Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Neurogastroenterol Motil ; 35(11): e14676, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37772676

RESUMO

BACKGROUND: Early-life events impact maturation of the gut microbiome, enteric nervous system, and gastrointestinal motility. We examined three regions of gastric tissue to determine how maternal separation and gut microbes influence the structure and motor function of specific regions of the neonatal mouse stomach. METHODS: Germ-free and conventionally housed C57BL/6J mouse pups underwent timed maternal separation (TmSep) or nursed uninterrupted (controls) until 14 days of life. We assessed gastric emptying by quantifying the progression of gavaged fluorescein isothiocyanate (FITC)-dextran. With isolated rings of forestomach, corpus, and antrum, we measured tone and contractility by force transduction, gastric wall thickness by light microscopy, and myenteric plexus neurochemistry by whole-mount immunostaining. KEY RESULTS: Regional gastric sampling revealed site-specific differences in contractile patterns and myenteric plexus structure. In neonatal mice, TmSep prolonged gastric emptying. In the forestomach, TmSep increased contractile responses to carbachol, decreased muscularis externa and mucosa thickness, and increased the relative proportion of myenteric plexus nNOS+ neurons. Germ-free conditions did not appreciably alter the structure or function of the neonatal mouse stomach and did not impact the changes caused by TmSep. CONCLUSIONS AND INFERENCES: A regional sampling approach facilitates site-specific investigations of murine gastric motor physiology and histology to identify site-specific alterations that may impact gastrointestinal function. Delayed gastric emptying in TmSep is associated with a thinner muscle wall, exaggerated cholinergic contractile responses, and increased proportions of inhibitory myenteric plexus nNOS+ neurons in the forestomach. Gut microbes do not profoundly affect the development of the neonatal mouse stomach or the gastric pathophysiology that results from TmSep.


Assuntos
Gastroparesia , Camundongos , Animais , Animais Recém-Nascidos , Privação Materna , Camundongos Endogâmicos C57BL , Estômago , Plexo Mientérico/patologia , Modelos Animais de Doenças , Esvaziamento Gástrico
2.
Res Sq ; 2023 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-36778495

RESUMO

Acute gastrointestinal intestinal GVHD (aGI-GVHD) is a serious complication of allogeneic hematopoietic stem cell transplantation, and the intestinal microbiota is known to impact on its severity. However, an association between treatment response of aGI-GVHD and the intestinal microbiota has not been well-studied. In a cohort of patients with aGI-GVHD (n=37), we found that non-response to standard therapy with corticosteroids was associated with prior treatment with carbapenem antibiotics and loss of Bacteroides ovatus from the microbiome. In a mouse model of carbapenem-aggravated GVHD, introducing Bacteroides ovatus reduced severity of GVHD and improved survival. Bacteroides ovatus reduced degradation of colonic mucus by another intestinal commensal, Bacteroides thetaiotaomicron, via its ability to metabolize dietary polysaccharides into monosaccharides, which then inhibit mucus degradation by Bacteroides thetaiotaomicron and reduce GVHD-related mortality.

3.
Front Nutr ; 9: 831879, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35419389

RESUMO

The nutrient sensing nuclear receptor peroxisome proliferator-activated receptor-α (PPARα) regulates the host response to short-term fasting by inducing hepatic transcriptional programming of ketogenesis, fatty acid oxidation and transport, and autophagy. This adaptation is ineffective in chronically undernourished individuals, among whom dyslipidemia and hepatic steatosis are common. We recently reported that hepatic PPARα protein is profoundly depleted in male mice undernourished by a low-protein, low-fat diet. Here, we identify PPARα as a deacetylation target of the NAD-dependent deacetylase sirtuin-1 (SIRT1) and link this to the decrease in PPARα protein levels in undernourished liver. Livers from undernourished male mice expressed high levels of SIRT1, with decreased PPARα acetylation and strongly decreased hepatic PPARα protein. In cultured hepatocytes, PPARα protein levels were decreased by transiently transfecting constitutively active SIRT1 or by treating cells with the potent SIRT1 activator resveratrol, while silencing SIRT1 increased PPARα protein levels. SIRT1 expression is correlated with increased PPARα ubiquitination, suggesting that protein loss is due to proteasomal degradation. In accord with these findings, the dramatic loss of hepatic PPARα in undernourished male mice was completely restored by treating mice with the proteasome inhibitor bortezomib. Similarly, treating undernourished mice with the SIRT1 inhibitor selisistat/EX-527 completely restored hepatic PPARα protein. These data suggest that induction of SIRT1 in undernutrition results in hepatic PPARα deacetylation, ubiquitination, and degradation, highlighting a new mechanism that mediates the liver's failed adaptive metabolic responses in chronic undernutrition.

4.
Front Immunol ; 12: 693462, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34691016

RESUMO

Background: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first reported in December 2019 in Wuhan, China, and then rapidly spread causing an unprecedented pandemic. A robust serological assay is needed to evaluate vaccine candidates and better understand the epidemiology of coronavirus disease (COVID-19). Methods: We used the full-length spike (S) protein of SARS-CoV-2 for the development of qualitative and quantitative IgG and IgA anti-S enzyme linked immunosorbent assays (ELISA). A total of 320 sera used for assay development were comprised of pandemic sera from SARS-CoV-2 infected adults (n=51) and pre-pandemic sera (n=269) including sera from endemic human coronavirus infected adults. Reverse cumulative curves and diagnostic test statistics were evaluated to define the optimal serum dilution and OD cutoff value for IgG anti-S and IgA anti-S ELISAs. The IgG and IgA anti-S, and three functional antibodies (ACE-2 receptor blocking antibody, lentipseudovirus-S neutralizing antibody, and SARS-CoV-2 neutralizing antibody) were measured using additional SARS-CoV-2 PCR positive sera (n=76) and surveillance sera (n=25). Lastly, the IgG and IgA anti-S levels were compared in different demographic groups. Results: The optimal serum dilution for the qualitative IgG anti-S ELISA was at 1:1024 yielding a 99.6% specificity, 92.2% sensitivity, 92.9% positive predictive value (PPV), and 99.6% negative predictive value (NPV) at a SARS-CoV-2 seroprevalence of 5%. The optimal serum dilution for the qualitative IgA anti-S ELISA was at 1:128 yielding a 98.9% specificity, 76.5% sensitivity, 78.3% PPV, and 98.8% NPV at the same seroprevalence. Significant correlations were demonstrated between the IgG and IgA (r=0.833 for concentrations, r=0.840 for titers) as well as between IgG and three functional antibodies (r=0.811-0.924 for concentrations, r=0.795-0.917 for titers). The IgG and IgA anti-S levels were significantly higher in males than females (p<0.05), and in adults with moderate/severe symptoms than in adults with mild/moderate symptoms (p<0.001). Conclusion: We developed a highly specific and sensitive IgG anti-S ELISA assay to SARS-CoV-2 using full length S protein. The IgG anti-S antibody level was strongly associated with IgA and functional antibody levels in adults with SARS-CoV-2 infection. Gender and disease severity, rather than age, play an important role in antibody levels.


Assuntos
Anticorpos Antivirais/imunologia , COVID-19/imunologia , Imunoglobulina A/imunologia , Imunoglobulina G/imunologia , SARS-CoV-2/imunologia , Adulto , COVID-19/diagnóstico , Teste Sorológico para COVID-19 , Feminino , Células HEK293 , Humanos
5.
Hepatol Commun ; 4(12): 1835-1850, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33305154

RESUMO

Liver dysfunction, including coagulopathy, is a prominent feature of protein-energy malnutrition. To identify mechanisms underlying malnutrition-associated coagulopathy, we administered a low-protein low-fat diet to lactating dams and examined hepatic transcription and plasma coagulation parameters in young adult weanlings. Malnutrition impacted body composition to a greater extent in male versus female mice. Transcriptional profiles suggested opposing effects of nutrient-sensing nuclear receptors, namely induction of peroxisome proliferator-activated receptor α (PPARα) targets and repression of farnesoid-X-receptor (FXR) targets. Coagulopathy with decreased synthesis of fibrinogen-α (FGA) and factor 11 (F11) was observed in malnourished male animals but not female animals. In primary mouse hepatocytes, FXR agonist increased and PPARα agonist decreased Fga and F11 messenger RNA expression. Nuclear receptor DNA response elements were identified in the Fga and F11 gene regulatory regions, and opposing effects of FXR and PPARα were confirmed with luciferase assays. Unexpectedly, hepatic PPARα protein was markedly depleted in malnourished male liver and was not enriched on Fga or F11 response elements. Rather, there was loss of FXR binding at these response elements. Reduced PPARα protein was associated with loss of hepatocyte peroxisomes, which are necessary for bile acid biosynthesis, and with decreased concentrations of bile acids that function as FXR ligands, most notably the FXR agonist chenodeoxycholic acid. Conclusion: Malnutrition impairs growth and liver synthetic function more severely in male mice than in female mice. Malnourished male mice are coagulopathic and exhibit decreased hepatocyte peroxisomes, FXR agonist bile acids, FXR binding on Fga and F11 gene regulatory elements, and coagulation factor synthesis. These effects are absent in female mice, which have low baseline levels of PPARα, suggesting that nutrient-sensing nuclear receptors regulate coagulation factor synthesis in response to host nutritional status in a sex-specific manner.

6.
Neurogastroenterol Motil ; 32(12): e13936, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33021011

RESUMO

BACKGROUND: Slow gastrointestinal (GI) transit occurs in moderate-to-severe malnutrition. Mechanisms underlying malnutrition-associated dysmotility remain unknown, partially due to lack of animal models. This study sought to characterize GI dysmotility in mouse models of malnutrition. METHODS: Neonatal mice were malnourished by timed maternal separation. Alternatively, low-protein, low-fat diet was administered to dams, with malnourished neonates tested at two weeks or weaned to the same chow and tested as young adults. We determined total GI transit time by carmine red gavage, colonic motility by rectal bead latency, and both gastric emptying and small bowel motility with fluorescein isothiocyanate-conjugated dextran. We assessed histology with light microscopy, ex vivo contractility and permeability with force-transduction and Ussing chamber studies, and gut microbiota composition by 16S rDNA sequencing. KEY RESULTS: Both models of neonatal malnutrition and young adult malnourished males but not females exhibited moderate growth faltering, stunting, and grossly abnormal stomachs. Progression of fluorescent dye was impaired in both neonatal models of malnutrition, whereas gastric emptying was delayed only in maternally separated pups and malnourished young adult females. Malnourished young adult males but not females had atrophic GI mucosa, exaggerated intestinal contractile responses, and increased gut barrier permeability. These sex-specific abnormalities were associated with altered gut microbial communities. CONCLUSIONS & INFERENCES: Multiple models of early-life malnutrition exhibit delayed upper GI transit. Malnutrition affects young adult males more profoundly than females. These models will facilitate future studies to identify mechanisms underlying malnutrition-induced pathophysiology and sex-specific regulatory effects.


Assuntos
Gastroenteropatias/fisiopatologia , Motilidade Gastrointestinal/fisiologia , Desnutrição/fisiopatologia , Privação Materna , Caracteres Sexuais , Fatores Etários , Animais , Animais Recém-Nascidos , Feminino , Gastroenteropatias/etiologia , Gastroenteropatias/psicologia , Trânsito Gastrointestinal/fisiologia , Masculino , Desnutrição/complicações , Desnutrição/psicologia , Camundongos , Camundongos Endogâmicos C57BL
7.
Clin Infect Dis ; 70(10): 2095-2102, 2020 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-31253983

RESUMO

BACKGROUND: Clostridioides (Clostridium) difficile colonization is common among infants. Serological sequelae of infant C. difficile colonization are poorly understood. METHODS: In this prospective cohort study of healthy infants, stools serially collected between ages 1-2 and 9-12 months were tested for non-toxigenic and toxigenic C. difficile (TCD). Cultured isolates underwent whole-genome sequencing. Serum collected at 9-12 months underwent measurement of IgA, IgG, and IgM against TCD toxins A and B and neutralizing antibody (NAb) titers against toxin B. For comparison, antitoxin IgG and NAb were measured in cord blood from 50 mothers unrelated to study infants. RESULTS: Among 32 infants, 16 (50%) were colonized with TCD; 12 were first colonized >1 month before serology measurements. A variety of sequence types were identified, and there was evidence of putative in-home (enrolled siblings) and outpatient clinic transmission. Infants first colonized with TCD >1 month prior had significantly greater serum antitoxin IgA and IgG against toxins A (P = .02 for both) and B (P = .009 and .008, respectively) compared with non-TCD-colonized infants, and greater IgG compared with unrelated cord blood (P = .005). Five of 12 (42%) colonized infants had detectable NAb titers compared with zero non-TCD-colonized infants (P = .02). Breastfeeding was not associated with differences in serological measurements. CONCLUSIONS: TCD colonization is associated with a humoral immune response against toxins A and B, with evidence of toxin B neutralization in vitro. The extent and duration of protection against CDI later in life afforded by natural C. difficile immunization events require further investigation.


Assuntos
Clostridioides difficile , Infecções por Clostridium , Clostridioides , Infecções por Clostridium/prevenção & controle , Feminino , Humanos , Imunização , Lactente , Estudos Prospectivos
8.
Nat Commun ; 10(1): 3650, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31409776

RESUMO

The intestinal immune system is emerging as an important contributor to obesity-related insulin resistance, but the role of intestinal B cells in this context is unclear. Here, we show that high fat diet (HFD) feeding alters intestinal IgA+ immune cells and that IgA is a critical immune regulator of glucose homeostasis. Obese mice have fewer IgA+ immune cells and less secretory IgA and IgA-promoting immune mediators. HFD-fed IgA-deficient mice have dysfunctional glucose metabolism, a phenotype that can be recapitulated by adoptive transfer of intestinal-associated pan-B cells. Mechanistically, IgA is a crucial link that controls intestinal and adipose tissue inflammation, intestinal permeability, microbial encroachment and the composition of the intestinal microbiome during HFD. Current glucose-lowering therapies, including metformin, affect intestinal-related IgA+ B cell populations in mice, while bariatric surgery regimen alters the level of fecal secretory IgA in humans. These findings identify intestinal IgA+ immune cells as mucosal mediators of whole-body glucose regulation in diet-induced metabolic disease.


Assuntos
Imunoglobulina A/imunologia , Resistência à Insulina , Obesidade/imunologia , Tecido Adiposo/imunologia , Animais , Linfócitos B/imunologia , Estudos de Coortes , Fezes/microbiologia , Microbioma Gastrointestinal , Glucose/metabolismo , Humanos , Intestinos/imunologia , Masculino , Camundongos , Obesidade/metabolismo , Obesidade/microbiologia
9.
Neurogastroenterol Motil ; 31(9): e13654, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31157504

RESUMO

BACKGROUND: An important limitation of gastrointestinal motility testing is high variability. Conditions that could contribute to variability, including the duration of pretest fasting and time of day, are rarely reported and have not been examined systematically. This study aimed to explore whether these conditions, as well as age, sex, and strain of mice, affect the results of a standard laboratory test of upper gastrointestinal motility. METHODS: Male and female 8-week-old C57BL/6J mice received a gastric gavage of fluorescein isothiocyanate (FITC)-conjugated dextran. FITC-dextran distribution was measured 30 minutes later. Mean geometric centers (MGCs) were calculated to determine the effects of short versus prolonged fasting and morning versus afternoon testing. The influence of age was assessed in 2- to 10-week-old animals, and the influence of strain was determined in C57BL/6J, BALB/c, and CD-1 mice. KEY RESULTS: Motility was sexually dimorphic. MGC progressed 19% further in 8-week-old C57BL/6J males versus females when tested in the morning after a short fast. Similar patterns were observed in morning or afternoon testing after overnight fasting. In males, motility was unaffected by time of day; however, MGC progressed 31% further in females tested in the afternoon versus morning after a short fast. Sex differences also were present in CD-1 but not BALB/c mice. Testing in neonates revealed strikingly low variability and no sex differences. CONCLUSIONS & INFERENCES: Fasting duration, time of day, age, sex, and strain of mice all influence upper gastrointestinal motility testing. Sex differences are not present in neonatal pups, but develop soon after weaning.


Assuntos
Jejum/fisiologia , Motilidade Gastrointestinal/fisiologia , Caracteres Sexuais , Fatores Etários , Animais , Animais Recém-Nascidos , Dextranos/administração & dosagem , Feminino , Fluoresceína-5-Isotiocianato/administração & dosagem , Fluoresceína-5-Isotiocianato/análogos & derivados , Motilidade Gastrointestinal/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Especificidade da Espécie , Fatores de Tempo
10.
Science ; 363(6430): 993-998, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30819965

RESUMO

Immunoglobulin A (IgA) is the major secretory immunoglobulin isotype found at mucosal surfaces, where it regulates microbial commensalism and excludes luminal factors from contacting intestinal epithelial cells (IECs). IgA is induced by both T cell-dependent and -independent (TI) pathways. However, little is known about TI regulation. We report that IEC endoplasmic reticulum (ER) stress induces a polyreactive IgA response, which is protective against enteric inflammation. IEC ER stress causes TI and microbiota-independent expansion and activation of peritoneal B1b cells, which culminates in increased lamina propria and luminal IgA. Increased numbers of IgA-producing plasma cells were observed in healthy humans with defective autophagy, who are known to exhibit IEC ER stress. Upon ER stress, IECs communicate signals to the peritoneum that induce a barrier-protective TI IgA response.


Assuntos
Estresse do Retículo Endoplasmático , Células Epiteliais/imunologia , Imunidade nas Mucosas , Imunoglobulina A/imunologia , Mucosa Intestinal/imunologia , Animais , Autofagia , Proteínas Relacionadas à Autofagia/genética , Humanos , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasmócitos/imunologia , Técnicas de Cultura de Tecidos , Proteína 1 de Ligação a X-Box/genética
11.
Proc Natl Acad Sci U S A ; 114(4): E570-E579, 2017 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-28069942

RESUMO

The intestinal epithelium can limit enteric pathogens by producing antiviral cytokines, such as IFNs. Type I IFN (IFN-α/ß) and type III IFN (IFN-λ) function at the epithelial level, and their respective efficacies depend on the specific pathogen and site of infection. However, the roles of type I and type III IFN in restricting human enteric viruses are poorly characterized as a result of the difficulties in cultivating these viruses in vitro and directly obtaining control and infected small intestinal human tissue. We infected nontransformed human intestinal enteroid cultures from multiple individuals with human rotavirus (HRV) and assessed the host epithelial response by using RNA-sequencing and functional assays. The dominant transcriptional pathway induced by HRV infection is a type III IFN-regulated response. Early after HRV infection, low levels of type III IFN protein activate IFN-stimulated genes. However, this endogenous response does not restrict HRV replication because replication-competent HRV antagonizes the type III IFN response at pre- and posttranscriptional levels. In contrast, exogenous IFN treatment restricts HRV replication, with type I IFN being more potent than type III IFN, suggesting that extraepithelial sources of type I IFN may be the critical IFN for limiting enteric virus replication in the human intestine.


Assuntos
Interferons/genética , Intestino Delgado/imunologia , Infecções por Rotavirus/genética , Animais , Linhagem Celular , Chlorocebus aethiops , Humanos , Imunidade Inata , Interferons/imunologia , Rotavirus/fisiologia , Infecções por Rotavirus/imunologia , Análise de Sequência de RNA , Replicação Viral
12.
Anaerobe ; 40: 54-7, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27180007

RESUMO

Lifeway(®) kefir, a fermented milk product containing 12 probiotic organisms, is reported to show promise as an alternative to fecal microbiota transplantation for recurrent Clostridium difficile infection (CDI). We employed a murine CDI model to study the probiotic protective mechanisms and unexpectedly determined that kefir drastically increased disease severity. Our results emphasize the need for further independent clinical testing of kefir as alternative therapy in recurrent CDI.


Assuntos
Clostridioides difficile/efeitos dos fármacos , Enterocolite Pseudomembranosa/patologia , Kefir/efeitos adversos , Probióticos/efeitos adversos , Animais , Clostridioides difficile/crescimento & desenvolvimento , Clostridioides difficile/patogenicidade , Contagem de Colônia Microbiana , Modelos Animais de Doenças , Progressão da Doença , Enterocolite Pseudomembranosa/microbiologia , Enterocolite Pseudomembranosa/mortalidade , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Índice de Gravidade de Doença , Análise de Sobrevida
13.
J Nutr ; 146(5): 940-8, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27052538

RESUMO

BACKGROUND: Protein-energy undernutrition during early development confers a lifelong increased risk of obesity-related metabolic disease. Mechanisms by which metabolic abnormalities persist despite catch-up growth are poorly understood. OBJECTIVE: We sought to determine whether abnormal metabolomic and intestinal microbiota profiles from undernourished neonatal mice remain altered during catch-up growth. METHODS: Male and female CD1 mouse pups were undernourished by timed separation from lactating dams for 4 h at 5 d of age, 8 h at 6 d of age, and 12 h/d from 7 to 15 d of age, then resumed ad libitum nursing, whereas controls fed uninterrupted. Both groups were weaned simultaneously to a standard unpurified diet. At 3 time points (0, 1, and 3 wk after ending feed deprivation), metabolites in urine, plasma, and stool were identified with the use of mass spectrometry, and fecal microbes were identified with the use of 16S metagenomic sequencing. RESULTS: Undernourished mice completely recovered deficits of 36% weight and 9% length by 3 wk of refeeding, at which time they had 1.4-fold higher plasma phenyllactate and 2.0-fold higher urinary p-cresol sulfate concentrations than did controls. Plasma serotonin concentrations in undernourished mice were 25% lower at 0 wk but 1.5-fold higher than in controls at 3 wk. Whereas most urine and plasma metabolites normalized with refeeding, 117 fecal metabolites remained altered at 3 wk, including multiple N-linked glycans. Microbiota profiles from undernourished mice also remained distinct, with lower mean proportions of Bacteroidetes (67% compared with 83%) and higher proportions of Firmicutes (26% compared with 16%). Abundances of the mucolytic organisms Akkermansia muciniphila and Mucispirillum schaedleri were altered at 0 and 1 wk. Whereas microbiota from undernourished mice at 0 wk contained 11% less community diversity (P = 0.015), refed mice at 3 wk harbored 1.2-fold greater diversity (P = 0.0006) than did controls. CONCLUSION: Microbial-derived metabolites and intestinal microbiota remain altered during catch-up growth in undernourished neonatal mice.


Assuntos
Animais Recém-Nascidos , Bactérias/metabolismo , Microbioma Gastrointestinal , Crescimento , Intestinos/microbiologia , Desnutrição Proteico-Calórica/microbiologia , Aumento de Peso , Animais , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Bacteroidetes/crescimento & desenvolvimento , Bacteroidetes/metabolismo , Biomarcadores/metabolismo , Cresóis/urina , Fezes , Feminino , Firmicutes/crescimento & desenvolvimento , Firmicutes/metabolismo , Mucosa Intestinal/metabolismo , Lactatos/sangue , Masculino , Metagenômica , Camundongos , Obesidade/etiologia , Obesidade/microbiologia , Polissacarídeos/metabolismo , Desnutrição Proteico-Calórica/dietoterapia , Desnutrição Proteico-Calórica/metabolismo , Serotonina/sangue , Ésteres do Ácido Sulfúrico/urina , Desmame
14.
J Virol ; 90(1): 43-56, 2016 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-26446608

RESUMO

UNLABELLED: Human gastrointestinal tract research is limited by the paucity of in vitro intestinal cell models that recapitulate the cellular diversity and complex functions of human physiology and disease pathology. Human intestinal enteroid (HIE) cultures contain multiple intestinal epithelial cell types that comprise the intestinal epithelium (enterocytes and goblet, enteroendocrine, and Paneth cells) and are physiologically active based on responses to agonists. We evaluated these nontransformed, three-dimensional HIE cultures as models for pathogenic infections in the small intestine by examining whether HIEs from different regions of the small intestine from different patients are susceptible to human rotavirus (HRV) infection. Little is known about HRVs, as they generally replicate poorly in transformed cell lines, and host range restriction prevents their replication in many animal models, whereas many animal rotaviruses (ARVs) exhibit a broader host range and replicate in mice. Using HRVs, including the Rotarix RV1 vaccine strain, and ARVs, we evaluated host susceptibility, virus production, and cellular responses of HIEs. HRVs infect at higher rates and grow to higher titers than do ARVs. HRVs infect differentiated enterocytes and enteroendocrine cells, and viroplasms and lipid droplets are induced. Heterogeneity in replication was seen in HIEs from different patients. HRV infection and RV enterotoxin treatment of HIEs caused physiological lumenal expansion detected by time-lapse microscopy, recapitulating one of the hallmarks of rotavirus-induced diarrhea. These results demonstrate that HIEs are a novel pathophysiological model that will allow the study of HRV biology, including host restriction, cell type restriction, and virus-induced fluid secretion. IMPORTANCE: Our research establishes HIEs as nontransformed cell culture models to understand human intestinal physiology and pathophysiology and the epithelial response, including host restriction of gastrointestinal infections such as HRV infection. HRVs remain a major worldwide cause of diarrhea-associated morbidity and mortality in children ≤5 years of age. Current in vitro models of rotavirus infection rely primarily on the use of animal rotaviruses because HRV growth is limited in most transformed cell lines and animal models. We demonstrate that HIEs are novel, cellularly diverse, and physiologically relevant epithelial cell cultures that recapitulate in vivo properties of HRV infection. HIEs will allow the study of HRV biology, including human host-pathogen and live, attenuated vaccine interactions; host and cell type restriction; virus-induced fluid secretion; cell-cell communication within the epithelium; and the epithelial response to infection in cultures from genetically diverse individuals. Finally, drug therapies to prevent/treat diarrheal disease can be tested in these physiologically active cultures.


Assuntos
Intestino Delgado/virologia , Modelos Teóricos , Técnicas de Cultura de Órgãos/métodos , Infecções por Rotavirus/patologia , Infecções por Rotavirus/virologia , Rotavirus/fisiologia , Replicação Viral , Humanos , Intestino Delgado/fisiologia
15.
J Nutr Biochem ; 26(10): 1050-7, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26070414

RESUMO

Undernutrition remains one of the key global health challenges facing children today. Distinct microbial profiles have been associated with obesity and undernutrition, although mechanisms behind these associations are unknown. We sought to understand how protein-energy undernutrition alters the microbiome and to propose mechanisms by which these alterations influence the malnourished phenotype. Outbred CD1 neonatal mice were undernourished by timed separation from lactating dams, while control animals nursed ad libitum. 16S rRNA gene sequencing and compositional analysis identified microbes from luminal contents of ileum, cecum and colon, while whole metagenome shotgun sequencing identified microbial gene content. Our results suggest that the most important determinant of microbiome composition is body compartment; communities derived from ileum are distinct from those from cecum and colon as observed by phylogenetic clustering analysis. However, within each compartment, microbiota from undernourished and control mice cluster separately. At the phylum level, undernourished mice harbor more Verrucomicrobia and less Bacteroidetes in the distal intestine; these changes are driven by an increase in Akkermansia muciniphila and decreases in Bacteroides and Alistipes. Undernourished mice have an overall loss of microbial community richness and diversity and are deficient in multiple microbial genetic pathways including N-glycan, inositol phosphate and one-carbon metabolism. Losses in these microbial genes may confer less efficient extraction of energy from nondigestible dietary components including glycans and phytates, whereas epigenetic alterations provide a means of persistently altering metabolism even after adequate nutrition is restored. Thus, the microbiome of an undernourished host may perpetuate states of poor nutrition via multiple mechanisms.


Assuntos
Animais Recém-Nascidos/microbiologia , Microbioma Gastrointestinal/fisiologia , Desnutrição/microbiologia , Animais , Bactérias/classificação , Bactérias/genética , DNA Bacteriano/análise , Camundongos , Desnutrição Proteico-Calórica/microbiologia , RNA Ribossômico 16S/genética
16.
J Nutr ; 144(3): 273-81, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24381221

RESUMO

Undernutrition contributes to half of all childhood deaths under the age of 5 y, and confers upon survivors a life-long predisposition to obesity, type 2 diabetes, and cardiovascular disease. Mechanisms underlying the link between early nutrient deprivation and noncommunicable diseases are unknown. Using outbred CD1 neonatal mice, we measured metabolic profile differences between conventionally reared mice given unrestricted access to nursing, the control group, and undernourished mice subjected to protein-calorie deprivation through timed separation from lactating mothers. After 11 d of undernutrition, urine, plasma, liver, ileal fluid, cecal fluid, and stool were harvested from 8 pools of 4 neonatal mice per group. The metabolome was identified using a multiplatform mass spectrometry-based approach, and random forest metrics were used to identify the most important metabolites that distinguished the undernourished from the control group. Our data reveal striking metabolic changes in undernourished mice consistent with the known mammalian response to starvation, including evidence of muscle and fat catabolism and increased reliance on the tricarboxylic acid cycle for energy. However, we also revealed evidence of liver and biliary injury, anomalies in bile acid metabolism, oxidative stress and inflammation, accelerated heme breakdown, and altered regulation of DNA methylation. Among the metabolites that most strongly distinguished the 2 groups were 2-hydroxyisobutyrate, increased 3-fold in plasma of undernourished mice (P = 2.19 × 10(-11)); urobilinogen, increased 11-fold in urine of undernourished mice (P = 4.22 × 10(-7)); deoxycholate, decreased 94% in stool of undernourished mice (P = 3.0 × 10(-4)); and 12 different products of the enzyme γ-glutamyltransferase, increased in all 6 compartments of undernourished mice. This model of the undernourished neonatal metabolome illustrates the wide range of pathways disrupted by undernutrition in early development, and suggests mechanistic links between early starvation and persistent metabolic diseases.


Assuntos
Doenças Biliares/patologia , Inflamação/patologia , Hepatopatias/patologia , Desnutrição/patologia , Metaboloma , Estresse Oxidativo , Animais , Animais Recém-Nascidos , Doenças Biliares/etiologia , Biomarcadores/sangue , Biomarcadores/urina , Metilação de DNA , Feminino , Hidroxibutiratos/sangue , Inflamação/etiologia , Hepatopatias/etiologia , Masculino , Desnutrição/complicações , Camundongos , gama-Glutamiltransferase/metabolismo
17.
Am J Physiol Gastrointest Liver Physiol ; 306(5): G445-53, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24407593

RESUMO

Intestinal intussusception (ISS) commonly causes intestinal obstruction in children. One mechanism that has been proposed to cause ISS is inflammation-induced alteration of intestinal motility. We investigated whether innate inflammatory factors or altered motility is required for induction of ISS by LPS. We compared rates of ISS among BALB/c and C57BL/6 mice, mice lacking lymphocytes or depleted of phagocytes, or mice with defects in the Toll-like receptor 4 (TLR4) signaling pathway following administration of LPS or the Ca(2+) analog MnCl2. At 6 or 2 h after administration of LPS or MnCl2, respectively, mice underwent image analysis to assess intestinal contraction rate or laparotomy to identify ISS. LPS-induced ISS (LPS-ISS) was observed in BALB/c mice, but not in C57BL/6 mice or any BALB/c mice with disruptions of TLR4 signaling. LPS-induced serum TNF-α, IL-6, and nitric oxide (NO) and intestinal NO levels were similar in BALB/c and C57BL/6 mice. The rate of LPS-ISS was significantly reduced in phagocyte-depleted, but not lymphocyte-deficient, mice. Intestinal contraction rates were reduced in LPS-ISS-susceptible BALB/c mice, but not in LPS-ISS-resistant C57BL/6 or TLR4 mutant mice, suggesting a role for reduced intestinal contraction rate in LPS-ISS susceptibility. This was tested with MnCl2, a Ca(2+) antagonist that reduced intestinal contraction rates and induced ISS, irrespective of mouse strain. Therefore, LPS-ISS is initiated by innate immune signaling that requires TLR4 and phagocytes but may be independent of TNF-α, IL-6, and NO levels. Furthermore, alteration of intestinal motility, specifically, reduced intestinal contraction rate, is a key factor in the development of ISS.


Assuntos
Cloretos/toxicidade , Motilidade Gastrointestinal/efeitos dos fármacos , Imunidade Inata/fisiologia , Intussuscepção/induzido quimicamente , Lipopolissacarídeos/toxicidade , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Feminino , Regulação da Expressão Gênica/imunologia , Imunidade Inata/efeitos dos fármacos , Intussuscepção/imunologia , Intussuscepção/metabolismo , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/metabolismo , Masculino , Compostos de Manganês , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
18.
Microbes Infect ; 16(1): 67-72, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24095866

RESUMO

Regulatory T cells produce TGF-ß that contributes to IgA induction by intestinal commensal bacteria but their importance in IgA responses to pathogens has not been determined. Immunity against the enteropathogen, rotavirus, is dependent on intestinal IgA, but whether FoxP3(+) regulatory T cells contribute to this IgA is unknown. Infection with rotavirus increased the numbers of intestinal FoxP3(+) regulatory T cells. Depletion of FoxP3(+) regulatory T cells altered leukocyte activation but did not significantly alter rotavirus clearance or specific antibody levels. These data suggest FoxP3(+) regulatory T cells are not critical for the early antibody response to rotavirus infection.


Assuntos
Infecções por Rotavirus/imunologia , Infecções por Rotavirus/virologia , Rotavirus/imunologia , Linfócitos T Reguladores/imunologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Feminino , Fatores de Transcrição Forkhead/metabolismo , Leucócitos/imunologia , Leucócitos/metabolismo , Leucócitos/virologia , Contagem de Linfócitos , Depleção Linfocítica , Masculino , Camundongos , Linfócitos T Reguladores/metabolismo
19.
Front Immunol ; 4: 402, 2013 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-24348474

RESUMO

Viral gastroenteritis is one of the leading causes of diseases that kill ~2.2 million people worldwide each year. IgA is one of the major immune effector products present in the gastrointestinal tract yet its importance in protection against gastrointestinal viral infections has been difficult to prove. In part this has been due to a lack of small and large animal models in which pathogenesis of and immunity to gastrointestinal viral infections is similar to that in humans. Much of what we have learned about the role of IgA in the intestinal immune response has been obtained from experimental animal models of rotavirus infection. Rotavirus-specific intestinal IgA appears to be one of the principle effectors of long term protection against rotavirus infection. Thus, there has been a focus on understanding the immunological pathways through which this virus-specific IgA is induced during infection. In addition, the experimental animal models of rotavirus infection provide excellent systems in which new areas of research on viral-specific intestinal IgA including the long term maintenance of viral-specific IgA.

20.
J Virol ; 87(1): 524-30, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23097456

RESUMO

Rotavirus is a major cause of pediatric diarrheal illness worldwide. To explore the role of organized intestinal lymphoid tissues in infection by and immunity to rotavirus, lymphotoxin alpha-deficient (LTα(-/-)) mice that lack Peyer's patches and mesenteric lymph nodes were orally infected with murine rotavirus. Systemic rotavirus was cleared within 10 days in both LTα(-/-) and wild-type mice, and both strains developed early and sustained serum antirotavirus antibody responses. However, unlike wild-type mice, which resolved the intestinal infection within 10 days, LTα(-/-) mice shed fecal virus for approximately 50 days after inoculation. The resolution of fecal virus shedding occurred concurrently with induction of intestinal rotavirus-specific IgA in both mouse strains. Induction of intestinal rotavirus-specific IgA in LTα(-/-) mice correlated with the (late) appearance of IgA-producing plasma cells in the small intestine. This, together with the absence of rotavirus-specific serum IgA, implies that secretory rotavirus-specific IgA was produced locally. These findings indicate that serum IgG responses are insufficient and imply that local intestinal IgA responses are important for the clearance of rotavirus from intestinal tissues. Furthermore, they show that while LTα-dependent lymphoid tissues are important for the generation of IgA-producing B cells in the intestine, they are not absolutely required in the setting of rotavirus infection. Moreover, the induction of local IgA-producing B cell responses can occur late after infection and in an LTα-independent manner.


Assuntos
Imunidade nas Mucosas , Imunoglobulina A/imunologia , Linfotoxina-alfa/deficiência , Infecções por Rotavirus/imunologia , Rotavirus/imunologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Linfócitos B/imunologia , Fezes/virologia , Feminino , Imunoglobulina A/sangue , Intestino Delgado/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Plasmócitos/imunologia , Fatores de Tempo , Eliminação de Partículas Virais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA