O.); (D.F.R.C.)
| | | | - Daniel F. R. Cleary
- CESAM & Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal; (A.L.); (D.A.M.S.); (V.O.); (D.F.R.C.)
| | - Newton C. M. Gomes
- CESAM & Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal; (A.L.); (D.A.M.S.); (V.O.); (D.F.R.C.)
| |
Collapse
|
9
|
Guo Z, Lei Y, Wang Q. Chinese expert consensus on standard technical specifications for a gut microecomics laboratory (Review). Exp Ther Med 2024; 28:403. [PMID: 39234587 PMCID: PMC11372251 DOI: 10.3892/etm.2024.12692] [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: 03/21/2024] [Accepted: 07/11/2024] [Indexed: 09/06/2024] Open
Abstract
The intestinal microbiota is a complex ecosystem that not only affects various physiological functions, such as metabolism, inflammation and the immune response, but also has an important effect on the development of tumors and response to treatment. The detection of intestinal flora enables the timely identification of disease-related flora abnormalities, which has significant implications for both disease prevention and treatment. In the field of basic and clinical research targeting gut microbiome, there is a need to recognize and understand the laboratory assays for gut microbiomics. Currently, there is no unified standard for the experimental procedure, quality management and report interpretation of intestinal microbiome assay technology. In order to clarify the process, the Tumor and Microecology Committee of China Anti-Cancer Association and the Tumor and Microecology Committee of Hubei Provincial Immunology Society organized relevant experts to discuss and put forward the standard technical specifications for gut microecomics laboratories, which provides a basis for further in-depth research in the field of intestinal microecomics.
Collapse
Affiliation(s)
- Zhi Guo
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong 518052, P.R. China
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, Hubei 430065, P.R. China
| | - Yumeng Lei
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, Hubei 430065, P.R. China
| | - Qiang Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, Hubei 430065, P.R. China
| |
Collapse
|
10
|
Li X, He E, Chen G, Cao X, Zhao L, Xu X, Fu Z, Qiu H. Intergenerational neurotoxicity of polystyrene nanoplastics in offspring mice is mediated by dysfunctional microbe-gut-brain axis. ENVIRONMENT INTERNATIONAL 2024; 192:109026. [PMID: 39321539 DOI: 10.1016/j.envint.2024.109026] [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: 05/21/2024] [Revised: 08/28/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
Nanoplastics (NPs) are ubiquitous in daily life, posing potential risks to the environment and human. While their negative effects on parental organisms have been extensively studied, intergenerational effects are still in the early stages of investigation. Here, we aimed to investigate the impact of maternal exposure to an environmentally relevant level of polystyrene NPs (PSNPs, 100 nm) during gestation and lactation (∼32 days, 50 μg/mouse/day) on neurotoxicity mediated by the microbe-gut-brain axis in offspring mice. Maternal PSNPs exposure significantly increased brain TNF-α level and microglia by 1.43 and 1.48 folds respectively, compared to control, accompanied by nuclear pyknosis and cell vacuolization in cortex and hippocampus. Targeted neurotransmitter metabolomics analysis revealed dysregulation in dopamine and serotonin metabolism. Specifically, dopamine levels increased significantly from 0.007 ng/L to 0.015 ng/L, while N-acetylseroton and 3,4-dihydroxyphenylacetic acid decreased significantly from 0.002 and 0.929 ng/L to 0.001 and 0.680 ng/L, respectively. Through a combination of 16S rRNA sequencing and biochemical analysis, we discovered that maternal PSNPs exposure led to a depletion of anti-inflammatory bacteria and an enrichment of pro-inflammatory bacteria resulting in intestinal barrier damage, elevated levels of lipopolysaccharide in blood, and subsequent activation of neuroinflammation. Meanwhile, gut bacteria dysbiosis interfered with communication between gut and brain by dysregulating neurotransmitter synthesis, as evidenced by significant associations between neurotransmitter-related bacteria (Akkermansia, Family_XIII_AD3011_group, Lachnoclostridium) and dopamine/serotonin related metabolites. Furthermore, transcriptional alterations in dopamine and serotonin related pathways were observed in the enteric nervous system, suggesting abnormal signal transduction from gut to brain contributes to neurotoxicity. This study provides new insights into NPs-induced neurotoxicity within the context of microbe-gut-brain axis and highlights the risk of cerebral dysfunction in offspring with maternal NPs exposure.
Collapse
Affiliation(s)
- Xing Li
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Erkai He
- School of Geographic Sciences, East China Normal University, Shanghai 200241, China
| | - Guangquan Chen
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 201204, China
| | - Xinde Cao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ling Zhao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoyun Xu
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhuozhong Fu
- School of Geographic Sciences, East China Normal University, Shanghai 200241, China
| | - Hao Qiu
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
11
|
Traidl-Hoffmann C, Afghani J, Akdis CA, Akdis M, Aydin H, Bärenfaller K, Behrendt H, Bieber T, Bigliardi P, Bigliardi-Qi M, Bonefeld CM, Bösch S, Brüggen MC, Diemert S, Duchna HW, Fähndrich M, Fehr D, Fellmann M, Frei R, Garvey LH, Gharbo R, Gökkaya M, Grando K, Guillet C, Guler E, Gutermuth J, Herrmann N, Hijnen DJ, Hülpüsch C, Irvine AD, Jensen-Jarolim E, Kong HH, Koren H, Lang CCV, Lauener R, Maintz L, Mantel PY, Maverakis E, Möhrenschlager M, Müller S, Nadeau K, Neumann AU, O'Mahony L, Rabenja FR, Renz H, Rhyner C, Rietschel E, Ring J, Roduit C, Sasaki M, Schenk M, Schröder J, Simon D, Simon HU, Sokolowska M, Ständer S, Steinhoff M, Piccirillo DS, Taïeb A, Takaoka R, Tapparo M, Teixeira H, Thyssen JP, Traidl S, Uhlmann M, van de Veen W, van Hage M, Virchow C, Wollenberg A, Yasutaka M, Zink A, Schmid-Grendelmeier P. Navigating the evolving landscape of atopic dermatitis: Challenges and future opportunities: The 4th Davos declaration. Allergy 2024; 79:2605-2624. [PMID: 39099205 DOI: 10.1111/all.16247] [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: 02/09/2024] [Revised: 06/18/2024] [Accepted: 07/04/2024] [Indexed: 08/06/2024]
Abstract
The 4th Davos Declaration was developed during the Global Allergy Forum in Davos which aimed to elevate the care of patients with atopic dermatitis (AD) by uniting experts and stakeholders. The forum addressed the high prevalence of AD, with a strategic focus on advancing research, treatment, and management to meet the evolving challenges in the field. This multidisciplinary forum brought together top leaders from research, clinical practice, policy, and patient advocacy to discuss the critical aspects of AD, including neuroimmunology, environmental factors, comorbidities, and breakthroughs in prevention, diagnosis, and treatment. The discussions were geared towards fostering a collaborative approach to integrate these advancements into practical, patient-centric care. The forum underlined the mounting burden of AD, attributing it to significant environmental and lifestyle changes. It acknowledged the progress in understanding AD and in developing targeted therapies but recognized a gap in translating these innovations into clinical practice. Emphasis was placed on the need for enhanced awareness, education, and stakeholder engagement to address this gap effectively and to consider environmental and lifestyle factors in a comprehensive disease management strategy. The 4th Davos Declaration marks a significant milestone in the journey to improve care for people with AD. By promoting a holistic approach that combines research, education, and clinical application, the Forum sets a roadmap for stakeholders to collaborate to improve patient outcomes in AD, reflecting a commitment to adapt and respond to the dynamic challenges of AD in a changing world.
Collapse
Affiliation(s)
- Claudia Traidl-Hoffmann
- Institute of Environmental Medicine and Integrative Health, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Jamie Afghani
- Institute of Environmental Medicine and Integrative Health, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Cezmi A Akdis
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Mübecel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | | | - Katja Bärenfaller
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Heidrun Behrendt
- Center for Allergy and Environment (ZAUM), Technische Universität München, Germany
| | - Thomas Bieber
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Davos Biosciences, Davos, Switzerland
| | | | | | - Charlotte Menné Bonefeld
- Department of Immunology and Microbiology, The LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| | - Stefanie Bösch
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
- Faculty of Medicine, University of Zürich, Zürich, Switzerland
| | - Marie Charlotte Brüggen
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
- Faculty of Medicine, University of Zürich, Zürich, Switzerland
| | | | - Hans-Werner Duchna
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Hochgebirgsklinik Davos, Davos, Switzerland
| | | | - Danielle Fehr
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
- Faculty of Medicine, University of Zürich, Zürich, Switzerland
| | | | - Remo Frei
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Bern University Hospital, Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Lena H Garvey
- Department of Dermatology and Allergy, Allergy Clinic, Copenhagen University Hospital-Herlev and Gentofte, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Raschid Gharbo
- Psychosomatic Department, Hochgebirgsklinik, Davos, Switzerland
| | - Mehmet Gökkaya
- Institute of Environmental Medicine and Integrative Health, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany
| | - Karin Grando
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
- Faculty of Medicine, University of Zürich, Zürich, Switzerland
| | - Carole Guillet
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
- Faculty of Medicine, University of Zürich, Zürich, Switzerland
| | | | | | - Nadine Herrmann
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - Dirk Jan Hijnen
- Diakonessenhuis Utrecht Zeist Doorn Locatie Utrecht, Erasmus MC, University Medical Center Utrecht, Utrecht, Netherlands
| | - Claudia Hülpüsch
- Institute of Environmental Medicine and Integrative Health, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Alan D Irvine
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| | - Erika Jensen-Jarolim
- Center of Pathophysiology, Infectiology and Immunology, Institute of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
- The interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Heidi H Kong
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Hillel Koren
- Environmental Health, LLC, Durham, North Carolina, USA
| | - Claudia C V Lang
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Immunology and Microbiology, The LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
| | - Roger Lauener
- Ostschweizer Kinderspital St. Gallen, St.Gallen, Switzerland
| | - Laura Maintz
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - Pierre-Yves Mantel
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Emanuel Maverakis
- Department of Dermatology, University of California Davis, Sacramento, California, USA
| | | | - Svenja Müller
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - Kari Nadeau
- Stanford University School of Medicine, Stanford, California, USA
| | - Avidan U Neumann
- Institute of Environmental Medicine and Integrative Health, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany
| | - Liam O'Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Medicine and School of Microbiology, University College Cork, Cork, Ireland
| | | | - Harald Renz
- Institute of Laboratory Medicine, Philipps University, Marburg, Germany
| | - Claudio Rhyner
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Ernst Rietschel
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Johannes Ring
- Klinik und Poliklinik für Dermatologie und Allergologie am Biederstein, Technische Universität München, Munich, Germany
| | - Caroline Roduit
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Bern University Hospital, Bern, Switzerland
- Ostschweizer Kinderspital St. Gallen, St.Gallen, Switzerland
| | - Mari Sasaki
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Bern University Hospital, Bern, Switzerland
| | - Mirjam Schenk
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Jens Schröder
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsklinikum Schleswig-Holstein (UK-SH), Kiel, Germany
| | - Dagmar Simon
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany
| | - Milena Sokolowska
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Sonja Ständer
- Center for Chronic Pruritus and Department of Dermatology, University Hospital Münster, Münster, Germany
| | - Martin Steinhoff
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- School of Medicine, Weill Cornell Medicine-Qatar, Ar-Rayyan, Qatar
- College of Medicine, Qatar University, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Department of Dermatology, Weill Cornell Medicine, New York, New York, USA
| | - Doris Straub Piccirillo
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Alain Taïeb
- INSERM 1312, University of Bordeaux, Bordeaux, France
| | - Roberto Takaoka
- Department of Dermatology, Faculdade de Medicina, Hospital das Clínicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | | | | | - Jacob Pontoppidan Thyssen
- Department of Dermatology and Venerology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Stephan Traidl
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Miriam Uhlmann
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Marianne van Hage
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institute and Karolinska University Hospital Stockholm, Solna, Sweden
| | - Christian Virchow
- Department of Pneumology, Intensive Care Medicine, Center for Internal Medicine, Universitätsmedizin Rostock, Rostock, Germany
| | - Andreas Wollenberg
- Department of Dermatology and Allergy, Ludwig-Maximilian-University, Munich, Germany
- Department of Dermatology and Allergy, University Hospital Augsburg, Augsburg, Germany
- Comprehensive Center of Inflammation Medicine, University Hospital Schleswig Holstein Campus Luebeck, Lubeck, Germany
| | - Mitamura Yasutaka
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Alexander Zink
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany
- Department of Medicine Solna, Division of Dermatology and Venereology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Schmid-Grendelmeier
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Immunology and Microbiology, The LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
| |
Collapse
|
12
|
Xiong ZY, Li HM, Qiu CS, Tang XL, Liao DQ, Du LY, Lai SM, Huang HX, Zhang BY, Kuang L, Li ZH. Investigating Causal Associations between the Gut Microbiota and Dementia: A Mendelian Randomization Study. Nutrients 2024; 16:3312. [PMID: 39408279 PMCID: PMC11479048 DOI: 10.3390/nu16193312] [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: 08/29/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Background: The causal association of specific gut microbiota with dementia remains incompletely understood. We aimed to access the causal relationships in which one or more gut microbiota account for dementia. Method: Using data from the MiBioGen and FinnGen consortia, we employed multiple Mendelian randomization (MR) approaches including two-sample MR (TSMR), multivariable MR (MVMR), and Bayesian model averaging MR to comprehensively evaluate the causal associations between 119 genera and dementia, and to prioritize the predominant bacterium. Result: We identified 21 genera that had causal effects on dementia and suggested Barnesiella (OR = 0.827, 95%CI = 0.722-0.948, marginal inclusion probability [MIP] = 0.464; model-averaged causal estimate [MACE] = -0.068) and Allisonella (OR = 0.770, 95%CI = 0.693-0.855, MIP = 0.898, MACE = -0.204) as the predominant genera for AD and all-cause dementia. Conclusions: These findings confirm the causal relationships between specific gut microbiota and dementia, highlighting the necessity of multiple MR approaches in gut microbiota analysis, and provides promising genera as potential novel biomarkers for dementia risk.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Zhi-Hao Li
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou 510515, China; (Z.-Y.X.); (H.-M.L.); (C.-S.Q.); (X.-L.T.); (D.-Q.L.); (L.-Y.D.); (S.-M.L.); (H.-X.H.); (B.-Y.Z.); (L.K.)
| |
Collapse
|
13
|
Xue Q, Lai H, Zhang H, Li G, Pi F, Wu Q, Liu S, Yang F, Chen T. Selenium Attenuates Radiation Colitis by Regulating cGAS-STING Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2403918. [PMID: 39348242 DOI: 10.1002/advs.202403918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/12/2024] [Indexed: 10/02/2024]
Abstract
Radiation colitis is one of the most common complications in patients undergoing pelvic radiotherapy and there is no effective treatment in the clinic. Therefore, searching for effective agents for the treatment of radiation colitis is urgently needed. Herein, it is found that the essential element selenium (Se) is protective against radiation colitis through inhibiting X-ray-induced apoptosis, cell cycle arrest, and inflammation with the involvement of balancing the generation of reactive oxygen species after the irradiation. Mechanistically, Se, especially for selenium nanoparticles (SeNPs), induced selenoprotein expression and then functioned to effectively restrain DNA damage response, which reduced X-ray-induced intestinal injury. Additionally, SeNPs treatment also restrained the cyclic GMP-AMP synthas (cGAS)- stimulator of interferon genes (STING)-TBK1-IRF3 signaling pathway cascade, thereby blocking the transcription of inflammatory cytokine gene, IL-6 and TNF-α, and thus alleviating inflammation. Moreover, inducing selenoprotein expression, such as GPX4, with SeNPs in vivo can regulate intestinal microenvironment immunity and gut microbiota to attenuate radiation-induced colitis by inhibiting oxidative stress and maintaining microenvironment immunity homeostasis. Together, these results unravel a previously unidentified modulation role that SeNPs restrained radiation colitis with the involvement of inducing selenoprotein expression but suppressing cGAS-STING-TBK1-IRF3 cascade.
Collapse
Affiliation(s)
- Qian Xue
- Department of Oncology of the First Affiliated Hospital, Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggalibility Assessment, Key laboratory of Viral Pathogenesis & Infection Prevention and Control, Ministry of Education, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China
| | - Haoqiang Lai
- Department of Oncology of the First Affiliated Hospital, Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggalibility Assessment, Key laboratory of Viral Pathogenesis & Infection Prevention and Control, Ministry of Education, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China
| | - Haimei Zhang
- Department of Oncology of the First Affiliated Hospital, Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggalibility Assessment, Key laboratory of Viral Pathogenesis & Infection Prevention and Control, Ministry of Education, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China
| | - Guizhen Li
- Department of Oncology of the First Affiliated Hospital, Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggalibility Assessment, Key laboratory of Viral Pathogenesis & Infection Prevention and Control, Ministry of Education, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China
| | - Fen Pi
- Department of Oncology of the First Affiliated Hospital, Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggalibility Assessment, Key laboratory of Viral Pathogenesis & Infection Prevention and Control, Ministry of Education, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China
| | - Qifeng Wu
- Department of Oncology of the First Affiliated Hospital, Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggalibility Assessment, Key laboratory of Viral Pathogenesis & Infection Prevention and Control, Ministry of Education, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China
| | - Siwei Liu
- Department of Oncology of the First Affiliated Hospital, Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggalibility Assessment, Key laboratory of Viral Pathogenesis & Infection Prevention and Control, Ministry of Education, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China
| | - Fang Yang
- Department of Oncology of the First Affiliated Hospital, Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggalibility Assessment, Key laboratory of Viral Pathogenesis & Infection Prevention and Control, Ministry of Education, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China
| | - Tianfeng Chen
- Department of Oncology of the First Affiliated Hospital, Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggalibility Assessment, Key laboratory of Viral Pathogenesis & Infection Prevention and Control, Ministry of Education, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
14
|
Wu Y, Chen B, Wu H, Gao J, Meng X, Chen H. How maternal factors shape the immune system of breastfed infants to alleviate food allergy: A systematic and updated review. Immunology 2024. [PMID: 39344356 DOI: 10.1111/imm.13864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 09/06/2024] [Indexed: 10/01/2024] Open
Abstract
What infants eat early in life may shape the immune system and have long-standing consequences on the health of the host during later life. In the early months post-birth, breast milk serves as the exclusive and optimal nourishment for infants, facilitating crucial molecular exchanges between mother and infant. Recent advances have uncovered that some maternal factors influence breastfed infant outcomes, including the risk of food allergy (FA). To date, accumulated data show that breastfed infants have a lower risk of FA. However, the issue remains disputed, some reported preventive allergy effects, while others did not confirm such effects, or if identified, protective effects were limited to early childhood. The disputed outcomes may be attributed to the maternal status, as it determines the compounds of the breast milk that breastfed infants are exposed to. In this review, we first detail the compounds in breast milk and their roles in infant FA. Then, we present maternal factors resulting in alterations in breast milk compounds, such as maternal health status, maternal diet intake, and maternal food allergen intake, which subsequently impact FA in breastfed infants. Finally, we analyze how these compounds in breast milk alleviated the infant FA by mother-to-infant transmission. Altogether, the mechanisms are primarily linked to the synergetic and direct effects of compounds in breast milk, via promoting the colonization of gut microbiota and the development of the immune system in infants.
Collapse
Affiliation(s)
- Yuhong Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science and Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
| | - Bihua Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science and Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
| | - Huan Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science and Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
| | - Jinyan Gao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science and Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
| | - Xuanyi Meng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
- Jiangxi-OAI Joint Research Institute, Nanchang University, Nanchang, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
- Jiangxi-OAI Joint Research Institute, Nanchang University, Nanchang, China
| |
Collapse
|
15
|
Ma Z, Zuo T, Frey N, Rangrez AY. A systematic framework for understanding the microbiome in human health and disease: from basic principles to clinical translation. Signal Transduct Target Ther 2024; 9:237. [PMID: 39307902 PMCID: PMC11418828 DOI: 10.1038/s41392-024-01946-6] [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: 03/05/2024] [Revised: 07/03/2024] [Accepted: 08/01/2024] [Indexed: 09/26/2024] Open
Abstract
The human microbiome is a complex and dynamic system that plays important roles in human health and disease. However, there remain limitations and theoretical gaps in our current understanding of the intricate relationship between microbes and humans. In this narrative review, we integrate the knowledge and insights from various fields, including anatomy, physiology, immunology, histology, genetics, and evolution, to propose a systematic framework. It introduces key concepts such as the 'innate and adaptive genomes', which enhance genetic and evolutionary comprehension of the human genome. The 'germ-free syndrome' challenges the traditional 'microbes as pathogens' view, advocating for the necessity of microbes for health. The 'slave tissue' concept underscores the symbiotic intricacies between human tissues and their microbial counterparts, highlighting the dynamic health implications of microbial interactions. 'Acquired microbial immunity' positions the microbiome as an adjunct to human immune systems, providing a rationale for probiotic therapies and prudent antibiotic use. The 'homeostatic reprogramming hypothesis' integrates the microbiome into the internal environment theory, potentially explaining the change in homeostatic indicators post-industrialization. The 'cell-microbe co-ecology model' elucidates the symbiotic regulation affecting cellular balance, while the 'meta-host model' broadens the host definition to include symbiotic microbes. The 'health-illness conversion model' encapsulates the innate and adaptive genomes' interplay and dysbiosis patterns. The aim here is to provide a more focused and coherent understanding of microbiome and highlight future research avenues that could lead to a more effective and efficient healthcare system.
Collapse
Affiliation(s)
- Ziqi Ma
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Tao Zuo
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Ashraf Yusuf Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
16
|
Qazi KR, Govindaraj D, Martí M, de Jong Y, Jensen GB, Abrahamsson T, Jenmalm MC, Sverremark-Ekström E. Impact of Extreme Prematurity, Chorioamnionitis, and Sepsis on Neonatal Monocyte Characteristics and Functions. J Innate Immun 2024; 16:470-488. [PMID: 39278208 PMCID: PMC11521501 DOI: 10.1159/000541468] [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: 04/19/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024] Open
Abstract
INTRODUCTION The innate branch of the immune system is important in early life, in particular for infants born preterm. METHODS We performed a longitudinal analysis of the peripheral monocyte compartment in extremely preterm children from a randomized, placebo-controlled study of probiotic supplementation. PBMCs and fecal samples were collected at several timepoints during the first months of life. Monocyte characteristics were analyzed by flow cytometry, and LPS-stimulated PBMC culture supernatants were analyzed by Luminex or ELISA. Plasma cytokines and gut microbiota composition were analyzed by ELISA and 16S rRNA-sequencing, respectively. RESULTS The extremely preterm infants had persistent alterations in their monocyte characteristics that were further aggravated in chorioamnionitis cases. They showed a markedly reduced TLR4 expression and hampered LPS-stimulated cytokine responses 14 days after birth. Notably, at later timepoints, TLR4 expression and LPS responses no longer correlated. Sepsis during the first weeks of life strongly associated with increased pro-inflammatory, and reduced IL-10, responses also at postmenstrual week 36. Further, we report a correlation between gut microbiota features and monocyte phenotype and responses, but also that probiotic supplementation associated with distinct monocyte phenotypic characteristics, without significantly influencing their responsiveness. CONCLUSION Extremely preterm infants have monocyte characteristics and functional features that deviate from infants born full-term. Some of these differences persist until they reach an age corresponding to full-term, potentially making them more vulnerable to microbial exposures during the first months of life.
Collapse
Affiliation(s)
- Khaleda Rahman Qazi
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Dhanapal Govindaraj
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Magalí Martí
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ymke de Jong
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Georg Bach Jensen
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Crown Princess Victoria Children’s Hospital, Linköping, Sweden
| | - Thomas Abrahamsson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Crown Princess Victoria Children’s Hospital, Linköping, Sweden
| | - Maria C. Jenmalm
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Eva Sverremark-Ekström
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
17
|
Cerovic V, Pabst O, Mowat AM. The renaissance of oral tolerance: merging tradition and new insights. Nat Rev Immunol 2024:10.1038/s41577-024-01077-7. [PMID: 39242920 DOI: 10.1038/s41577-024-01077-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 09/09/2024]
Abstract
Oral tolerance is the process by which feeding of soluble proteins induces antigen-specific systemic immune unresponsiveness. Oral tolerance is thought to have a central role in suppressing immune responses to 'harmless' food antigens, and its failure can lead to development of pathologies such as food allergies or coeliac disease. However, on the basis of long-standing experimental observations, the relevance of oral tolerance in human health has achieved new prominence recently following the discovery that oral administration of peanut proteins prevents the development of peanut allergy in at-risk human infants. In this Review, we summarize the new mechanistic insights into three key processes necessary for the induction of tolerance to oral antigens: antigen uptake and transport across the small intestinal epithelial barrier to the underlying immune cells; the processing, transport and presentation of fed antigen by different populations of antigen-presenting cells; and the development of immunosuppressive T cell populations that mediate antigen-specific tolerance. In addition, we consider how related but distinct processes maintain tolerance to bacterial antigens in the large intestine. Finally, we outline the molecular mechanisms and functional consequences of failure of oral tolerance and how these may be modulated to enhance clinical outcomes and prevent disease.
Collapse
Affiliation(s)
- Vuk Cerovic
- Institute of Molecular Medicine, RWTH Aachen University, Aachen, Germany.
| | - Oliver Pabst
- Institute of Molecular Medicine, RWTH Aachen University, Aachen, Germany
| | - Allan McI Mowat
- School of Infection and Immunity, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
18
|
Sardar P, Almeida A, Pedicord VA. Integrating functional metagenomics to decipher microbiome-immune interactions. Immunol Cell Biol 2024; 102:680-691. [PMID: 38952337 DOI: 10.1111/imcb.12798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/04/2024] [Accepted: 06/13/2024] [Indexed: 07/03/2024]
Abstract
Microbial metabolites can be viewed as the cytokines of the microbiome, transmitting information about the microbial and metabolic environment of the gut to orchestrate and modulate local and systemic immune responses. Still, many immunology studies focus solely on the taxonomy and community structure of the gut microbiota rather than its functions. Early sequencing-based microbiota profiling approaches relied on PCR amplification of small regions of bacterial and fungal genomes to facilitate identification of the microbes present. However, recent microbiome analysis methods, particularly shotgun metagenomic sequencing, now enable culture-independent profiling of microbiome functions and metabolites in addition to taxonomic characterization. In this review, we showcase recent advances in functional metagenomics methods and applications and discuss the current limitations and potential avenues for future development. Importantly, we highlight a few examples of key areas of opportunity in immunology research where integrating functional metagenomic analyses of the microbiome can substantially enhance a mechanistic understanding of microbiome-immune interactions and their contributions to health and disease states.
Collapse
Affiliation(s)
- Puspendu Sardar
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Alexandre Almeida
- Department of Veterinary Medicine, University of Cambridge School of Biological Sciences, Cambridge, UK
| | - Virginia A Pedicord
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| |
Collapse
|
19
|
Melki R, Litvak Y. From vacant to vivid: The nutritional landscape drives infant gut microbiota establishment. Mol Microbiol 2024; 122:347-356. [PMID: 39044538 DOI: 10.1111/mmi.15296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 06/25/2024] [Accepted: 07/03/2024] [Indexed: 07/25/2024]
Abstract
From the moment of birth, the newborn gastrointestinal tract is infiltrated by various bacteria originating from both maternal and environmental sources. These colonizing bacteria form a complex microbiota community that undergoes continuous changes until adulthood and plays an important role in infant health. The maturation of the infant gut microbiota is driven by many factors and follows a distinct patterned trajectory, with specific bacterial taxa establish in the intestine in accordance with developmental milestones as the infant grows. In this review, we highlight how elements such as diet and host physiology select for specific microbial functions and shape the composition of the bacterial community in the large intestine.
Collapse
Affiliation(s)
- Reut Melki
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yael Litvak
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
20
|
Xu Z, Chen M, Ng SC. Metabolic Regulation of Microbiota and Tissue Response. Gastroenterol Clin North Am 2024; 53:399-412. [PMID: 39068002 DOI: 10.1016/j.gtc.2024.01.003] [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: 07/30/2024]
Abstract
The microbiota in our gut regulates the sophisticated metabolic system that the human body has, essentially converting food into energy and the building blocks for various bodily functions. In this review, we discuss the multifaceted impact of the microbiota on host nutritional status by producing short-chain fatty acids, influencing gut hormones and mediating bile acid metabolism, and the key role in maintaining intestinal barrier integrity and immune homeostasis. Understanding and leveraging the power of the gut microbiome holds tremendous potential for enhancing human health and preventing various diseases.
Collapse
Affiliation(s)
- Zhilu Xu
- Microbiota I-Center (MagIC), Hong Kong SAR, China; Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Manman Chen
- Microbiota I-Center (MagIC), Hong Kong SAR, China; Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Siew Chien Ng
- Microbiota I-Center (MagIC), Hong Kong SAR, China; Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
21
|
Huang Y, Cao J, Zhu M, Wang Z, Jin Z, Xiong Z. Nontoxigenic Bacteroides fragilis: A double-edged sword. Microbiol Res 2024; 286:127796. [PMID: 38870618 DOI: 10.1016/j.micres.2024.127796] [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: 09/23/2023] [Revised: 04/12/2024] [Accepted: 05/31/2024] [Indexed: 06/15/2024]
Abstract
The contribution of commensal microbes to human health and disease is unknown. Bacteroides fragilis (B. fragilis) is an opportunistic pathogen and a common colonizer of the human gut. Nontoxigenic B. fragilis (NTBF) and enterotoxigenic B. fragilis (ETBF) are two kinds of B. fragilis. NTBF has been shown to affect the host immune system and interact with gut microbes and pathogenic microbes. Previous studies indicated that certain strains of B. fragilis have the potential to serve as probiotics, based on their observed relationship with the immune system. However, several recent studies have shown detrimental effects on the host when beneficial gut bacteria are found in the digestive system or elsewhere. In some pathological conditions, NTBF may have adverse reactions. This paper presents a comprehensive analysis of NTBF ecology from the host-microbe perspective, encompassing molecular disease mechanisms analysis, bacteria-bacteria interaction, bacteria-host interaction, and the intricate ecological context of the gut. Our review provides much-needed insights into the precise application of NTBF.
Collapse
Affiliation(s)
- Yumei Huang
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiali Cao
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mengpei Zhu
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ziwen Wang
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ze Jin
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhifan Xiong
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
22
|
Mamun AA, Zou C, Lishman H, Stenlund S, Xie M, Chuang E, Patrick DM. Association between antibiotic usage during infancy and asthma incidence among children: a population-level ecological study in British Columbia, Canada. FRONTIERS IN ALLERGY 2024; 5:1456077. [PMID: 39286476 PMCID: PMC11403638 DOI: 10.3389/falgy.2024.1456077] [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: 06/27/2024] [Accepted: 08/08/2024] [Indexed: 09/19/2024] Open
Abstract
Background This study follows published associations in BC to 2014 (updated in 2019) to model the predicted incidence of asthma in BC children attributable to antibiotic use within the context of reduced antibiotic use and increased breastfeeding in BC infants from 2000 to 2019. Methods A population-based ecological study was conducted in BC from 2000 to 2019, using outpatient antibiotic prescription data from BC PharmaNet and asthma diagnoses from the Chronic Disease Registry. Breastfeeding estimates were calculated using the Canadian Community Health Survey (CCHS). Population attributable risk (PAR) was calculated using a blended relative risk (RR) of asthma in antibiotic-exposed children who were and were not breastfed. PAR was used to calculate predicted vs. actual asthma incidence in 2019. Negative binomial regression was used to estimate the association between the average antibiotic prescription rate in infants under 1 and asthma incidence in 1-4 year olds, stratified by periods between 2000-2014 and 2015-2019. Results In BC, antibiotic prescribing decreased by 77% in infants under 1 and asthma incidence decreased by 41% in children 1-4 years from 2000 to 2019. BC breastfeeding rates increased from 46% in the 2005 CCHS to 71% in the 2017/18 CCHS. After calculating the PAR using a blended RR, the predicted asthma incidence in 2019 was 18.8/1,000 population. This was comparable to the observed asthma incidence in children 1-4 years of 16.6/1,000 population in 2019. During 2000-2014, adjusted incidence risk ratio (aIRR) for children under Quintile 5 of average antibiotic prescribing was 1.75 (95% CI: 1.63-1.88, P < 0.0001) times higher than that for Quintile 1. However, between 2015 and 2019, this association weakened (as expected because of increasing prevalence of breastfeeding), with the expected asthma incidence for Quintile 5 only 11% (aIRR 1.11, 95% CI: 0.78-1.57) higher than for Quintile 1. Conclusion We identified that over the past 20 years, antibiotic exposure in infants under 1 and asthma incidence in children 1-4 years has decreased significantly. Decreasing antibiotic exposure and increasing breastfeeding (which further mitigates risk associated with antibiotics) are of sufficient scale to explain much of this population trend. Changes in environmental, social and other exposures remain relevant to this complicated etiological pathway.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
- British Columbia Centre for Disease Control, Vancouver, BC, Canada
| | - Carl Zou
- British Columbia Centre for Disease Control, Vancouver, BC, Canada
| | - Hannah Lishman
- British Columbia Centre for Disease Control, Vancouver, BC, Canada
| | - Säde Stenlund
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
- British Columbia Centre for Disease Control, Vancouver, BC, Canada
| | - Max Xie
- British Columbia Centre for Disease Control, Vancouver, BC, Canada
| | - Erica Chuang
- British Columbia Centre for Disease Control, Vancouver, BC, Canada
| | - David M Patrick
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
- British Columbia Centre for Disease Control, Vancouver, BC, Canada
| |
Collapse
|
23
|
Cherayil BJ, Jain N. From Womb to World: Exploring the Immunological Connections between Mother and Child. Immunohorizons 2024; 8:552-562. [PMID: 39172025 PMCID: PMC11374749 DOI: 10.4049/immunohorizons.2400032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
Mother and child are immunologically interconnected by mechanisms that we are only beginning to understand. During pregnancy, multiple molecular and cellular factors of maternal origin are transferred across the placenta and influence the development and function of the fetal and newborn immune system. Altered maternal immune states arising from pregnancy-associated infections or immunizations have the potential to program offspring immune function in ways that may have long-term health consequences. In this study, we review current literature on the impact of prenatal infection and vaccination on the developing immune system, highlight knowledge gaps, and look to the horizon to envision maternal interventions that could benefit both the mother and her child.
Collapse
Affiliation(s)
- Bobby J Cherayil
- Mucosal Immunology and Biology Research Center, Mass General for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Nitya Jain
- Mucosal Immunology and Biology Research Center, Mass General for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Center for Computational and Integrative Biology, Mass General Brigham, Boston, MA
| |
Collapse
|
24
|
Yao H, Chen J, Wang Y, Li Y, Jiang Q. Assessing causal relationships between gut microbiota and abortion: evidence from two sample Mendelian randomization analysis. Front Endocrinol (Lausanne) 2024; 15:1415730. [PMID: 39050566 PMCID: PMC11266152 DOI: 10.3389/fendo.2024.1415730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Background While some studies have suggested a link between gut microbiota (GM) and abortion, the causal relationship remains unclear. Methods To explore the causal relationship between GM and abortion, including spontaneous abortion (SA) and habitual abortion (HA), we performed a two-sample Mendelian randomization (MR) analysis. We used summary statistics data from MiBioGen and FinnGen for genome-wide association studies (GWAS), with GM data as the exposure variable and abortion data as the outcome variable. Results In the absence of heterogeneity and horizontal pleiotropy, the inverse-variance weighted (IVW) method identified five genetically predicted GM genera linked to the risk of abortions. Lactococcus was negatively correlated with the risk of SA, whereas the Eubacterium fissicatena group was positively correlated with the risk of SA. Genetic predictions of Coprococcus3 and Odoribacter were linked to a reduced risk of HA, while the Eubacterium ruminantium group was associated with an increased risk of HA. Conclusion Our study suggests a genetic causal relationship between specific GM and two types of abortions, improving our understanding of the pathological relationship between GM and abortion.
Collapse
Affiliation(s)
- Hang Yao
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, China
| | - Jiahao Chen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu Wang
- Graduate School of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Yuxin Li
- Graduate School of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Qingling Jiang
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, China
| |
Collapse
|
25
|
Xue K, Li J, Huang R. The immunoregulatory role of gut microbiota in the incidence, progression, and therapy of breast cancer. Front Cell Infect Microbiol 2024; 14:1411249. [PMID: 39035351 PMCID: PMC11257971 DOI: 10.3389/fcimb.2024.1411249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/14/2024] [Indexed: 07/23/2024] Open
Abstract
Breast cancer (BrCa) is the most prevalent malignant tumor in women and one of the leading causes of female mortality. Its occurrence and progression are influenced by various factors, including genetics, environment, lifestyle, and hormones. In recent years, the gut microbiota has been identified as a significant factor affecting BrCa. The gut microbiota refers to the collective population of various microorganisms in the human gastrointestinal tract. Gut microbiota is closely associated with human health and disease development, participating in crucial physiological functions such as digestion, metabolism, immune response, and neural regulation. It has been found to influence the occurrence and treatment of BrCa through a variety of mechanisms. This article aims to review the immunomodulatory role of the gut microbiota in the development and treatment of BrCa.
Collapse
Affiliation(s)
| | | | - Ruijie Huang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
26
|
Zhao Y, Li C, Wu K, Chen H, Wang Q, Xiao Y, Yao S, Hong A, Zhang M, Lei S, Yang W, Zhong S, Umar A, Huang J, Yu Z. Exploring the Impact of Short Term Travel on Gut Microbiota and Probiotic Bacteria Mediated Stability. Biomedicines 2024; 12:1378. [PMID: 39061954 PMCID: PMC11274169 DOI: 10.3390/biomedicines12071378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 07/28/2024] Open
Abstract
Although travelers are frequently accompanied by abdominal discomfort and even diarrhea, not every trip can cause this issue. Many studies have reported that intestinal microbes play an important role in it. However, little is known about the reason for the dynamics of these intestinal microbes. Here, we delved into the effects of short-term travel on the gut microbiota of 12 healthy individuals. A total of 72 fecal samples collected before and after one-week travel, alongside non-traveling controls, underwent amplicon sequencing and a series of bioinformatic analyses. We found that travel significantly increased intra-individual gut microbiota fluctuations without diarrhea symptoms. In addition, the initial composition of the gut microbiota before travel emerged as a crucial factor in understanding these fluctuations. Travelers with stable microbiota exhibited an enrichment of specific probiotic bacteria (Agathobaculum, Faecalibacterium, Bifidobacterium, Roseburia, Lactobacillus) before travel. Another batch of data validated their predictive role in distinguishing travelers with and without the gut microbial disorder. This work provided valuable insights into understanding the relationship between gut microbiota and travel. It offered a microbiota-centric perspective and a potential avenue for interventions to preserve gut health during travel.
Collapse
Affiliation(s)
- Yiming Zhao
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| | - Chunyan Li
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| | - Kaijuan Wu
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha 410013, China; (K.W.); (A.H.); (W.Y.); (S.Z.); (J.H.)
| | - Hao Chen
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha 410013, China; (K.W.); (A.H.); (W.Y.); (S.Z.); (J.H.)
| | - Qingqun Wang
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| | - Ying Xiao
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| | - Siqi Yao
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| | - Ao Hong
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha 410013, China; (K.W.); (A.H.); (W.Y.); (S.Z.); (J.H.)
| | - Man Zhang
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| | - Shibo Lei
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| | - Wenyu Yang
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha 410013, China; (K.W.); (A.H.); (W.Y.); (S.Z.); (J.H.)
| | - Shukun Zhong
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha 410013, China; (K.W.); (A.H.); (W.Y.); (S.Z.); (J.H.)
| | - Abdulrahim Umar
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| | - Jing Huang
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha 410013, China; (K.W.); (A.H.); (W.Y.); (S.Z.); (J.H.)
| | - Zheng Yu
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| |
Collapse
|
27
|
Shama S, Asbury MR, O'Connor DL. From parent to progeny. Cell Host Microbe 2024; 32:947-949. [PMID: 38870905 DOI: 10.1016/j.chom.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 06/15/2024]
Abstract
How infants acquire their gut microbial communities and the various factors influencing these dynamics remain unclear. In this issue of Cell Host & Microbe, Selma-Royo et al. and Dubois et al. use shotgun metagenomic sequencing to understand the transmission of microbes from parents to infants and delve into factors modifying this process.
Collapse
Affiliation(s)
- Sara Shama
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, Canada
| | - Michelle R Asbury
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Canada; Department of Pediatrics, University of Calgary, Calgary, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada; Alberta Children's Hospital Research Institute, Calgary, Canada
| | - Deborah L O'Connor
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, Canada; Joannah and Brian Lawson Centre for Child Nutrition, University of Toronto, Toronto, Canada; Rogers Hixon Ontario Human Milk Bank, Toronto, Canada.
| |
Collapse
|
28
|
Schüller SS, Barman S, Mendez-Giraldez R, Soni D, Daley J, Baden LR, Levy O, Dowling DJ. Immune profiling of age and adjuvant-specific activation of human blood mononuclear cells in vitro. Commun Biol 2024; 7:709. [PMID: 38851856 PMCID: PMC11162429 DOI: 10.1038/s42003-024-06390-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 05/27/2024] [Indexed: 06/10/2024] Open
Abstract
Vaccination reduces morbidity and mortality due to infections, but efficacy may be limited due to distinct immunogenicity at the extremes of age. This raises the possibility of employing adjuvants to enhance immunogenicity and protection. Early IFNγ production is a hallmark of effective vaccine immunogenicity in adults serving as a biomarker that may predict effective adjuvanticity. We utilized mass cytometry (CyTOF) to dissect the source of adjuvant-induced cytokine production in human blood mononuclear cells (BMCs) from newborns (~39-week-gestation), adults (~18-63 years old) and elders (>65 years of age) after stimulation with pattern recognition receptors agonist (PRRa) adjuvants. Dimensionality reduction analysis of CyTOF data mapped the BMC compartment, elucidated age-specific immune responses and profiled PRR-mediated activation of monocytes and DCs upon adjuvant stimulation. Furthermore, we demonstrated PRRa adjuvants mediated innate IFNγ induction and mapped NK cells as the key source of TLR7/8 agonist (TLR7/8a) specific innate IFNγ responses. Hierarchical clustering analysis revealed age and TLR7/8a-specific accumulation of innate IFNγ producing γδ T cells. Our study demonstrates the application of mass cytometry and cutting-edge computational approaches to characterize immune responses across immunologically distinct age groups and may inform identification of the bespoke adjuvantation systems tailored to enhance immunity in distinct vulnerable populations.
Collapse
Affiliation(s)
- Simone S Schüller
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Neonatal Directorate, Child and Adolescent Health Service, Perth, Australia
| | - Soumik Barman
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Dheeraj Soni
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Sanofi, Cambridge, MA, USA
| | - John Daley
- Dana Farber CyTOF Core Facility, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Lindsey R Baden
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT & Harvard, Cambridge, MA, USA.
| | - David J Dowling
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
29
|
Deng Q, Liu L. One allergy: one exposure and one pathway. Pediatr Res 2024; 95:1683-1684. [PMID: 38307926 DOI: 10.1038/s41390-024-03060-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 01/07/2024] [Indexed: 02/04/2024]
Affiliation(s)
- Qihong Deng
- School of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| | - Le Liu
- School of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
30
|
Rook GAW. Evolution and the critical role of the microbiota in the reduced mental and physical health associated with low socioeconomic status (SES). Neurosci Biobehav Rev 2024; 161:105653. [PMID: 38582194 DOI: 10.1016/j.neubiorev.2024.105653] [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: 11/28/2023] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
The evolution of the gut-microbiota-brain axis in animals reveals that microbial inputs influence metabolism, the regulation of inflammation and the development of organs, including the brain. Inflammatory, neurodegenerative and psychiatric disorders are more prevalent in people of low socioeconomic status (SES). Many aspects of low SES reduce exposure to the microbial inputs on which we are in a state of evolved dependence, whereas the lifestyle of wealthy citizens maintains these exposures. This partially explains the health deficit of low SES, so focussing on our evolutionary history and on environmental and lifestyle factors that distort microbial exposures might help to mitigate that deficit. But the human microbiota is complex and we have poor understanding of its functions at the microbial and mechanistic levels, and in the brain. Perhaps its composition is more flexible than the microbiota of animals that have restricted habitats and less diverse diets? These uncertainties are discussed in relation to the encouraging but frustrating results of attempts to treat psychiatric disorders by modulating the microbiota.
Collapse
Affiliation(s)
- Graham A W Rook
- Centre for Clinical Microbiology, Department of infection, UCL (University College London), London, UK.
| |
Collapse
|
31
|
Nelson BN, Friedman JE. Developmental Programming of the Fetal Immune System by Maternal Western-Style Diet: Mechanisms and Implications for Disease Pathways in the Offspring. Int J Mol Sci 2024; 25:5951. [PMID: 38892139 PMCID: PMC11172957 DOI: 10.3390/ijms25115951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Maternal obesity and over/undernutrition can have a long-lasting impact on offspring health during critical periods in the first 1000 days of life. Children born to mothers with obesity have reduced immune responses to stimuli which increase susceptibility to infections. Recently, maternal western-style diets (WSDs), high in fat and simple sugars, have been associated with skewing neonatal immune cell development, and recent evidence suggests that dysregulation of innate immunity in early life has long-term consequences on metabolic diseases and behavioral disorders in later life. Several factors contribute to abnormal innate immune tolerance or trained immunity, including changes in gut microbiota, metabolites, and epigenetic modifications. Critical knowledge gaps remain regarding the mechanisms whereby these factors impact fetal and postnatal immune cell development, especially in precursor stem cells in bone marrow and fetal liver. Components of the maternal microbiota that are transferred from mothers consuming a WSD to their offspring are understudied and identifying cause and effect on neonatal innate and adaptive immune development needs to be refined. Tools including single-cell RNA-sequencing, epigenetic analysis, and spatial location of specific immune cells in liver and bone marrow are critical for understanding immune system programming. Considering the vital role immune function plays in offspring health, it will be important to understand how maternal diets can control developmental programming of innate and adaptive immunity.
Collapse
Affiliation(s)
- Benjamin N. Nelson
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Department of Physiology and Biochemistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Pediatrics, Section of Diabetes and Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
32
|
Rheinheimer N, Vacaru SV, van Immerseel JC, Kühn S, de Weerth C. Infant Care: Predictors of Outdoor Walking, Infant Carrying and Infant Outdoor Sleeping. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:694. [PMID: 38928940 PMCID: PMC11203610 DOI: 10.3390/ijerph21060694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Although spending time outdoors is beneficial for development, little is known about outdoor time during infancy. The aim of this study was to assess frequencies and durations of (1a) outdoor walking and carrying in mother-infant dyads and (1b) infant outdoor sleeping in a stationary cot or pram. We furthermore aimed to identify associations of (2a) outdoor walking and carrying and (2b) infant outdoor sleeping, with infant, maternal and environmental sample characteristics. METHODS An online survey was distributed among mothers of 0- to 12-month-old infants. Initially, 1453 mothers were recruited, of which 1275 were included in the analyses. With respect to (1a) the outcomes of interest were: mother-infant dyads' total weekly duration of walking in minutes, frequency of walking on weekdays, as well as weekends, and the frequency of using an infant carrier during walks, as well as the daily duration of carrying in hours (indoors and outdoors together). With respect to (1b) the outcome variables were: placing the infant outdoors to sleep (yes/no), the total weekly duration of outdoor sleeping and the weekly frequency of outdoor sleeping. For aim 2, associations of the outcome variables with infant (i.e., age), maternal (i.e., working status) and environmental (i.e., house type) sample characteristics were assessed. RESULTS Mother-infant dyads engaged in walks for a total weekly duration of 201 min, for approximately one to three walks over weekdays (Monday through Friday), as well as one to three walks on the weekend. The infant carrier was used by 22% of mothers at least half of the time during outdoor walks, and 18% reported a daily duration of infant carrying of one hour or more. Among other associations, infant and maternal enjoyment of outdoor walking correlated positively with the duration as well as the frequency of walking during weekdays and during the weekend. Furthermore, employed mothers walked for a shorter duration and less frequently on weekdays as compared to mothers on maternity leave or mothers without a paid job. The availability of nearby recreational areas correlated positively with the weekly duration and frequency of walks. The infant carrier was used more frequently during outdoor walks if more than one child lived in the household. Infant carrying during outdoor walks was also related to infant behavior at night. Roughly a third of the mothers (29%) regularly had their infant sleep outdoors for a weekly duration of four hours and a weekly frequency of approximately one to two times. Younger infants, infants of mothers with higher education and infants living in detached houses were more likely to be placed outdoors to sleep. DISCUSSION We identified associations of infant, maternal and environmental characteristics with outdoor time spent during infancy. These results lay the foundation for future research on the effects of the outdoors on child development as well as on facilitators and barriers for caregivers.
Collapse
Affiliation(s)
- Nicole Rheinheimer
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Stefania V. Vacaru
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Department of Psychology, New York University-Abu Dhabi, Abu Dhabi P.O. Box 129188, United Arab Emirates
- Department of Clinical Child and Family Studies & Amsterdam Public Health, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Julie C. van Immerseel
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Simone Kühn
- Center for Environmental Neuroscience, Max Planck Institute for Human Development, 14195 Berlin, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, 20249 Hamburg, Germany
| | - Carolina de Weerth
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
33
|
Sobel AL, Melamed J, Haas D, LeBlanc G, Cirone A, Constantinides MG. Antibiotic use in early life subsequently impairs MAIT cell-mediated immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.593643. [PMID: 38798453 PMCID: PMC11118404 DOI: 10.1101/2024.05.10.593643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Mucosal-associated invariant T (MAIT) cells are predominantly located in barrier tissues where they rapidly respond to pathogens and commensals by recognizing microbial derivatives of riboflavin synthesis. Early-life exposure to these metabolites imprints the abundance of MAIT cells within tissues, so we hypothesized that antibiotic use during this period may abrogate their development. We identified antibiotics that deplete riboflavin-synthesizing commensals and revealed an early period of susceptibility during which antibiotic administration impaired MAIT cell development. The reduction in MAIT cell abundance rendered mice more susceptible to pneumonia, while MAIT cell-deficient mice were unaffected by early-life antibiotics. Concomitant administration of a riboflavin-synthesizing commensal during antibiotic treatment was sufficient to restore MAIT cell development and immunity. Our work demonstrates that transient depletion of riboflavin-synthesizing commensals in early life can adversely affect responses to subsequent infections.
Collapse
|
34
|
Wen M, Chen S, Zhang Y, Liu Y, Tang C, Zhang J, Sun J, Li X, Ding Y, Lu L, Long K, Nie Y, Li X, Li M, Ge L, Ma J. Diversity and host interaction of the gut microbiota in specific pathogen-free pigs. Front Microbiol 2024; 15:1402807. [PMID: 38800748 PMCID: PMC11122924 DOI: 10.3389/fmicb.2024.1402807] [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: 03/18/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Pigs are widely used as animal models in various studies related to humans. The interaction between the gut microbiota and the host has significant effects on the host's health and disease status. However, although there have been many studies investigating the pig gut microbiota, the findings have been inconsistent due to variations in rearing conditions. Interactions between the gut microbiota and host have not been fully explored in pigs. Specific pathogen-free (SPF) pigs are ideal non-primate large animals to study the interactions between the gut microbiota and the host. In this study, we performed high-throughput sequencing analysis of the gut microbiota and the gut tissue transcriptome of six SPF pigs to provide a systematic understanding of the composition, function, and spatial distribution of gut microbiota in SPF pigs. We identified significant differences in microbial diversity and functionality among different gastrointestinal tract sites. Metagenomics data analysis revealed significant differences in alpha diversity and beta diversity of microbiota in different gastrointestinal sites of SPF pigs. Additionally, transcriptomic data indicated significant differences in gene expression as well as KEGG and GO functional enrichment between the small intestine and large intestine. Furthermore, by combining microbial metagenomics and host transcriptomics analyses, specific correlations were found between gut microbiota and host genes. These included a negative correlation between the TCN1 gene and Prevotella dentalis, possibly related to bacterial metabolic pathways involving vitamin B12, and a positive correlation between the BDH1 gene and Roseburia hominis, possibly because both are involved in fatty acid metabolism. These findings lay the groundwork for further exploration of the co-evolution between the microbiota and the host, specifically in relation to nutrition, metabolism, and immunity. In conclusion, we have elucidated the diversity of the gut microbiota in SPF pigs and conducted a detailed investigation into the interactions between the gut microbiota and host gene expression. These results contribute to our understanding of the intricate dynamics between the gut microbiota and the host, offering important references for advancements in life science research, bioproduct production, and sustainable development in animal husbandry.
Collapse
Affiliation(s)
- Mingxing Wen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Shuangshuang Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yali Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yan Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Chuang Tang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jinwei Zhang
- Chongqing Academy of Animal Sciences, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
- Ministry of Agriculture Key Laboratory of Pig Sciences, Chongqing Key Laboratory of Pig Sciences, Chongqing, China
| | - Jing Sun
- Chongqing Academy of Animal Sciences, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
- Ministry of Agriculture Key Laboratory of Pig Sciences, Chongqing Key Laboratory of Pig Sciences, Chongqing, China
| | - Xiaokai Li
- National Center of Technology Innovation for Pigs, Chongqing, China
- Ministry of Agriculture Key Laboratory of Pig Sciences, Chongqing Key Laboratory of Pig Sciences, Chongqing, China
| | - Yuchun Ding
- Chongqing Academy of Animal Sciences, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
- Ministry of Agriculture Key Laboratory of Pig Sciences, Chongqing Key Laboratory of Pig Sciences, Chongqing, China
| | - Lu Lu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Keren Long
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yong Nie
- College of Engineering, Peking University, Beijing, China
| | - Xuewei Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Mingzhou Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Liangpeng Ge
- Chongqing Academy of Animal Sciences, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
- Ministry of Agriculture Key Laboratory of Pig Sciences, Chongqing Key Laboratory of Pig Sciences, Chongqing, China
| | - Jideng Ma
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
35
|
Paller AS, Scharschmidt TC, Kezic S, Irvine AD. Preclinical Atopic Dermatitis Skin in Infants: An Emerging Research Area. J Invest Dermatol 2024; 144:1001-1009. [PMID: 38573278 DOI: 10.1016/j.jid.2024.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 04/05/2024]
Abstract
Whereas clinically apparent atopic dermatitis (AD) can be confirmed by validated diagnostic criteria, the preclinical phenotype of infants who eventually develop AD is less well-characterized. Analogous to unaffected or nonlesional skin in established AD, clinically normal-appearing skin in infants who will develop clinical AD has distinct changes. Prospective studies have revealed insights into this preclinical AD phenotype. In this study, we review the structural, immunologic, and microbiome nature of the preclinical AD phenotype. Determination of markers that predict the development of AD will facilitate targeting of interventions to prevent the development or reduce the severity of AD in infants.
Collapse
Affiliation(s)
- Amy S Paller
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| | - Tiffany C Scharschmidt
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Sanja Kezic
- Department of Public and Occupational Health, Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Alan D Irvine
- Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
36
|
Xu Y, Li Y. Association between lipid-lowering drugs and allergic diseases: A Mendelian randomization study. World Allergy Organ J 2024; 17:100899. [PMID: 38623319 PMCID: PMC11017355 DOI: 10.1016/j.waojou.2024.100899] [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: 08/08/2023] [Revised: 02/28/2024] [Accepted: 03/18/2024] [Indexed: 04/17/2024] Open
Abstract
Background Several observational studies suggest a possible link between lipid-lowering drugs and allergic diseases. However, inferring causality from these studies can be challenging due to issues such as bias, reverse causation, and residual confounding. To investigate the potential causal effect of lipid-lowering drugs, proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors and 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR) inhibitors, on allergic diseases (allergic asthma, allergic conjunctivitis, atopic dermatitis, allergic rhinitis, and allergic urticaria), we performed a Mendelian randomization (MR)-based study. Methods We employed MR and summary-data-based MR (SMR), analyzing genome-wide association study (GWAS) data from people of European descent. Single nucleotide polymorphisms (SNPs) were employed as instrumental variables. We selected 2 types of genetic measures to represent the impact of lipid-lowering drugs, including genetic variants near or within drug target genes correlated with low-density lipoprotein cholesterol (LDL-C), and expression quantitative trait loci of drug target genes. The inverse-variance weighted (IVW)-MR approach was the primary utilized MR method, while sensitivity analyses were used to test the robustness of the results. We used SMR analysis as a supplementary analytical method, applying the heterogeneity in dependent instruments (HEIDI) test to assess if the observed correlation between gene expression and outcome was due to a linkage situation. Results The IVW-MR analysis revealed significant evidence for an association between PCSK9-mediated LDL-C reduction and a decrease in the risk of allergic asthma (odds ratio [OR] = 1.31, 95% confidence interval [CI] = 1.11-1.56; P < 0.01). Likewise, SMR analysis discovered an augmented expression of PCSK9 being linked with a heightened susceptibility to allergic asthma (OR = 1.21, 95% CI = 1.03-1.43; P = 0.02). No consistent evidence was found for other associations in either analysis. Conclusion Our findings support a potential causal relationship between PCSK9 activity and an increased risk of allergic asthma. Thus, PCSK9 inhibitors, which reduce PCSK9 activity, might be considered a priority in future clinical trials investigating drugs for allergic asthma prevention or treatment.
Collapse
Affiliation(s)
- Yinsong Xu
- Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
- Ya'an Polytechnic College, Ya'an, 625000, Sichuan, China
| | - Yuanzhi Li
- Department of Anorectal Surgery, Shenzhen TCM Anorectal Hospital (Futian), Shenzhen, 518000, China
- Department of Traditional Chinese Medicine, The Afliated Hospital of Southwest Medical University, Luzhou, 646000, China
| |
Collapse
|
37
|
侯 林, 马 紫, 晁 爽, 李 中, 张 宇, 刘 毅, 张 俊, 武 文, 黄山 雅, 刘 捷. [Risk factors for cow's milk protein allergy in infants: a multicenter prospective nested case-control study]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:230-235. [PMID: 38557373 PMCID: PMC10986379 DOI: 10.7499/j.issn.1008-8830.2309105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/29/2023] [Indexed: 04/04/2024]
Abstract
OBJECTIVES To explore the risk factors associated with cow's milk protein allergy (CMPA) in infants. METHODS This study was a multicenter prospective nested case-control study conducted in seven medical centers in Beijing, China. Infants aged 0-12 months were included, with 200 cases of CMPA infants and 799 control infants without CMPA. Univariate and multivariate logistic regression analyses were used to investigate the risk factors for the occurrence of CMPA. RESULTS Univariate logistic regression analysis showed that preterm birth, low birth weight, birth from the first pregnancy, firstborn, spring birth, summer birth, mixed/artificial feeding, and parental history of allergic diseases were associated with an increased risk of CMPA in infants (P<0.05). Multivariate logistic regression analysis revealed that firstborn (OR=1.89, 95%CI: 1.14-3.13), spring birth (OR=3.42, 95%CI: 1.70-6.58), summer birth (OR=2.29, 95%CI: 1.22-4.27), mixed/artificial feeding (OR=1.57, 95%CI: 1.10-2.26), parental history of allergies (OR=2.13, 95%CI: 1.51-3.02), and both parents having allergies (OR=3.15, 95%CI: 1.78-5.56) were risk factors for CMPA in infants (P<0.05). CONCLUSIONS Firstborn, spring birth, summer birth, mixed/artificial feeding, and a family history of allergies are associated with an increased risk of CMPA in infants.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - 文艳 武
- 北京华信医院(清华大学第一附属医院)儿科北京100016
| | | | | |
Collapse
|
38
|
Bernstein JA, Bernstein JS, Makol R, Ward S. Allergic Rhinitis: A Review. JAMA 2024; 331:866-877. [PMID: 38470381 DOI: 10.1001/jama.2024.0530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Importance Allergic rhinitis affects an estimated 15% of the US population (approximately 50 million individuals) and is associated with the presence of asthma, eczema, chronic or recurrent sinusitis, cough, and both tension and migraine headaches. Observations Allergic rhinitis occurs when disruption of the epithelial barrier allows allergens to penetrate the mucosal epithelium of nasal passages, inducing a T-helper type 2 inflammatory response and production of allergen-specific IgE. Allergic rhinitis typically presents with symptoms of nasal congestion, rhinorrhea, postnasal drainage, sneezing, and itching of the eyes, nose, and throat. In an international study, the most common symptoms of allergic rhinitis were rhinorrhea (90.38%) and nasal congestion (94.23%). Patients with nonallergic rhinitis present primarily with nasal congestion and postnasal drainage frequently associated with sinus pressure, ear plugging, muffled sounds and pain, and eustachian tube dysfunction that is less responsive to nasal corticosteroids. Patients with seasonal allergic rhinitis typically have physical examination findings of edematous and pale turbinates. Patients with perennial allergic rhinitis typically have erythematous and inflamed turbinates with serous secretions that appear similar to other forms of chronic rhinitis at physical examination. Patients with nonallergic rhinitis have negative test results for specific IgE aeroallergens. Intermittent allergic rhinitis is defined as symptoms occurring less than 4 consecutive days/week or less than 4 consecutive weeks/year. Persistent allergic rhinitis is defined as symptoms occurring more often than 4 consecutive days/week and for more than 4 consecutive weeks/year. Patients with allergic rhinitis should avoid inciting allergens. In addition, first-line treatment for mild intermittent or mild persistent allergic rhinitis may include a second-generation H1 antihistamine (eg, cetirizine, fexofenadine, desloratadine, loratadine) or an intranasal antihistamine (eg, azelastine, olopatadine), whereas patients with persistent moderate to severe allergic rhinitis should be treated initially with an intranasal corticosteroid (eg, fluticasone, triamcinolone, budesonide, mometasone) either alone or in combination with an intranasal antihistamine. In contrast, first-line therapy for patients with nonallergic rhinitis consists of an intranasal antihistamine as monotherapy or in combination with an intranasal corticosteroid. Conclusions and Relevance Allergic rhinitis is associated with symptoms of nasal congestion, sneezing, and itching of the eyes, nose, and throat. Patients with allergic rhinitis should be instructed to avoid inciting allergens. Therapies include second-generation H1 antihistamines (eg, cetirizine, fexofenadine, desloratadine, loratadine), intranasal antihistamines (eg, azelastine, olopatadine), and intranasal corticosteroids (eg, fluticasone, triamcinolone, budesonide, mometasone) and should be selected based on the severity and frequency of symptoms and patient preference.
Collapse
Affiliation(s)
- Jonathan A Bernstein
- Division of Rheumatology, Allergy, and Immunology, Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Joshua S Bernstein
- Division of Rheumatology, Allergy, and Immunology, Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Richika Makol
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Stephanie Ward
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
39
|
Ananthakrishnan AN. Inflammatory bowel diseases: are we ready to recommend a preventive diet for infants? Gut 2024; 73:559-560. [PMID: 38290831 DOI: 10.1136/gutjnl-2023-331849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Affiliation(s)
- Ashwin N Ananthakrishnan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
40
|
Kahhaleh FG, Barrientos G, Conrad ML. The gut-lung axis and asthma susceptibility in early life. Acta Physiol (Oxf) 2024; 240:e14092. [PMID: 38251788 DOI: 10.1111/apha.14092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/06/2023] [Accepted: 01/01/2024] [Indexed: 01/23/2024]
Abstract
Asthma is the most common chronic disease among children, with more than 300 million cases worldwide. Over the past several decades, asthma incidence has grown, and epidemiological studies identify the modernized lifestyle as playing a strong contributing role in this phenomenon. In particular, lifestyle factors that modify the maternal gut microbiome during pregnancy, or the infant microbiome in early life, can act as developmental programming events which determine health or disease susceptibility later in life. Microbial colonization of the gut begins at birth, and factors such as delivery mode, breastfeeding, diet, antibiotic use, and exposure to environmental bacteria influence the development of the infant microbiome. Colonization of the gut microbiome is crucial for proper immune system development and disruptions to this process can predispose a child to asthma development. Here, we describe the importance of early-life events for shaping immune responses along the gut-lung axis and why they may provide a window of opportunity for asthma prevention.
Collapse
Affiliation(s)
- Fariz G Kahhaleh
- Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gabriela Barrientos
- Laboratory of Experimental Medicine, Hospital Alemán, Buenos Aires, Argentina
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Melanie L Conrad
- Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
41
|
Zubeldia-Varela E, Ibáñez-Sandín MD, Gomez-Casado C, Pérez-Gordo M. Allergy-associated biomarkers in early life identified by Omics techniques. FRONTIERS IN ALLERGY 2024; 5:1359142. [PMID: 38464396 PMCID: PMC10920277 DOI: 10.3389/falgy.2024.1359142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/12/2024] [Indexed: 03/12/2024] Open
Abstract
The prevalence and severity of allergic diseases have increased over the last 30 years. Understanding the mechanisms responsible for these diseases is a major challenge in current allergology, as it is crucial for the transition towards precision medicine, which encompasses predictive, preventive, and personalized strategies. The urge to identify predictive biomarkers of allergy at early stages of life is crucial, especially in the context of major allergic diseases such as food allergy and atopic dermatitis. Identifying these biomarkers could enhance our understanding of the immature immune responses, improve allergy handling at early ages and pave the way for preventive and therapeutic approaches. This minireview aims to explore the relevance of three biomarker categories (proteome, microbiome, and metabolome) in early life. First, levels of some proteins emerge as potential indicators of mucosal health and metabolic status in certain allergic diseases. Second, bacterial taxonomy provides insight into the composition of the microbiota through high-throughput sequencing methods. Finally, metabolites, representing the end products of bacterial and host metabolic activity, serve as early indicators of changes in microbiota and host metabolism. This information could help to develop an extensive identification of biomarkers in AD and FA and their potential in translational personalized medicine in early life.
Collapse
Affiliation(s)
- Elisa Zubeldia-Varela
- Institute of Applied Molecular Medicine Nemesio Díez (IMMA), Department of Basic Medical Sciences, Facultad de Medicina. Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - María Dolores Ibáñez-Sandín
- Department of Allergy, H. Infantil Universitario Niño Jesús, FibHNJ, ARADyAL- RETICs Instituto de Salud Carlos III, IIS-P, Madrid, Spain
| | - Cristina Gomez-Casado
- Department of Dermatology, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, Germany
| | - Marina Pérez-Gordo
- Institute of Applied Molecular Medicine Nemesio Díez (IMMA), Department of Basic Medical Sciences, Facultad de Medicina. Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| |
Collapse
|
42
|
Loh JS, Mak WQ, Tan LKS, Ng CX, Chan HH, Yeow SH, Foo JB, Ong YS, How CW, Khaw KY. Microbiota-gut-brain axis and its therapeutic applications in neurodegenerative diseases. Signal Transduct Target Ther 2024; 9:37. [PMID: 38360862 PMCID: PMC10869798 DOI: 10.1038/s41392-024-01743-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 01/02/2024] [Accepted: 01/14/2024] [Indexed: 02/17/2024] Open
Abstract
The human gastrointestinal tract is populated with a diverse microbial community. The vast genetic and metabolic potential of the gut microbiome underpins its ubiquity in nearly every aspect of human biology, including health maintenance, development, aging, and disease. The advent of new sequencing technologies and culture-independent methods has allowed researchers to move beyond correlative studies toward mechanistic explorations to shed light on microbiome-host interactions. Evidence has unveiled the bidirectional communication between the gut microbiome and the central nervous system, referred to as the "microbiota-gut-brain axis". The microbiota-gut-brain axis represents an important regulator of glial functions, making it an actionable target to ameliorate the development and progression of neurodegenerative diseases. In this review, we discuss the mechanisms of the microbiota-gut-brain axis in neurodegenerative diseases. As the gut microbiome provides essential cues to microglia, astrocytes, and oligodendrocytes, we examine the communications between gut microbiota and these glial cells during healthy states and neurodegenerative diseases. Subsequently, we discuss the mechanisms of the microbiota-gut-brain axis in neurodegenerative diseases using a metabolite-centric approach, while also examining the role of gut microbiota-related neurotransmitters and gut hormones. Next, we examine the potential of targeting the intestinal barrier, blood-brain barrier, meninges, and peripheral immune system to counteract glial dysfunction in neurodegeneration. Finally, we conclude by assessing the pre-clinical and clinical evidence of probiotics, prebiotics, and fecal microbiota transplantation in neurodegenerative diseases. A thorough comprehension of the microbiota-gut-brain axis will foster the development of effective therapeutic interventions for the management of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jian Sheng Loh
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Wen Qi Mak
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Li Kar Stella Tan
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
- Digital Health & Medical Advancements, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
| | - Chu Xin Ng
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
| | - Hong Hao Chan
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Shiau Hueh Yeow
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
- Digital Health & Medical Advancements, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
| | - Yong Sze Ong
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Chee Wun How
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia.
| | - Kooi Yeong Khaw
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
43
|
Jin W, Peng J, Dai J, Tang R, Guo J, Zhao H, Wang J, Zhang S, Gao Y. Bacterial load in meconium. IMETA 2024; 3:e173. [PMID: 38868517 PMCID: PMC10989067 DOI: 10.1002/imt2.173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/19/2024] [Accepted: 01/19/2024] [Indexed: 06/14/2024]
Abstract
The spike-in plasmid method was utilized to perform an analysis on meconium and second-pass feces, yielding both relative and absolute quantitative results. With the absolute quantitative data, the abundance of bacteria in 17 meconium samples and 17 second-pass fecal samples were found to be 1.14 × 107 and 1.59 × 109 copies/g, respectively. The mode of delivery can significantly influence the alterations and compositions of gut bacteria in a newborn within 72 h.
Collapse
Affiliation(s)
- Wen‐Yu Jin
- The Center for Microbes, Development and Health, Shanghai Institute of Immunity and InfectionChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jing Peng
- Obstetrics and Gynecology Hospital of Fudan UniversityShanghaiChina
| | - Jinping Dai
- Obstetrics and Gynecology Hospital of Fudan UniversityShanghaiChina
| | - Rongkang Tang
- The Center for Microbes, Development and Health, Shanghai Institute of Immunity and InfectionChinese Academy of SciencesShanghaiChina
| | - Jia‐Xin Guo
- The Center for Microbes, Development and Health, Shanghai Institute of Immunity and InfectionChinese Academy of SciencesShanghaiChina
| | - Huan Zhao
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jielin Wang
- Hongqiao International Institute of Medicine, Tongren HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shu Zhang
- Department of Gynecological OncologyFudan University Shanghai Cancer CenterShanghaiChina
| | - Yi‐Zhou Gao
- The Center for Microbes, Development and Health, Shanghai Institute of Immunity and InfectionChinese Academy of SciencesShanghaiChina
| |
Collapse
|
44
|
Lin C, Lin Y, Wang S, Wang J, Mao X, Zhou Y, Zhang H, Chen W, Wang G. Bifidobacterium animalis subsp. lactis boosts neonatal immunity: unravelling systemic defences against Salmonella. Food Funct 2024; 15:236-254. [PMID: 38054827 DOI: 10.1039/d3fo03686c] [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: 12/07/2023]
Abstract
Bifidobacterium animalis subsp. lactis may be a useful probiotic intervention for regulating neonatal intestinal immune responses and counteracting Salmonella infection. However, recent research has focused on intestinal immunity, leaving uncertainties regarding the central, peripheral, and neural immune responses in neonates. Therefore, this study investigated the role and mechanisms of B. animalis subsp. lactis in the systemic immune responses of neonatal rats following Salmonella infection. Through extremely early pretreatment with B. animalis subsp. lactis (6 hours postnatal), the neonatal rat gut microbiota was effectively reshaped, especially the Bifidobacterium community. In the rats pretreated with B. animalis subsp. lactis, Salmonella was less prevalent in the blood, liver, spleen, and intestines following infection. The intervention promoted T lymphocyte subset balance in the spleen and thymus and fostered neurodevelopment and neuroimmune balance in the brain. Furthermore, metabolic profiling showed a strong correlation between the metabolites in the serum and colon, supporting the view that B. animalis subsp. lactis pretreatment influences the systemic immune response by modifying the composition and metabolism of the gut microbiota. Overall, the results imply that B. animalis subsp. lactis pretreatment, through the coordinated regulation of colonic and serum metabolites, influences the systemic immune responses of neonatal rats against Salmonella infection.
Collapse
Affiliation(s)
- Chunxiu Lin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
| | - Yugui Lin
- Microbiology Laboratory, Zhongshan Bo'ai Hospital, Southern Medical University, Zhongshan 528400, P. R. China
| | - Shunhe Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, P. R. China
| | - Jialiang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
| | - Xuhua Mao
- Department of Clinical Laboratory, Yixing People's Hospital, Wuxi 214200, P. R. China
| | - Yonghua Zhou
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, P. R. China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, P. R. China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, P. R. China
| | - Gang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, P. R. China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, P. R. China
| |
Collapse
|
45
|
Lee E, Lee SY, Kim HB, Yang SI, Yoon J, Suh DI, Oh HY, Ahn K, Kim KW, Shin YH, Hong SJ. Insights from the COCOA birth cohort: The origins of childhood allergic diseases and future perspectives. Allergol Int 2024; 73:3-12. [PMID: 37752021 DOI: 10.1016/j.alit.2023.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 08/31/2023] [Indexed: 09/28/2023] Open
Abstract
The ongoing COhort for Childhood Origin of Asthma and allergic diseases (COCOA) study is a prospective birth cohort investigating the origin and natural courses of childhood allergic diseases, including atopic dermatitis, food allergy, allergic rhinitis and asthma, with long-term prognosis. Initiated under the premise that allergic diseases result from a complex interplay of immune development alterations, environmental exposures, and host susceptibility, the COCOA study explores these dynamic interactions during prenatal and postnatal periods, framed within the hygiene and microbial hypotheses alongside the developmental origins of health and disease (DOHaD) hypothesis. The scope of the COCOA study extends to genetic predispositions, indoor and outdoor environmental variables affecting mothers and their offsprings such as outdoor and indoor air pollution, psychological factors, diets, and the microbiomes of skin, gut, and airway. We have embarked on in-depth investigations of diverse risk factors and the pathophysiological underpinnings of allergic diseases. By employing multi-omics approaches-proteomics, transcriptomics, and metabolomics-we gain deeper insights into the distinct pathophysiological processes across various endotypes of childhood allergic diseases, incorporating the exposome using extensive resources within the COCOA study. Integration with large-scale datasets, such as national health insurance records, enhances robustness and mitigates potential limitations inherent to birth cohort studies. As part of global networks focused on childhood allergic diseases, the COCOA study fosters collaborative research across multiple cohorts. The findings from the COCOA study are instrumental in informing precision medicine strategies for childhood allergic diseases, underpinning the establishment of disease trajectories.
Collapse
Affiliation(s)
- Eun Lee
- Department of Pediatrics, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, South Korea
| | - So-Yeon Lee
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyo-Bin Kim
- Department of Pediatrics, Inje University Sanggye Paik Hospital, Seoul, South Korea
| | - Song-I Yang
- Department of Pediatrics, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, South Korea
| | - Jisun Yoon
- Department of Pediatrics, Chung-Ang University College of Medicine, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong, South Korea
| | - Dong In Suh
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, South Korea
| | - Hea Young Oh
- Department of Medicine, University of Ulsan College of Medicine, Seoul, South Korea
| | - Kangmo Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyung Won Kim
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Youn Ho Shin
- Department of Pediatrics, The Catholic University of Korea, Yeouido St. Mary's Hospital, Seoul, South Korea
| | - Soo-Jong Hong
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.
| |
Collapse
|
46
|
Dapa T, Xavier KB. Effect of diet on the evolution of gut commensal bacteria. Gut Microbes 2024; 16:2369337. [PMID: 38904092 PMCID: PMC11195494 DOI: 10.1080/19490976.2024.2369337] [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] [Received: 12/07/2023] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
The gut microbiota, comprising trillions of diverse microorganisms inhabiting the intestines of animals, forms a complex and indispensable ecosystem with profound implications for the host's well-being. Its functions include contributing to developing the host's immune response, aiding in nutrient digestion, synthesizing essential compounds, acting as a barrier against pathogen invasion, and influencing the development or regression of various pathologies. The dietary habits of the host directly impact this intricate community of gut microbes. Diet influences the composition and function of the gut microbiota through alterations in gene expression, enzymatic activity, and metabolome. While the impact of diet on gut ecology is well-established, the investigation into the relationship between dietary consumption and microbial genotypic diversity has been limited. This review provides an overview of the relationship between diet and gut microbiota, emphasizing the impact of host nutrition on both short- and long-term evolution in the mammalian gut. It is evident that the evolution of the gut microbiota occurs even on short timescales through the acquisition of novel mutations, within the gut bacteria of individual hosts. Consequently, we discuss the importance of considering alterations in bacterial genomic diversity when analyzing microbiota-dependent effects on host physiology. Future investigations into the various microbiota-related traits shall greatly benefit from a deeper understanding of commensal bacterial evolutionary adaptation.
Collapse
Affiliation(s)
- Tanja Dapa
- Andalusian Center for Developmental Biology (CABD), Department of Molecular Biology and Biochemical Engineering, Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | |
Collapse
|
47
|
Monaco MH, Wang M, Hauser J, Yan J, Dilger RN, Donovan SM. Formula supplementation with human and bovine milk oligosaccharides modulates blood IgG and T-helper cell populations, and ex vivo LPS-stimulated cytokine production in a neonatal preclinical model. Front Immunol 2023; 14:1327853. [PMID: 38179055 PMCID: PMC10765566 DOI: 10.3389/fimmu.2023.1327853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/06/2023] [Indexed: 01/06/2024] Open
Abstract
Introduction Human milk contains structurally diverse oligosaccharides (HMO), which are multifunctional modulators of neonatal immune development. Our objective was to investigate formula supplemented with fucosylated (2'FL) + neutral (lacto-N-neotetraose, LNnt) oligosaccharides and/or sialylated bovine milk oligosaccharides (BMOS) on immunological outcomes. Methods Pigs (n=46) were randomized at 48h of age to four diets: sow milk replacer formula (CON), BMOS (CON + 6.5 g/L BMOS), HMO (CON + 1.0 g/L 2'FL + 0.5 g/L LNnT), or BMOS+HMO (CON + 6.5 g/L BMOS + 1.0 g/L 2'FL + 0.5 g/L LNnT). Blood and tissues were collected on postnatal day 33 for measurement of cytokines and IgG, phenotypic identification of immune cells, and ex vivo lipopolysaccharide (LPS)-stimulation of immune cells. Results Serum IgG was significantly lower in the HMO group than BMOS+HMO but did not differ from CON or BMOS. The percentage of PBMC T-helper cells was lower in BMOS+HMO than the other groups. Splenocytes from the BMOS group secreted more IL-1β when stimulated ex vivo with LPS compared to CON or HMO groups. For PBMCs, a statistical interaction of BMOS*HMO was observed for IL-10 secretion (p=0.037), with BMOS+HMO and HMO groups differing at p=0.1. Discussion The addition of a mix of fucosylated and sialylated oligosaccharides to infant formula provides specific activities in the immune system that differ from formulations supplemented with one oligosaccharide structure.
Collapse
Affiliation(s)
- Marcia H. Monaco
- Department of Food Science and Human Nutrition, University of Illinois at Urbana Champaign, Urbana, IL, United States
| | - Mei Wang
- Department of Food Science and Human Nutrition, University of Illinois at Urbana Champaign, Urbana, IL, United States
| | - Jonas Hauser
- Brain Health Department, Nestlé Institute of Health Sciences, Société des Produits Nestlé SA, Lausanne, Switzerland
| | - Jian Yan
- Nestlé Product Technology Center Nutrition, Vevey, Switzerland
| | - Ryan N. Dilger
- Department of Animal Sciences, University of Illinois at Urbana Champaign, Urbana, IL, United States
| | - Sharon M. Donovan
- Department of Food Science and Human Nutrition, University of Illinois at Urbana Champaign, Urbana, IL, United States
| |
Collapse
|
48
|
Galeana-Cadena D, Gómez-García IA, Lopez-Salinas KG, Irineo-Moreno V, Jiménez-Juárez F, Tapia-García AR, Boyzo-Cortes CA, Matías-Martínez MB, Jiménez-Alvarez L, Zúñiga J, Camarena A. Winds of change a tale of: asthma and microbiome. Front Microbiol 2023; 14:1295215. [PMID: 38146448 PMCID: PMC10749662 DOI: 10.3389/fmicb.2023.1295215] [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: 09/15/2023] [Accepted: 11/15/2023] [Indexed: 12/27/2023] Open
Abstract
The role of the microbiome in asthma is highlighted, considering its influence on immune responses and its connection to alterations in asthmatic patients. In this context, we review the variables influencing asthma phenotypes from a microbiome perspective and provide insights into the microbiome's role in asthma pathogenesis. Previous cohort studies in patients with asthma have shown that the presence of genera such as Bifidobacterium, Lactobacillus, Faecalibacterium, and Bacteroides in the gut microbiome has been associated with protection against the disease. While, the presence of other genera such as Haemophilus, Streptococcus, Staphylococcus, and Moraxella in the respiratory microbiome has been implicated in asthma pathogenesis, indicating a potential link between microbial dysbiosis and the development of asthma. Furthermore, respiratory infections have been demonstrated to impact the composition of the upper respiratory tract microbiota, increasing susceptibility to bacterial diseases and potentially triggering asthma exacerbations. By understanding the interplay between the microbiome and asthma, valuable insights into disease mechanisms can be gained, potentially leading to the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- David Galeana-Cadena
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | - Itzel Alejandra Gómez-García
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Karen Gabriel Lopez-Salinas
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Valeria Irineo-Moreno
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Fabiola Jiménez-Juárez
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Alan Rodrigo Tapia-García
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Red de Medicina para la Educación, el Desarrollo y la Investigación Científica de Iztacala, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Carlos Alberto Boyzo-Cortes
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | - Melvin Barish Matías-Martínez
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Luis Jiménez-Alvarez
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | - Joaquín Zúñiga
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Angel Camarena
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| |
Collapse
|
49
|
Ren X, Wang L, Wang Z, Wang L, Kong Y, Guo Y, Sun L. Association between parental occupational exposure and the risk of asthma in offspring: A meta-analysis and systematic review. Medicine (Baltimore) 2023; 102:e36345. [PMID: 38050266 PMCID: PMC10695554 DOI: 10.1097/md.0000000000036345] [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] [Received: 09/26/2023] [Accepted: 11/06/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Previous epidemiological studies have shown inconsistent results regarding the relation between the risk of asthma in offspring and parental occupational exposure. Therefore, we conducted a comprehensive and systematic collection of currently available epidemiological data to quantify the correlation between the 2. METHODS Related studies published before March 2023 were identified through searches of the Cochrane Library, Embase, PubMed, and Web of Science databases. The quality of included studies was assessed using the Newcastle-Ottawa Scale, while pooled odds ratios (ORs) with 95% confidence intervals (CIs) were computed using fixed-effect or random-effects models. RESULTS This systematic review included 10 cohort studies, with a total of 89,571 parent-child pairs included in the quantitative analysis. The results exhibited a substantial association between parental occupational exposure to allergens (OR = 1.11; 95% CI: 1.00, 1.23; P = .051) and irritants (OR = 1.19; 95% CI: 1.07, 1.32; P = .001) and an increased risk of asthma in offspring. This association was also observed in the analysis of wheezing (OR = 1.22; 95% CI: 1.11, 1.35; P < .001 and OR = 1.19; 95% CI: 1.08, 1.32; P = .001). Subgroup analysis demonstrated that maternal occupational exposure to allergens (OR = 1.07; 95% CI: 1.02, 1.12; P = .008) and irritants (OR = 1.13; 95% CI: 1.05, 1.21; P = .001) significantly increased the risk of childhood asthma. Furthermore, parental postnatal occupational exposure to allergens (OR = 1.26; 95% CI: 1.10, 1.46; P = .001) and irritants (OR = 1.26; 95% CI: 1.06, 1.49; P = .009) had a more pronounced impact on childhood asthma. Higher levels of exposure (OR = 1.26; 95% CI: 1.10, 1.46; P = .001 and OR = 1.30; 95% CI: 1.16, 1.47; P < .001) were recognized as significant risk factors for childhood asthma. CONCLUSION Parental occupational exposure to allergens and irritants increases the risk of asthma and wheezing in offspring, with maternal exposure, postnatal exposure, and high-dose exposure being the primary risk factors for childhood asthma.
Collapse
Affiliation(s)
- Xiaoting Ren
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Lie Wang
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun Jilin, China
| | - Zhongtian Wang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Lei Wang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yibu Kong
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun Jilin, China
| | - Yinan Guo
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun Jilin, China
| | - Liping Sun
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun Jilin, China
| |
Collapse
|
50
|
Dhariwala MO, DeRogatis AM, Okoro JN, Weckel A, Tran VM, Habrylo I, Ojewumi OT, Tammen AE, Leech JM, Merana GR, Carale RO, Barrere-Cain R, Hiam-Galvez KJ, Spitzer MH, Scharschmidt TC. Commensal myeloid crosstalk in neonatal skin regulates long-term cutaneous type 17 inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.560039. [PMID: 37873143 PMCID: PMC10592812 DOI: 10.1101/2023.09.29.560039] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Early life microbe-immune interactions at barrier surfaces have lasting impacts on the trajectory towards health versus disease. Monocytes, macrophages and dendritic cells are primary sentinels in barrier tissues, yet the salient contributions of commensal-myeloid crosstalk during tissue development remain poorly understood. Here, we identify that commensal microbes facilitate accumulation of a population of monocytes in neonatal skin. Transient postnatal depletion of these monocytes resulted in heightened IL-17A production by skin T cells, which was particularly sustained among CD4+ T cells into adulthood and sufficient to exacerbate inflammatory skin pathologies. Neonatal skin monocytes were enriched in expression of negative regulators of the IL-1 pathway. Functional in vivo experiments confirmed a key role for excessive IL-1R1 signaling in T cells as contributing to the dysregulated type 17 response in neonatal monocyte-depleted mice. Thus, a commensal-driven wave of monocytes into neonatal skin critically facilitates long-term immune homeostasis in this prominent barrier tissue.
Collapse
Affiliation(s)
- Miqdad O. Dhariwala
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
| | - Andrea M. DeRogatis
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
| | - Joy N. Okoro
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
- Biomedical Sciences Program, University of California San Francisco; San Francisco, CA USA
| | - Antonin Weckel
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
| | - Victoria M. Tran
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
- Biomedical Sciences Program, University of California San Francisco; San Francisco, CA USA
| | - Irek Habrylo
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
- Biomedical Sciences Program, University of California San Francisco; San Francisco, CA USA
| | | | - Allison E. Tammen
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
| | - John M. Leech
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
| | - Geil R. Merana
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
- Biomedical Sciences Program, University of California San Francisco; San Francisco, CA USA
| | - Ricardo O. Carale
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
| | - Rio Barrere-Cain
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
| | - Kamir J. Hiam-Galvez
- Biomedical Sciences Program, University of California San Francisco; San Francisco, CA USA
- Department of Otolaryngology-Head and Neck Surgery, Department of Microbiology and Immunology, Parker Institute for Cancer Immunotherapy, University of California San Francisco; San Francisco, CA USA
| | - Matthew H. Spitzer
- Department of Otolaryngology-Head and Neck Surgery, Department of Microbiology and Immunology, Parker Institute for Cancer Immunotherapy, University of California San Francisco; San Francisco, CA USA
| | | |
Collapse
|