Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Allergy Clin Immunol ; 143(5): 1865-1877.e4, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30439403

RESUMEN

BACKGROUND: Psychological stress and heightened mast cell (MC) activation are linked with important immunologic disorders, including allergy, anaphylaxis, asthma, and functional bowel diseases, but the mechanisms remain poorly defined. We have previously demonstrated that activation of the corticotropin-releasing factor (CRF) system potentiates MC degranulation responses during IgE-mediated anaphylaxis and psychological stress through corticotropin-releasing factor receptor subtype 1 (CRF1) expressed on MCs. OBJECTIVE: In this study we investigated the role of corticotropin-releasing factor receptor subtype 2 (CRF2) as a modulator of stress-induced MC degranulation and associated disease pathophysiology. METHODS: In vitro MC degranulation assays were performed with bone marrow-derived mast cells (BMMCs) derived from wild-type (WT) and CRF2-deficient (CRF2-/-) mice and RBL-2H3 MCs transfected with CRF2-overexpressing plasmid or CRF2 small interfering RNA. In vivo MC responses and associated pathophysiology in IgE-mediated passive systemic anaphylaxis and acute psychological restraint stress were measured in WT, CRF2-/-, and MC-deficient KitW-sh/W-sh knock-in mice. RESULTS: Compared with WT mice, CRF2-/- mice exhibited greater serum histamine levels and exacerbated IgE-mediated anaphylaxis and colonic permeability. In addition, CRF2-/- mice exhibited increased serum histamine levels and colonic permeability after acute restraint stress. Experiments with BMMCs and RBL-2H3 MCs demonstrated that CRF2 expressed on MCs suppresses store-operated Ca2+ entry signaling and MC degranulation induced by diverse MC stimuli. Experiments with MC-deficient KitW-sh/W-sh mice systemically engrafted with WT and CRF2-/- BMMCs demonstrated the functional importance of MC CRF2 in modulating stress-induced pathophysiology. CONCLUSIONS: MC CRF2 is a negative global modulator of stimuli-induced MC degranulation and limits the severity of IgE-mediated anaphylaxis and stress-related disease pathogenesis.


Asunto(s)
Anafilaxia/inmunología , Mucosa Intestinal/metabolismo , Mastocitos/fisiología , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Estrés Psicológico/inmunología , Uniones Estrechas/metabolismo , Animales , Degranulación de la Célula , Línea Celular , Modelos Animales de Enfermedad , Femenino , Liberación de Histamina/genética , Humanos , Inmunoglobulina E/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Receptores de Hormona Liberadora de Corticotropina/genética
2.
Am J Physiol Regul Integr Comp Physiol ; 310(11): R1244-57, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27053649

RESUMEN

We investigated whether corticotropin-releasing factor receptor 2 (CRF2) and its high-affinity agonist urocortin 1 (Ucn1) mediate sex-specific signaling and immune responses. Intrarectal trinitrobenzene sulfonic acid was used to induce experimental colitis in wild-type, CRF2 knockout (CRF2KO), and heterozygous (CRF2Ht) mice of both sexes. Changes in plasma extravasation, organ weight, survival, immune cell numbers, inflammatory cytokines, and the MAPK signaling pathway were assessed. Stored intestinal biopsies from patients with Crohn's disease (CD) and age- and sex-matched individuals without inflammatory bowel disease (IBD) were examined by immunofluorescence and confocal microscopy to characterize Ucn1 and CRF receptor expression. CRF2Ht mice of both sexes showed decreased survival during colitis compared with other genotypes. Ucn1 improved survival in male mice alone. Ucn1 restored colon length and spleen and adrenal weight and decreased colonic TNF-α, IL-6, and IL-1ß levels in male CRF2Ht mice alone. CRF2Ht mice of both sexes showed decreased phosphorylation of MAPK p38 and heat shock protein 27 (Hsp27) levels. Ucn1 restored p-Hsp27 levels in male CRF2Ht mice alone. Expression of the chaperone protein Hsp90 decreased during colitis, except in male CRF2Ht mice. Taken together, our data indicate that sex shows significant interaction with genotype and Ucn1 during colitis. Human duodenal and colonic biopsies revealed that sex-specific differences exist in levels of CRF receptors and Ucn1 expression in patients with CD compared with the matched non-IBD subjects. To conclude, Ucn1 mediates sex-specific immune and cellular signaling responses via CRF2, emphasizing the need for inclusion of females in preclinical studies.


Asunto(s)
Colitis/inmunología , Citocinas/inmunología , Mediadores de Inflamación/inmunología , Inflamación/inmunología , Receptores de Hormona Liberadora de Corticotropina/inmunología , Urocortinas/inmunología , Animales , Femenino , Masculino , Ratones , Caracteres Sexuales
3.
Proc Natl Acad Sci U S A ; 110(2): 731-6, 2013 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-23267070

RESUMEN

Enteritis caused by Clostridium difficile toxin (Tx) is a nosocomial disease of increasing clinical concern, but the local mediators of C. difficile TxA inflammation are unknown. The potent vasodilator calcitonin gene-related peptide mediates neurogenic inflammation via the calcitonin receptor-like receptor (CLR). Here we examined the ileum-specific effects of reducing CLR on TxA ileitis by local preinjection of double-stranded RNAs. Treatment with CLR dsRNA for 7 d decreased CLR immunoreactivity, whereas treatment with non-CLR dsRNA did not. Subsequent injection of TxA in the same location increased CLR in rats treated with non-CLR dsRNA but not in rats treated with CLR dsRNA, documenting that local injection of dsRNA is effective in preventing the increase in CLR immunoreactivity in response to local TxA. After non-CLR dsRNA pretreatment, TxA induced robust intestinal secretion, myeloperoxidase activity, and histopathologic indications of inflammation including epithelial damage, congestion, neutrophil infiltration, loss of mucin from goblet cells, and increase in mast cell numbers. After CLR dsRNA pretreatment, TxA-induced changes in intestinal secretion and histopathologic inflammation were improved, including normal mucin staining and fewer resident mast cells. Loss of CLR prevented TxA-mediated activation of NF-κB and concomitant increases in pERK1/2 and TNF-α mRNA. Locally produced CLR plays a proinflammatory role in TxA ileitis via MAPK signaling and TNF-α. The results reported here strongly suggest that a local injection of dsRNA targeting CLR could be an effective local therapeutic approach at the inflammation site in the treatment of a growing, clinically relevant hospital-acquired disease, C. difficile infection.


Asunto(s)
Toxinas Bacterianas/toxicidad , Proteína Similar al Receptor de Calcitonina/metabolismo , Enterotoxinas/toxicidad , Ileítis/inducido químicamente , Ileítis/tratamiento farmacológico , ARN Bicatenario/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Western Blotting , Proteína Similar al Receptor de Calcitonina/administración & dosificación , Proteína Similar al Receptor de Calcitonina/inmunología , Células Caliciformes/efectos de los fármacos , Masculino , Mastocitos/efectos de los fármacos , Microscopía Fluorescente , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mucinas/metabolismo , FN-kappa B/metabolismo , Infiltración Neutrófila/efectos de los fármacos , Peroxidasa/metabolismo , Interferencia de ARN , ARN Bicatenario/administración & dosificación , ARN Bicatenario/metabolismo , Ratas , Ratas Sprague-Dawley , Estadísticas no Paramétricas , Factor de Necrosis Tumoral alfa/metabolismo
4.
Sci Rep ; 14(1): 15256, 2024 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-38956202

RESUMEN

Posttraumatic stress disorder (PTSD) can develop after trauma exposure. Some studies report that women develop PTSD at twice the rate of men, despite greater trauma exposure in men. Lipids and their metabolites (lipidome) regulate a myriad of key biological processes and pathways such as membrane integrity, oxidative stress, and neuroinflammation in the brain by maintaining neuronal connectivity and homeostasis. In this study, we analyzed the lipidome of 40 adults with PTSD and 40 trauma-exposed non-PTSD individuals (n = 20/sex/condition; 19-39 years old). Plasma samples were analyzed for lipidomics using Quadrupole Time-of-Flight (QToF) mass spectrometry. Additionally, ~ 90 measures were collected, on sleep, and mental and physical health indices. Poorer sleep quality was associated with greater PTSD severity in both sexes. The lipidomics analysis identified a total of 348 quantifiable known lipid metabolites and 1951 lipid metabolites that are yet unknown; known metabolites were part of 13 lipid subclasses. After adjusting for BMI and sleep quality, in women with PTSD, only one lipid subclass, phosphatidylethanolamine (PE) was altered, whereas, in men with PTSD, 9 out of 13 subclasses were altered compared to non-PTSD women and men, respectively. Severe PTSD was associated with 22% and 5% of altered lipid metabolites in men and women, respectively. Of the changed metabolites, only 0.5% measures (2 PEs and cholesterol) were common between women and men with PTSD. Several sphingomyelins, PEs, ceramides, and triglycerides were increased in men with severe PTSD. The correlations between triglycerides and ceramide metabolites with cholesterol metabolites and systolic blood pressure were dependent upon sex and PTSD status. Alterations in triglycerides and ceramides are linked with cardiac health and metabolic function in humans. Thus, disturbed sleep and higher body mass may have contributed to changes in the lipidome found in PTSD.


Asunto(s)
Lipidómica , Trastornos por Estrés Postraumático , Humanos , Trastornos por Estrés Postraumático/metabolismo , Trastornos por Estrés Postraumático/sangre , Masculino , Femenino , Adulto , Lipidómica/métodos , Adulto Joven , Lípidos/sangre , Estudios de Cohortes , Metabolismo de los Lípidos
5.
bioRxiv ; 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38464224

RESUMEN

Posttraumatic stress disorder (PTSD) can develop after trauma exposure. Some studies report that women develop PTSD at twice the rate of men, despite greater trauma exposure in men. Lipids and their metabolites (lipidome) regulate a myriad of key biological processes and pathways such as membrane integrity, oxidative stress, and neuroinflammation in the brain by maintaining neuronal connectivity and homeostasis. In this study, we analyzed the lipidome of 40 individuals with PTSD and 40 trauma-exposed non-PTSD individuals. Plasma samples were analyzed for lipidomics using Quadrupole Time-of-Flight (QToF) mass spectrometry. Additionally, ~ 90 measures were collected, on sleep, mental and physical health indices. Sleep quality worsened as PTSD severity increased in both sexes. The lipidomics analysis identified a total of 348 quantifiable known lipid metabolites and 1951 lipid metabolites that are yet unknown; known metabolites were part of 13 classes of lipids. After adjusting for sleep quality, in women with PTSD, only one lipid subclass, phosphatidylethanolamine (PE) was altered, whereas, in men with PTSD, 9 out of 13 subclasses were altered compared to non-PTSD women and men, respectively. Severe PTSD was associated with 22% and 5% of altered lipid metabolites in men and women, respectively. Of the changed metabolites, only 0.5% measures (2 PEs and cholesterol) were common between women and men with PTSD. Several sphingomyelins, PEs, ceramides, and triglycerides were increased in men with severe PTSD. The triglycerides and ceramide metabolites that were most highly increased were correlated with cholesterol metabolites and systolic blood pressure in men but not always in women with PTSD. Alterations in triglycerides and ceramides are linked with cardiac health and metabolic function in humans. Thus, disturbed sleep and higher weight may have contributed to changes in the lipidome found in PTSD.

6.
Mol Med ; 19: 212-22, 2013 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-23835907

RESUMEN

Although females suffer twice as much as males from stress-related disorders, sex-specific participating and pathogenic cellular stress mechanisms remain uncharacterized. Using corticotropin-releasing factor receptor 2-deficient (Crhr2-/-) and wild-type (WT) mice, we show that CRF receptor type 2 (CRF2) and its high-affinity ligand, urocortin 1 (Ucn1), are key mediators of the endoplasmic reticulum (ER) stress response in a murine model of acute pancreatic inflammation. Ucn1 was expressed de novo in acinar cells of male, but not female WT mice during acute inflammation. Upon insult, acinar Ucn1 induction was markedly attenuated in male but not female Crhr2-/- mice. Crhr2-/- mice of both sexes show exacerbated acinar cell inflammation and necrosis. Electron microscopy showed mild ER damage in WT male mice and markedly distorted ER structure in Crhr2-/- male mice during pancreatitis. WT and Crhr2-/- female mice showed similarly distorted ER ultrastructure that was less severe than distortion seen in Crhr2-/- male mice. Damage in ER structure was accompanied by increased ubiquitination, peIF2, and mistargeted localization of vimentin in WT mice that was further exacerbated in Crhr2-/- mice of both sexes during pancreatitis. Exogenous Ucn1 rescued many aspects of histological damage and cellular stress response, including restoration of ER structure in male WT and Crhr2-/- mice, but not in females. Instead, females often showed increased damage. Thus, specific cellular pathways involved in coping and resolution seem to be distinct to each sex. Our results demonstrate the importance of identifying sex-specific pathogenic mechanisms and their value in designing effective therapeutics.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , Pancreatitis/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Células Acinares/metabolismo , Amilasas/metabolismo , Animales , Línea Celular , Ceruletida , Femenino , Masculino , Ratones , Ratones Transgénicos , Pancreatitis/inducido químicamente , Factores Sexuales , Urocortinas/metabolismo
7.
Vitam Horm ; 123: 27-65, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37717988

RESUMEN

Stress responses to physical, psychological, environmental, or cellular stressors, has two arms: initiation and recovery. Corticotropin-releasing factor (CRF) is primarily responsible for regulating and/or initiating stress responses via, whereas urocortins (UCNs) are involved in the recovery response to stress via feedback inhibition. Stress is a loaded, polysemous word and is experienced in a myriad of ways. Some stressors are good for an individual, in fact essential, whereas other stressors are associated with bad outcomes. Perceived stress, like beauty, lies in the eye of the beholder, and hence the same stressor can result in individual-specific outcomes. In mammals, there are two main biological sexes with reproduction as primary function. Reproduction and nutrition can also be viewed as stressors; based on a body of work from my laboratory, we propose that the functions of all other organs have co-evolved to optimize and facilitate an individual's nutritional and reproductive functions. Hence, sex differences in physiologically relevant outcomes are innate and occur at all levels- molecular, endocrine, immune, and (patho)physiological. CRF and three UCNs are peptide hormones that mediate their physiological effects by binding to two known G protein-coupled receptors (GPCRs), CRF1 and CRF2. Expression and function of CRF family of hormones and their receptors is likely to be sexually dimorphic in all organs. In this chapter, based on the large body of work from others and my laboratory, an overview of the CRF family with special emphasis on sex-specific actions of peripherally expressed CRF2 receptor in health and disease is provided.


Asunto(s)
Hormona Liberadora de Corticotropina , Transducción de Señal , Femenino , Masculino , Animales , Humanos , Reproducción , Caracteres Sexuales , Cognición , Mamíferos
8.
Cells ; 12(22)2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37998327

RESUMEN

COVID-19-associated intensive care unit (ICU) admissions were recognized as critical health issues that contributed to morbidity and mortality in SARS-CoV-2-infected patients. Severe symptoms in COVID-19 patients are often accompanied by cytokine release syndrome. Here, we analyzed publicly available data from the Yale IMPACT cohort to address immunological misfiring and sex differences in early COVID-19 patients. In 2020, SARS-CoV-2 was considered far more pathogenic and lethal than other circulating respiratory viruses, and the inclusion of SARS-CoV-2 negative patients in IMPACT cohorts confounds many findings. We ascertained the impact of several important biological variables such as days from symptom onset (DFSO); pre-existing risk factors, including obesity; and early COVID-19 treatments on significantly changed immunological measures in ICU-admitted COVID-19 patients that survived versus those that did not. Deceased patients had 19 unique measures that were not shared with ICU patients including increased granzyme-B-producing GzB+CD8+ T cells and interferon-γ. Male COVID-19 patients in ICU experienced many more changes in immunological and clinical measures than female ICU patients (25% vs. ~16%, respectively). A total of 13/124 measures including CCL5, CCL17, IL-18, IFNα2, Fractalkine, classical monocytes, T cells, and CD4Temra exhibited significant sex differences in female vs. male COVID-19 patients. A total of nine measures including IL-21, CCL5, and CD4Temra differed significantly between female and male healthy controls. Immunosuppressed patients experienced the most decreases in CD4Temra and CD8Tem cell numbers. None of the early COVID-19 treatments were effective in reducing levels of IL-6, a major component of the cytokine storm. Obesity (BMI >30) was the most impactful risk factor for COVID-19-related deaths and worst clinical outcomes. Our analysis highlights the contribution of biological sex, risk factors, and early treatments with respect to COVID-19-related ICU admission and progression to morbidity and mortality.


Asunto(s)
COVID-19 , Humanos , Femenino , Masculino , SARS-CoV-2 , Linfocitos T CD8-positivos , Caracteres Sexuales , Obesidad/epidemiología
9.
AIMS Public Health ; 10(1): 105-115, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37063355

RESUMEN

Backgrounds: Data on the association between comorbid diabetes mellitus (DM) and acute pancreatitis (AP) remains limited. Utilizing a large, nationwide database, we aimed to examine the impact of comorbid diabetes mellitus on patients admitted for acute pancreatitis. Methods: This was a retrospective case-control study of adult patients with AP utilizing the National Inpatient Sample from 2015-2018, using ICD-10 codes. Hospitalization outcomes of patients admitted for AP with comorbid DM were compared to those without comorbid DM at the time of admission. The primary outcome was a mortality difference between the cohorts. Multivariable-adjusted cox proportional hazards model analysis was performed. Data was analyzed as both sex aggregated, and sex segregated. Results: 940,789 adult patients with AP were included, of which 256,330 (27.3%) had comorbid DM. Comorbid DM was associated with a 31% increased risk of inpatient mortality (aOR: 1.31; p = 0.004), a 53% increased risk of developing sepsis (aOR: 1.53; p = 0.002), increased hospital length of stay (LOS) (4.5 days vs. 3.7 days; p < 0.001), and hospital costs ($9934 vs. $8486; p < 0.001). Whites admitted for AP with comorbid DM were at a 49% increased risk of mortality as compared to Hispanics (aOR: 1.49; p < 0.0001). Different comorbidities had sex-specific risks; men admitted for AP with comorbid DM were at a 28% increased risk of mortality (aOR: 1.28; p < 0.0001) as compared to women. Men with comorbid DM plus obesity or hypertension were also at increased risk of mortality as compared to women, whereas women with comorbid DM plus renal failure were at greater risk of mortality as compared to men. Conclusions: Comorbid DM appears to be a risk factor for adverse hospitalization outcomes in patients admitted for AP with male sex and race as additional risk factors. Future prospective studies are warranted to confirm these findings to better risk stratify this patient population.

10.
bioRxiv ; 2023 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-37662303

RESUMEN

Lung immune tone, i.e. the immune state of the lung, can vary between individuals and over a single individual's lifetime, and its basis and regulation in the context of inflammatory responses to injury is poorly understood. The gut microbiome, through the gut-lung axis, can influence lung injury outcomes but how the diet and microbiota affect lung immune tone is also unclear. We hypothesized that lung immune tone would be influenced by the presence of fiber-fermenting short-chain fatty acid (SCFA)-producing gut bacteria. To test this hypothesis, we conducted a fiber diet intervention study followed by lung injury in mice and profiled gut microbiota using 16S sequencing, metabolomics, and lung immune tone. We also studied germ-free mice to evaluate lung immune tone in the absence of microbiota and performed in vitro mechanistic studies on immune tone and metabolic programming of alveolar macrophages exposed to the SCFA propionate (C3). Mice on high-fiber diet were protected from sterile lung injury compared to mice on a fiber-free diet. This protection strongly correlated with lower lung immune tone, elevated propionate levels and enrichment of specific fecal microbiota taxa; conversely, lower levels of SCFAs and an increase in other fatty acid metabolites and bacterial taxa correlated with increased lung immune tone and increased lung injury in the fiber-free group. In vitro , C3 reduced lung alveolar macrophage immune tone (through suppression of IL-1ß and IL-18) and metabolically reprogrammed them (switching from glycolysis to oxidative phosphorylation after LPS challenge). Overall, our findings reveal that the gut-lung axis, through dietary fiber intake and enrichment of SCFA-producing gut bacteria, can regulate innate lung immune tone via IL-1ß and IL-18 pathways. These results provide a rationale for the therapeutic development of dietary interventions to preserve or enhance specific aspects of host lung immunity.

11.
Med Sci Sports Exerc ; 55(12): 2328-2360, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37772882

RESUMEN

ABSTRACT: Biological sex is a primary determinant of athletic performance because of fundamental sex differences in anatomy and physiology dictated by sex chromosomes and sex hormones. Adult men are typically stronger, more powerful, and faster than women of similar age and training status. Thus, for athletic events and sports relying on endurance, muscle strength, speed, and power, males typically outperform females by 10%-30% depending on the requirements of the event. These sex differences in performance emerge with the onset of puberty and coincide with the increase in endogenous sex steroid hormones, in particular testosterone in males, which increases 30-fold by adulthood, but remains low in females. The primary goal of this consensus statement is to provide the latest scientific knowledge and mechanisms for the sex differences in athletic performance. This review highlights the differences in anatomy and physiology between males and females that are primary determinants of the sex differences in athletic performance and in response to exercise training, and the role of sex steroid hormones (particularly testosterone and estradiol). We also identify historical and nonphysiological factors that influence the sex differences in performance. Finally, we identify gaps in the knowledge of sex differences in athletic performance and the underlying mechanisms, providing substantial opportunities for high-impact studies. A major step toward closing the knowledge gap is to include more and equitable numbers of women to that of men in mechanistic studies that determine any of the sex differences in response to an acute bout of exercise, exercise training, and athletic performance.


Asunto(s)
Rendimiento Atlético , Medicina Deportiva , Adulto , Humanos , Femenino , Masculino , Estados Unidos , Caracteres Sexuales , Rendimiento Atlético/fisiología , Testosterona , Congéneres de la Testosterona , Hormonas Esteroides Gonadales
12.
Biochem J ; 433(1): 51-63, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20961289

RESUMEN

ERK (extracellular-signal-regulated kinase) activation leads to disruption of tight junctions in some epithelial monolayers, whereas it prevents disruption of tight junctions in other epithelia. The factors responsible for such contrasting influences of ERK on tight junction integrity are unknown. The present study investigated the effect of the state of cell differentiation on ERK-mediated regulation of tight junctions in Caco-2 cell monolayers. EGF (epidermal growth factor) potentiated H2O2-induced tight junction disruption in under-differentiated cell monolayers, which was attenuated by the MEK [MAPK (mitogen-activated protein kinase)/ERK kinase] inhibitor U0126. In contrast, EGF prevented H2O2-induced disruption of tight junctions in differentiated cell monolayers, which was also attenuated by U0126. Knockdown of ERK1/2 enhanced tight junction integrity and accelerated assembly of tight junctions in under-differentiated cell monolayers, whereas it had the opposite effect in differentiated cell monolayers. Regulated expression of wild-type and constitutively active MEK1 disrupted tight junctions, and the expression of dominant-negative MEK1 enhanced tight junction integrity in under-differentiated cells, whereas contrasting responses were recorded in differentiated cells. EGF prevented both H2O2-induced association of PP2A (protein phosphatase 2A), and loss of association of PKCζ (protein kinase Cζ), with occludin by an ERK-dependent mechanism in differentiated cell monolayers, but not in under-differentiated cell monolayers. Active ERK was distributed in the intracellular compartment in under-differentiated cell monolayers, whereas it was localized mainly in the perijunctional region in differentiated cell monolayers. Thus ERK may exhibit its contrasting influences on tight junction integrity in under-differentiated and differentiated epithelial cells by virtue of differences in its subcellular distribution and ability to regulate the association of PKCζ and PP2A with tight junction proteins.


Asunto(s)
Diferenciación Celular/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Uniones Estrechas , Células CACO-2 , Células Epiteliales/citología , Células Epiteliales/fisiología , Quinasas MAP Reguladas por Señal Extracelular/análisis , Humanos , MAP Quinasa Quinasa 1/fisiología , Unión Proteica , Proteína Quinasa C/metabolismo , Proteína Fosfatasa 2/metabolismo , Distribución Tisular
13.
Cells ; 11(14)2022 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-35883660

RESUMEN

Glucose uptake by peripheral organs is essential for maintaining blood glucose levels within normal range. Impaired glucose uptake is a hallmark of type 2 diabetes (T2D) and metabolic syndrome and is characterized by insulin resistance. Male sex is an independent risk factor for the development of T2D. We tested whether sex and diet are independent variables for differential glucose uptake by various organs. Here, in a longitudinal study, we used 18F-fluorodeoxyglucose (FDG) and positron emission tomography (PET) to determine baseline differences in whole-body glucose uptake in young male and female mice on chow and high-fat diets. We report that sex and diet are important independent variables that account for differential glucose uptake in brown fat, skeletal muscle, liver, heart, kidney, and the stomach, but not the brain, lungs, pancreas, small intestine, or perigonadal adipose. Of the seven organs analyzed, two organs, namely brown fat, and the heart had the highest concentrations of FDG, followed by the brain, kidneys, and skeletal muscle on chow diet. Young female mice had 47% greater FDG uptake in the brown fat compared to male mice, whereas skeletal muscle FDG uptake was 49% greater in male mice. The high-fat diet inhibited FDG uptake in brown fat, skeletal muscle, and the heart, three major organs involved in uptake, whereas brain uptake was enhanced in both sexes. These foundational and groundbreaking findings suggest that mechanisms of glucose homeostasis are context- and organ-dependent and highlight the need to study sex-specific outcomes and mechanisms for diseases such as T2D, obesity, and metabolic syndrome.


Asunto(s)
Diabetes Mellitus Tipo 2 , Síndrome Metabólico , Animales , Dieta Alta en Grasa , Femenino , Fluorodesoxiglucosa F18/metabolismo , Glucosa , Estudios Longitudinales , Masculino , Ratones
14.
Am J Physiol Gastrointest Liver Physiol ; 300(5): G884-94, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21330446

RESUMEN

Urocortins (UCNs) and their receptors are potent immunoregulators in the gastrointestinal (GI) tract, where they can exert both pro- and anti-inflammatory effects. We examined the contribution of Ucn1 and its receptors to the pathogenesis, progression, and resolution of colitis. Trinitrobenzene sulfonic acid was used to induce colitis in rats. Ucn1 mRNA and immunoreactivity (IR) were ubiquitously expressed throughout the GI tract under basal conditions. During colitis, Ucn1 mRNA levels fell below basal levels on day 1 then increased again by day 6, in association with an increase in the number of Ucn1-IR inflammatory cells. Ucn1-IR cells were also numerous in proliferating granulation tissue. In contrast to Ucn1 expression, average phosphorylated ERK1/2 (pERK1/2) expression rose above controls levels on day 1 and was very low on day 6 of colitis. Knockdown of corticotropin-releasing factor 2 (CRF(2)) but not CRF(1) by RNA interference during colitis significantly decreased the macroscopic lateral spread of ulceration compared with uninjected controls or animals with CRF(1) knockdown. After knockdown of CRF(2), but not of CRF(1) during colitis, edema resolution assessed microscopically was slowed, and myeloperoxidase activity remained elevated even at day 6. Ucn1 and TNF-α mRNA peaked earlier, whereas pERK1/2 activation was attenuated after CRF(2) knockdown. Thus we conclude that local CRF(2) and pERK1/2 activation is pivotal for macroscopic spread of colitis and resolution of edema. Elimination of CRF(2), but not CRF(1), results in uncoordinated immune and pERK1/2 signaling responses.


Asunto(s)
Colitis/fisiopatología , Receptores de Hormona Liberadora de Corticotropina/fisiología , Urocortinas/fisiología , Animales , Western Blotting , Colitis/inmunología , Colitis/patología , Progresión de la Enfermedad , Edema/etiología , Edema/patología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Inmunohistoquímica , Cinética , Masculino , Peroxidasa/metabolismo , Fosforilación , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Ácido Trinitrobencenosulfónico
15.
Cells ; 10(6)2021 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-34205372

RESUMEN

Pain is an unpleasant sensation that alerts one to the presence of obnoxious stimuli or sensations. These stimuli are transferred by sensory neurons to the dorsal root ganglia-spinal cord and finally to the brain. Glial cells in the peripheral nervous system, astrocytes in the brain, dorsal root ganglia, and immune cells all contribute to the development, maintenance, and resolution of pain. Both innate and adaptive immune responses modulate pain perception and behavior. Neutrophils, microglial, and T cell activation, essential components of the innate and adaptive immune responses, can play both excitatory and inhibitory roles and are involved in the transition from acute to chronic pain. Immune responses may also exacerbate pain perception by modulating the function of the cortical-limbic brain regions involved in behavioral and emotional responses. The link between an emotional state and pain perception is larger than what is widely acknowledged. In positive psychological states, perception of pain along with other somatic symptoms decreases, whereas in negative psychological states, these symptoms may worsen. Sex differences in mechanisms of pain perception are not well studied. In this review, we highlight what is known, controversies, and the gaps in this field.


Asunto(s)
Corteza Cerebral/inmunología , Sistema Límbico/inmunología , Microglía/inmunología , Neuronas/inmunología , Dolor/inmunología , Animales , Astrocitos/inmunología , Astrocitos/patología , Corteza Cerebral/patología , Humanos , Sistema Límbico/patología , Activación de Linfocitos , Microglía/patología , Neuronas/patología , Activación Neutrófila , Neutrófilos/inmunología , Neutrófilos/patología , Dolor/patología , Linfocitos T/inmunología , Linfocitos T/patología
16.
Cell Mol Gastroenterol Hepatol ; 12(2): 427-441, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33895424

RESUMEN

Differences in pancreatic anatomy, size, and function exist in men and women. The anatomical differences could contribute to the increase in complications associated with pancreatic surgery in women. Although diagnostic criteria for pancreatitis are the same in men and women, major sex differences in etiology are reported. Alcohol and tobacco predominate in men, whereas idiopathic and obstructive etiologies predominate in women. Circulating levels of estrogens, progesterone, and androgens contribute significantly to overall health outcomes; premenopausal women have lower prevalence of cardiovascular and pancreatic diseases suggesting protective effects of estrogens, whereas androgens promote growth of normal and cancerous cells. Sex chromosomes and gonadal and nongonadal hormones together determine an individual's sex, which is distinct from gender or gender identity. Human pancreatic disease etiology, outcomes, and sex-specific mechanisms are largely unknown. In rodents of both sexes, glucocorticoids and estrogens from the adrenal glands influence pancreatic secretion and acinar cell zymogen granule numbers. Lack of corticotropin-releasing factor receptor 2 function, a G protein-coupled receptor whose expression is regulated by both estrogens and glucocorticoids, causes sex-specific changes in pancreatic histopathology, zymogen granule numbers, and endoplasmic reticulum ultrastructure changes in acute pancreatitis model. Here, we review existing literature on sex differences in the normal exocrine pancreas and mechanisms that operate at homeostasis and diseased states in both sexes. Finally, we review pregnancy-related pancreatic diseases and discuss the effects of sex differences on proposed treatments in pancreatic disease.


Asunto(s)
Páncreas Exocrino/patología , Enfermedades Pancreáticas/patología , Caracteres Sexuales , COVID-19/patología , Femenino , Hormonas/metabolismo , Humanos , Masculino , Embarazo
17.
Neurogastroenterol Motil ; 33(4): e14021, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33118282

RESUMEN

BACKGROUND: Stress increases intestinal secretion and exacerbates symptoms of irritable bowel syndrome (IBS). Peripherally derived corticotropin-releasing factor (CRF) is known to mediate stress-induced intestinal secretion, presumably by activation of CRF1 receptors in the gut. The present study aimed to ascertain the role of CRF2 activation in intestinal secretion by three other members of CRF peptide family, urocortin (UCN) 1-3, in wild type (WT) and CRF2 knockout (Crhr2-/- ) mice. METHODS: Mucosal/submucosal preparations from proximal colon of WT and Crhr2-/- mice of both sexes were mounted in Ussing chambers for measurement of short-circuit current (Isc ) as an indicator of ion secretion. KEY RESULTS: Male mice demonstrated a significantly higher baseline Isc than female in both WT and Crhr2-/- genotypes. CRF and UCN1-3 (1 µM) caused greater increases in colonic Isc (ΔIsc ) in male than female. Colonic Isc response to the selective CRF1 agonist, stressin1, was similar in both sexes. In male mice, the selective CRF2 agonists (UCN2 and UCN3) caused significantly greater ΔIsc than CRF and stressin1. UCN2- and UCN3-evoked ΔISC was significantly reduced in preparations pretreated with the selective CRF2 antagonist antisauvagine-30 and in Crhr2-/- mice. The prosecretory effects of urocortins were due to increases in Cl- secretion and involved enteric neurons and mast cells. CONCLUSIONS AND INFERENCE: The findings revealed sex differences in baseline colonic secretion and responses to stress-related peptides. CRF2 receptors play a more prominent role in colonic secretion in male mice. The greater baseline secretion and responses to UCNs may contribute to the higher prevalence of diarrhea-predominant IBS in males.


Asunto(s)
Cloruros/metabolismo , Colon/metabolismo , Hormona Liberadora de Corticotropina/farmacología , Caracteres Sexuales , Estrés Psicológico/metabolismo , Urocortinas/farmacología , Animales , Colon/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Cultivo de Órganos , Ratas , Receptores de Hormona Liberadora de Corticotropina/deficiencia , Estrés Psicológico/psicología
18.
PLoS One ; 16(6): e0246412, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34111125

RESUMEN

The etiology of necrotizing enterocolitis (NEC) is not known. Alterations in gut microbiome, mucosal barrier function, immune cell activation, and blood flow are characterized events in its development, with stress as a contributing factor. The hormone corticotropin-releasing factor (CRF) is a key mediator of stress responses and influences these aforementioned processes. CRF signaling is modulated by NEC's main risk factors of prematurity and formula feeding. Using an established neonatal rat model of NEC, we tested hypotheses that: (i) increased CRF levels-as seen during stress-promote NEC in formula-fed (FF) newborn rats, and (ii) antagonism of CRF action ameliorates NEC. Newborn pups were formula-fed to initiate gut inflammation and randomized to: no stress, no stress with subcutaneous CRF administration, stress (acute hypoxia followed by cold exposure-NEC model), or stress after pretreatment with the CRF peptide antagonist Astressin. Dam-fed unstressed and stressed littermates served as controls. NEC incidence and severity in the terminal ileum were determined using a histologic scoring system. Changes in CRF, CRF receptor (CRFRs), and toll-like receptor 4 (TLR4) expression levels were determined by immunofluorescence and immunoblotting, respectively. Stress exposure in FF neonates resulted in 40.0% NEC incidence, whereas exogenous CRF administration resulted in 51.7% NEC incidence compared to 8.7% in FF non-stressed neonates (p<0.001). Astressin prevented development of NEC in FF-stressed neonates (7.7% vs. 40.0%; p = 0.003). CRF and CRFR immunoreactivity increased in the ileum of neonates with NEC compared to dam-fed controls or FF unstressed pups. Immunoblotting confirmed increased TLR4 protein levels in FF stressed (NEC model) animals vs. controls, and Astressin treatment restored TLR4 to control levels. Peripheral CRF may serve as specific pharmacologic target for the prevention and treatment of NEC.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/patología , Estrés Fisiológico , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/fisiopatología , Fórmulas Infantiles
19.
Cells ; 10(2)2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33673157

RESUMEN

Maternal stress during pregnancy is linked to several negative birth outcomes. The placenta, a unique pregnancy-specific organ, not only nourishes and protects the fetus but is also the major source of progesterone and estrogens. As the placenta becomes the primary source of maternal progesterone (P4) and estradiol between 6-9 weeks of gestation, and these hormones are critical for maintaining pregnancy, maternal stress may modulate levels of these steroids to impact birth outcomes. The objective was to test whether maternal perceived stress crosses the placental barrier to modulate fetal steroids, including cortisol, which is a downstream indicator of maternal hypothalamic-pituitary-adrenal (HPA) axis regulation and is associated with negative fetal outcomes. Nulliparous women, 18 years or older, with no known history of adrenal or endocrine illness were recruited during their third trimester of pregnancy at the University of California San Francisco (UCSF) Mission Bay hospital obstetrics clinics. Simultaneous measurement of 10 steroid metabolites in maternal (plasma and hair) and fetal (cord blood and placenta) samples was performed using tandem mass spectrometry along with assessment of the perceived stress score and sociodemographic status. While the maternal perceived stress score (PSS) and sociodemographic status were positively associated with each other and each with the body mass index (BMI) (r = 0.73, p = 0.0008; r = 0.48, p = 0.05; r = 0.59, p = 0.014, respectively), PSS did not correlate with maternal or fetal cortisol, cortisone levels, or fetal birth weight. Regardless of maternal PSS or BMI, fetal steroid levels remained stable and unaffected. Progesterone was the only steroid analyte quantifiable in maternal hair and correlated positively with PSS (r = 0.964, p = 0.003), whereas cord estradiol was negatively associated with PSS (r = -0.94, p = 0.017). In conclusion, hair progesterone might serve as a better marker of maternal stress than cortisol or cortisone and maternal PSS negatively impacts fetal estradiol levels. Findings have implications for improved biomarkers of stress and targets for future research to identify factors that buffer the fetus from adverse effects of maternal stress.


Asunto(s)
Feto/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Placenta/metabolismo , Estrés Psicológico/fisiopatología , Adulto , Cortisona/sangre , Estradiol/sangre , Femenino , Humanos , Hidrocortisona/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Masculino , Embarazo
20.
JCI Insight ; 6(6)2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33591956

RESUMEN

Functional dyspepsia (FD) is associated with chronic gastrointestinal distress and with anxiety and depression. Here, we hypothesized that aberrant gastric signals, transmitted by the vagus nerve, may alter key brain regions modulating affective and pain behavior. Using a previously validated rat model of FD characterized by gastric hypersensitivity, depression-like behavior, and anxiety-like behavior, we found that vagal activity - in response to gastric distention - was increased in FD rats. The FD phenotype was associated with gastric mast cell hyperplasia and increased expression of corticotrophin-releasing factor (Crh) and decreased brain-derived neurotrophic factor genes in the central amygdala. Subdiaphragmatic vagotomy reversed these changes and restored affective behavior to that of controls. Vagotomy partially attenuated pain responses to gastric distention, which may be mediated by central reflexes in the periaqueductal gray, as determined by local injection of lidocaine. Ketotifen, a mast cell stabilizer, reduced vagal hypersensitivity, normalized affective behavior, and attenuated gastric hyperalgesia. In conclusion, vagal activity, partially driven by gastric mast cells, induces long-lasting changes in Crh signaling in the amygdala that may be responsible for enhanced pain and enhanced anxiety- and depression-like behaviors. Together, these results support a "bottom-up" pathway involving the gut-brain axis in the pathogenesis of both gastric pain and psychiatric comorbidity in FD.


Asunto(s)
Afecto , Amígdala del Cerebelo/fisiopatología , Eje Cerebro-Intestino , Dispepsia/fisiopatología , Dolor/fisiopatología , Transducción de Señal , Nervio Vago/metabolismo , Amígdala del Cerebelo/metabolismo , Animales , Dispepsia/metabolismo , Femenino , Dolor/metabolismo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA