Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
bioRxiv ; 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38948872

ABSTRACT

Monogenic skin disorders such as ichthyosis introduce multiple sources of disturbance to the skin, including the direct biochemical consequences of the genotype, the phenotypic changes in skin physiology, and an altered skin microbiome. The association between changes in the skin microbiome and the disease's genotypic and phenotypic effects are of both ecological and clinical interest but are historically obscured by 1) the limited resolution of metagenomic profiles, and 2) additional sources of variation such as age and topical/oral treatments. Here we characterize the skin microbiome from seven ichthyosis genotypes, at species, strain, and metabolic pathway levels. Critically, we assess the association between these microbiome features and the ichthyosis genotype and phenotype while adjusting for contextual host covariables. We show that the ichthyosis genotype, especially that caused by mutations in TGM1, and the ichthyosis phenotype, particularly transepidermal water loss (TEWL), and personal covariables, such as topical emollients and oral retinoids, collectively, and sometimes antagonistically, influence the species community, strain population, and metabolic potential of the skin microbiome.

2.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38828852

ABSTRACT

The cellular and genetic networks that contribute to the development of the zeugopod (radius and ulna of the forearm, tibia and fibula of the leg) are not well understood, although these bones are susceptible to loss in congenital human syndromes and to the action of teratogens such as thalidomide. Using a new fate-mapping approach with the Chameleon transgenic chicken line, we show that there is a small contribution of SHH-expressing cells to the posterior ulna, posterior carpals and digit 3. We establish that although the majority of the ulna develops in response to paracrine SHH signalling in both the chicken and mouse, there are differences in the contribution of SHH-expressing cells between mouse and chicken as well as between the chicken ulna and fibula. This is evidence that, although zeugopod bones are clearly homologous according to the fossil record, the gene regulatory networks that contribute to their development and evolution are not fixed.


Subject(s)
Animals, Genetically Modified , Chickens , Hedgehog Proteins , Animals , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Chickens/genetics , Mice , Biological Evolution , Chick Embryo , Ulna , Gene Expression Regulation, Developmental , Fibula/metabolism , Radius/metabolism , Humans , Extremities/embryology
3.
Nature ; 628(8006): 171-179, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38509360

ABSTRACT

The myriad microorganisms that live in close association with humans have diverse effects on physiology, yet the molecular bases for these impacts remain mostly unknown1-3. Classical pathogens often invade host tissues and modulate immune responses through interactions with human extracellular and secreted proteins (the 'exoproteome'). Commensal microorganisms may also facilitate niche colonization and shape host biology by engaging host exoproteins; however, direct exoproteome-microbiota interactions remain largely unexplored. Here we developed and validated a novel technology, BASEHIT, that enables proteome-scale assessment of human exoproteome-microbiome interactions. Using BASEHIT, we interrogated more than 1.7 million potential interactions between 519 human-associated bacterial strains from diverse phylogenies and tissues of origin and 3,324 human exoproteins. The resulting interactome revealed an extensive network of transkingdom connectivity consisting of thousands of previously undescribed host-microorganism interactions involving 383 strains and 651 host proteins. Specific binding patterns within this network implied underlying biological logic; for example, conspecific strains exhibited shared exoprotein-binding patterns, and individual tissue isolates uniquely bound tissue-specific exoproteins. Furthermore, we observed dozens of unique and often strain-specific interactions with potential roles in niche colonization, tissue remodelling and immunomodulation, and found that strains with differing host interaction profiles had divergent interactions with host cells in vitro and effects on the host immune system in vivo. Overall, these studies expose a previously unexplored landscape of molecular-level host-microbiota interactions that may underlie causal effects of indigenous microorganisms on human health and disease.


Subject(s)
Bacteria , Host Microbial Interactions , Microbiota , Phylogeny , Proteome , Symbiosis , Animals , Female , Humans , Mice , Bacteria/classification , Bacteria/immunology , Bacteria/metabolism , Bacteria/pathogenicity , Host Microbial Interactions/immunology , Host Microbial Interactions/physiology , Host Tropism , Microbiota/immunology , Microbiota/physiology , Organ Specificity , Protein Binding , Proteome/immunology , Proteome/metabolism , Reproducibility of Results
4.
J Am Soc Mass Spectrom ; 35(3): 518-526, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38308645

ABSTRACT

Aryl hydrocarbon receptor (AhR) is a transcription factor that regulates gene expression upon ligand activation, enabling microbiota-dependent induction, training, and function of the host immune system. A spectrum of metabolites, encompassing indole and tryptophan derivatives, have been recognized as activators. This work introduces an integrated, mass spectrometry-centric workflow that employs a bioassay-guided, fractionation-based methodology for the identification of AhR activators derived from human bacterial isolates. By leveraging the workflow efficiency, the complexities inherent in metabolomics profiling are significantly reduced, paving the way for an in-depth and focused mass spectrometry analysis of bioactive fractions isolated from bacterial culture supernatants. Validation of AhR activator candidates used multiple criteria─MS/MS of the synthetic reference compound, bioassay of AhR activity, and elution time confirmation using a C-13 isotopic reference─and was demonstrated for N-formylkynurenine (NFK). The workflow reported provides a roadmap update for improved efficiency of identifying bioactive metabolites using mass spectrometry-based metabolomics. Mass spectrometry datasets are accessible at the National Metabolomics Data Repository (PR001479, Project DOI: 10.21228/M8JM7Q).


Subject(s)
Receptors, Aryl Hydrocarbon , Tandem Mass Spectrometry , Humans , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism
5.
Biofabrication ; 15(4)2023 08 14.
Article in English | MEDLINE | ID: mdl-37536321

ABSTRACT

Progenitor human nasal epithelial cells (hNECs) are an essential cell source for the reconstruction of the respiratory pseudostratified columnar epithelium composed of multiple cell types in the context of infection studies and disease modeling. Hitherto, manual seeding has been the dominant method for creating nasal epithelial tissue models through biofabrication. However, this approach has limitations in terms of achieving the intricate three-dimensional (3D) structure of the natural nasal epithelium. 3D bioprinting has been utilized to reconstruct various epithelial tissue models, such as cutaneous, intestinal, alveolar, and bronchial epithelium, but there has been no attempt to use of 3D bioprinting technologies for reconstruction of the nasal epithelium. In this study, for the first time, we demonstrate the reconstruction of the nasal epithelium with the use of primary hNECs deposited on Transwell inserts via droplet-based bioprinting (DBB), which enabled high-throughput fabrication of the nasal epithelium in Transwell inserts of 24-well plates. DBB of progenitor hNECs ranging from one-tenth to one-half of the cell seeding density employed during the conventional cell seeding approach enabled a high degree of differentiation with the presence of cilia and tight-junctions over a 4 weeks air-liquid interface culture. Single cell RNA sequencing of these cultures identified five major epithelial cells populations, including basal, suprabasal, goblet, club, and ciliated cells. These cultures recapitulated the pseudostratified columnar epithelial architecture present in the native nasal epithelium and were permissive to respiratory virus infection. These results denote the potential of 3D bioprinting for high-throughput fabrication of nasal epithelial tissue models not only for infection studies but also for other purposes, such as disease modeling, immunological studies, and drug screening.


Subject(s)
Bioprinting , Humans , Nasal Mucosa/metabolism , Epithelial Cells , Respiratory Mucosa/metabolism , Cilia
6.
Exp Dermatol ; 32(10): 1624-1632, 2023 10.
Article in English | MEDLINE | ID: mdl-37350109

ABSTRACT

The gut microbiome is increasingly recognized to alter cancer risk, progression and response to treatments such as immunotherapy, especially in cutaneous melanoma. However, whether the microbiome influences immune checkpoint inhibitor (ICI) immunotherapy response to non-melanoma skin cancer has not yet been defined. As squamous cell carcinomas (SCC) are in closest proximity to the skin microbiome, we hypothesized that the skin microbiome, which regulates cutaneous immunity, might affect SCC-associated anti-PD1 immunotherapy treatment response. We used ultraviolet radiation to induce SCC in SKH1 hairless mice. We then treated the mice with broad-band antibiotics to deplete the microbiome, followed by colonisation by candidate skin and gut bacteria or persistent antibiotic treatment, all in parallel with ICI treatment. We longitudinally monitored skin and gut microbiome dynamics by 16S rRNA gene sequencing and tumour burden by periodic tumour measurements and histologic assessment. Our study revealed that antibiotics-induced abrogation of the microbiome reduced the tumour burden, suggesting a functional role of the microbiome in non-melanoma skin cancer therapy response.


Subject(s)
Carcinoma, Squamous Cell , Gastrointestinal Microbiome , Immunotherapy , Melanoma , Skin Neoplasms , Animals , Mice , Anti-Bacterial Agents/therapeutic use , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/therapy , Immunotherapy/methods , Melanoma/therapy , Microbiota , RNA, Ribosomal, 16S/genetics , Skin Neoplasms/immunology , Skin Neoplasms/therapy , Ultraviolet Rays , Gastrointestinal Microbiome/immunology
7.
Exp Dermatol ; 32(9): 1546-1556, 2023 09.
Article in English | MEDLINE | ID: mdl-37350224

ABSTRACT

Two major arms of skin ageing are changes in the skin's biophysical conditions and alterations in the skin microbiome. This work partitioned both arms to study their interaction in detail. Leveraging the resolution provided by shotgun metagenomics, we explored how skin microbial species, strains and gene content interact with the biophysical traits of the skin during ageing. With a dataset well-controlled for confounding factors, we found that skin biophysical traits, especially the collagen diffusion coefficient, are associated with the composition and the functional potential of the skin microbiome, including the abundance of bacterial strains found in nosocomial infections and the abundance of antibiotic resistance genes. Our findings reveal important associations between skin biophysical features and ageing-related changes in the skin microbiome and generate testable hypotheses for the mechanisms of such associations.


Subject(s)
Microbiota , Skin Aging , Microbiota/genetics , Bacteria , Anti-Bacterial Agents , Skin/microbiology
8.
bioRxiv ; 2023 Mar 30.
Article in English | MEDLINE | ID: mdl-37034627

ABSTRACT

Human nasal epithelial cells (hNECs) are an essential cell source for the reconstruction of the respiratory pseudostratified columnar epithelium composed of multiple cell types in the context of infection studies and disease modeling. Hitherto, manual seeding has been the dominant method for creating nasal epithelial tissue models. However, the manual approach is slow, low-throughput and has limitations in terms of achieving the intricate 3D structure of the natural nasal epithelium in a uniform manner. 3D Bioprinting has been utilized to reconstruct various epithelial tissue models, such as cutaneous, intestinal, alveolar, and bronchial epithelium, but there has been no attempt to use of 3D bioprinting technologies for reconstruction of the nasal epithelium. In this study, for the first time, we demonstrate the reconstruction of the nasal epithelium with the use of primary hNECs deposited on Transwell inserts via droplet-based bioprinting (DBB), which enabled high-throughput fabrication of the nasal epithelium in Transwell inserts of 24-well plates. DBB of nasal progenitor cells ranging from one-tenth to one-half of the cell seeding density employed during the conventional cell seeding approach enabled a high degree of differentiation with the presence of cilia and tight-junctions over a 4-week air-liquid interface culture. Single cell RNA sequencing of these cultures identified five major epithelial cells populations, including basal, suprabasal, goblet, club, and ciliated cells. These cultures recapitulated the pseudostratified columnar epithelial architecture present in the native nasal epithelium and were permissive to respiratory virus infection. These results denote the potential of 3D bioprinting for high-throughput fabrication of nasal epithelial tissue models not only for infection studies but also for other purposes such as disease modeling, immunological studies, and drug screening.

9.
Res Sq ; 2023 Mar 13.
Article in English | MEDLINE | ID: mdl-36993518

ABSTRACT

Purpose: Patients with non-infectious complications have worse clinical outcomes in common variable immunodeficiency (CVID) than those with infections-only. Non-infectious complications are associated with gut microbiome aberrations, but there are no reductionist animal models that emulate CVID. Our aim in this study was to uncover potential microbiome roles in the development of non-infectious complications in CVID. Methods: We examined fecal whole genome shotgun sequencing from patients CVID, and non-infectious complications, infections-only, and their household controls. We also performed Fecal Microbiota transplant from CVID patients to Germ-Free Mice. Results: We found potentially pathogenic microbes Streptococcus parasanguinis and Erysipelatoclostridium ramosum were enriched in gut microbiomes of CVID patients with non-infectious complications. In contrast, Fusicatenibacter saccharivorans and Anaerostipes hadrus, known to suppress inflammation and promote healthy metabolism, were enriched in gut microbiomes of infections-only CVID patients. Fecal microbiota transplant from non-infectious complications, infections-only, and their household controls into germ-free mice revealed gut dysbiosis patterns in recipients from CVID patients with non-infectious complications, but not infections-only CVID, or household controls recipients. Conclusion: Our findings provide a proof of concept that fecal microbiota transplant from CVID patients with non-infectious complications to Germ-Free mice recapitulates microbiome alterations observed in the donors.

10.
bioRxiv ; 2023 Jan 25.
Article in English | MEDLINE | ID: mdl-36747869

ABSTRACT

The gut microbiome is increasingly recognized to alter cancer risk, progression, and response to treatments such as immunotherapy, especially in cutaneous melanoma. However, whether the microbiome influences immune checkpoint inhibitor (ICI) immunotherapy response to non-melanoma skin cancer has not yet been defined. As squamous cell carcinomas (SCC) are in closest proximity to the skin microbiome, we hypothesized that the skin microbiome, which regulates cutaneous immunity, might affect SCC-associated anti-PD1 immunotherapy treatment response. We used ultraviolet radiation to induce SCC in SKH1 hairless mice. We then treated the mice with broad-band antibiotics to deplete the microbiome, followed by colonization by candidate skin and gut bacteria or persistent antibiotic treatment, all in parallel with ICI treatment. We longitudinally monitored skin and gut microbiome dynamics by 16S rRNA gene sequencing, and tumor burden by periodic tumor measurements and histologic assessment. Our study revealed that antibiotics-induced abrogation of the microbiome reduced tumor burden, suggesting a functional role of the microbiome in non-melanoma skin cancer therapy response.

11.
Cell Host Microbe ; 31(2): 273-287.e5, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36758521

ABSTRACT

Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a complex, debilitating disorder manifesting as severe fatigue and post-exertional malaise. The etiology of ME/CFS remains elusive. Here, we present a deep metagenomic analysis of stool combined with plasma metabolomics and clinical phenotyping of two ME/CFS cohorts with short-term (<4 years, n = 75) or long-term disease (>10 years, n = 79) compared with healthy controls (n = 79). First, we describe microbial and metabolomic dysbiosis in ME/CFS patients. Short-term patients showed significant microbial dysbiosis, while long-term patients had largely resolved microbial dysbiosis but had metabolic and clinical aberrations. Second, we identified phenotypic, microbial, and metabolic biomarkers specific to patient cohorts. These revealed potential functional mechanisms underlying disease onset and duration, including reduced microbial butyrate biosynthesis and a reduction in plasma butyrate, bile acids, and benzoate. In addition to the insights derived, our data represent an important resource to facilitate mechanistic hypotheses of host-microbiome interactions in ME/CFS.


Subject(s)
Fatigue Syndrome, Chronic , Gastrointestinal Microbiome , Humans , Fatigue Syndrome, Chronic/metabolism , Dysbiosis , Metabolomics , Feces
12.
PLoS One ; 17(12): e0276795, 2022.
Article in English | MEDLINE | ID: mdl-36520793

ABSTRACT

The prevalence and virulence of pathogens such as methicillin-resistant Staphylococcus (S.) aureus (MRSA), which can cause recurrent skin infections, are of significant clinical concern. Prolonged antibiotic exposure to treat or decolonize S. aureus contributes to development of antibiotic resistance, as well as depletion of the microbiome, and its numerous beneficial functions. We hypothesized an engineered skin probiotic with the ability to selectively deliver antimicrobials only in the presence of the target organism could provide local bioremediation of pathogen colonization. We constructed a biosensing S. epidermidis capable of detecting the presence of S. aureus quorum sensing autoinducer peptide and producing lysostaphin in response. Here, we demonstrate in vitro activity of this biosensor and present and discuss challenges to deployment of this and other engineered topical skin probiotics.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Probiotics , Staphylococcal Infections , Humans , Staphylococcus aureus/physiology , Anti-Bacterial Agents/therapeutic use , Virulence , Probiotics/therapeutic use , Staphylococcal Infections/drug therapy , Microbial Sensitivity Tests
13.
Nat Aging ; 2(10): 941-955, 2022 10.
Article in English | MEDLINE | ID: mdl-36398033

ABSTRACT

Older adults represent a vulnerable population with elevated risk for numerous morbidities. To explore the association of the microbiome with aging and age-related susceptibilities including frailty and infectious disease risk, we conducted a longitudinal study of the skin, oral, and gut microbiota in 47 community- or skilled nursing facility-dwelling older adults vs. younger adults. We found that microbiome changes were not associated with chronological age so much as frailty: we identified prominent changes in microbiome features associated with susceptibility to pathogen colonization and disease risk, including diversity, stability, heterogeneity, and biogeographic determinism, which were moreover associated with a loss of Cutibacterium (C.) acnes in the skin microbiome. Strikingly, the skin microbiota were also the primary reservoir for antimicrobial resistance, clinically important pathobionts, and nosocomial strains, suggesting a potential role particularly for the skin microbiome in disease risk and dissemination of multidrug resistant pathogens.


Subject(s)
Frailty , Gastrointestinal Microbiome , Infections , Microbiota , Humans , Aged , Frailty/epidemiology , Longitudinal Studies , Disease Susceptibility/microbiology
14.
mBio ; 13(6): e0263222, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36409086

ABSTRACT

Staphylococcus epidermidis is a ubiquitous human commensal skin bacterium that is also one of the most prevalent nosocomial pathogens. The genetic factors underlying this remarkable lifestyle plasticity are incompletely understood, mainly due to the difficulties of genetic manipulation, precluding high-throughput functional profiling of this species. To probe the versatility of S. epidermidis to survive across a diversity of environmental conditions, we developed a large-scale CRISPR interference (CRISPRi) screen complemented by transcriptional profiling (RNA sequencing) across 24 diverse conditions and piloted a droplet-based CRISPRi approach to enhance throughput and sensitivity. We identified putative essential genes, importantly revealing amino acid metabolism as crucial to survival across diverse environments, and demonstrated the importance of trace metal uptake for survival under multiple stress conditions. We identified pathways significantly enriched and repressed across our range of stress and nutrient-limited conditions, demonstrating the considerable plasticity of S. epidermidis in responding to environmental stressors. Additionally, we postulate a mechanism by which nitrogen metabolism is linked to lifestyle versatility in response to hyperosmotic challenges, such as those encountered on human skin. Finally, we examined the survival of S. epidermidis under acid stress and hypothesize a role for cell wall modification as a vital component of the survival response under acidic conditions. Taken together, this study integrates large-scale CRISPRi and transcriptomics data across multiple environments to provide insights into a keystone member of the human skin microbiome. Our results additionally provide a valuable benchmarking analysis for CRISPRi screens and are a rich resource for other staphylococcal researchers. IMPORTANCE Staphylococcus epidermidis is a bacteria that broadly inhabits healthy human skin, yet it is also a common cause of skin infections and bloodstream infections associated with implanted medical devices. Because human skin has many different types of S. epidermidis, each containing different genes, our goal is to determine how these different genes allow S. epidermidis to switch from healthy growth in the skin to being an infectious pathogen. Understanding this switch is critical to developing new strategies to prevent and treat S. epidermidis infections.


Subject(s)
Staphylococcal Infections , Humans , Staphylococcal Infections/microbiology , Staphylococcus epidermidis/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Transcriptome , Skin/microbiology
15.
Curr Opin Microbiol ; 70: 102222, 2022 12.
Article in English | MEDLINE | ID: mdl-36242896

ABSTRACT

An extraordinary biodiversity of bacteria, fungi, viruses, and even small multicellular eukaryota inhabit the human skin. Genomic innovations have accelerated characterization of this biodiversity both at a species as well as the subspecies, or strain level, which further imparts a tremendous genetic diversity to an individual's skin microbiome. In turn, these advances portend significant species- and strain-specificity in the skin microbiome's functional impact on cutaneous immunity, barrier integrity, aging, and other skin physiologic processes. Future advances in defining strain diversity, spatial distribution, and metabolic diversity for major skin species will be foundational for understanding the microbiome's essentiality to the skin ecosystem and for designing topical therapeutics that leverage or target the skin microbiome.


Subject(s)
Microbiota , Humans , Microbiota/physiology , Bacteria/genetics , Skin/microbiology , Biodiversity , Fungi/genetics
16.
J Hand Surg Eur Vol ; 47(10): 1085-1088, 2022 11.
Article in English | MEDLINE | ID: mdl-36165432
17.
Comput Struct Biotechnol J ; 20: 4315-4324, 2022.
Article in English | MEDLINE | ID: mdl-36016717

ABSTRACT

A gene expression compendium is a heterogeneous collection of gene expression experiments assembled from data collected for diverse purposes. The widely varied experimental conditions and genetic backgrounds across samples creates a tremendous opportunity for gaining a systems level understanding of the transcriptional responses that influence phenotypes. Variety in experimental design is particularly important for studying microbes, where the transcriptional responses integrate many signals and demonstrate plasticity across strains including response to what nutrients are available and what microbes are present. Advances in high-throughput measurement technology have made it feasible to construct compendia for many microbes. In this review we discuss how these compendia are constructed and analyzed to reveal transcriptional patterns.

18.
J Invest Dermatol ; 142(10): 2773-2782.e16, 2022 10.
Article in English | MEDLINE | ID: mdl-35390349

ABSTRACT

The skin microbiome plays a critical role in skin homeostasis and disorders. UVR is the major cause of nonmelanoma skin cancer, but other risk factors, including immune suppression, chronic inflammation, and antibiotic usage, suggest the microbiome as an additional, unexplored risk factor and potential disease biomarker. The overarching goal was to study the skin microbiome in squamous cell carcinoma (SCC) and premalignant actinic keratosis compared with that in healthy skin to identify skin cancer‒associated changes in the skin microbiome. We performed a high-resolution analysis of shotgun metagenomes of actinic keratosis and SCC in healthy skin, revealing the microbial community shifts specific to actinic keratosis and SCC. Most prominently, the relative abundance of pathobiont Staphylococcus aureus was increased at the expense of commensal Cutibacterium acnes in SCC compared with that in healthy skin, and enrichment of functional pathways in SCC reflected this shift. Notably, C. acnes associated with lesional versus healthy skin differed at the strain level, suggesting the specific functional changes associated with its depletion in SCC. Our study revealed a transitional microbial dysbiosis from healthy skin to actinic keratosis to SCC, supporting further investigation of the skin microbiome for use as a biomarker and providing hypotheses for studies investigating how these microbes might influence skin cancer progression.


Subject(s)
Carcinoma, Squamous Cell , Keratosis, Actinic , Microbiota , Skin Neoplasms , Anti-Bacterial Agents , Carcinoma, Squamous Cell/pathology , Humans , Keratosis, Actinic/pathology , Skin Neoplasms/pathology
19.
J Invest Dermatol ; 142(5): 1260-1264, 2022 05.
Article in English | MEDLINE | ID: mdl-34688614

ABSTRACT

Human skin hosts a diversity of microbiota. Advances in sequencing and analytical methods have increasingly illuminated the importance of the finest resolution in understanding the genetic diversity of the skin microbiota, highlighting strain-level differences and their functional implications. Such genetic diversity, which exists within an individual and is strongly individual specific underscores the difficulty in elucidating functionality. Integrated investigations of the microbial strain diversity through sequencing and culture-based approaches with host immunology and physiology will be critical in expanding our understanding of the roles of the skin microbiome.


Subject(s)
Internship and Residency , Microbiota , Humans , Microbiota/genetics , Skin
20.
Nat Aging ; 2(10): 876-877, 2022 10.
Article in English | MEDLINE | ID: mdl-36909933

ABSTRACT

Our longitudinal study comparing the skin, gut and oral microbiomes of community-dwelling older adults and nursing home residents showed striking changes known to be linked to antibiotic resistance and disease risk. Such shifts were associated with frailty, not chronological age, and were most pronounced in the skin, the primary reservoir for infection risk.


Subject(s)
Frailty , Gastrointestinal Microbiome , Microbiota , Humans , Aged , Frailty/epidemiology , Longitudinal Studies , Dysbiosis/epidemiology , Nursing Homes , Aging
SELECTION OF CITATIONS
SEARCH DETAIL
...