Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Biochem J ; 474(5): 647-665, 2017 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-28008134

RESUMEN

M3 muscarinic receptor (M3R) expression is increased in colon cancer; M3R activation stimulates colon cancer cell invasion via cross-talk with epidermal growth factor receptors (EGFR), post-EGFR activation of mitogen-activated protein kinase (MAPK) extracellular signal-related kinase 1/2 (ERK1/2), and induction of matrix metalloproteinase-1 (MMP1) expression. MMP1 expression is strongly associated with tumor metastasis and adverse outcomes. Here, we asked whether other MAPKs regulate M3R agonist-induced MMP1 expression. In addition to activating ERK1/2, we found that treating colon cancer cells with acetylcholine (ACh) stimulated robust time- and dose-dependent phosphorylation of p38 MAPK. Unlike ERK1/2 activation, ACh-induced p38 phosphorylation was EGFR-independent and blocked by inhibiting protein kinase C-α (PKC-α). Inhibiting activation of PKC-α, EGFR, ERK1/2, or p38-α/ß alone attenuated, but did not abolish ACh-induced MMP1 expression, a finding that predicted potentiating interactions between these pathways. Indeed, ACh-induced MMP1 expression was abolished by incubating cells with either an EGFR or MEK/ERK1/2 inhibitor combined with a p38-α/ß inhibitor. Activating PKC-α and EGFR directly with the combination of phorbol 12-myristate 13-acetate (PMA) and EGF potentiated MMP1 gene and protein expression, and cell invasion. PMA- and ACh-induced MMP1 expression were strongly diminished by inhibiting Src and abolished by concurrently inhibiting both p38-α/ß and Src, indicating that Src mediates the cross-talk between PKC-α and EGFR signaling. Using siRNA knockdown, we identified p38-α as the relevant p38 isoform. Collectively, these studies uncover novel functional interactions between post-muscarinic receptor signaling pathways that augment MMP1 expression and drive colon cancer cell invasion; targeting these potentiating interactions has therapeutic potential.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Metaloproteinasa 1 de la Matriz/metabolismo , Receptor Muscarínico M3/genética , Transducción de Señal/genética , Acetilcolina/farmacología , Células CACO-2 , Línea Celular Tumoral , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Células HT29 , Humanos , Metaloproteinasa 1 de la Matriz/genética , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Proteína Quinasa C-alfa/genética , Proteína Quinasa C-alfa/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptor Muscarínico M3/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
2.
J Clin Microbiol ; 53(5): 1672-6, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25762766

RESUMEN

Candida species are common causes of bloodstream infections (BSI), with high mortality. Four species cause >90% of Candida BSI: C. albicans, C. glabrata, C. parapsilosis, and C. tropicalis. Differentiation of Candida spp. is important because of differences in virulence and antimicrobial susceptibility. Candida QuickFISH BC, a multicolor, qualitative nucleic acid hybridization assay for the identification of C. albicans (green fluorescence), C. glabrata (red fluorescence), and C. parapsilosis (yellow fluorescence), was tested on Bactec and BacT/Alert blood culture bottles which signaled positive on automated blood culture devices and were positive for yeast by Gram stain at seven study sites. The results were compared to conventional identification. A total of 419 yeast-positive blood culture bottles were studied, consisting of 258 clinical samples (89 C. glabrata, 79 C. albicans, 23 C. parapsilosis, 18 C. tropicalis, and 49 other species) and 161 contrived samples inoculated with clinical isolates (40 C. glabrata, 46 C. albicans, 36 C. parapsilosis, 19 C. tropicalis, and 20 other species). A total of 415 samples contained a single fungal species, with C. glabrata (n = 129; 30.8%) being the most common isolate, followed by C. albicans (n = 125; 29.8%), C. parapsilosis (n = 59; 14.1%), C. tropicalis (n = 37; 8.8%), and C. krusei (n = 17; 4.1%). The overall agreement (with range for the three major Candida species) between the two methods was 99.3% (98.3 to 100%), with a sensitivity of 99.7% (98.3 to 100%) and a specificity of 98.0% (99.4 to 100%). This study showed that Candida QuickFISH BC is a rapid and accurate method for identifying C. albicans, C. glabrata, and C. parapsilosis, the three most common Candida species causing BSI, directly from blood culture bottles.


Asunto(s)
Sangre/microbiología , Candida/clasificación , Candida/aislamiento & purificación , Candidemia/diagnóstico , Candidemia/microbiología , Hibridación Fluorescente in Situ/métodos , Técnicas Microbiológicas/métodos , Técnicas de Diagnóstico Molecular/métodos , Candida/genética , Humanos , Sensibilidad y Especificidad
3.
J Clin Microbiol ; 53(1): 319-22, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25378578

RESUMEN

(1-3)-ß-d-Glucan (BDG) from cerebrospinal fluid (CSF) is a promising marker for diagnostic and prognostic aid of central nervous system (CNS) fungal infection, but its relationship to serum values has not been studied. Herein, we detected BDG from CSF at levels 2-fold lower than those in serum in patients without evidence of fungal disease but 25-fold higher than those in in serum in noncryptococcal CNS fungal infections. CSF BDG may be a useful biomarker in the evaluation of fungal CNS disease.


Asunto(s)
Biomarcadores/líquido cefalorraquídeo , Infecciones Fúngicas del Sistema Nervioso Central/líquido cefalorraquídeo , Infecciones Fúngicas del Sistema Nervioso Central/diagnóstico , Infecciones Fúngicas del Sistema Nervioso Central/epidemiología , beta-Glucanos/líquido cefalorraquídeo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteoglicanos , Adulto Joven
4.
J Clin Microbiol ; 53(9): 3063-7, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26109445

RESUMEN

We report an aggressive fungal keratitis caused by a putatively novel species of Lophotrichus in a patient with traumatic injury to the cornea from a dog paw. The organism was isolated from the patient's necrotic cornea, which perforated despite coverage with hourly fortified broad-spectrum topical antibiotic therapy. This report represents the first case of human infection caused by this species.


Asunto(s)
Ascomicetos/aislamiento & purificación , Úlcera de la Córnea/diagnóstico , Úlcera de la Córnea/patología , Micosis/diagnóstico , Micosis/patología , Animales , Lesiones de la Cornea/complicaciones , Úlcera de la Córnea/microbiología , ADN de Hongos/química , ADN de Hongos/genética , ADN Espaciador Ribosómico/química , ADN Espaciador Ribosómico/genética , Perros , Femenino , Histocitoquímica , Humanos , Técnicas Microbiológicas , Microscopía , Persona de Mediana Edad , Datos de Secuencia Molecular , Micosis/microbiología , Análisis de Secuencia de ADN
5.
J Clin Microbiol ; 52(12): 4407-11, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25232159

RESUMEN

Angioinvasive fungal infections (AFIs) are an important cause of morbidity and mortality among immunocompromised patients. However, clinicomicrobiological characteristics and treatment of many AFI agents remain poorly defined. We report the first human case of infection with Westerdykella dispersa, an emergent cause of AFI, which was successfully treated in a neutropenic pediatric patient.


Asunto(s)
Ascomicetos/aislamiento & purificación , Micosis/diagnóstico , Micosis/patología , Neutropenia/complicaciones , Vasculitis/diagnóstico , Vasculitis/patología , Ascomicetos/clasificación , Ascomicetos/genética , Niño , ADN de Hongos/química , ADN de Hongos/genética , ADN Espaciador Ribosómico/química , ADN Espaciador Ribosómico/genética , Histocitoquímica , Humanos , Huésped Inmunocomprometido , Inyecciones/efectos adversos , Masculino , Técnicas Microbiológicas , Microscopía , Datos de Secuencia Molecular , Micosis/microbiología , Radiografía Torácica , Análisis de Secuencia de ADN , Tomografía Computarizada por Rayos X , Vasculitis/microbiología
6.
Hum Mol Genet ; 20(1): 165-75, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20940146

RESUMEN

Patients with genetic defects of the cyclic (c) adenosine-monophosphate (AMP)-signaling pathway and those with neonatal-onset multisystem inflammatory disease (NOMID) develop tumor-like lesions of the long bones. The molecular basis of this similarity is unknown. NOMID is caused by inappropriate caspase-1 activity, which in turn activates the inflammasome. The present study demonstrates that NOMID bone lesions are derived from the same osteoblast progenitor cells that form fibroblastoid tumors in mice and humans with defects that lead to increased cAMP-dependent protein kinase A (PKA) signaling. NOMID tumor cells showed high PKA activity, and an increase in their cAMP signaling led to PKA-specific activation of caspase-1. Increased PKA led to inflammation-independent activation of caspase-1 via over-expression of the proto-oncogene (and early osteoblast factor) Ets-1. In NOMID tumor cells, as in cells with defective PKA regulation, increased prostaglandin E2 (PGE2) led to increased cAMP levels and activation of Wnt signaling, like in other states of inappropriate PKA activity. Caspase-1 and PGE2 inhibition led to a decrease in cell proliferation of both NOMID and cells with abnormal PKA. These data reveal a previously unsuspected link between abnormal cAMP signaling and defective regulation of the inflammasome and suggest that caspase-1 and PGE2 inhibition may be therapeutic targets in bone lesions associated with defects of these two pathways.


Asunto(s)
Síndromes Periódicos Asociados a Criopirina/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Osteoblastos/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Células Madre/metabolismo , Animales , Huesos/patología , Caspasa 1/genética , Células Cultivadas , Síndromes Periódicos Asociados a Criopirina/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Dinoprostona/genética , Dinoprostona/metabolismo , Humanos , Inflamasomas/genética , Inflamasomas/metabolismo , Ratones , Proto-Oncogenes Mas , Proteína Proto-Oncogénica c-ets-1/genética , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Células del Estroma/metabolismo , Activación Transcripcional/genética , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
7.
Proc Natl Acad Sci U S A ; 107(19): 8683-8, 2010 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-20421483

RESUMEN

A population of stromal cells that retains osteogenic capacity in adult bone (adult bone stromal cells or aBSCs) exists and is under intense investigation. Mice heterozygous for a null allele of prkar1a (Prkar1a(+/-)), the primary receptor for cyclic adenosine monophosphate (cAMP) and regulator of protein kinase A (PKA) activity, developed bone lesions that were derived from cAMP-responsive osteogenic cells and resembled fibrous dysplasia (FD). Prkar1a(+/-) mice were crossed with mice that were heterozygous for catalytic subunit Calpha (Prkaca(+/-)), the main PKA activity-mediating molecule, to generate a mouse model with double heterozygosity for prkar1a and prkaca (Prkar1a(+/-)Prkaca(+/-)). Unexpectedly, Prkar1a(+/-)Prkaca(+/-) mice developed a greater number of osseous lesions starting at 3 months of age that varied from the rare chondromas in the long bones and the ubiquitous osteochondrodysplasia of vertebral bodies to the occasional sarcoma in older animals. Cells from these lesions originated from an area proximal to the growth plate, expressed osteogenic cell markers, and showed higher PKA activity that was mostly type II (PKA-II) mediated by an alternate pattern of catalytic subunit expression. Gene expression profiling confirmed a preosteoblastic nature for these cells but also showed a signature that was indicative of mesenchymal-to-epithelial transition and increased Wnt signaling. These studies show that a specific subpopulation of aBSCs can be stimulated in adult bone by alternate PKA and catalytic subunit activity; abnormal proliferation of these cells leads to skeletal lesions that have similarities to human FD and bone tumors.


Asunto(s)
Envejecimiento/patología , Huesos/enzimología , Huesos/patología , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/metabolismo , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Animales , Huesos/diagnóstico por imagen , Calcificación Fisiológica , Dominio Catalítico , Heterocigoto , Mesodermo/metabolismo , Ratones , Hidrolasas Diéster Fosfóricas/metabolismo , Espectrometría Raman , Células del Estroma/enzimología , Células del Estroma/patología , Tomografía Computarizada por Rayos X
8.
Am J Physiol Renal Physiol ; 302(6): F762-73, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22160774

RESUMEN

Although T cells have been shown to play a direct role in kidney ischemia-reperfusion injury (IRI), little is known about the underlying mechanisms. We hypothesized that studying the transcriptional responses in kidney-infiltrating T cells would help elucidate novel therapeutic targets for kidney IRI. Unilateral renal pedicle clamping for 45 min was performed in male C57BL/6 mice, and CD3(+) T cells were isolated from the kidney and purified. Transcriptional activities of T cell were measured by array-based PCR compared between ischemic kidneys and contralateral nonischemic kidneys. Among total of 89 genes analyzed, 24, 22, 24, and 37 genes were significantly changed at 6 h, day 3, day 10, and day 28 after IRI. Genes associated with cytokines, chemokines, and costimulatory molecules were upregulated. Pathway analysis identified CC motif chemokine receptor 5 (CCR5) as a candidate pathophysiological pathway. CCR5 upregulation was validated at the protein level, and CCR5 blockade improved renal function after kidney IRI. Using discovery techniques to identify transcriptional responses in purified kidney-infiltrating cells enabled the elucidation of novel mechanisms and therapeutic targets for IRI.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Riñón/lesiones , Riñón/patología , Receptores CCR5/metabolismo , Daño por Reperfusión/fisiopatología , Linfocitos T/metabolismo , Animales , Anticuerpos , Complejo CD3/genética , Complejo CD3/metabolismo , Quimiocinas/genética , Quimiocinas/metabolismo , Análisis por Conglomerados , Ensayo de Inmunoadsorción Enzimática , Perfilación de la Expresión Génica , Riñón/citología , Enfermedades Renales/metabolismo , Enfermedades Renales/fisiopatología , Masculino , Ratones , Receptores CCR5/genética , Daño por Reperfusión/metabolismo , Organismos Libres de Patógenos Específicos
9.
Hum Mol Genet ; 19(8): 1387-98, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20080939

RESUMEN

PRKAR1A inactivation leads to dysregulated cAMP signaling and Carney complex (CNC) in humans, a syndrome associated with skin, endocrine and other tumors. The CNC phenotype is not easily explained by the ubiquitous cAMP signaling defect; furthermore, Prkar1a(+/-) mice did not develop skin and other CNC tumors. To identify whether a Prkar1a defect is truly a generic but weak tumorigenic signal that depends on tissue-specific or other factors, we investigated Prkar1a(+/-) mice when bred within the Rb1(+/-) or Trp53(+/-) backgrounds, or treated with a two-step skin carcinogenesis protocol. Prkar1a(+/-) Trp53(+/-) mice developed more sarcomas than Trp53(+/-) mice (P < 0.05) and Prkar1a(+/-) Rb1(+/-) mice grew more (and larger) pituitary and thyroid tumors than Rb1(+/-) mice. All mice with double heterozygosity had significantly reduced life-spans compared with their single-heterozygous counterparts. Prkar1a(+/-) mice also developed more papillomas than wild-type animals. A whole-genome transcriptome profiling of tumors produced by all three models identified Wnt signaling as the main pathway activated by abnormal cAMP signaling, along with cell cycle abnormalities; all changes were confirmed by qRT-PCR array and immunohistochemistry. siRNA down-regulation of Ctnnb1, E2f1 or Cdk4 inhibited proliferation of human adrenal cells bearing a PRKAR1A-inactivating mutation and Prkar1a(+/-) mouse embryonic fibroblasts and arrested both cell lines at the G0/G1 phase of the cell cycle. In conclusion, Prkar1a haploinsufficiency is a relatively weak tumorigenic signal that can act synergistically with other tumor suppressor gene defects or chemicals to induce tumors, mostly through Wnt-signaling activation and cell cycle dysregulation, consistent with studies in human neoplasms carrying PRKAR1A defects.


Asunto(s)
Ciclo Celular , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Neoplasias/genética , Neoplasias/patología , Proteína de Retinoblastoma/genética , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteínas Wnt/metabolismo , Animales , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Haploidia , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias/metabolismo , Neoplasias/fisiopatología , Procesos Neoplásicos , Papiloma/inducido químicamente , Papiloma/genética , Papiloma/metabolismo , Papiloma/fisiopatología , Proteína de Retinoblastoma/metabolismo , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/fisiopatología , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Wnt/genética
10.
Clin Transplant ; 25(5): 766-75, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21114535

RESUMEN

INTRODUCTION: Molecular biomarkers validated previously in animal models are increasingly being studied in conjunction with traditional clinical endpoints in therapeutic trials. PATIENT AND METHODS: We hypothesized that human kidneys would exhibit a brisk, gene-specific inflammatory response during ischemia reperfusion injury (IRI), which would be modified by anti-adhesive therapy. Forty deceased-donor kidneys were biopsied prior to implantation and ∼1 h after reperfusion during an intervention trial with the selectin antagonist YSPSL (recombinant P-selectin glycoprotein ligand Ig). Ten inflammatory genes were measured by RT-PCR and normalized to three housekeeping genes. RESULTS: Pre-implantation kidney biopsies were already significantly inflamed relative to healthy tissue, with transcripts encoding IL-6, IL-8, and CD25 > 10-fold elevated. After reperfusion, IL-6 and IL-8 increased additional 60- and 120-fold (p < 0.05), while already elevated CD25-levels remained stable. Furthermore, transcripts encoding MCP-1, E-selectin, and TNFα were also induced significantly upon reperfusion (p < 0.0005). Systemic treatment of the recipient with YSPSL pre-reperfusion, with or without pre-implantation YSPSL flush of the donor organ, attenuated the post-reperfusion increase in MCP-1 and TGFß (p < 0.05), E-selectin and hemoxygenase 1 transcripts (p < 0.1). CONCLUSIONS: Our data in humans demonstrate a robust increase in inflammatory gene transcript levels during kidney transplantation IRI and reduction thereof by inhibition of leukocyte adhesion.


Asunto(s)
Biomarcadores/análisis , Citocinas/genética , Riñón/fisiopatología , Glicoproteínas de Membrana/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Daño por Reperfusión/diagnóstico , Daño por Reperfusión/prevención & control , Donantes de Tejidos , Adulto , Anciano , Cadáver , Estudios de Cohortes , Método Doble Ciego , Femenino , Estudios de Seguimiento , Humanos , Trasplante de Riñón , Masculino , Persona de Mediana Edad , Selectina-P/antagonistas & inhibidores , Pronóstico , Reacción en Cadena en Tiempo Real de la Polimerasa , Reperfusión , Daño por Reperfusión/genética , Trasplante Homólogo
11.
Arthritis Rheum ; 62(6): 1744-54, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20155833

RESUMEN

OBJECTIVE: Wegener's granulomatosis (WG) is a systemic inflammatory disease that is associated with substantial morbidity. The aim of this study was to understand the biology underlying WG and to discover markers of disease activity that would be useful for prognosis and treatment guidance. METHODS: Gene expression profiling was performed using total RNA from peripheral blood mononuclear cells (PBMCs) and granulocyte fractions from 41 patients with WG and 23 healthy control subjects. Gene set enrichment analysis (GSEA) was performed to search for candidate WG-associated molecular pathways and disease activity biomarkers. Principal components analysis was used to visualize relationships between subgroups of WG patients and controls. Longitudinal changes in proteinase 3 (PR3) gene expression were evaluated using reverse transcription-polymerase chain reaction, and clinical outcomes, including remission status and disease activity, were determined using the Birmingham Vasculitis Activity Score for WG (BVAS-WG). RESULTS: Eighty-six genes in WG PBMCs and 40 in WG polymorphonuclear neutrophils (PMNs) were significantly up-regulated relative to controls. Genes up-regulated in WG PBMCs were involved in myeloid differentiation, and these included the WG autoantigen PR3. The coordinated regulation of myeloid differentiation genes was confirmed by GSEA. The median expression values of the 86 up-regulated genes in WG PBMCs were associated with disease activity (P = 1.3 x 10(-4)), and WG patients with low-level expression of the WG signature genes showed expression profiles that were only modestly different from that in healthy controls (P = 0.07). PR3 transcription was significantly up-regulated in WG PBMCs (P = 1.3 x 10(-5), false discovery rate [FDR] 0.002), but not in WG PMNs (P = 0.03, FDR 0.28), and a preliminary longitudinal analysis showed that the fold change in PR3 RNA levels in WG PBMCs corresponded to changes in the BVAS-WG score over time. CONCLUSION: Transcription of PR3 and related myeloid differentiation genes in PBMCs may represent novel markers of disease activity in WG.


Asunto(s)
Granulomatosis con Poliangitis/genética , Leucocitos Mononucleares/metabolismo , Mieloblastina/genética , Mielopoyesis/genética , Adulto , Anciano , Autoanticuerpos/genética , Autoanticuerpos/metabolismo , Femenino , Perfilación de la Expresión Génica , Granulomatosis con Poliangitis/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Mieloblastina/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de Componente Principal , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Índice de Severidad de la Enfermedad , Transcripción Genética/genética
12.
J Allergy Clin Immunol ; 125(1): 153-159.e28, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20109744

RESUMEN

BACKGROUND: Eczema vaccinatum (EV), a disseminated viral skin infection, is a life-threatening complication of vaccinia virus (VV) inoculation in patients with atopic dermatitis (AD) and is thought to be associated with a defective innate immune response. However, the precise mechanism or mechanisms and key factor or factors of EV are unknown. OBJECTIVE: Given that patients with psoriasis, another inflammatory skin disorder, are not susceptible to EV, we compared the global transcriptional response of skin to VV in healthy subjects, patients with psoriasis, and patients with AD, focusing on AD-specific genes. We hypothesized that differences in the transcriptional response to VV between patients with AD and patients with psoriasis or healthy subjects would identify a defective pathway or pathways that might be associated with the development of EV. METHODS: Gene expression profiling of sham-treated and VV-treated unaffected skin explants from patients with AD (n = 12), patients with psoriasis (n = 12), or healthy subjects (n = 13) were generated with U133_Plus2 (54,613 probe sets) GeneChips and analyzed with the GCOS_1.4/SAM_2.1/MAPPFinder_2.0 pipeline. RESULTS: Sixty-seven genes were significantly affected by VV in AD skin but not in psoriatic and healthy skin. Genes associated with defense response, response to wounding, and immune response were the most affected by VV in AD skin. All genes in these ontologies were downregulated, including the innate immunity genes leukotriene B(4) receptor (LTB4R), orosomucoid 1 (ORM1), coagulation factor II (thrombin) receptor (F2R), complement component 9 (C9), and LPS-binding protein (LBP). These findings were confirmed by means of real-time PCR and validated by means of PubMatrix analysis. ORM1, Toll-like receptor 4 (TLR4), and NLR family pyrin domain containing 1 (NLRP1) genes were also linked to AD severity. CONCLUSION: This study identified groups of innate immunity genes that are associated with the aberrant response of AD skin to VV and represent potential targets for EV pathogenesis.


Asunto(s)
Dermatitis Atópica/virología , Perfilación de la Expresión Génica , Inmunidad Innata/genética , Proteínas/metabolismo , Virus Vaccinia/patogenicidad , Adulto , Dermatitis Atópica/complicaciones , Dermatitis Atópica/fisiopatología , Femenino , Genómica , Humanos , Erupción Variceliforme de Kaposi/inmunología , Erupción Variceliforme de Kaposi/virología , Masculino , Persona de Mediana Edad , Proteínas/genética , Psoriasis/inmunología , Psoriasis/virología , Índice de Severidad de la Enfermedad , Piel/metabolismo , Piel/fisiopatología , Piel/virología , Virus Vaccinia/genética , Adulto Joven
13.
Am J Physiol Renal Physiol ; 298(6): F1472-83, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20181666

RESUMEN

Acute kidney injury (AKI) is being increasingly shown to be a risk factor for chronic kidney disease (CKD), but little is known about the possible mechanistic links. We hypothesized that analysis of the genomic signature in the repair stage after AKI would reveal pathways that could link AKI and CKD. Unilateral renal pedicle clamping for 45 min was performed in male C57BL/6J mice. Mice were euthanized at 3, 10, and 28 days after ischemia-reperfusion injury (IRI). Total RNA was isolated from kidney and analyzed using an Illumina mouse array. Among 24,600 tested genes, 242, 146, and 46 genes were upregulated at days 3, 10, and 28 after IRI, and 85, 35, and 0 genes were downregulated, respectively. Gene ontology analysis showed that gene expression changes were primarily related to immune and inflammatory pathways both early and late after AKI. The most highly upregulated genes late after AKI were hepatitis A virus cellular receptor 1 (Havcr1) and lipocalin 2 (Lcn2), which code for kidney injury molecule-1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL), respectively. This was unexpected since they are both primarily potential biomarkers of the early stage of AKI. Furthermore, increases observed in gene expression in amiloride binding protein 1, vascular cell adhesion molecule-1, and endothelin 1 could explain the salt-sensitive hypertension that can follow AKI. These data suggested that 1) persistent inflammation and immune responses late after AKI could contribute to the pathogenesis of CKD, 2) late upregulation of KIM-1 and NGAL could be a useful marker for sustained renal injury after AKI, and 3) hypertension-related gene changes could underlie mechanisms for persistent renal and vascular injury after AKI.


Asunto(s)
Proteínas de Fase Aguda/genética , Perfilación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Enfermedades Renales/genética , Riñón/enzimología , Lipocalinas/genética , Proteínas de la Membrana/genética , Proteínas Oncogénicas/genética , Daño por Reperfusión/genética , Transcripción Genética , Enfermedad Aguda , Proteínas de Fase Aguda/metabolismo , Animales , Biomarcadores/metabolismo , Enfermedad Crónica , Modelos Animales de Enfermedad , Fibrosis , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Receptor Celular 1 del Virus de la Hepatitis A , Inflamación/genética , Inflamación/inmunología , Riñón/inmunología , Riñón/patología , Enfermedades Renales/enzimología , Enfermedades Renales/inmunología , Enfermedades Renales/patología , Lipocalina 2 , Lipocalinas/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Oncogénicas/metabolismo , Daño por Reperfusión/enzimología , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Regulación hacia Arriba
14.
Front Genet ; 11: 699, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32695145

RESUMEN

Mucormycoses are deadly invasive infections caused by several fungal species belonging to the subphylum Mucoromycotina, order Mucorales. Hallmarks of disease progression include angioinvasion and tissue necrosis that aid in fungal dissemination through the blood stream, causing deeper infections and resulting in poor penetration of antifungal agents to the site of infection. In the absence of surgical removal of the infected focus, antifungal therapy alone is rarely curative. Even when surgical debridement is combined with high-dose antifungal therapy, the mortality associated with mucormycoses is >50%. The unacceptably high mortality rate, limited options for therapy and the extreme morbidity of highly disfiguring surgical therapy provide a clear mandate to understand the molecular mechanisms that govern pathogenesis with the hopes of developing alternative strategies to treat and prevent mucormycoses. In the absence of robust forward and reverse genetic systems available for this taxonomic group of fungi, unbiased next generation sequence (NGS)-based approaches have provided much needed insights into our understanding of many aspects of Mucormycoses, including genome structure, drug resistance, diagnostic development, and fungus-host interactions. Here, we will discuss the specific contributions that NGS-based approaches have made to the field and discuss open questions that can be addressed using similar approaches.

16.
Microb Genom ; 4(2)2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29345613

RESUMEN

Aspergillus fumigatus is an opportunistic, ubiquitous, saprophytic mould that can cause severe allergic responses in atopic individuals as well as life-threatening infections in immunocompromised patients. A critical step in the establishment of infection is the invasion of airway epithelial cells by the inhaled fungi. Understanding how A. fumigatus senses and responds to airway cells is important to understand the pathogenesis of invasive pulmonary aspergillosis. Here, we analysed the transcriptomes of two commonly used clinical isolates, Af293 and CEA10, during infection of the A549 type II pneumocyte cell line in vitro. We focused our RNA-seq analysis on the core set of genes that are present in the genomes of the two strains. Our results suggest that: (a) A. fumigatus does not mount a conserved transcriptional response to airway epithelial cells in our in vitro model and (b) strain background and time spent in the tissue culture media have a greater impact on the transcriptome than the presence of host cells. Our analyses reveal both common and strain-specific transcriptional programmes that allow for the generation of hypotheses about gene function as it pertains to pathogenesis and the significant phenotypic heterogeneity that is observed among A. fumigatus isolates.


Asunto(s)
Aspergillus fumigatus/genética , Aspergillus fumigatus/patogenicidad , Células Epiteliales/microbiología , Sistema Respiratorio/microbiología , Transcriptoma , Células A549 , Células Epiteliales Alveolares/microbiología , Aspergillus fumigatus/crecimiento & desarrollo , Regulación Fúngica de la Expresión Génica , Genes Fúngicos/genética , Interacciones Huésped-Patógeno/genética , Humanos , Fenotipo , Aspergilosis Pulmonar/microbiología , Análisis de Secuencia , Esporas Fúngicas
17.
mBio ; 9(4)2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30108171

RESUMEN

Mucormycosis is a life-threatening, invasive fungal infection that is caused by various species belonging to the order Mucorales. Rhizopus species are the most common cause of the disease, responsible for approximately 70% of all cases of mucormycosis. During pulmonary mucormycosis, inhaled Rhizopus spores must adhere to and invade airway epithelial cells in order to establish infection. The molecular mechanisms that govern this interaction are poorly understood. We performed an unbiased survey of the host transcriptional response during early stages of Rhizopus arrhizus var. delemar (R. delemar) infection in a murine model of pulmonary mucormycosis using transcriptome sequencing (RNA-seq). Network analysis revealed activation of the host's epidermal growth factor receptor (EGFR) signaling. Consistent with the RNA-seq results, EGFR became phosphorylated upon in vitro infection of human alveolar epithelial cells with several members of the Mucorales, and this phosphorylated, activated form of EGFR colocalized with R. delemar spores. Inhibition of EGFR signaling with cetuximab or gefitinib, specific FDA-approved inhibitors of EGFR, significantly reduced the ability of R. delemar to invade and damage airway epithelial cells. Furthermore, gefitinib treatment significantly prolonged survival of mice with pulmonary mucormycosis, reduced tissue fungal burden, and attenuated the activation of EGFR in response to pulmonary mucormycosis. These results indicate EGFR represents a novel host target to block invasion of alveolar epithelial cells by R. delemar, and inhibition of EGFR signaling provides a novel approach for treating mucormycosis by repurposing an FDA-approved drug.IMPORTANCE Mucormycosis is an increasingly common, highly lethal fungal infection with very limited treatment options. Using a combination of in vivo animal models, transcriptomics, cell biology, and pharmacological approaches, we have demonstrated that Mucorales fungi activate EGFR signaling to induce fungal uptake into airway epithelial cells. Inhibition of EGFR signaling with existing FDA-approved drugs significantly increased survival following R. arrhizus var. delemar infection in mice. This study enhances our understanding of how Mucorales fungi invade host cells during the establishment of pulmonary mucormycosis and provides a proof-of-concept for the repurposing of FDA-approved drugs that target EGFR function.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Interacciones Huésped-Patógeno , Pulmón/microbiología , Mucormicosis/prevención & control , Células A549 , Animales , Cetuximab/farmacología , Modelos Animales de Enfermedad , Receptores ErbB/metabolismo , Gefitinib/farmacología , Redes Reguladoras de Genes , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Mucormicosis/microbiología , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Rhizopus/efectos de los fármacos , Rhizopus/patogenicidad , Análisis de Secuencia de ARN , Transducción de Señal/efectos de los fármacos
18.
Cell Signal ; 18(9): 1396-407, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16376521

RESUMEN

Our earlier studies have shown that vitamin C at pharmacological doses (mM) induces loss of redox-dependent viability in bovine lung microvascular endothelial cells (BLMVECs) that is mediated by oxidative stress. Therefore, here, we investigated the vitamin C-induced activation of the lipid signaling enzyme, phospholipase D (PLD) in BLMVECs. Monolayer cultures of BLMVECs were treated with vitamin C (0-10 mM) for different time periods (0-2 h) and the activity of PLD was determined. Vitamin C induced activation of PLD in BLMVECs in a time- and dose-dependent fashion that was significantly attenuated by antioxidants, p38 mitogen-activated protein kinase (p38 MAPK)-specific inhibitor (SB203580), extracellular signal-regulated protein kinase (ERK)-specific inhibitor (PD98059), and transient transfection of cells with dominant-negative (DN)-p38 MAPK and DN-ERK1/ERK2. Vitamin C also induced phosphorylation and enhanced the activities of p38 MAPK and ERK in BLMVECs in a time-dependent fashion. It was also evident that vitamin C induced translocation of PLD(1) and PLD(2), association of p38 MAPK and ERK with PLD(1) and PLD(2), threonine phosphorylation of PLD(1) and PLD(2) and SB203580- and PD98059-inhibitable threonine phosphorylation of PLD(1) in BLMVECs. Transient transfection of BLMVECs with DN-p38 MAPK and DN-ERK1/ERK2 resulted in marked attenuation of vitamin C-induced phosphorylation of threonine in PLD(1) and PLD(2). We, for the first time, showed that vitamin C at pharmacological doses, activated PLD in the lung microvascular ECs through oxidative stress and MAPK activation.


Asunto(s)
Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfolipasa D/metabolismo , Animales , Bovinos , Relación Dosis-Respuesta a Droga , Células Endoteliales/citología , Activación Enzimática , Proteínas Quinasas Activadas por Mitógenos/genética , Fosfolipasa D/genética , Fosforilación , Inhibidores de Proteínas Quinasas/metabolismo , Treonina/metabolismo
19.
Biochem J ; 393(Pt 3): 657-68, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16197369

RESUMEN

HBEpCs (human bronchial epithelial cells) contribute to airway inflammation by secreting a variety of cytokines and chemokines in response to allergens, pathogens, viruses and environmental toxins and pollutants. The potent neutrophil chemoattractant, IL-8 (interleukin-8), is a major cytokine secreted by HBEpCs. We have recently demonstrated that LPA (lysophosphatidic acid) stimulated IL-8 production in HBEpCs via protein kinase C delta dependent signal transduction. However, mechanisms of IL-8 expression and secretion are complex and involve multiple protein kinases and transcriptional factors. The present study was undertaken to investigate MAPK (mitogen-activated protein kinase) signalling in the transcriptional regulation of IL-8 expression and secretion in HBEpCs. Exposure of HBEpCs to LPA (1 microM) enhanced expression and secretion of IL-8 by 5-8-fold and stimulated threonine/tyrosine phosphorylation of ERK (extracellular-signal-regulated kinase), p38 MAPK and JNK (c-Jun N-terminal kinase). The LPA-induced secretion of IL-8 was blocked by the p38 MAPK inhibitor SB203580, by p38 MAPK siRNA (small interfering RNA), and by the JNK inhibitor JNK(i) II, but not by the MEK (MAPK/ERK kinase) inhibitor, PD98059. LPA enhanced the transcriptional activity of the IL-8 gene; that effect relied on activation of the transcriptional factors NF-kappaB (nuclear factor kappaB) and AP-1 (activator protein-1). Furthermore, SB203580 attenuated LPA-dependent phosphorylation of IkappaB (inhibitory kappaB), NF-kappaB and phospho-p38 translocation to the nucleus, NF-kappaB transcription and IL-8 promoter-mediated luciferase reporter activity, without affecting the JNK pathway and AP-1 transcription. Similarly, JNK(i) II only blocked LPA-mediated phosphorylation of JNK and c-Jun, AP-1 transcription and IL-8 promoter-mediated luciferase reporter activity, without blocking p38 MAPK-dependent NF-kappaB transcription. Additionally, siRNA for LPA(1-3) receptors partially blocked LPA-induced IL-8 production and activation of MAPKs. The LPA1 and LPA3 receptors, as compared with LPA2, were most efficient in transducing LPA-mediated IL-8 production. These results show an independent role for p38 MAPK and JNK in LPA-induced IL-8 expression and secretion via NF-kappaB and AP-1 transcription respectively in HBEpCs.


Asunto(s)
Bronquios/citología , Células Epiteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-8/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lisofosfolípidos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Humanos , Interleucina-8/genética , FN-kappa B/metabolismo , Fosforilación , Transporte de Proteínas , Mucosa Respiratoria/citología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/enzimología , Factor de Transcripción AP-1/metabolismo , Transcripción Genética/efectos de los fármacos
20.
Biochem J ; 385(Pt 2): 493-502, 2005 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-15461590

RESUMEN

LPA (lysophosphatidic acid), a potent bioactive phospholipid, elicits diverse cellular responses through activation of the G-protein-coupled receptors LPA1-LPA4. LPA-mediated signalling is partially regulated by LPPs (lipid phosphate phosphatases; LPP-1, -2 and -3) that belong to the phosphatase superfamily. This study addresses the role of LPPs in regulating LPA-mediated cell signalling and IL-8 (interleukin-8) secretion in HBEpCs (human bronchial epithelial cells). Reverse transcription-PCR and Western blotting revealed the presence and expression of LPP-1-3 in HBEpCs. Exogenous [3H]oleoyl LPA was hydrolysed to [3H]-mono-oleoylglycerol. Infection of HBEpCs with an adenoviral construct of human LPP-1 for 48 h enhanced the dephosphorylation of exogenous LPA by 2-3-fold compared with vector controls. Furthermore, overexpression of LPP-1 partially attenuated LPA-induced increases in the intracellular Ca2+ concentration, phosphorylation of IkappaB (inhibitory kappaB) and translocation of NF-kappaB (nuclear factor-kappaB) to the nucleus, and almost completely prevented IL-8 secretion. Infection of cells with an adenoviral construct of the mouse LPP-1 (R217K) mutant partially attenuated LPA-induced IL-8 secretion without altering LPA-induced changes in intracellular Ca2+ concentration, phosphorylation of IkappaB, NF-kappaB activation or IL-8 gene expression. Our results identify LPP-1 as a key regulator of LPA signalling and IL-8 secretion in HBEpCs. Thus LPPs could represent potential targets in regulating leucocyte infiltration and airway inflammation.


Asunto(s)
Bronquios/citología , Calcio/metabolismo , Células Epiteliales/enzimología , Células Epiteliales/metabolismo , Interleucina-8/metabolismo , Lisofosfolípidos/metabolismo , FN-kappa B/metabolismo , Fosfatidato Fosfatasa/fisiología , Adenoviridae , Arginina/genética , Arginina/fisiología , Extractos Celulares/química , Células Cultivadas , Células Epiteliales/virología , Humanos , Trasplante de Pulmón , Lisina/genética , Lisina/fisiología , Mutación Missense/genética , Mutación Missense/fisiología , Fosfatidato Fosfatasa/biosíntesis , Fosfatidato Fosfatasa/genética , Receptores del Ácido Lisofosfatídico/agonistas , Donantes de Tejidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA