Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Immunol ; 210(6): 721-731, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36695771

RESUMEN

Besides antiviral functions, type I IFN expresses potent anti-inflammatory properties and is being widely used to treat certain autoimmune conditions, such as multiple sclerosis. In a murine model of multiple sclerosis, experimental autoimmune encephalomyelitis, administration of IFN-ß effectively attenuates the disease development. However, the precise mechanisms underlying IFN-ß-mediated treatment remain elusive. In this study, we report that IFN-induced protein with tetratricopeptide repeats 2 (Ifit2), a type I and type III IFN-stimulated gene, plays a previously unrecognized immune-regulatory role during autoimmune neuroinflammation. Mice deficient in Ifit2 displayed greater susceptibility to experimental autoimmune encephalomyelitis and escalated immune cell infiltration in the CNS. Ifit2 deficiency was also associated with microglial activation and increased myeloid cell infiltration. We also observed that myelin debris clearance and the subsequent remyelination were substantially impaired in Ifit2-/- CNS tissues. Clearing myelin debris is an important function of the reparative-type myeloid cell subset to promote remyelination. Indeed, we observed that bone marrow-derived macrophages, CNS-infiltrating myeloid cells, and microglia from Ifit2-/- mice express cytokine and metabolic genes associated with proinflammatory-type myeloid cell subsets. Taken together, our findings uncover a novel regulatory function of Ifit2 in autoimmune inflammation in part by modulating myeloid cell function and metabolic activity.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Animales , Ratones , Inflamación , Ratones Endogámicos C57BL , Microglía , Células Mieloides , Repeticiones de Tetratricopéptidos , Interferones/farmacología
2.
J Biol Chem ; 299(11): 105299, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37777156

RESUMEN

Microbes living in the intestine can regulate key signaling processes in the central nervous system that directly impact brain health. This gut-brain signaling axis is partially mediated by microbe-host-dependent immune regulation, gut-innervating neuronal communication, and endocrine-like small molecule metabolites that originate from bacteria to ultimately cross the blood-brain barrier. Given the mounting evidence of gut-brain crosstalk, a new therapeutic approach of "psychobiotics" has emerged, whereby strategies designed to primarily modify the gut microbiome have been shown to improve mental health or slow neurodegenerative diseases. Diet is one of the most powerful determinants of gut microbiome community structure, and dietary habits are associated with brain health and disease. Recently, the metaorganismal (i.e., diet-microbe-host) trimethylamine N-oxide (TMAO) pathway has been linked to the development of several brain diseases including Alzheimer's, Parkinson's, and ischemic stroke. However, it is poorly understood how metaorganismal TMAO production influences brain function under normal physiological conditions. To address this, here we have reduced TMAO levels by inhibiting gut microbe-driven choline conversion to trimethylamine (TMA), and then performed comprehensive behavioral phenotyping in mice. Unexpectedly, we find that TMAO is particularly enriched in the murine olfactory bulb, and when TMAO production is blunted at the level of bacterial choline TMA lyase (CutC/D), olfactory perception is altered. Taken together, our studies demonstrate a previously underappreciated role for the TMAO pathway in olfactory-related behaviors.


Asunto(s)
Percepción Olfatoria , Animales , Ratones , Bacterias/metabolismo , Colina/metabolismo , Metilaminas/metabolismo , Femenino , Ratones Endogámicos C57BL
3.
J Virol ; 97(8): e0074923, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37504572

RESUMEN

Interferon-induced protein with tetratricopeptide repeats 2, Ifit2, is critical in restricting neurotropic murine-ß-coronavirus, RSA59 infection. RSA59 intracranial injection of Ifit2-deficient (-/-) compared to wild-type (WT) mice results in impaired acute microglial activation, reduced CX3CR1 expression, limited migration of peripheral lymphocytes into the brain, and impaired virus control followed by severe morbidity and mortality. While the protective role of Ifit2 is established for acute viral encephalitis, less is known about its influence during the chronic demyelinating phase of RSA59 infection. To understand this, RSA59 infected Ifit2-/- and Ifit2+/+ (WT) were observed for neuropathological outcomes at day 5 (acute phase) and 30 post-infection (chronic phase). Our study demonstrates that Ifit2 deficiency causes extensive RSA59 spread throughout the spinal cord gray and white matter, associated with impaired CD4+ T and CD8+ T cell infiltration. Further, the cervical lymph nodes of RSA59 infected Ifit2-/- mice showed reduced activation of CD4+ T cells and impaired IFNγ expression during acute encephalomyelitis. Interestingly, BBB integrity was better preserved in Ifit2-/- mice, as evidenced by tight junction protein Claudin-5 and adapter protein ZO-1 expression surrounding the meninges and blood vessels and decreased Texas red dye uptake, which may be responsible for reduced leukocyte infiltration. In contrast to sparse myelin loss in WT mice, the chronic disease phase in Ifit2-/- mice was associated with severe demyelination and persistent viral load, even at low inoculation doses. Overall, our study highlights that Ifit2 provides antiviral functions by promoting acute neuroinflammation and thereby aiding virus control and limiting severe chronic demyelination. IMPORTANCE Interferons execute their function by inducing specific genes collectively termed as interferon-stimulated genes (ISGs), among which interferon-induced protein with tetratricopeptide repeats 2, Ifit2, is known for restricting neurotropic viral replication and spread. However, little is known about its role in viral spread to the spinal cord and its associated myelin pathology. Toward this, our study using a neurotropic murine ß-coronavirus and Ifit2-deficient mice demonstrates that Ifit2 deficiency causes extensive viral spread throughout the gray and white matter of the spinal cord accompanied by impaired microglial activation and T cell infiltration. Furthermore, infected Ifit2-deficient mice showed impaired activation of T cells in the cervical lymph node and relatively intact blood-brain barrier integrity. Overall, Ifit2 plays a crucial role in mounting host immunity against neurotropic murine coronavirus in the acute phase while preventing mice from developing viral-induced severe chronic neuroinflammatory demyelination, the characteristic feature of human neurological disease multiple sclerosis (MS).


Asunto(s)
Infecciones por Coronavirus , Esclerosis Múltiple , Virus de la Hepatitis Murina , Sustancia Blanca , Ratones , Humanos , Animales , Sustancia Blanca/patología , Virus de la Hepatitis Murina/fisiología , Vaina de Mielina , Interferones , Proteínas/genética , Médula Espinal/patología , Esclerosis Múltiple/patología , Ratones Endogámicos C57BL , Proteínas de Unión al ARN/genética , Proteínas Reguladoras de la Apoptosis/genética
4.
J Biol Chem ; 297(4): 101184, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34509474

RESUMEN

The deubiquitinating enzyme USP37 is known to contribute to timely onset of S phase and progression of mitosis. However, it is not clear if USP37 is required beyond S-phase entry despite expression and activity of USP37 peaking within S phase. We have utilized flow cytometry and microscopy to analyze populations of replicating cells labeled with thymidine analogs and monitored mitotic entry in synchronized cells to determine that USP37-depleted cells exhibited altered S-phase kinetics. Further analysis revealed that cells depleted of USP37 harbored increased levels of the replication stress and DNA damage markers γH2AX and 53BP1 in response to perturbed replication. Depletion of USP37 also reduced cellular proliferation and led to increased sensitivity to agents that induce replication stress. Underlying the increased sensitivity, we found that the checkpoint kinase 1 is destabilized in the absence of USP37, attenuating its function. We further demonstrated that USP37 deubiquitinates checkpoint kinase 1, promoting its stability. Together, our results establish that USP37 is required beyond S-phase entry to promote the efficiency and fidelity of replication. These data further define the role of USP37 in the regulation of cell proliferation and contribute to an evolving understanding of USP37 as a multifaceted regulator of genome stability.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Endopeptidasas/metabolismo , Fase S , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Daño del ADN , Replicación del ADN , Endopeptidasas/genética , Estabilidad de Enzimas , Inestabilidad Genómica , Células HCT116 , Células HeLa , Histonas , Humanos , Células MCF-7 , Ubiquitinación
5.
PLoS Pathog ; 16(11): e1009034, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33253295

RESUMEN

The interferon-induced tetratricopeptide repeat protein (Ifit2) protects mice from lethal neurotropic viruses. Neurotropic coronavirus MHV-RSA59 infection of Ifit2-/- mice caused pronounced morbidity and mortality accompanied by rampant virus replication and spread throughout the brain. In spite of the higher virus load, induction of many cytokines and chemokines in the brains of infected Ifit2-/- mice were similar to that in wild-type mice. In contrast, infected Ifit2-/- mice revealed significantly impaired microglial activation as well as reduced recruitment of NK1.1 T cells and CD4 T cells to the brain, possibly contributing to the lack of viral clearance. These two deficiencies were associated with a lower level of microglial expression of CX3CR1, the receptor of the CX3CL1 (Fractalkine) chemokine, which plays a critical role in both microglial activation and leukocyte recruitment. The above results uncovered a new potential role of an interferon-induced protein in immune protection.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Movimiento Celular/inmunología , Infecciones por Coronavirus/virología , Leucocitos/virología , Virus de la Hepatitis Murina/patogenicidad , Proteínas de Unión al ARN/metabolismo , Replicación Viral/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/deficiencia , Infecciones por Coronavirus/inmunología , Citocinas/metabolismo , Interferones/metabolismo , Leucocitos/citología , Leucocitos/metabolismo , Ratones Endogámicos C57BL , Microglía/metabolismo , Virus de la Hepatitis Murina/metabolismo
6.
Arterioscler Thromb Vasc Biol ; 38(1): 218-231, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29074585

RESUMEN

OBJECTIVE: Human genetic variants near the FADS (fatty acid desaturase) gene cluster (FADS1-2-3) are strongly associated with cardiometabolic traits including dyslipidemia, fatty liver, type 2 diabetes mellitus, and coronary artery disease. However, mechanisms underlying these genetic associations are unclear. APPROACH AND RESULTS: Here, we specifically investigated the physiological role of the Δ-5 desaturase FADS1 in regulating diet-induced cardiometabolic phenotypes by treating hyperlipidemic LDLR (low-density lipoprotein receptor)-null mice with antisense oligonucleotides targeting the selective knockdown of Fads1. Fads1 knockdown resulted in striking reorganization of both ω-6 and ω-3 polyunsaturated fatty acid levels and their associated proinflammatory and proresolving lipid mediators in a highly diet-specific manner. Loss of Fads1 activity promoted hepatic inflammation and atherosclerosis, yet was associated with suppression of hepatic lipogenesis. Fads1 knockdown in isolated macrophages promoted classic M1 activation, whereas suppressing alternative M2 activation programs, and also altered systemic and tissue inflammatory responses in vivo. Finally, the ability of Fads1 to reciprocally regulate lipogenesis and inflammation may rely in part on its role as an effector of liver X receptor signaling. CONCLUSIONS: These results position Fads1 as an underappreciated regulator of inflammation initiation and resolution, and suggest that endogenously synthesized arachidonic acid and eicosapentaenoic acid are key determinates of inflammatory disease progression and liver X receptor signaling.


Asunto(s)
Aorta/enzimología , Enfermedades de la Aorta/enzimología , Aterosclerosis/enzimología , Dislipidemias/enzimología , Ácido Graso Desaturasas/metabolismo , Mediadores de Inflamación/metabolismo , Inflamación/enzimología , Lipogénesis , Animales , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Ácido Araquidónico/metabolismo , Aterosclerosis/genética , Aterosclerosis/patología , Células Cultivadas , delta-5 Desaturasa de Ácido Graso , Modelos Animales de Enfermedad , Dislipidemias/genética , Dislipidemias/patología , Ácido Eicosapentaenoico/metabolismo , Ácido Graso Desaturasas/genética , Inflamación/genética , Inflamación/patología , Hígado/metabolismo , Receptores X del Hígado/metabolismo , Activación de Macrófagos , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Placa Aterosclerótica , Receptores de LDL/deficiencia , Receptores de LDL/genética
7.
J Biol Chem ; 289(16): 11367-11373, 2014 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-24596092

RESUMEN

The spindle assembly checkpoint (SAC) ensures the faithful segregation of the genome during mitosis by ensuring that sister chromosomes form bipolar attachments with microtubules of the mitotic spindle. p31(Comet) is an antagonist of the SAC effector Mad2 and promotes silencing of the SAC and mitotic progression. However, p31(Comet) interacts with Mad2 throughout the cell cycle. We show that p31(Comet) binds Mad2 solely in an inhibitory manner. We demonstrate that attenuating the affinity of p31(Comet) for Mad2 by phosphorylation promotes SAC activity in mitosis. Specifically, phosphorylation of Ser-102 weakens p31(Comet)-Mad2 binding and enhances p31(Comet)-mediated bypass of the SAC. Our results provide the first evidence for regulation of p31(Comet) and demonstrate a previously unknown event controlling SAC activity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Puntos de Control de la Fase M del Ciclo Celular/fisiología , Proteínas Mad2/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas de Ciclo Celular/genética , Células HeLa , Humanos , Proteínas Mad2/genética , Proteínas Nucleares/genética , Fosforilación/fisiología , Unión Proteica/fisiología
9.
Front Physiol ; 15: 1371096, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38694206

RESUMEN

Introduction: The Aster-C protein (encoded by the Gramd1c gene) is an endoplasmic reticulum (ER) resident protein that has been reported to transport cholesterol from the plasma membrane to the ER. Although there is a clear role for the closely-related Aster-B protein in cholesterol transport and downstream esterification in the adrenal gland, the specific role for Aster-C in cholesterol homeostasis is not well understood. Here, we have examined whole body cholesterol balance in mice globally lacking Aster-C under low or high dietary cholesterol conditions. Method: Age-matched Gramd1c +/+ and Gramd1c -/- mice were fed either low (0.02%, wt/wt) or high (0.2%, wt/wt) dietarycholesterol and levels of sterol-derived metabolites were assessed in the feces, liver, and plasma. Results: Compared to wild type controls (Gramd1c +/+) mice, mice lackingGramd1c (Gramd1c -/-) have no significant alterations in fecal, liver, or plasma cholesterol. Given the potential role for Aster C in modulating cholesterol metabolism in diverse tissues, we quantified levels of cholesterol metabolites such as bile acids, oxysterols, and steroid hormones. Compared to Gramd1c +/+ controls, Gramd1c -/- mice had modestly reduced levels of select bile acid species and elevated cortisol levels, only under low dietary cholesterol conditions. However, the vast majority of bile acids, oxysterols, and steroid hormones were unaltered in Gramd1c -/- mice. Bulk RNA sequencing in the liver showed that Gramd1c -/- mice did not exhibit alterations in sterol-sensitive genes, but instead showed altered expression of genes in major urinary protein and cytochrome P450 (CYP) families only under low dietary cholesterol conditions. Discussion: Collectively, these data indicate nominal effects of Aster-C on whole body cholesterol transport and metabolism under divergent dietary cholesterol conditions. These results strongly suggest that Aster-C alone is not sufficient to control whole body cholesterol balance, but can modestly impact circulating cortisol and bile acid levels when dietary cholesterol is limited.

10.
J Biol Chem ; 287(46): 39021-9, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-23027877

RESUMEN

Ubiquitin-mediated proteolysis is a key regulatory process in cell cycle progression. The Skp1-Cul1-F-box (SCF) and anaphase-promoting complex (APC) ubiquitin ligases target numerous components of the cell cycle machinery for destruction. Throughout the cell cycle, these ligases cooperate to maintain precise levels of key regulatory proteins, and indirectly, each other. Recently, we have identified the deubiquitinase USP37 as a regulator of the cell cycle. USP37 expression is cell cycle-regulated, being expressed in late G(1) and ubiquitinated by APC(Cdh1) in early G(1). Here we report that in addition to destruction at G(1), a major fraction of USP37 is degraded at the G(2)/M transition, prior to APC substrates and similar to SCF(ßTrCP) substrates. Consistent with this hypothesis, USP37 interacts with components of the SCF in a ßTrCP-dependent manner. Interaction with ßTrCP and subsequent degradation is phosphorylation-dependent and is mediated by the Polo-like kinase (Plk1). USP37 is stabilized in G(2) by depletion of ßTrCP as well as chemical or genetic manipulation of Plk1. Similarly, mutation of the phospho-sites abolishes ßTrCP binding and renders USP37 resistant to Plk1 activity. Expression of this mutant hinders the G(2)/M transition. Our data demonstrate that tight regulation of USP37 levels is required for proper cell cycle progression.


Asunto(s)
Proteínas Cullin/metabolismo , Endopeptidasas/química , Regulación Enzimológica de la Expresión Génica , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Fase G2 , Células HEK293 , Células HeLa , Humanos , Mitosis , Mutación , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Ubiquitina/química , Quinasa Tipo Polo 1
11.
Elife ; 112022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-35072627

RESUMEN

Obesity has repeatedly been linked to reorganization of the gut microbiome, yet to this point obesity therapeutics have been targeted exclusively toward the human host. Here, we show that gut microbe-targeted inhibition of the trimethylamine N-oxide (TMAO) pathway protects mice against the metabolic disturbances associated with diet-induced obesity (DIO) or leptin deficiency (Lepob/ob). Small molecule inhibition of the gut microbial enzyme choline TMA-lyase (CutC) does not reduce food intake but is instead associated with alterations in the gut microbiome, improvement in glucose tolerance, and enhanced energy expenditure. We also show that gut microbial CutC inhibition is associated with reorganization of host circadian control of both phosphatidylcholine and energy metabolism. This study underscores the relationship between microbe and host metabolism and provides evidence that gut microbe-derived trimethylamine (TMA) is a key regulator of the host circadian clock. This work also demonstrates that gut microbe-targeted enzyme inhibitors have potential as anti-obesity therapeutics.


Asunto(s)
Colina/análogos & derivados , Ritmo Circadiano/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Obesidad/metabolismo , Animales , Colina/administración & dosificación , Colina/metabolismo , Dieta Alta en Grasa , Inhibidores Enzimáticos/farmacología , Leptina/deficiencia , Liasas/efectos de los fármacos , Masculino , Metilaminas/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/genética , Obesidad/microbiología
12.
Brain Disord ; 4: 100021, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34514445

RESUMEN

Coronaviruses have emerged as alarming pathogens owing to their inherent ability of genetic variation and cross-species transmission. Coronavirus infection burdens the endoplasmic reticulum (ER.), causes reactive oxygen species production and induces host stress responses, including unfolded protein response (UPR) and antioxidant system. In this study, we have employed a neurotropic murine ß-coronavirus (M-CoV) infection in the Central Nervous System (CNS) of experimental mice model to study the role of host stress responses mediated by interplay of DJ-1 and XBP1. DJ-1 is an antioxidant molecule with established functions in neurodegeneration. However, its regulation in virus-induced cellular stress response is less explored. Our study showed that M-CoV infection activated the glial cells and induced antioxidant and UPR genes during the acute stage when the viral titer peaks. As the virus particles decreased and acute neuroinflammation diminished at day ten p.i., a significant up-regulation in UPR responsive XBP1, antioxidant DJ-1, and downstream signaling molecules, including Nrf2, was recorded in the brain tissues. Additionally, preliminary in silico analysis of the binding between the DJ-1 promoter and a positively charged groove of XBP1 is also investigated, thus hinting at a mechanism behind the upregulation of DJ-1 during MHV-infection. The current study thus attempts to elucidate a novel interplay between the antioxidant system and UPR in the outcome of coronavirus infection.

13.
Ann Otol Rhinol Laryngol ; 119(4): 270-8, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20433028

RESUMEN

OBJECTIVES: We sought to construct and partially characterize complementary DNA (cDNA) libraries prepared from the middle ear mucosa (MEM) of chinchillas to better understand pathogenic aspects of infection and inflammation, particularly with respect to leukotriene biogenesis and response. METHODS: Chinchilla MEM was harvested from controls and after middle ear inoculation with nontypeable Haemophilus influenzae. RNA was extracted to generate cDNA libraries. Randomly selected clones were subjected to sequence analysis to characterize the libraries and to provide DNA sequence for phylogenetic analyses. Reverse transcription-polymerase chain reaction of the RNA pools was used to generate cDNA sequences corresponding to genes associated with leukotriene biosynthesis and metabolism. RESULTS: Sequence analysis of 921 randomly selected clones from the uninfected MEM cDNA library produced approximately 250,000 nucleotides of almost entirely novel sequence data. Searches of the GenBank database with the Basic Local Alignment Search Tool provided for identification of 515 unique genes expressed in the MEM and not previously described in chinchillas. In almost all cases, the chinchilla cDNA sequences displayed much greater homology to human or other primate genes than with rodent species. Genes associated with leukotriene metabolism were present in both normal and infected MEM. CONCLUSIONS: Based on both phylogenetic comparisons and gene expression similarities with humans, chinchilla MEM appears to be an excellent model for the study of middle ear inflammation and infection. The higher degree of sequence similarity between chinchillas and humans compared to chinchillas and rodents was unexpected. The cDNA libraries from normal and infected chinchilla MEM will serve as useful molecular tools in the study of otitis media and should yield important information with respect to middle ear pathogenesis.


Asunto(s)
ADN Complementario/análisis , Oído Medio/química , Infecciones por Haemophilus/genética , Haemophilus influenzae , Membrana Mucosa/química , Animales , Chinchilla , Biblioteca de Genes , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
iScience ; 23(6): 101237, 2020 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-32629605

RESUMEN

Metastasis is the leading cause of death for patients with cancer. Consequently it is imperative that we improve our understanding of the molecular mechanisms that underlie progression of tumor growth toward malignancy. Advances in genome characterization technologies have been very successful in identifying commonly mutated or misregulated genes in a variety of human cancers. However, the difficulty in evaluating whether these candidates drive tumor progression remains a major challenge. Using the genetic amenability of Drosophila melanogaster we generated tumors with specific genotypes in the living animal and carried out a detailed systematic loss-of-function analysis to identify conserved genes that enhance or suppress epithelial tumor progression. This enabled the discovery of functional cooperative regulators of invasion and the establishment of a network of conserved invasion suppressors. This includes constituents of the cohesin complex, whose loss of function either promotes individual or collective cell invasion, depending on the severity of effect on cohesin complex function.

15.
Cytokine ; 41(1): 38-43, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18063379

RESUMEN

OBJECTIVES: Middle ear mucins are associated with otitis media (OM), contribute to hearing loss and are regulated by cytokines. This work investigates the regulation of mucin secretion from human middle ear epithelial cells (HMEEC) by inflammatory cytokines interleukin-1beta (IL-1beta), tumor necrosis factor-alpha (TNF-alpha) and cytokine inhibitors interleukin-1 receptor antagonist (IL-1ra) and anti-tumor necrosis factor-alpha antibody (TNFab). METHODS: HMEEC were exposed to IL-1beta and TNF-alpha in a dose- and time-dependent manner. Cytokine stimulated HMEEC were also exposed to IL-1ra and TNFab in a dose-dependent manner. Mucin secretion was characterized by exclusion chromatography and liquid scintillation. RESULTS: HMEEC exposed to IL-1beta and TNF-alpha demonstrated significant upregulation of mucin secretion in a dose-dependent fashion. Cultures exposed to IL-1beta at 100ng/ml and TNF-alpha at 200ng/ml showed increased mucin secretion in time-dependent experiments at 16h (P=0.00008) for TNF-alpha and 8 (P=0.028) and 16h (P=0.00001) for IL-1beta. IL-1ra and TNFab inhibited the effects of increased mucin secretion by IL-1beta and TNF-alpha. CONCLUSIONS: IL-1beta and TNF-alpha upregulate mucin secretion from HMEEC in a dose- and time-dependant manner and these effects can be inhibited by cytokine blockade. Improved understanding of these mechanisms has the potential to alter the approach and management of OM and lead to novel therapeutic interventions.


Asunto(s)
Oído Medio/metabolismo , Células Epiteliales/metabolismo , Interleucina-1beta/farmacología , Mucinas/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Anticuerpos/farmacología , Línea Celular Transformada , Relación Dosis-Respuesta a Droga , Oído Medio/patología , Células Epiteliales/patología , Pérdida Auditiva/metabolismo , Pérdida Auditiva/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/metabolismo , Factores de Tiempo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
16.
Arch Otolaryngol Head Neck Surg ; 134(1): 39-44, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18209134

RESUMEN

OBJECTIVE: To compare levels of expression of mucin gene 2 (MUC2), a major secretory mucin, in the middle ear of patients with otitis media (OM) and control patients. DESIGN: Case-control study. SETTING: Children's Hospital of Wisconsin, Milwaukee. PATIENTS: Nineteen patients aged 6 months to 15 years undergoing routine ventilation tube insertion for recurrent OM or chronic OM with effusion and 8 controls with no history of OM undergoing cochlear implantation. INTERVENTIONS: Biopsy of middle ear epithelium for RNA extraction. MAIN OUTCOME MEASURE: Expression of MUC2 by real-time reverse transcription-polymerase chain reaction. RESULTS: Twenty-seven OM samples (17 recurrent and 10 with effusion) from 19 patients were analyzed and compared with 9 control samples from 8 patients. The mean MUC2 expression was 6.12 (95% confidence interval, 3.32-8.89) times that of the controls in the OM samples overall, 5.00 (95% confidence interval, 2.79-7.21) times that of controls in the recurrent OM samples, and 7.98 (95% confidence interval, 1.58-14.38) times that of controls in the OM with effusion samples. CONCLUSIONS: Levels of MUC2 expression in human middle ear epithelium are significantly increased in patients with OM overall, patients with recurrent OM, and patients with OM with effusion compared with controls. Mucins are fundamentally important in the middle ear, controlling viscoelastic properties of secretions and providing mucosal protection and bacterial clearance. Demonstration of these differences between patient groups highlights the need for greater understanding of molecular responses in OM, which may provide novel interventions for this common problem.


Asunto(s)
Oído Medio/metabolismo , Mucinas/genética , Otitis Media/metabolismo , Estudios de Casos y Controles , Preescolar , Oído Medio/patología , Femenino , Humanos , Masculino , Mucina 2 , Mucinas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
17.
Cell Rep ; 19(12): 2451-2461, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28636934

RESUMEN

Emerging evidence suggests that microbes resident in the human intestine represent a key environmental factor contributing to obesity-associated disorders. Here, we demonstrate that the gut microbiota-initiated trimethylamine N-oxide (TMAO)-generating pathway is linked to obesity and energy metabolism. In multiple clinical cohorts, systemic levels of TMAO were observed to strongly associate with type 2 diabetes. In addition, circulating TMAO levels were associated with obesity traits in the different inbred strains represented in the Hybrid Mouse Diversity Panel. Further, antisense oligonucleotide-mediated knockdown or genetic deletion of the TMAO-producing enzyme flavin-containing monooxygenase 3 (FMO3) conferred protection against obesity in mice. Complimentary mouse and human studies indicate a negative regulatory role for FMO3 in the beiging of white adipose tissue. Collectively, our studies reveal a link between the TMAO-producing enzyme FMO3 and obesity and the beiging of white adipose tissue.


Asunto(s)
Metilaminas/sangre , Obesidad/enzimología , Oxigenasas/fisiología , Grasa Subcutánea/enzimología , Adipocitos Beige/enzimología , Animales , Diabetes Mellitus Tipo 2/sangre , Femenino , Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/sangre , Obesidad/patología , Grasa Subcutánea/patología , Grasa Subcutánea/fisiopatología
18.
Laryngoscope ; 116(2): 207-11, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16467705

RESUMEN

OBJECTIVES: The objectives of this study were to investigate the role of the phosphatidylcholine-specific phospholipase C (PC-PLC), protein kinase C (PKC), and nitric oxide synthase (NOS) pathways during upregulation of mucin secretion by middle ear epithelium after exposure to interleukin-1beta and to examine the ability of a specific interleukin-1 receptor antagonist (IL-1betara) to block this increased secretion. MATERIALS AND METHODS: Primary chinchilla middle ear epithelial cultures were established and exposed to IL-1beta. Specific inhibitors of calmodulin, PC-PLC, PKC, and NOS pathways were used to investigate the potential role of these pathways leading to increased epithelial mucin secretion after exposure to IL-1beta. Mucin secretion was characterized by exclusion chromatography and liquid scintillation. RESULTS: Epithelial cultures exposed to IL-1beta demonstrate an increase in mucin secretion that is blocked by specific inhibitors of PC-PLC, PKC, and NOS, but not by inhibitors of calmodulin. In addition, mucin secretion stimulated by IL-1beta was reversible with use of a specific IL-1betara. CONCLUSIONS: IL-1beta stimulates mucin secretion from middle ear epithelium and its effects can be reversed by IL-1betara. PC-PLC, PKC, and NOS pathways play a role in the increased secretion of mucin in middle ear epithelial cells after exposure to IL-1beta.


Asunto(s)
Oído Medio/metabolismo , Interleucina-1/farmacología , Mucinas/metabolismo , Animales , Calmodulina/antagonistas & inhibidores , Calmodulina/fisiología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Chinchilla , Oído Medio/efectos de los fármacos , Interleucina-1/antagonistas & inhibidores , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/fisiología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/fisiología , Receptores de Interleucina-1/antagonistas & inhibidores , Transducción de Señal , Fosfolipasas de Tipo C/antagonistas & inhibidores , Fosfolipasas de Tipo C/fisiología , Regulación hacia Arriba
19.
Int J Pediatr Otorhinolaryngol ; 70(8): 1449-56, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16672162

RESUMEN

OBJECTIVES: To identify mucin genes in chinchilla middle ear epithelium and characterize complimentary deoxyribonucleic acid (cDNA) sequences to facilitate further investigations into mucin physiology and pathophysiology on a molecular level using the chinchilla model. METHODS: Chinchilla mucin gene exploration and cDNA characterization was accomplished using reverse transcriptase-polymerase chain reactions (RT-PCR). Forward and reverse primer pairs were designed using consensus sequences available for human and rodent species. Chinchilla middle ear epithelium was harvested and primary cell cultures (CMEEC) were established. The CMEEC were explored for the expression of chinchilla mucin genes 1, 2, 4 and 5AC (cMuc1, cMuc2, cMuc4 and cMuc5AC). Identified cDNA amplicons for each of these genes was sequenced and homology compared to previously published human and rodent sequences. RESULTS: CMEEC express all four of the mucin genes cMuc1, cMuc2, cMuc4 and cMuc5AC. cDNA amplicons for each of the genes were able to be sequenced with lengths ranging from 66 to 362 base pairs. Each of the chinchilla cDNA sequences expressed significant homology with published human and rodent cDNA for these mucin genes. A cDNA sequence for the housekeeping gene, beta-actin, was also identified. CONCLUSIONS: Chinchilla middle ear epithelium grown in culture expresses the mucin genes 1, 2, 4 and 5AC, which have been identified as important in mucin regulation in the middle ear. cDNA sequences corresponding to these mucin genes were identified and may serve as important molecular tools in future studies of otitis media using the chinchilla model.


Asunto(s)
Chinchilla/genética , ADN Complementario/análisis , Oído Medio/metabolismo , Epitelio/metabolismo , Mucinas/genética , Análisis de Secuencia de ADN , Actinas/genética , Animales , Células Cultivadas , Humanos , Modelos Animales , Datos de Secuencia Molecular , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
JAMA ; 296(2): 202-11, 2006 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-16835426

RESUMEN

CONTEXT: Chronic otitis media (OM) is a common pediatric infectious disease. Previous studies demonstrating that metabolically active bacteria exist in culture-negative pediatric middle-ear effusions and that experimental infection with Haemophilus influenzae in the chinchilla model of otitis media results in the formation of adherent mucosal biofilms suggest that chronic OM may result from a mucosal biofilm infection. OBJECTIVE: To test the hypothesis that chronic OM in humans is biofilm-related. DESIGN, SETTING, AND PATIENTS: Middle-ear mucosa (MEM) biopsy specimens were obtained from 26 children (mean age, 2.5 [range, 0.5-14] years) undergoing tympanostomy tube placement for treatment of otitis media with effusion (OME) and recurrent OM and were analyzed using microbiological culture, polymerase chain reaction (PCR)-based diagnostics, direct microscopic examination, fluorescence in situ hybridization, and immunostaining. Uninfected (control) MEM specimens were obtained from 3 children and 5 adults undergoing cochlear implantation. Patients were enrolled between February 2004 and April 2005 from a single US tertiary referral otolaryngology practice. MAIN OUTCOME MEASURES: Confocal laser scanning microscopic (CLSM) images were obtained from MEM biopsy specimens and were evaluated for biofilm morphology using generic stains and species-specific probes for H influenzae, Streptococcus pneumoniae, and Moraxella catarrhalis. Effusions, when present, were evaluated by PCR and culture for evidence of pathogen-specific nucleic acid sequences and bacterial growth, respectively. RESULTS: Of the 26 children undergoing tympanostomy tube placement, 13 (50%) had OME, 20 (77%) had recurrent OM, and 7 (27%) had both diagnoses; 27 of 52 (52%) of the ears had effusions, 24 of 24 effusions were PCR-positive for at least 1 OM pathogen, and 6 (22%) of 27 effusions were culture-positive for any pathogen. Mucosal biofilms were visualized by CLSM on 46 (92%) of 50 MEM specimens from children with OME and recurrent OM using generic and pathogen-specific probes. Biofilms were not observed on 8 control MEM specimens obtained from the patients undergoing cochlear implantation. CONCLUSION: Direct detection of biofilms on MEM biopsy specimens from children with OME and recurrent OM supports the hypothesis that these chronic middle-ear disorders are biofilm-related.


Asunto(s)
Biopelículas , Oído Medio/microbiología , Otitis Media/microbiología , Adolescente , Biopelículas/crecimiento & desarrollo , Niño , Preescolar , Enfermedad Crónica , Oído Medio/patología , Femenino , Humanos , Hibridación Fluorescente in Situ , Lactante , Masculino , Microscopía Confocal , Membrana Mucosa/microbiología , Membrana Mucosa/patología , Otitis Media/patología , Otitis Media con Derrame/microbiología , Otitis Media con Derrame/patología , ARN Bacteriano , ARN Ribosómico 16S
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA