Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biomater Sci ; 11(6): 2065-2079, 2023 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-36723072

RESUMEN

Prolonged maintenance of therapeutically-relevant levels of broadly neutralizing antibodies (bnAbs) is necessary to enable passive immunization against infectious disease. Unfortunately, protection only lasts for as long as these bnAbs remain present at a sufficiently high concentration in the body. Poor pharmacokinetics and burdensome administration are two challenges that need to be addressed in order to make pre- and post-exposure prophylaxis with bnAbs feasible and effective. In this work, we develop a supramolecular hydrogel as an injectable, subcutaneous depot to encapsulate and deliver antibody drug cargo. This polymer-nanoparticle (PNP) hydrogel exhibits shear-thinning and self-healing properties that are required for an injectable drug delivery vehicle. In vitro drug release assays and diffusion measurements indicate that the PNP hydrogels prevent burst release and slow the release of encapsulated antibodies. Delivery of bnAbs against SARS-CoV-2 from PNP hydrogels is compared to standard routes of administration in a preclinical mouse model. We develop a multi-compartment model to understand the ability of these subcutaneous depot materials to modulate the pharmacokinetics of released antibodies; the model is extrapolated to explore the requirements needed for novel materials to successfully deliver relevant antibody therapeutics with different pharmacokinetic characteristics.


Asunto(s)
COVID-19 , Hidrogeles , Ratones , Animales , Hidrogeles/farmacocinética , SARS-CoV-2 , Anticuerpos ampliamente neutralizantes , Sistemas de Liberación de Medicamentos , Polímeros , Anticuerpos
2.
Proc Natl Acad Sci U S A ; 119(35): e2211310119, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-35994674

RESUMEN

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS). Astrocytes are the most abundant glial cells in the CNS, and their dysfunction contributes to the pathogenesis of MS and its animal model, experimental autoimmune encephalomyelitis (EAE). Recent advances highlight the pivotal role of cellular metabolism in programming immune responses. However, the underlying immunometabolic mechanisms that drive astrocyte pathogenicity remain elusive. Nicotinamide adenine dinucleotide (NAD+) is a vital coenzyme involved in cellular redox reactions and a substrate for NAD+-dependent enzymes. Cellular NAD+ levels are dynamically controlled by synthesis and degradation, and dysregulation of this balance has been associated with inflammation and disease. Here, we demonstrate that cell-autonomous generation of NAD+ via the salvage pathway regulates astrocyte immune function. Inhibition of nicotinamide phosphoribosyltransferase (NAMPT), a key enzyme in the salvage pathway, results in depletion of NAD+, inhibits oxidative phosphorylation, and limits astrocyte inflammatory potential. We identified CD38 as the main NADase up-regulated in reactive mouse and human astrocytes in models of neuroinflammation and MS. Genetic or pharmacological blockade of astrocyte CD38 activity augmented NAD+ levels, suppressed proinflammatory transcriptional reprogramming, impaired chemotactic potential to inflammatory monocytes, and ameliorated EAE. We found that CD38 activity is mediated via calcineurin/NFAT signaling in mouse and human reactive astrocytes. Thus, NAMPT-NAD+-CD38 circuitry in astrocytes controls their ability to meet their energy demands and drives the expression of proinflammatory transcriptional modules, contributing to CNS pathology in EAE and, potentially, MS. Our results identify candidate therapeutic targets in MS.


Asunto(s)
ADP-Ribosil Ciclasa 1 , Astrocitos , Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , NAD , ADP-Ribosil Ciclasa 1/metabolismo , Animales , Astrocitos/inmunología , Astrocitos/metabolismo , Autoinmunidad , Sistema Nervioso Central/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Humanos , Ratones , Esclerosis Múltiple/inmunología , NAD/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo
3.
bioRxiv ; 2022 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-35665002

RESUMEN

Prolonged maintenance of therapeutically-relevant levels of broadly neutralizing antibodies (bnAbs) is necessary to enable passive immunization against infectious disease. Unfortunately, protection only lasts for as long as these bnAbs remain present at a sufficiently high concentration in the body. Poor pharmacokinetics and burdensome administration are two challenges that need to be addressed in order to make pre- and post-exposure prophylaxis with bnAbs feasible and effective. In this work, we develop a supramolecular hydrogel as an injectable, subcutaneous depot to encapsulate and deliver antibody drug cargo. This polymer-nanoparticle (PNP) hydrogel exhibits shear-thinning and self-healing properties that are required for an injectable drug delivery vehicle. In vitro drug release assays and diffusion measurements indicate that the PNP hydrogels prevent burst release and slow the release of encapsulated antibodies. Delivery of bnAbs against SARS-CoV-2 from PNP hydrogels is compared to standard routes of administration in a preclinical mouse model. We develop a multi-compartment model to understand the ability of these subcutaneous depot materials to modulate the pharmacokinetics of released antibodies; the model is extrapolated to explore the requirements needed for novel materials to successfully deliver relevant antibody therapeutics with different pharmacokinetic characteristics.

4.
J Cancer Educ ; 37(3): 843-851, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-33219500

RESUMEN

Breast cancer ranks highest in incidence and mortality among females and second among both genders. Lebanon has the second highest rate of breast cancer worldwide for those 35-39 years old and the highest for those 40-49. Mastectomy often results in deceased shoulder and arm mobility and decreased quality of life. The objective of this study was to assess the effect of an educational program of therapeutic exercises on the quality of life and functional ability in women after a mastectomy. Sixty women undergoing a mastectomy were randomly assigned to either an intervention or control group. The intervention group received extensive pre-surgery education as well as training on therapeutic exercises. Follow-up phone calls to the intervention group were made to ensure that the exercises were being done. Both groups were visited at home at two and four weeks to obtain the outcome variables. The Breast Cancer Patient Version was used to assess quality of life, and the "Goniometer" was used to assess the range of motion of the affected shoulder. At two and four weeks after surgery, women in the intervention group had significant improvements in their shoulder range of motion: flexion, extension, and abduction were significantly different between the control and intervention group at p = 0.04-0.00. For quality of life, physical, psychological, psychological, social, and spiritual well-being were significantly higher for the intervention group at both two and four weeks after surgery, p < 0.001. In a middle-income country, one-to-one education provided by a nurse, which included demonstrations, back demonstrations, and weekly phone calls had a positive impact on women's shoulder range of motion and quality of life. NCT04184102.


Asunto(s)
Neoplasias de la Mama , Mastectomía , Adulto , Neoplasias de la Mama/cirugía , Terapia por Ejercicio/métodos , Femenino , Humanos , Masculino , Calidad de Vida , Rango del Movimiento Articular , Hombro/cirugía
5.
Nat Commun ; 12(1): 3661, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-34135340

RESUMEN

SARS-CoV-2, the virus responsible for COVID-19, has caused a global pandemic. Antibodies can be powerful biotherapeutics to fight viral infections. Here, we use the human apoferritin protomer as a modular subunit to drive oligomerization of antibody fragments and transform antibodies targeting SARS-CoV-2 into exceptionally potent neutralizers. Using this platform, half-maximal inhibitory concentration (IC50) values as low as 9 × 10-14 M are achieved as a result of up to 10,000-fold potency enhancements compared to corresponding IgGs. Combination of three different antibody specificities and the fragment crystallizable (Fc) domain on a single multivalent molecule conferred the ability to overcome viral sequence variability together with outstanding potency and IgG-like bioavailability. The MULTi-specific, multi-Affinity antiBODY (Multabody or MB) platform thus uniquely leverages binding avidity together with multi-specificity to deliver ultrapotent and broad neutralizers against SARS-CoV-2. The modularity of the platform also makes it relevant for rapid evaluation against other infectious diseases of global health importance. Neutralizing antibodies are a promising therapeutic for SARS-CoV-2.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/química , SARS-CoV-2/inmunología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/química , Anticuerpos Antivirales/inmunología , Especificidad de Anticuerpos , Apoferritinas/química , Disponibilidad Biológica , Mapeo Epitopo , Humanos , Inmunoglobulina G/inmunología , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ingeniería de Proteínas/métodos , Subunidades de Proteína/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Distribución Tisular
6.
J Virol ; 94(22)2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-32900822

RESUMEN

Animal models recapitulating human COVID-19 disease, especially severe disease, are urgently needed to understand pathogenesis and to evaluate candidate vaccines and therapeutics. Here, we develop novel severe-disease animal models for COVID-19 involving disruption of adaptive immunity in Syrian hamsters. Cyclophosphamide (CyP) immunosuppressed or RAG2 knockout (KO) hamsters were exposed to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) by the respiratory route. Both the CyP-treated and RAG2 KO hamsters developed clinical signs of disease that were more severe than those in immunocompetent hamsters, notably weight loss, viral loads, and fatality (RAG2 KO only). Disease was prolonged in transiently immunosuppressed hamsters and was uniformly lethal in RAG2 KO hamsters. We evaluated the protective efficacy of a neutralizing monoclonal antibody and found that pretreatment, even in immunosuppressed animals, limited infection. Our results suggest that functional B and/or T cells are not only important for the clearance of SARS-CoV-2 but also play an early role in protection from acute disease.IMPORTANCE Syrian hamsters are in use as a model of disease caused by SARS-CoV-2. Pathology is pronounced in the upper and lower respiratory tract, and disease signs and endpoints include weight loss and viral RNA and/or infectious virus in swabs and organs (e.g., lungs). However, a high dose of virus is needed to produce disease, and the disease resolves rapidly. Here, we demonstrate that immunosuppressed hamsters are susceptible to low doses of virus and develop more severe and prolonged disease. We demonstrate the efficacy of a novel neutralizing monoclonal antibody using the cyclophosphamide transient suppression model. Furthermore, we demonstrate that RAG2 knockout hamsters develop severe/fatal disease when exposed to SARS-CoV-2. These immunosuppressed hamster models provide researchers with new tools for evaluating therapies and vaccines and understanding COVID-19 pathogenesis.


Asunto(s)
Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Modelos Animales de Enfermedad , Mesocricetus , Neumonía Viral/inmunología , Neumonía Viral/patología , Inmunidad Adaptativa , Animales , Animales Modificados Genéticamente , Betacoronavirus/fisiología , COVID-19 , Ciclofosfamida , Proteínas de Unión al ADN/genética , Técnicas de Inactivación de Genes , Inmunosupresores , Pandemias , SARS-CoV-2 , Índice de Severidad de la Enfermedad
7.
Mol Cancer Ther ; 19(10): 2105-2116, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32847983

RESUMEN

Development of antagonistic mAbs that specifically target the immune checkpoint receptor, programmed cell death protein-1 (PD-1), is of great interest for cancer immunotherapy. Here, we report the biophysical characteristics and nonclinical antagonistic activities of sasanlimab (PF-06801591), a humanized anti-PD-1 antibody of IgG4 isotype. We show that sasanlimab binds selectively and with similar high potency to human and cynomolgus monkey PD-1 receptor and blocks its interaction with PD-L1 and PD-L2, with no detectable Fc-dependent effector function. The binding of sasanlimab to human and cynomolgus PD-1 is associated with the formation of a stable complex, which is likely to be the main driver of this high-affinity interaction. In vitro, sasanlimab significantly augmented T-cell proliferation and cytokine production in mixed lymphocyte reaction and superantigen stimulation assays. In vivo, sasanlimab accelerated the incidence of GvHD by enhancing T-cell proliferation and cytokine secretion in a xenogeneic model of acute GvHD and halted the growth of MC-38 colon adenocarcinoma tumors in human PD-1 knock-in mice. Pharmacokinetic and toxicokinetic findings from cynomolgus monkey showed that sasanlimab was active and well-tolerated. Taken together, the data presented here support the clinical development of sasanlimab for the treatment of patients with advanced cancers as a single agent or in combination with other immunotherapies.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Animales , Línea Celular Tumoral , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones
9.
J Neuroinflammation ; 11: 29, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24498870

RESUMEN

Oral atorvastatin has prevented or reversed paralysis in the multiple sclerosis (MS) model experimental autoimmune encephalomyelitis (EAE), and reduced development of new MS lesions in clinical trials. Besides inhibiting development of encephalitogenic T cells, atorvastatin treatment of EAE has been associated with an induction of anti-inflammatory myelin-reactive T-helper type (Th)-2 cells. To investigate the clinical significance of atorvastatin-mediated Th2 differentiation, we first evaluated atorvastatin treatment in interleukin (IL)-4 green fluorescent protein-enhanced transcript (4-GET) reporter mice. Atorvastatin treatment failed to induce IL-4-producing Th2 cells in vivo; however, when T cells from atorvastatin-treated 4-GET mice were reactivated in vitro, T cells preferentially differentiated into Th2 cells, while antigen-specific T-cell proliferation and secretion of proinflammatory cytokines (interferon gamma, IL-17, tumor necrosis factor and IL-12) were reduced. Oral atorvastatin also prevented or reversed EAE in signal transducer and activator of transcription 6-deficient (STAT6-/-) mice, which cannot generate IL-4-producing Th2 cells. Further, atorvastatin treatment did not induce or expand Foxp3+ regulatory T cells in either wild-type or STAT6-/- mice. In vivo proliferation of T cells, as measured by incorporation of bromodeoxyuridine, was inhibited in atorvastatin-treated wild-type and STAT6-/- mice. These data imply that atorvastatin ameliorates central nervous system autoimmune disease primarily by inhibiting proliferation of proinflammatory encephalitogenic T cells, and not simply through induction of anti-inflammatory Th2 cells. This cytostatic effect may be a relevant mechanism of action when considering use of statins in MS and other inflammatory conditions.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Factores de Transcripción Forkhead/metabolismo , Ácidos Heptanoicos/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Pirroles/uso terapéutico , Linfocitos T Reguladores/fisiología , Células Th2/fisiología , Adenilil Ciclasas/genética , Animales , Atorvastatina , Diferenciación Celular , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/genética , Femenino , Ácidos Heptanoicos/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Interleucina-4/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Pirroles/farmacología , Ratas , Factor de Transcripción STAT6/deficiencia
10.
Mol Cell Biol ; 31(17): 3653-69, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21746882

RESUMEN

A new class of inflammatory CD4(+) T cells that produce interleukin-17 (IL-17) (termed Th17) has been identified, which plays a critical role in numerous inflammatory conditions and autoimmune diseases. The active form of vitamin D, 1,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)], has a direct repressive effect on the expression of IL-17A in both human and mouse T cells. In vivo treatment of mice with ongoing experimental autoimmune encephalomyelitis (EAE; a mouse model of multiple sclerosis) diminishes paralysis and progression of the disease and reduces IL-17A-secreting CD4(+) T cells in the periphery and central nervous system (CNS). The mechanism of 1,25(OH)(2)D(3) repression of IL-17A expression was found to be transcriptional repression, mediated by the vitamin D receptor (VDR). Transcription assays, gel shifting, and chromatin immunoprecipitation (ChIP) assays indicate that the negative effect of 1,25(OH)(2)D(3) on IL-17A involves blocking of nuclear factor for activated T cells (NFAT), recruitment of histone deacetylase (HDAC), sequestration of Runt-related transcription factor 1 (Runx1) by 1,25(OH)(2)D(3)/VDR, and a direct effect of 1,25(OH)(2)D(3) on induction of Foxp3. Our results describe novel mechanisms and new concepts with regard to vitamin D and the immune system and suggest therapeutic targets for the control of autoimmune diseases.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Interleucina-17/inmunología , Células Th17/inmunología , Vitamina D/análogos & derivados , Secuencia de Aminoácidos , Animales , Autoinmunidad/inmunología , Western Blotting , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Femenino , Células HEK293 , Humanos , Interleucina-17/genética , Interleucina-17/metabolismo , Células Jurkat , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Transgénicos , Datos de Secuencia Molecular , Receptores de Calcitriol/genética , Receptores de Calcitriol/inmunología , Receptores de Calcitriol/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Linfocitos T/metabolismo , Células Th17/metabolismo , Transcripción Genética/efectos de los fármacos , Vitamina D/farmacología , Vitaminas/farmacología
11.
GM Crops ; 1(2): 87-93, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21865876

RESUMEN

The objective of the present study is to develop an efficient protocol for regeneration of transgenic wheat plants using Agrobacterium- mediated transformation of mature embryos of hexaploid bread wheat (Triticum aestivum) and tetraploid pasta wheat (Triticum durum). The data indicated that embryogenic calli were formed within 7 days in the presence of 2 mgl-1 2,4-D. Adventitious shoots emerged from the embryonic calli in the presence of 2 mgl-1 BA. Shoot regeneration frequency varied between wheat cultivars according to their genetic background differences. Regeneration frequency was higher in the cultivar Gemmiza 10 (95 %) compared with the other cultivars tested. Mature embryos derived callus of the cultivars Gemmiza 10 and Gemmiza 9 were co-cultivated with A. tumefaciens strain LBA4404 harboring a binary vector pBI-121 containing the neomycin phosphotransferase-II gene (npt-II). The resulted putative transgenic plantlets were able to grow on kanamycin containing medium. A successful integration of the transgene was confirmed by analyzing the T0 plantlets using Southern hybridization and PCR amplification. The gus gene expression can be detected only in the transgenic plants. The reported protocol is reproducible and can be used to regenerate transgenic wheat plants expressing the genes present in A. tumifaciens binary vectors.


Asunto(s)
Plantas Modificadas Genéticamente/genética , Semillas/genética , Transformación Genética , Triticum/genética , Ácido 2,4-Diclorofenoxiacético/farmacología , Agrobacterium/genética , Southern Blotting , ADN Bacteriano/genética , Relación Dosis-Respuesta a Droga , Vectores Genéticos/genética , Genotipo , Glucuronidasa/genética , Glucuronidasa/metabolismo , Ácidos Indolacéticos/farmacología , Kanamicina Quinasa/genética , Reguladores del Crecimiento de las Plantas/farmacología , Brotes de la Planta/genética , Brotes de la Planta/fisiología , Plantas Modificadas Genéticamente/fisiología , Reacción en Cadena de la Polimerasa , Poliploidía , Regeneración/efectos de los fármacos , Semillas/fisiología , Especificidad de la Especie , Técnicas de Cultivo de Tejidos , Transgenes/genética , Triticum/clasificación , Triticum/fisiología
12.
Proc Natl Acad Sci U S A ; 106(35): 14948-53, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19706421

RESUMEN

The renin-angiotensin-aldosterone system (RAAS) is a major regulator of blood pressure. The octapeptide angiotensin II (AII) is proteolytically processed from the decapeptide AI by angiotensin-converting enzyme (ACE), and then acts via angiotensin type 1 and type 2 receptors (AT1R and AT2R). Inhibitors of ACE and antagonists of the AT1R are used in the treatment of hypertension, myocardial infarction, and stroke. We now show that the RAAS also plays a major role in autoimmunity, exemplified by multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Using proteomics, we observed that RAAS is up-regulated in brain lesions of MS. AT1R was induced in myelin-specific CD4+ T cells and monocytes during autoimmune neuroinflammation. Blocking AII production with ACE inhibitors or inhibiting AII signaling with AT1R blockers suppressed autoreactive TH1 and TH17 cells and promoted antigen-specific CD4+FoxP3+ regulatory T cells (Treg cells) with inhibition of the canonical NF-kappaB1 transcription factor complex and activation of the alternative NF-kappaB2 pathway. Treatment with ACE inhibitors induces abundant CD4+FoxP3+ T cells with sufficient potency to reverse paralytic EAE. Modulation of the RAAS with inexpensive, safe pharmaceuticals used by millions worldwide is an attractive therapeutic strategy for application to human autoimmune diseases.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Animales , Encefalomielitis Autoinmune Experimental/enzimología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Factores de Transcripción Forkhead/inmunología , Humanos , Interleucina-17/inmunología , Ratones , Peptidil-Dipeptidasa A/metabolismo , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/enzimología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/enzimología
13.
Immunology ; 127(1): 18-25, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19191903

RESUMEN

The 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase inhibitors (statins) interfere with the mevalonate pathway. While initially developed for their lipid-lowering properties, statins have been extensively investigated with respect to their impact on autoantigen and alloantigen driven immune responses. Mechanistically it was shown that statins modify immune responses on several levels, including effects on dendritic cells, endothelial cells, macrophages, B cells and T cells. Several lines of evidence suggest that statins act in a disease-specific manner and are not effective in each immune disorder. This review discusses possible modes of action of statins in modulating immunity towards autoantigens and alloantigens.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Factores Inmunológicos/farmacología , Ácido Mevalónico/metabolismo , Animales , Enfermedades Autoinmunes/inmunología , Autoinmunidad/efectos de los fármacos , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Factores Inmunológicos/uso terapéutico , Transducción de Señal/efectos de los fármacos
14.
Anticancer Res ; 29(12): 4901-8, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20044596

RESUMEN

BACKGROUND: Statins are thought to have tumorolytic properties, reducing angiogenesis by inhibiting pro-angiogenic factors and inducing apoptosis of mural pericytes within the tumor vascular tree. MATERIALS AND METHODS: An orthotopic mouse glioblastoma (GL-26) model was used to investigate the effect of simvastatin on glioblastoma vasculature in vivo. GL-26 cells were implanted into the striatum of C5LKa mice treated with either control, low- or high-dose simvastatin. Brains were analyzed for necrotic volume, apoptosis, morphology and pericytic cells within the vascular tree. RESULTS: Low-dose simvastatin increased necrosis and apoptosis compared to both control and high-dose simvastatin groups. High-dose simvastatin increased vessel caliber by reducing pericytic cells along the tumor vessel wall compared to both control and low-dose simvastatin groups. CONCLUSION: Simvastatin has a dual effect on tumorigenesis. At high doses, it may worsen instead of 'normalizing' tumor angio-architecture, albeit low doses affect tumor cell survival by promoting necrosis and apoptosis.


Asunto(s)
Acilcoenzima A/antagonistas & inhibidores , Apoptosis , Neoplasias Encefálicas/patología , Glioblastoma/patología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Simvastatina/farmacología , Acilcoenzima A/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Glioblastoma/metabolismo , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos C57BL , Necrosis , Neoplasias Experimentales , Neovascularización Fisiológica/efectos de los fármacos
15.
J Immunol ; 181(9): 6536-45, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18941244

RESUMEN

The IL-23/IL-17 pathway plays an important role in chronic inflammatory diseases, including inflammatory bowel disease. In inflammatory bowel disease, intestinal epithelial cells are an important source of chemokines that recruit inflammatory cells. We examined the effect of IL-17 on chemokine expression of HT-29 colonic epithelial cells. IL-17 strongly repressed TNF-alpha-stimulated expression of CXCL10, CXCL11, and CCL5, but synergized with TNF-alpha for induction of CXCL8, CXCL1, and CCL20 mRNAs. For CXCL10, IL-17 strongly inhibited promoter activity but had no effect on mRNA stability. In contrast, for CXCL8, IL-17 slightly decreased promoter activity but stabilized its normally unstable mRNA, leading to a net increase in steady-state mRNA abundance. IL-17 synergized with TNF-alpha in transactivating the epidermal growth factor receptor (EGFR) and in activating ERK and p38 MAPK. The p38 and ERK pathway inhibitors SB203580 and U0126 reversed the repressive effect of IL-17 on CXCL10 mRNA abundance and promoter activity and also reversed the inductive effect of IL-17 on CXCL8 mRNA, indicating that MAPK signaling mediates both the transcriptional repression of CXCL10 and the stabilization of CXCL8 mRNA by IL-17. The EGFR kinase inhibitor AG1478 partially reversed the effects of IL-17 on CXCL8 and CXCL10 mRNA, demonstrating a role for EGFR in downstream IL-17 signaling. The overall results indicate a positive effect of IL-17 on chemokines that recruit neutrophils (CXCL8 and CXCL1), and Th17 cells (CCL20). In contrast, IL-17 represses expression of CXCL10, CXCL11, and CCR5, three chemokines that selectively recruit Th1 but not other effector T cells.


Asunto(s)
Quimiocinas/metabolismo , Colon/inmunología , Colon/metabolismo , Interleucina-17/fisiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Adyuvantes Inmunológicos/fisiología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Quimiocina CCL20/biosíntesis , Quimiocina CCL5/antagonistas & inhibidores , Quimiocina CXCL1/biosíntesis , Quimiocina CXCL10/antagonistas & inhibidores , Quimiocina CXCL11/antagonistas & inhibidores , Quimiocinas/antagonistas & inhibidores , Quimiocinas/biosíntesis , Colon/citología , Regulación hacia Abajo/inmunología , Células HT29 , Humanos , Interleucina-8/biosíntesis , Mucosa Intestinal/citología , Proteínas Represoras/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Regulación hacia Arriba/inmunología
16.
Immunity ; 29(1): 90-100, 2008 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-18585064

RESUMEN

gammadelta T cells uniquely contribute to host immune defense, but how this is accomplished remains unclear. Here, we analyzed the nonclassical major histocompatibility complex class I T10 and T22-specific gammadelta T cells in mice and found that encountering antigen in the thymus was neither required nor inhibitory for their development. But when triggered through the T cell receptor, ligand-naive lymphoid-gammadelta T cells produced IL-17, whereas ligand-experienced cells made IFN-gamma. Immediately after immunization, a large fraction of IL-17(+) gammadelta T cells were found in the draining lymph nodes days before the appearance of antigen-specific IL-17(+) *beta T cells. Thus, thymic selection determines the effector fate of gammadelta T cells rather than constrains their antigen specificities. The swift IL-17 response mounted by antigen-naive gammadelta T cells suggests a critical role for these cells at the onset of an acute inflammatory response to novel antigens.


Asunto(s)
Diferenciación Celular/inmunología , Interferón gamma/biosíntesis , Interleucina-17/biosíntesis , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Subgrupos de Linfocitos T/citología , Animales , Antígenos/inmunología , Antígenos de Superficie/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Citometría de Flujo , Antígenos de Histocompatibilidad Clase I/inmunología , Ratones , Subgrupos de Linfocitos T/inmunología , Timo/citología , Timo/inmunología
17.
Nat Cell Biol ; 10(5): 575-83, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18425116

RESUMEN

Transplanted bone marrow-derived cells (BMDCs) have been reported to fuse with cells of diverse tissues, but the extremely low frequency of fusion has led to the view that such events are biologically insignificant. Nonetheless, in mice with a lethal recessive liver disease (tyrosinaemia), transplantation of wild-type BMDCs restored liver function by cell fusion and prevented death, indicating that cell fusion can have beneficial effects. Here we report that chronic inflammation resulting from severe dermatitis or autoimmune encephalitis leads to robust fusion of BMDCs with Purkinje neurons and formation of hundreds of binucleate heterokaryons per cerebellum, a 10-100-fold higher frequency than previously reported. Single haematopoietic stem-cell transplants showed that the fusogenic cell is from the haematopoietic lineage and parabiosis experiments revealed that fusion can occur without irradiation. Transplantation of rat bone marrow into mice led to activation of dormant rat Purkinje neuron-specific genes in BMDC nuclei after fusion with mouse Purkinje neurons, consistent with nuclear reprogramming. The precise neurological role of these heterokaryons awaits elucidation, but their frequency in brain after inflammation is clearly much higher than previously appreciated.


Asunto(s)
Células de la Médula Ósea/fisiología , Fusión Celular , Dermatitis/inmunología , Células Madre Hematopoyéticas/fisiología , Inflamación/metabolismo , Células de Purkinje/fisiología , Animales , Células de la Médula Ósea/citología , Dermatitis/patología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Hematopoyéticas/citología , Lipopolisacáridos/inmunología , Trasplante de Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos C57BL , Células de Purkinje/citología , Ratas , Ratas Sprague-Dawley , Quimera por Trasplante
18.
Blood ; 110(13): 4588-98, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17827390

RESUMEN

We investigated whether atorvastatin (AT) was capable of protecting animals from acute graft-versus-host disease (aGVHD) across major histocompatibility complex (MHC) mismatch barriers. AT treatment of the donor induced a Th-2 cytokine profile in the adoptively transferred T cells and reduced their in vivo expansion, which translated into significantly reduced aGVHD lethality. Host treatment down-regulated costimulatory molecules and MHC class II expression on recipient antigen-presenting cells (APCs) and enhanced the protective statin effect, without impacting graft-versus-leukemia (GVL) activity. The AT effect was partially reversed in STAT6(-/-) donors and abrogated by L-mevalonate, indicating the relevance of STAT6 signaling and the L-mevalonate pathway for AT-mediated aGVHD protection. AT reduced prenylation levels of GTPases, abolished T-bet expression, and increased c-MAF and GATA-3 protein in vivo. Thus, AT has significant protective impact on aGVHD lethality by Th-2 polarization and inhibition of an uncontrolled Th-1 response while maintaining GVL activity, which is of great clinical relevance given the modest toxicity profile of AT.


Asunto(s)
Quimioprevención , Enfermedad Injerto contra Huésped/prevención & control , Efecto Injerto vs Leucemia , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Células Th2/citología , Traslado Adoptivo , Animales , Presentación de Antígeno/efectos de los fármacos , Atorvastatina , Citocinas/genética , Ácidos Heptanoicos , Ácido Mevalónico/metabolismo , Ratones , Ratones Noqueados , Premedicación , Pirroles , Factor de Transcripción STAT6/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/trasplante , Células Th2/inmunología
19.
Nat Med ; 13(8): 935-43, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17676050

RESUMEN

Treatment with glatiramer acetate (GA, copolymer-1, Copaxone), a drug approved for multiple sclerosis (MS), in a mouse model promoted development of anti-inflammatory type II monocytes, characterized by increased secretion of interleukin (IL)-10 and transforming growth factor (TGF)-beta, and decreased production of IL-12 and tumor necrosis factor (TNF). This anti-inflammatory cytokine shift was associated with reduced STAT-1 signaling. Type II monocytes directed differentiation of T(H)2 cells and CD4+CD25+FoxP3+ regulatory T cells (T(reg)) independent of antigen specificity. Type II monocyte-induced regulatory T cells specific for a foreign antigen ameliorated experimental autoimmune encephalomyelitis (EAE), indicating that neither GA specificity nor recognition of self-antigen was required for their therapeutic effect. Adoptive transfer of type II monocytes reversed EAE, suppressed T(H)17 cell development and promoted both T(H)2 differentiation and expansion of T(reg) cells in recipient mice. This demonstration of adoptive immunotherapy by type II monocytes identifies a central role for these cells in T cell immune modulation of autoimmunity.


Asunto(s)
Monocitos/citología , Monocitos/inmunología , Esclerosis Múltiple/inmunología , Linfocitos T/inmunología , Animales , Diferenciación Celular/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Acetato de Glatiramer , Ratones , Ratones Transgénicos , Monocitos/clasificación , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/patología , Vaina de Mielina/metabolismo , Péptidos/uso terapéutico , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo
20.
Blood ; 110(7): 2674-84, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17622571

RESUMEN

Statins are a class of drugs that inhibit 3-hydroxy-3-methylglutaryl-coenzyme A (HMGcoA) reductase, a critical enzyme in the mevalonate pathway. Several reports document that statins may prevent different human cancers. However, whether or not statins can prevent cancer is controversial due to discordant results. One possible explanation for these conflicting conclusions is that only some tumors or specific statins may be effective. Here, we demonstrate in an in vivo transgenic model in which atorvastatin reverses and prevents the onset of MYC-induced lymphomagenesis, but fails to reverse or prevent tumorigenesis in the presence of constitutively activated K-Ras (G12D). Using phosphoprotein fluorescence-activated cell sorter (FACS) analysis, atorvastatin treatment was found to result in the inactivation of the Ras and ERK1/2 signaling pathways associated with the dephosphorylation and inactivation of MYC. Correspondingly, tumors with a constitutively activated K-Ras (G12D) did not exhibit dephosphorylation of ERK1/2 and MYC. Atorvastatin's effects on MYC were specific to the inhibition of HMGcoA reductase, as treatment with mevalonate, the product of HMG-CoA reductase activity, abrogated these effects and inhibited the ability of atorvastatin to reverse or suppress tumorigenesis. Also, RNAi directed at HMGcoA reductase was sufficient to abrogate the neoplastic properties of MYC-induced tumors. Thus, atorvastatin, by inhibiting HMGcoA reductase, induces changes in phosphoprotein signaling that in turn prevent MYC-induced lymphomagenesis.


Asunto(s)
Transformación Celular Neoplásica/patología , Ácidos Heptanoicos/farmacología , Hidroximetilglutaril-CoA Reductasas/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Linfoma/metabolismo , Linfoma/prevención & control , Proteína Oncogénica p55(v-myc)/metabolismo , Pirroles/farmacología , Animales , Atorvastatina , Supervivencia Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Citometría de Flujo , Perfilación de la Expresión Génica , Glicina/genética , Glicina/metabolismo , Humanos , Linfoma/patología , Ratones , Ratones Transgénicos , Mutación/genética , Proteína Oncogénica p55(v-myc)/genética , Fosfoproteínas/metabolismo , Fosforilación , Lesiones Precancerosas/patología , Transducción de Señal , Tasa de Supervivencia , Proteínas ras/genética , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...