Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Brain Behav Immun ; 115: 308-318, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37914098

RESUMO

Maternal stress during pregnancy is prevalent and associated with increased risk of neurodevelopmental disorders in the offspring. Maternal and offspring immune dysfunction has been implicated as a potential mechanism by which prenatal stress shapes offspring neurodevelopment; however, the impact of prenatal stress on the developing immune system has yet to be elucidated. Furthermore, there is evidence that the chemokine C-C motif chemokine ligand 2 (CCL2) plays a key role in mediating the behavioral sequelae of prenatal stress. Here, we use an established model of prenatal restraint stress in mice to investigate alterations in the fetal immune system, with a focus on CCL2. In the placenta, stress led to a reduction in CCL2 and Ccr2 expression with a concomitant decrease in leukocyte number. However, the fetal liver exhibited an inflammatory phenotype, with upregulation of Ccl2, Il6, and Lbp expression, along with an increase in pro-inflammatory Ly6CHi monocytes. Prenatal stress also disrupted chemokine signaling and increased the number of monocytes and microglia in the fetal brain. Furthermore, stress increased Il1b expression by fetal brain CD11b+ microglia and monocytes. Finally, intra-amniotic injections of recombinant mouse CCL2 partially recapitulated the social behavioral deficits in the adult offspring previously observed in the prenatal restraint stress model. Altogether, these data suggest that prenatal stress led to fetal inflammation, and that fetal CCL2 plays a role in shaping offspring social behavior.


Assuntos
Quimiocina CCL2 , Efeitos Tardios da Exposição Pré-Natal , Animais , Feminino , Humanos , Camundongos , Gravidez , Quimiocina CCL2/metabolismo , Quimiocinas/metabolismo , Inflamação/metabolismo , Ligantes , Monócitos/metabolismo , Comportamento Social
2.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34161260

RESUMO

Individuals who are minoritized as a result of race, sexual identity, gender, or socioeconomic status experience a higher prevalence of many diseases. Understanding the biological processes that cause and maintain these socially driven health inequities is essential for addressing them. The gut microbiome is strongly shaped by host environments and affects host metabolic, immune, and neuroendocrine functions, making it an important pathway by which differences in experiences caused by social, political, and economic forces could contribute to health inequities. Nevertheless, few studies have directly integrated the gut microbiome into investigations of health inequities. Here, we argue that accounting for host-gut microbe interactions will improve understanding and management of health inequities, and that health policy must begin to consider the microbiome as an important pathway linking environments to population health.


Assuntos
Microbioma Gastrointestinal , Disparidades nos Níveis de Saúde , Doença , Saúde , Humanos , Saúde Mental , Publicações
3.
Psychosom Med ; 84(7): 749-756, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35797533

RESUMO

OBJECTIVE: Emerging research has connected abundances of specific bacteria to differences in psychosocial behaviors in animals and adult humans. However, research assessing mind-microbiome associations in children is sparse with extant work primarily focused on populations with autism, making it unclear whether links are also present in typically developing children. The current study fills this gap by examining associations between prosocial-self-regulating temperaments (effortful control; EC) and the gut microbiome in typically developing children. METHODS: Maternal ratings of temperament were assessed in 77 toddlers 18 to 27 months of age (46.7% female, mean age = 23.14 months). Next-generation pyrosequencing of the V1-V3 region of the 16S rRNA gene was used to classify children's gut microbial composition from fecal samples. EC included the following subcategories: cuddliness, attentional focusing, attentional shifting, inhibitory control, and low-intensity pleasure. RESULTS: After adjusting for covariates, EC was positively associated with relative abundances of Akkermansia (Δ R2 = 0.117, b = 0.022, SE = 0.007, p = .002), with cuddliness (i.e., joy and ease of being held) driving the relation. Furthermore, attentional focusing was negatively associated with Alistipes (Δ R2 = 0.062, b = -0.011, SE = 0.005, p = .028). Permutational analysis of variance revealed no significant differences in community structure between high and low EC groups on the phylum level ( R2 = 0.00372, p = .745) or the genus level ( R2 = 0.01559, p = .276). CONCLUSIONS: Findings suggest that certain microbes may be linked to prosocial behaviors used to regulate emotion in typically developing children. Further research is needed to test whether these observations replicate in larger samples.


Assuntos
Microbioma Gastrointestinal , Adulto , Bactérias/genética , Pré-Escolar , Fezes/microbiologia , Feminino , Humanos , Lactente , Masculino , RNA Ribossômico 16S/genética , Comportamento Social
4.
BMC Cancer ; 22(1): 245, 2022 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-35248004

RESUMO

BACKGROUND: Cancer patients experience gastrointestinal and behavioral symptoms, and are at increased risk of systemic infection and inflammation. These conditions are a major source of morbidity and decreased quality of life prior to cancer treatment, but poorly defined etiologies impede successful treatment. The gastrointestinal microbiota shape inflammation, influence cancer progression and treatment, and colonize tumors. However, research has not directly determined if peripheral tumors influence the microbiome and intestinal physiology, thus influencing gastrointestinal and behavioral symptoms. Therefore, the purpose of this study was to examine consequences of orthotopic, syngeneic mammary tumor implantation, growth, and resection on fecal bacteriome composition and intestinal barrier function in relation to systemic inflammation and enteric bacterial translocation in mice. METHODS: Female mice were randomized to 3 experimental groups: sham surgical control, tumor recipients, and tumor recipients later receiving tumor-resection. Mice were sacrificed three weeks after tumor implantation or resection for collection of stool, colon, spleen, and brain tissue and analysis. RESULTS: Tumor-bearing mice exhibited several markers of colonic barrier disruption, including dampened expression of tight junction proteins (Cldn1 and Ocln) and elevated circulating lipopolysaccharide binding protein (LBP). Compromised colonic barrier integrity was associated with altered fecal bacterial profiles in tumor-mice, including lower relative abundance of Lactobacillus, but higher Bacteroides. Consistent with colonic barrier disruption and altered microbiomes, tumor-mice displayed markers of systemic inflammation including splenomegaly, higher splenic bacterial load, and elevated splenic and brain pro-inflammatory cytokines. Several  bacteria cultured from spleens had 16S rRNA gene amplicons matching those in fecal samples, suggesting they were of intestinal origin. Fecal Lactobacillus was highly-interrelated to physiological parameters disrupted by tumors via correlation network analysis. Tumor resection ameliorated circulating LBP, splenomegaly, and splenic cytokines, but not other parameters associated with loss of colonic barrier integrity and bacterial translocation. CONCLUSIONS: Orthotopic mammary tumors alter the microbiome, reduce intestinal barrier function, increase translocation of enteric bacteria, and alter systemic inflammation. This provides insight into how tumors commence gastrointestinal and behavioral symptoms prior to treatment, and identify targets for future therapeutics, such as probiotic Lactobacillus supplementation.


Assuntos
Translocação Bacteriana , Neoplasias da Mama/microbiologia , Fezes/microbiologia , Microbioma Gastrointestinal , Mucosa Intestinal/microbiologia , Animais , Colo/microbiologia , Modelos Animais de Doenças , Feminino , Inflamação/microbiologia , Camundongos , RNA Ribossômico 16S/metabolismo
5.
Brain Behav Immun ; 100: 145-154, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34808291

RESUMO

BACKGROUND: Breast cancer survivors are prone to weakened gut barriers, allowing bacteria to migrate into the blood stream. Gut permeability fuels inflammation, which, among survivors, can elevate risk for comorbid disease development, cancer recurrence, and a poor quality of life; however, survivors' satisfying relationships can provide health benefits. This longitudinal study used a conceptual model addressing how intimate relationships is associated with health through changes in gut permeability and inflammation. METHOD: Breast cancer survivors (n = 139, stages 0-IIIC) completed a baseline visit before treatment and two follow-up visits 6 and 18 months after treatment ended. Women who had an abnormal breast cancer test followed by a benign diagnosis completed visits within a comparable timeframe (noncancer patient controls; n = 69). All women completed questionnaires assessing their relationship satisfaction and provided blood samples to assess two bacterial endotoxin biomarkers, lipopolysaccharide-binding protein (LBP) and soluble CD14 (sCD14), as well as C-reactive protein (CRP) and interleukin 6 (IL-6). RESULTS: Within-person multilevel mediation analyses showed that when a survivor's relationship satisfaction was higher than usual, her own LBP and LBP/sCD14 were lower, which was associated with lower than her own average CRP and IL-6 (95% CIs [-0.0104, -0.0002]). IL-6 was also higher when older survivors, but not younger survivors, experienced higher than usual intestinal permeability (p = .001). These effects of satisfying relationships held after accounting for cancer-related and behavioral factors. Post-hoc analyses showed LBP, sCD14, and LBP/sCD14 were associated with CRP for the cancer survivors, but only LBP and LBP/sCD14 were linked to CRP among the noncancer control patients. CONCLUSION: The gut environment is a new promising candidate for understanding a relationship's long-term health impact, particularly among those with elevated health risks. Survivors may reap multiple physiological benefits from satisfying relationships.


Assuntos
Neoplasias da Mama , Sobreviventes de Câncer , Feminino , Humanos , Inflamação , Estudos Longitudinais , Recidiva Local de Neoplasia , Permeabilidade , Satisfação Pessoal , Qualidade de Vida , Sobreviventes
6.
Int J Mol Sci ; 23(4)2022 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-35216112

RESUMO

Stressor exposure increases colonic inflammation. Because inflammation leads to anxiety-like behavior, we tested whether stressor exposure in mice recovering from dextran-sulfate-sodium (DSS)-induced colitis enhances anxiety-like behavior. Mice received 2% DSS for five consecutive days prior to being exposed to a social-disruption (SDR) stressor (or being left undisturbed). After stressor exposure, their behavior was tested and colitis was assessed via histopathology and via inflammatory-cytokine measurement in the serum and colon. Cytokine and chemokine mRNA levels in the colon, mesenteric lymph nodes (MLNs), hippocampus, and amygdala were measured with RT-PCR. SDR increased anxiety-like behaviors, which correlated with serum and hippocampal IL-17A. The stressor also reduced IL-1ß, CCL2, and iNOS in the colonic tissue, but increased iNOS, IFNγ, IL-17A, and TNFα in the MLNs. A network analysis indicated that reductions in colonic iNOS were related to elevated MLN iNOS and IFNγ. These inflammatory markers were related to serum and hippocampal IL-17A and associated with anxiety-like behavior. Our data suggest that iNOS may protect against extra-colonic inflammation, and when suppressed during stress it is associated with elevated MLN IFNγ, which may coordinate gut-to-brain inflammation. Our data point to hippocampal IL-17A as a key correlate of anxiety-like behavior.


Assuntos
Ansiedade/metabolismo , Colite/metabolismo , Citocinas/metabolismo , Hipocampo/metabolismo , Inflamação/metabolismo , Animais , Ansiedade/patologia , Colite/patologia , Colo/metabolismo , Colo/patologia , Modelos Animais de Doenças , Hipocampo/patologia , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
7.
J Pediatr Gastroenterol Nutr ; 73(4): 499-506, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34238825

RESUMO

OBJECTIVES: Polyethylene Glycol 3350 (PEG3350) is a laxative commonly used to treat constipation in children. The Food and Drug Administration has received reports of increased anxiety, aggression, and obsessive--compulsive behaviors in children administered PEG3350. Thus, we assessed whether daily administration of PEG3350 leads to anxiety-like behavior in mice. METHODS: Outbred CD-1 IGS mice were administered either a high or a low dose of PEG3350 via daily oral gavage for 2 weeks. As a laxative comparison and control, additional mice were given a high or low dose of magnesium citrate or vehicle (water). Weight and stool consistency were assessed after each gavage to determine laxative effectiveness. Anxiety-like behaviors were assessed using light/dark, open field, and elevated plus maze (EPM) tests at baseline, after 2 weeks of daily gavage, and after a 2 week washout in experiment 1, and after 2 weeks of daily gavage in experiment 2. Stool samples were collected for microbiome analysis in experiment 2 at baseline, after 2 weeks of daily gavage, and after 2 weeks washout. RESULTS: PEG3350 and magnesium citrate significantly changed stool consistency, as well as microbiome alpha and beta diversity. Anxiety-like behaviors were not, however, different in mice administered low or high doses of PEG3350 or magnesium citrate. CONCLUSIONS: Although changes in stool consistency and the gut microbiome occurred, administration of PEG3350 did not alter anxiety-like behaviors.


Assuntos
Microbioma Gastrointestinal , Laxantes , Animais , Camundongos , Polietilenoglicóis , Resultado do Tratamento
8.
Nurs Res ; 69(5S Suppl 1): S66-S78, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32555010

RESUMO

BACKGROUND: Neonatal exposure and subsequent withdrawal from maternal substance use disorder are a growing problem and consequence of the current opioid epidemic. Neonatal abstinence syndrome (NAS) is defined by a specified cluster of symptoms with treatment guided by the expression and severity of these symptoms. The mechanisms or pathophysiology contributing to the development of NAS symptoms are not well known, but one factor that may influence NAS symptoms is the gut microbiota. OBJECTIVES: The purpose of this integrative review was to examine evidence that might show if and how the gut microbiota influence expression and severity of symptoms similar to those seen in NAS. METHODS: Using published guidelines, a review of research studies that focused on the gut microbiome and symptoms similar to those seen in NAS was conducted, using the Cochrane, EMBASE, and Scopus databases, from 2009 through 2019. RESULTS: The review results included findings of aberrant microbial diversity, differences in microbial communities between study groups, and associations between specific taxa and symptoms. In studies involving interventions, there were reports of improved microbial diversity, community structure, and symptoms. DISCUSSION: The review findings provide evidence that the gut microbiota may play a role in modifying variability in the expression and severity of symptoms associated with NAS. Future research should focus on examining the gut microbiota in infants with and without the syndrome as well as exploring the relationship between symptom expression and aberrant gut microbiota colonization in infants with NAS.


Assuntos
Microbioma Gastrointestinal/fisiologia , Síndrome de Abstinência Neonatal/complicações , Síndrome , Correlação de Dados , Feminino , Humanos , Recém-Nascido , Síndrome de Abstinência Neonatal/fisiopatologia , Tratamento de Substituição de Opiáceos/métodos , Gravidez , Complicações na Gravidez/etiologia , Complicações na Gravidez/fisiopatologia
9.
J Am Soc Nephrol ; 30(8): 1385-1397, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31239387

RESUMO

BACKGROUND: Evidence suggests that antimicrobial peptides, components of the innate immune response, protect the kidneys and bladder from bacterial challenge. We previously identified ribonuclease 7 (RNase 7) as a human antimicrobial peptide that has bactericidal activity against uropathogenic Escherichia coli (UPEC). Functional studies assessing RNase 7's contributions to urinary tract defense are limited. METHODS: To investigate RNase 7's role in preventing urinary tract infection (UTI), we quantified urinary RNase 7 concentrations in 29 girls and adolescents with a UTI history and 29 healthy female human controls. To assess RNase 7's antimicrobial activity in vitro in human urothelial cells, we used siRNA to silence urothelial RNase 7 production and retroviral constructs to stably overexpress RNase 7; we then evaluated UPEC's ability to bind and invade these cells. For RNase 7 in vivo studies, we developed humanized RNase 7 transgenic mice, subjected them to experimental UTI, and enumerated UPEC burden in the urine, bladder, and kidneys. RESULTS: Compared with controls, study participants with a UTI history had 1.5-fold lower urinary RNase 7 concentrations. When RNase 7 was silenced in vitro, the percentage of UPEC binding or invading human urothelial cells increased; when cells overexpressed RNase 7, UPEC attachment and invasion decreased. In the transgenic mice, we detected RNase 7 expression in the kidney's intercalated cells and bladder urothelium. RNase 7 humanized mice exhibited marked protection from UPEC. CONCLUSIONS: These findings provide evidence that RNase 7 has a role in kidney and bladder host defense against UPEC and establish a foundation for investigating RNase 7 as a UTI prognostic marker or nonantibiotic-based therapy.


Assuntos
Infecções por Escherichia coli/enzimologia , Rim/enzimologia , Ribonucleases/genética , Bexiga Urinária/enzimologia , Infecções Urinárias/enzimologia , Infecções Urinárias/microbiologia , Escherichia coli Uropatogênica , Adolescente , Animais , Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/genética , Criança , Pré-Escolar , Feminino , Inativação Gênica , Humanos , Imunidade Inata , Lactente , Rim/microbiologia , Masculino , Camundongos , Camundongos Transgênicos , Fenótipo , Prognóstico , Bexiga Urinária/microbiologia , Urotélio/metabolismo , Urotélio/patologia , Adulto Jovem
10.
J Immunol ; 198(6): 2383-2393, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28167628

RESUMO

Studies have shown that exposure to psychological stressors leads to inflammation throughout the body. This has been widely studied using social disruption (SDR), a social stressor that involves repeated social defeat in subordinate mice. Exposure to SDR increases serum cytokine levels, results in accumulation of spleen CD11b+ myeloid cells, and primes macrophages for increased cytokine and microbicidal activity. Our previous studies showed that intestinal microbes are necessary for SDR-enhancement of innate immunity. In this study, we show that SDR increases spleen CD11b+Ly6CintermLy6G+ neutrophil and CD11b+Ly6ChiLy6G-monocyte numbers compared with control mice. Further, we found that neutrophils and monocytes from stressor-exposed mice expressed higher levels of IL-1ß mRNA. To determine whether bacterial translocation may contribute to these effects, bacterial 16S rRNA was quantified using quantitative real-time RT-PCR with bacterial group-specific primers. Exposure to the SDR stressor specifically increased Lactobacillus RNA in the spleen, which localized in spleen monocytes. The increased spleen levels of Lactobacillus 16S rRNA in SDR mice positively correlated with increased levels of IL-1ß and IL-23 mRNA. Our findings indicate that during stressor exposure, Lactobacillus spp. can translocate to the spleen and prime the innate immune system for enhanced reactivity.


Assuntos
Translocação Bacteriana , Imunidade Inata , Lactobacillus/fisiologia , Monócitos/imunologia , Neutrófilos/imunologia , Baço/imunologia , Estresse Psicológico/imunologia , Animais , Células Cultivadas , Exposição Ambiental/efeitos adversos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-23/genética , Interleucina-23/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Ribossômico 16S/genética , Baço/microbiologia , Estresse Psicológico/microbiologia , Simbiose , Regulação para Cima
11.
J Pediatr Gastroenterol Nutr ; 68(4): 533-540, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30540706

RESUMO

OBJECTIVES: Gastrointestinal disorders, such as inflammatory bowel diseases (IBDs) and functional gastrointestinal disorders (FGIDs), involve disrupted homeostatic interactions between the microbiota and the host. Both disorders are worsened during stress, and in laboratory mice, stress exposure has been shown to change the composition of the gut microbiome. Stress-induced changes to the microbiome exacerbate intestinal inflammation and alter intestinal motility in mice. It is, however, not yet known whether microbiota-derived short-chain fatty acids (butyrate, propionate, and acetate) and their receptors contribute to this effect. METHODS: Mice were exposed to a social disruption stress, or left undisturbed as a control. After the first stress exposure, mice were orally challenged with Citrobacter rodentium or with vehicle. The levels of short-chain fatty acids (SCFAs) were measured using gas chromatography-mass spectrometry. SCFA receptors were measured via real-time polymerase chain reaction. Microbial community composition was assessed using 16S rRNA gene sequencing. RESULTS: Stress exposure reduced colonic SCFA levels. Stress exposure and C rodentium, however, significantly increased SCFA levels and changed the expression of SCFA receptors. The levels of SCFAs did not correlate with the severity of colonic inflammation, but the colonic expression of the SCFA receptor GPR41 was positively associated with inflammatory cytokines and colonic histopathology scores. The relative abundances of several taxa of colonic bacteria were significantly changed by stress exposure, including SCFA producers. CONCLUSIONS: Social stress can have a significant effect on infection-induced colonic inflammation, and stress-induced changes in microbial-produced metabolites and their receptors may be involved.


Assuntos
Ansiedade , Doenças Inflamatórias Intestinais/psicologia , Estresse Psicológico , Animais , Modelos Animais de Doenças , Ácidos Graxos Voláteis/metabolismo , Microbioma Gastrointestinal , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
12.
Am J Primatol ; 81(10-11): e22969, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30941799

RESUMO

The gastrointestinal microbiome is recognized as a critical component in host immune function, physiology, and behavior. Early life experiences that alter diet and social contact also influence these outcomes. Despite the growing number of studies in this area, no studies to date have examined the contribution of early life experiences on the gut microbiome in infants across development. Such studies are important for understanding the biological and environmental factors that contribute to optimal gut microbial colonization and subsequent health. We studied infant rhesus monkeys (Macaca mulatta) across the first 6 months of life that were pseudo-randomly assigned to one of two different rearing conditions at birth: mother-peer-reared (MPR), in which infants were reared in social groups with many other adults and peers and nursed on their mothers, or nursery-reared (NR), in which infants were reared by human caregivers, fed formula, and given daily social contact with peers. We analyzed the microbiome from rectal swabs (total N = 97; MPR = 43, NR = 54) taken on the day of birth and at postnatal Days 14, 30, 90, and 180 using 16S rRNA gene sequencing. Bacterial composition differences were evident as early as 14 days, with MPR infants exhibiting a lower abundance of Bifidobacterium and a higher abundance of Bacteroides than NR infants. The most marked differences were observed at 90 days, when Bifidobacterium, Lactobacillus, Streptococcus, Bacteroides, Clostridium, and Prevotella differed across rearing groups. By Day 180, no differences in the relative abundances of the bacteria of interest were observed. These novel findings in developing primate neonates indicate that the early social environment as well as diet influence gut microbiota composition very early in life. These results also lay the groundwork for mechanistic studies examining the effects of early experiences on gut microbiota across development with the ultimate goal of understanding the clinical significance of developmental changes.


Assuntos
Microbioma Gastrointestinal , Macaca mulatta/microbiologia , Meio Social , Criação de Animais Domésticos , Animais , Animais Recém-Nascidos/microbiologia , Bactérias/classificação , Bactérias/genética , Dieta , Feminino , Masculino , Mães , Análise de Sequência de DNA
13.
Cancer ; 124(20): 3990-3999, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-29975400

RESUMO

Increasing scientific attention is focused on the gut-brain axis, including the ability of the gastrointestinal (GI) tract to modulate central nervous system function. Changes in the intestinal microbiome can influence affective-like behavior, cognitive performance, fatigue, and sleep in rodents and humans. Patients with cancer who are receiving chemotherapy experience similar negative behavioral changes and concurrent GI symptoms. These chemotherapy comorbidities can be long-lasting and may reduce patients' quality of life and motivation to comply with treatment. This review summarizes the clinical and preclinical evidence supporting a role for the intestinal microbiome in mediating behavioral comorbidities through peripheral immune activation in patients with cancer who are receiving chemotherapy. In addition, evidence suggesting that targeted modification of the intestinal microbiome during cancer treatment could ameliorate associated behavioral comorbidities is reviewed.


Assuntos
Antineoplásicos/efeitos adversos , Microbioma Gastrointestinal/fisiologia , Transtornos Mentais/induzido quimicamente , Neoplasias/tratamento farmacológico , Neuroimunomodulação/fisiologia , Encéfalo/fisiologia , Comunicação Celular/fisiologia , Comorbidade , Gastroenteropatias/induzido quimicamente , Gastroenteropatias/imunologia , Gastroenteropatias/microbiologia , Gastroenteropatias/terapia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/inervação , Trato Gastrointestinal/microbiologia , Humanos , Sistema Imunitário/fisiologia , Transtornos Mentais/epidemiologia , Transtornos Mentais/imunologia , Transtornos Mentais/microbiologia , Neoplasias/epidemiologia , Neoplasias/imunologia , Neoplasias/microbiologia
14.
Am J Physiol Gastrointest Liver Physiol ; 315(3): G408-G419, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29848024

RESUMO

One significant drawback of current probiotic therapy for the prevention of necrotizing enterocolitis (NEC) is the need for at least daily administration because of poor probiotic persistence after enteral administration, increasing the risk of the probiotic bacteria causing bacteremia or sepsis if the intestines are already compromised. We previously showed that the effectiveness of Lactobacillus reuteri ( Lr) in preventing NEC is enhanced when Lr is grown as a biofilm on the surface of dextranomer microspheres (DM). Here we sought to test the efficacy of Lr administration by manipulating the Lr biofilm state with the addition of biofilm-promoting substances (sucrose and maltose) to DM or by mutating the Lr gtfW gene (encoding an enzyme central to biofilm production). Using an animal model of NEC, we determined that Lr adhered to sucrose- or maltose-loaded DM significantly reduced histologic injury, improved host survival, decreased intestinal permeability, reduced intestinal inflammation, and altered the gut microbiome compared with Lr adhered to unloaded DM. These effects were abolished when DM or GtfW were absent from the Lr inoculum. This demonstrates that a single dose of Lr in its biofilm state decreases NEC incidence. Importantly, preloading DM with sucrose or maltose further enhances Lr protection against NEC in a GtfW-dependent fashion, demonstrating the tunability of the approach and the potential to use other cargos to enhance future probiotic formulations. NEW & NOTEWORTHY Previous clinical trials of probiotics to prevent necrotizing enterocolitis have had variable results. In these studies, probiotics were delivered in their planktonic, free-living form. We have developed a novel probiotic delivery system in which Lactobacillus reuteri (Lr) is delivered in its biofilm state. In a model of experimental necrotizing enterocolitis, this formulation significantly reduces intestinal inflammation and permeability, improves survival, and preserves the natural gut microflora compared with the administration of Lr in its free-living form.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Enterocolite Necrosante , Inflamação , Intestinos , Limosilactobacillus reuteri/fisiologia , Probióticos/farmacologia , Animais , Animais Recém-Nascidos , Biofilmes/crescimento & desenvolvimento , Dextranos/farmacologia , Enterocolite Necrosante/microbiologia , Enterocolite Necrosante/prevenção & controle , Inflamação/tratamento farmacológico , Inflamação/microbiologia , Intestinos/efeitos dos fármacos , Intestinos/microbiologia , Intestinos/fisiopatologia , Microesferas , Ratos , Ratos Sprague-Dawley
15.
Brain Behav Immun ; 66: 18-22, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28843452

RESUMO

The focus on the microbiome for the 2017 Named Series in Brain, Behavior, and Immunity reflects the rapidly growing interest in commensal microbes and the effects that they can have on physiological processes often studied in PsychoNeuroImmunology Research. The studies included in this Named Series show that commensal microbes can impact immune system activity, as well as brain and behavioral processes across the lifespan, and are involved in behavioral and immunological responses to social stresses. The studies also show that dietary effects on brain, behavior, and immunity often involve alterations of the gut microbiota. Thus, diet can be used therapeutically for diseases and conditions involving the brain, behavior, and immunity, as can treatment with both pre- and probiotics. While this has been widely tested in animal models, fewer studies have focused on pre- and probiotic treatment in humans. The studies in this Named Series highlight the challenges of probiotic research in human populations, but also highlight the future promise of probiotics for human health. While emotional disorders, such as anxiety and depression have been often been linked to alterations in the gut microbiota, studies in this Named Series identify new domains involving interactions between the microbiota, brain, behavior, and immunity, including schizophrenia, traumatic brain injury, and stroke. As a whole, this collection of work demonstrates the importance of the microbiome in regulating key aspects of immunity, brain, and behavior, and provides important rationale for extending the work so that findings can be translated into clinical practice.

16.
Brain Behav Immun ; 60: 44-50, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27633986

RESUMO

Exposure to a prolonged restraint stressor disrupts the colonic microbiota community composition, and is associated with an elevated inflammatory response to colonic pathogen challenge. Since the stability of the microbiota has been implicated in the development and modulation of mucosal immune responses, we hypothesized that the disruptive effect of the stressor upon the microbiota composition directly contributed to the stressor-induced exacerbation of pathogen-induced colitis. In order to establish a causative role for stressor-induced changes in the microbiota, conventional mice were exposed to prolonged restraint to change the microbiota. Germfree mice were then colonized by microbiota from either stressor-exposed or non-stressed control mice. One day after colonization, mice were infected with the colonic pathogen, Citrobacter rodentium. At six days post-infection, mice that received microbiota from stressor-exposed animals had significant increases in colonic pathology and pro-inflammatory cytokine (e.g. IL-1ß) and chemokine (e.g. CCL2) levels after C. rodentium infection in comparison with mice that received microbiota from non-stressed mice. 16S rRNA gene sequencing revealed that microbial communities from stressed mice did not have any detectable Bifidobacterium present, a stark contrast with the microbial communities from non-stressed mice, suggesting that stressor-induced alterations in commensal, immunomodulatory Bifidobacterium levels may predispose to an increased inflammatory response to pathogen challenge. This study demonstrates that the commensal microbiota directly contribute to excessive inflammatory responses to C. rodentium during stressor exposure, and may help to explain why gastrointestinal disorders are worsened during stressful experiences.


Assuntos
Citrobacter rodentium/imunologia , Mucosa Intestinal/imunologia , Microbiota/imunologia , Estresse Fisiológico/imunologia , Animais , Colo/imunologia , Colo/patologia , Suscetibilidade a Doenças/imunologia , Imunidade nas Mucosas/imunologia , Masculino , Camundongos , Simbiose/imunologia
17.
Brain Behav Immun ; 64: 50-58, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28027927

RESUMO

Recent studies demonstrate that exposure to stress changes the composition of the intestinal microbiota, which is associated with development of stress-induced changes to social behavior, anxiety, and depression. Stress during pregnancy has also been related to the emergence of these disorders; whether commensal microbes are part of a maternal intrauterine environment during prenatal stress is not known. Here, we demonstrate that microbiome changes are manifested in the mother, and also found in female offspring in adulthood, with a correlation between stressed mothers and female offspring. Alterations in the microbiome have been shown to alter immune responses, thus we examined cytokines in utero. IL-1ß was increased in placenta and fetal brain from offspring exposed to the prenatal stressor. Because IL-1ß has been shown to prevent induction of brain derived neurotrophic factor (BDNF), we examined BDNF and found a reduction in female placenta and adult amygdala, suggesting in utero impact on neurodevelopment extending into adulthood. Furthermore, gastrointestinal microbial communities were different in adult females born from stressed vs. non-stressed pregnancies. Adult female offspring also demonstrated increased anxiety-like behavior and alterations in cognition, suggesting a critical window where stress is able to influence the microbiome and the intrauterine environment in a deleterious manner with lasting behavioral consequences. The microbiome may be a key link between the intrauterine environment and adult behavioral changes.


Assuntos
Placenta/microbiologia , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Estresse Psicológico/microbiologia , Animais , Ansiedade , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Citocinas/metabolismo , Encefalite/metabolismo , Encefalite/microbiologia , Feminino , Microbioma Gastrointestinal , Inflamação/metabolismo , Inflamação/microbiologia , Mediadores da Inflamação/metabolismo , Interleucina-1beta/metabolismo , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural/metabolismo , Placenta/metabolismo , Gravidez , Complicações na Gravidez/microbiologia , Simbiose
18.
Horm Behav ; 88: 70-78, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27760302

RESUMO

Over the past decade, it has become increasingly evident that there are extensive bidirectional interactions between the body and its microbiota. These interactions are evident during stressful periods, where it is recognized that commensal microbiota community structure is significantly changed. Many different stressors, ranging from early life stressors to stressors administered during adulthood, lead to significant, community-wide differences in the microbiota. The mechanisms through which this occurs are not yet known, but it is known that commensal microbes can recognize, and respond to, mammalian hormones and neurotransmitters, including those that are involved with the physiological response to stressful stimuli. In addition, the physiological stress response also changes many aspects of gastrointestinal physiology that can impact microbial community composition. Thus, there are many routes through which microbial community composition might be disrupted during stressful periods. The implications of these disruptions in commensal microbial communities for host health are still not well understood, but the commensal microbiota have been linked to stressor-induced immunopotentiation. The role of the microbiota in stressor-induced immunopotentiation can be adaptive, such as when these microbes stimulate innate defenses against bacterial infection. However, the commensal microbiota can also lead to maladaptive immune responses during stressor-exposure. This is evident in animal models of colonic inflammation where stressor exposure increases the inflammation through mechanisms involving the microbiota. It is likely that during stressor exposure, immune cell functioning is regulated by combined effects of both neurotransmitters/hormones and commensal microbes. Defining this regulation should be a focus of future studies.


Assuntos
Imunomodulação/fisiologia , Microbiota/fisiologia , Estresse Fisiológico/fisiologia , Animais , Humanos , Inflamação/fisiopatologia
19.
Adv Exp Med Biol ; 874: 225-46, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26589222

RESUMO

Psychological stress is an intrinsic part of life that affects all organs of the body through direct nervous system innervation and the release of neuroendocrine hormones. The field of PsychoNeuroImmunology (PNI) has clearly demonstrated that the physiological response to psychological stressors can dramatically impact the functioning of the immune system, thus identifying one way in which susceptibility to or severity of diseases are exacerbated during stressful periods. This chapter describes research at the interface between the fields of PNI and Microbial Endocrinology to demonstrate that natural barrier defenses, such as those provided by the commensal microflora, can be disrupted by exposure to psychological stressors. These stress effects are evident in the development of the intestinal microflora in animals born from stressful pregnancy conditions, and in older animals with fully developed microbial populations. Moreover, data are presented demonstrating that exposure to different types of stressors results in the translocation of microflora from cutaneous and mucosal surfaces into regional lymph nodes. When considered together, a scenario emerges in which psychological stressors induce a neuroendocrine response that has the potential to directly or indirectly affect commensal microflora populations, the integrity of barrier defenses, and the internalization of microbes. Finally, a hypothesis is put forth in which stressor-induced alterations of the microflora contribute to the observed stressor-induced increases in inflammatory markers in the absence of overt infection.


Assuntos
Bactérias/isolamento & purificação , Intestinos/microbiologia , Estresse Psicológico/imunologia , Animais , Translocação Bacteriana , Humanos , Imunidade , Linfonodos/microbiologia , Estresse Psicológico/microbiologia
20.
Adv Exp Med Biol ; 874: 289-300, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26589225

RESUMO

The body harbors a vast array of microbes that are collectively known as the microbiota. Increasing attention is being paid to the role of the gut microbiota in the health of the host. Gut microbial communities are relatively resistant to change, though alterations in homeostasis can also significantly change gut microbial community structure. An important factor that has been demonstrated to alter the composition of the gut microbiota is exposure to psychological stressors. And, evidence indicates that the commensal microbiota are involved in stressor-induced immunomodulation. This chapter will discuss the impact of psychosocial stress on immunity, and present evidence that stressor-induced alterations in the composition of gut microbial communities contributes to stressor-induced immunomodulation and neurobiological sequelae. Finally, the role of the microbiota in the perinatal time period will be explored, and an integrative hypothesis of the role of the microbiome in health and stress response will be proposed.


Assuntos
Microbioma Gastrointestinal/fisiologia , Estresse Psicológico/imunologia , Estresse Psicológico/microbiologia , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA