Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Surg Res ; 179(1): 153-67, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23122671

RESUMEN

BACKGROUND: Complement is invariably activated during trauma and contributes to tissue injury. Recombinant human decay-accelerating factor (DAF), a complement regulatory protein that inhibits both classical and alternative pathways, improves survival and reduces tissue damage in animal models of tissue injury. The extent to which DAF may facilitate resuscitation in hemorrhaged large animals is not known. METHODS: Male Yorkshire swine assigned to one of six groups were subjected to controlled, isobaric hemorrhage over 15 min to a target mean arterial pressure (MAP) of 35 mm Hg. Hypotension was maintained for 20 min followed by a bolus intravenous injection of DAF or vehicle followed by Hextend resuscitation. Animals were observed for 3 h after hypotensive Hextend resuscitation. Survival, blood chemistry, and physiological parameters were recorded. Additionally, tissue from lung, small intestine, liver, and kidney were subjected to histopathologic evaluation and tissue deposition of complement proteins was determined by immunohistochemistry, dot-blot, and Western blot analyses. RESULTS: Administration of DAF (25 µg/kg) to animals subjected to hemorrhage prior to Hextend infusion significantly improved survival (73% versus 27%); protected gut, lung, liver, and kidney tissue from damage; and resulted in reduced resuscitation fluid requirements when compared with animals subjected to hemorrhage and resuscitation with Hextend alone. Animals treated with a higher dose of DAF (50 µg/kg) followed by Hextend fluid resuscitation did not experience the same benefit, suggesting a narrow therapeutic range for use of DAF as adjunct to Hextend fluid. CONCLUSION: DAF improved survival and reduced early Hextend fluid resuscitation requirements in swine subjected to hemorrhagic shock. These benefits are attributed to decreased complement deposition and limited organ damage.


Asunto(s)
Antígenos CD55/farmacología , Intestino Delgado/efectos de los fármacos , Hígado/efectos de los fármacos , Pulmón/efectos de los fármacos , Resucitación , Choque Hemorrágico/complicaciones , Animales , Relación Dosis-Respuesta a Droga , Fluidoterapia , Derivados de Hidroxietil Almidón/uso terapéutico , Intestino Delgado/patología , Riñón/efectos de los fármacos , Riñón/patología , Hígado/patología , Pulmón/patología , Masculino , Modelos Animales , Resucitación/métodos , Choque Hemorrágico/mortalidad , Tasa de Supervivencia , Porcinos , Resultado del Tratamiento
2.
Cell Mol Life Sci ; 68(4): 709-20, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20706768

RESUMEN

The recent approvals of anticancer therapeutic agents targeting the histone deacetylases and DNA methyltransferases have highlighted the important role that epigenetics plays in human diseases, and suggested that the factors controlling gene expression are novel drug targets. Protein arginine deiminase 4 (PAD4) is one such target because its effects on gene expression parallel those observed for the histone deacetylases. We demonstrated that F- and Cl-amidine, two potent PAD4 inhibitors, display micromolar cytotoxic effects towards several cancerous cell lines (HL-60, MCF7 and HT-29); no effect was observed in noncancerous lines (NIH 3T3 and HL-60 granulocytes). These compounds also induced the differentiation of HL-60 and HT29 cells. Finally, these compounds synergistically potentiated the cell killing effects of doxorubicin. Taken together, these findings suggest PAD4 inhibition as a novel epigenetic approach for the treatment of cancer, and suggest that F- and Cl-amidine are candidate therapeutic agents for this disease.


Asunto(s)
Amidinas/farmacología , Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Hidrolasas/genética , Hidrolasas/metabolismo , Neoplasias/tratamiento farmacológico , ADP-Ribosil Ciclasa 1/genética , Amidinas/química , Animales , Antibióticos Antineoplásicos/farmacología , Antineoplásicos/química , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Doxorrubicina/farmacología , Sinergismo Farmacológico , Inhibidores Enzimáticos/química , Epigenómica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias/enzimología , Neoplasias/genética , Arginina Deiminasa Proteína-Tipo 4 , Desiminasas de la Arginina Proteica
3.
Biochemistry ; 50(19): 3997-4010, 2011 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-21466234

RESUMEN

The protein arginine deiminases (PAD), which catalyze the hydrolysis of peptidyl-arginine to form peptidyl-citrulline, play important roles in a variety of cell signaling pathways, including apoptosis, differentiation, and transcriptional regulation. In addition to these important cellular roles, PAD activity is dysregulated in multiple human diseases [e.g., rheumatoid arthritis (RA), cancer, and colitis], and significantly, PAD inhibition with Cl-amidine has been shown to reduce disease severity in the collagen-induced arthritis model of RA. Although these enzymes play important roles in human cell signaling and disease, the mechanisms that regulate PAD activity under both physiological and pathological conditions are poorly understood. One possible mechanism for regulating PAD activity is autodeimination, to which PAD4 has been shown by us and others to be subjected in vitro and in vivo. Herein, we demonstrate that PAD4 autodeimination does not alter the activity, substrate specificity, or calcium dependence of this isozyme. However, the results of these studies indicate a novel role for autodeimination in modulating the ability of PAD4 to interact with histone deacetylase 1 (HDAC1), citrullinated histone H3 (Cit H3), and protein arginine methyltransferase 1 (PRMT1).


Asunto(s)
Hidrolasas/metabolismo , Iminas/metabolismo , Dominios y Motivos de Interacción de Proteínas , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Activación Enzimática/genética , Células HL-60 , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Histonas/metabolismo , Humanos , Hidrolasas/antagonistas & inhibidores , Hidrolasas/genética , Procesamiento Proteico-Postraduccional , Arginina Deiminasa Proteína-Tipo 4 , Desiminasas de la Arginina Proteica , Proteína-Arginina N-Metiltransferasas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Represoras/genética
4.
Front Physiol ; 12: 806574, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35095566

RESUMEN

Endometriosis is a prevalent gynecologic condition associated with pelvic pain and infertility characterized by the implantation and growth of endometrial tissue displaced into the pelvis via retrograde menstruation. The mouse is a molecularly well-annotated and cost-efficient species for modeling human disease in the therapeutic discovery pipeline. However, as a non-menstrual species with a closed tubo-ovarian junction, the mouse poses inherent challenges as a preclinical model for endometriosis research. Over the past three decades, numerous murine models of endometriosis have been described with varying degrees of fidelity in recapitulating the essential pathophysiologic features of the human disease. We conducted a search of the peer-reviewed literature to identify publications describing preclinical research using a murine model of endometriosis. Each model was reviewed according to a panel of ideal model parameters founded on the current understanding of endometriosis pathophysiology. Evaluated parameters included method of transplantation, cycle phase and type of tissue transplanted, recipient immune/ovarian status, iterative schedule of transplantation, and option for longitudinal lesion assessment. Though challenges remain, more recent models have incorporated innovative technical approaches such as in vivo fluorescence imaging and novel hormonal preparations to overcome the unique challenges posed by murine anatomy and physiology. These models offer significant advantages in lesion development and readout toward a high-fidelity mouse model for translational research in endometriosis.

5.
Sci Rep ; 7(1): 17883, 2017 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-29263436

RESUMEN

Preterm infants are at significantly increased risk for lifelong neurodevelopmental disability with male offspring disproportionately affected. Corticosteroids (such as betamethasone) and magnesium sulphate (MgSO4) are administered to women in preterm labor to reduce neurologic morbidity. Despite widespread use of MgSO4 in clinical practice, its effects on adult offspring are not well known nor have sex-specific differences in therapeutic response been explored. The objective of our study was to examine the long-term effects of perinatal neuroinflammation and the effectiveness of prenatal MgSO4/betamethasone treatments between males and females in a murine model via histologic and expression analyses. Our results demonstrate that male but not female offspring exposed to intrauterine inflammation demonstrated impaired performance in neurodevelopmental testing in early life assessed via negative geotaxis, while those exposed to injury plus treatment fared better. Histologic analysis of adult male brains identified a significant reduction in hippocampal neural density in the injured group compared to controls. Evaluation of key neural markers via qRT-PCR demonstrated more profound differences in gene expression in adult males exposed to injury and treatment compared to female offspring, which largely showed resistance to injury. Prenatal treatment with MgSO4/betamethasone confers long-term benefits beyond cerebral palsy prevention with sex-specific differences in response.


Asunto(s)
Betametasona/farmacología , Inflamación/tratamiento farmacológico , Sulfato de Magnesio/farmacología , Animales , Biomarcadores/metabolismo , Parálisis Cerebral/tratamiento farmacológico , Parálisis Cerebral/metabolismo , Modelos Animales de Enfermedad , Femenino , Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Inflamación/metabolismo , Masculino , Ratones , Embarazo , Atención Prenatal , Caracteres Sexuales
6.
Acta Neuropathol Commun ; 1: 52, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-24252631

RESUMEN

BACKGROUND: Blast-induced neurotrauma (BINT) is the signature life threatening injury of current military casualties. Neuroinflammation is a key pathological occurrence of secondary injury contributing to brain damage after blast injury. We have recently demonstrated that blast-triggered complement activation and cytokine release are associated with BINT. Here, we evaluated if administration of the complement inhibitor recombinant human decay-accelerating factor (rhDAF) is beneficial on neuroinflammation and neurodegeneration in a rat model of moderate BINT. Administration of rhDAF after exposure to moderate blast overpressure (BOP, 120 kPa) mitigated brain injury characterized by neuronal degeneration. rhDAF treatment reduced complement hemolytic activity at 3 hours and tissue complement deposition at 3, 24, and 48 hours as well as systemic and local cytokine release at 24 hours post BOP. Furthermore, rhDAF protected blood-brain barrier (BBB) integrity and reduced cytotoxic edema. Interaction between complement cleavage component, C3a and C3a receptor and tau phosphorylation were also attenuated in rhDAF treated animals at 3 and 24 hours after BOP. These novel findings suggest early complement targeted inhibition as a new therapeutic strategy to decrease neuroinflammation and neurodegeneration after blast TBI. RESULT: Administration of rhDAF after exposure to moderate blast overpressure (BOP, 120 kPa) mitigated brain injury characterized by neuronal degeneration. rhDAF treatment reduced complement hemolytic activity at 3 hours and tissue complement deposition at 3, 24, and 48 hours as well as systemic and local cytokine release at 24 hours post BOP. Furthermore, rhDAF protected blood-brain barrier (BBB) integrity and reduced cytotoxic edema. Interaction between complement cleavage component, C3a and C3a receptor and tau phosphorylation were also attenuated in rhDAF treated animals at 3 and 24 hours after BOP. CONCLUSION: These novel findings suggest early complement targeted inhibition as a new therapeutic strategy to decrease neuroinflammation and neurodegeneration after blast TBI.


Asunto(s)
Traumatismos por Explosión/complicaciones , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/etiología , Antígenos CD55/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiopatología , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/etiología , Edema Encefálico/fisiopatología , Lesiones Encefálicas/fisiopatología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Masculino , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/etiología , Neuroinmunomodulación/efectos de los fármacos , Presión , Distribución Aleatoria , Ratas Sprague-Dawley , Factores de Tiempo , Resultado del Tratamiento
7.
J Neurol Sci ; 318(1-2): 146-54, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22537900

RESUMEN

Blast-induced neurotrauma (BINT) is a major medical concern yet its etiology is largely undefined. Complement activation may play a role in the development of secondary injury following traumatic brain injury; however, its role in BINT is still undefined. The present study was designed to characterize the complement system and adaptive immune-inflammatory responses in a rat model of moderate BINT. Anesthetized rats were exposed to a moderate blast (120 kPa) using an air-driven shock tube. Brain tissue injury, systemic and local complement, cerebral edema, inflammatory cell infiltration, and pro-inflammatory cytokine production were measured at 0.5, 3, 48, 72, 120, and 168 h. Injury to brain tissue was evaluated by histological evaluation. Systemic complement was measured via ELSIA. The remaining measurements were determined by immunohistoflourescent staining. Moderate blast triggers moderate brain injuries, elevated levels of local brain C3/C5b-9 and systemic C5b-9, increased leukocyte infiltration, unregulated tumor necrosis factor alpha (TNFα), and aquaporin-4 in rat brain cortex at 3- and 48-hour post blast. Early immune-inflammatory response to BINT involves complement and TNFα, which correlates with hippocampus and cerebral cortex damage. Complement and TNFα activation may be a novel therapeutic target for reducing the damaging effects of BINT inflammation.


Asunto(s)
Traumatismos por Explosión/fisiopatología , Lesiones Encefálicas/fisiopatología , Activación de Complemento/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Traumatismos por Explosión/inmunología , Traumatismos por Explosión/patología , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Lesiones Encefálicas/inmunología , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
8.
ACS Chem Biol ; 7(1): 160-5, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22004374

RESUMEN

Protein arginine deiminase activity (PAD) is increased in cancer, rheumatoid arthritis, and ulcerative colitis. Although the link between abnormal PAD activity and disease is clear, the relative contribution of the individual PADs to human disease is not known; there are 5 PAD isozymes in humans. Building on our previous development of F- and Cl-amidine as potent pan-PAD irreversible inhibitors, we describe herein a library approach that was used to identify PAD-selective inhibitors. Specifically, we describe the identification of Thr-Asp-F-amidine (TDFA) as a highly potent PAD4 inactivator that displays ≥15-fold selectivity for PAD4 versus PAD1 and ≥50-fold versus PADs 2 and 3. This compound is active in cells and can be used to inhibit PAD4 activity in cellulo. The structure of the PAD4·TDFA complex has also been solved, and the structure and mutagenesis data indicate that the enhanced potency is due to interactions between the side chains of Q346, R374, and R639. Finally, we converted TDFA into a PAD4-selective ABPP and demonstrated that this compound, biotin-TDFA, can be used to selectively isolate purified PAD4 in vitro. In total, TDFA and biotin-TDFA represent PAD4-selective chemical probes that can be used to study the physiological roles of this enzyme.


Asunto(s)
Amidinas/síntesis química , Inhibidores Enzimáticos/síntesis química , Hidrocarburos Fluorados/síntesis química , Hidrolasas/antagonistas & inhibidores , Ornitina/análogos & derivados , Bibliotecas de Moléculas Pequeñas/síntesis química , Amidinas/farmacología , Biotinilación , Inhibidores Enzimáticos/farmacología , Humanos , Hidrocarburos Fluorados/farmacología , Hidrolasas/química , Hidrolasas/metabolismo , Isoenzimas/antagonistas & inhibidores , Isoenzimas/química , Isoenzimas/metabolismo , Cinética , Modelos Moleculares , Sondas Moleculares/síntesis química , Ornitina/síntesis química , Ornitina/farmacología , Estructura Terciaria de Proteína , Arginina Deiminasa Proteína-Tipo 4 , Desiminasas de la Arginina Proteica , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Especificidad por Sustrato
9.
ACS Chem Biol ; 6(5): 466-76, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21265574

RESUMEN

The protein arginine deiminases (PADs), which catalyze the hydrolysis of peptidyl-arginine to form peptidyl-citrulline, are potential targets for the development of a rheumatoid arthritis (RA) therapeutic, as well as other human diseases including colitis and cancer. Additionally, these enzymes, and in particular PAD4, appear to play important roles in a variety of cell signaling pathways including apoptosis, differentiation, and transcriptional regulation. To better understand the factors that regulate in vivo PAD4 activity, we set out to design and synthesize a series of activity-based protein profiling (ABPP) reagents that target this enzyme. Herein we describe the design, synthesis, and evaluation of six ABPPs including (i) FITC-conjugated F-amidine (FFA1 and 2) and Cl-amidine (FCA1 and 2), and (ii) biotin-conjugated F-amidine (BFA) and Cl-amidine (BCA). We further demonstrate the utility of these probes for labeling PAD4 in cells, as well as for isolating PAD4 and PAD4 binding proteins. These probes will undoubtedly prove to be powerful tools that can be used to dissect the factors controlling the dynamics of PAD4 expression, activity, and function.


Asunto(s)
Amidinas/farmacología , Hidrolasas/metabolismo , Biotina/análogos & derivados , Activación Enzimática , Colorantes Fluorescentes , Humanos , Hidrolasas/antagonistas & inhibidores , Límite de Detección , Análisis por Matrices de Proteínas , Arginina Deiminasa Proteína-Tipo 4 , Desiminasas de la Arginina Proteica , Triazoles/química
10.
J Med Chem ; 54(19): 6919-35, 2011 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-21882827

RESUMEN

Protein arginine deiminase (PAD) activity is upregulated in a number of human diseases, including rheumatoid arthritis, ulcerative colitis, and cancer. These enzymes, there are five in humans (PADs 1-4 and 6), regulate gene transcription, cellular differentiation, and the innate immune response. Building on our successful generation of F- and Cl-amidine, which irreversibly inhibit all of the PADs, a structure-activity relationship was performed to develop second generation compounds with improved potency and selectivity. Incorporation of a carboxylate ortho to the backbone amide resulted in the identification of N-α-(2-carboxyl)benzoyl-N(5)-(2-fluoro-1-iminoethyl)-l-ornithine amide (o-F-amidine) and N-α-(2-carboxyl)benzoyl-N(5)-(2-chloro-1-iminoethyl)-l-ornithine amide (o-Cl-amidine), as PAD inactivators with improved potency (up to 65-fold) and selectivity (up to 25-fold). Relative to F- and Cl-amidine, the compounds also show enhanced potency in cellulo. As such, these compounds will be versatile chemical probes of PAD function.


Asunto(s)
Amidinas/síntesis química , Hidrolasas/antagonistas & inhibidores , Ornitina/análogos & derivados , Amidinas/química , Amidinas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Disponibilidad Biológica , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cristalografía por Rayos X , Doxorrubicina/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Humanos , Hidrolasas/química , Cinética , Estructura Molecular , Ornitina/síntesis química , Ornitina/química , Ornitina/farmacología , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA