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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
J Exp Med ; 204(2): 321-30, 2007 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-17261635

RESUMEN

Peroxisome proliferator-activated receptor (PPAR)alpha is a nuclear receptor that mediates gender differences in lipid metabolism. PPARalpha also functions to control inflammatory responses by repressing the activity of nuclear factor kappaB (NF-kappaB) and c-jun in immune cells. Because PPARalpha is situated at the crossroads of gender and immune regulation, we hypothesized that this gene may mediate sex differences in the development of T cell-mediated autoimmune disease. We show that PPARalpha is more abundant in male as compared with female CD4(+) cells and that its expression is sensitive to androgen levels. Genetic ablation of this gene selectively removed the brake on NF-kappaB and c-jun activity in male T lymphocytes, resulting in higher production of interferon gamma and tumor necrosis factor (but not interleukin 17), and lower production of T helper (Th)2 cytokines. Upon induction of experimental autoimmune encephalomyelitis, male but not female PPARalpha(-/-) mice developed more severe clinical signs that were restricted to the acute phase of disease. These results suggest that males are less prone to develop Th1-mediated autoimmunity because they have higher T cell expression of PPARalpha.


Asunto(s)
Autoinmunidad/inmunología , Linfocitos T CD4-Positivos/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , PPAR alfa/metabolismo , Traslado Adoptivo , Andrógenos/sangre , Andrógenos/metabolismo , Animales , Western Blotting , Linfocitos T CD4-Positivos/inmunología , Sistema Nervioso Central/patología , Citocinas/biosíntesis , Cartilla de ADN , Femenino , Citometría de Flujo , Interferón gamma/biosíntesis , Masculino , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , PPAR alfa/genética , PPAR alfa/inmunología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores Sexuales , Estadísticas no Paramétricas , Factor de Necrosis Tumoral alfa/biosíntesis
8.
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
9.
J Exp Med ; 203(2): 401-12, 2006 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-16476765

RESUMEN

3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase is a critical enzyme in the mevalonate pathway that regulates the biosynthesis of cholesterol as well as isoprenoids that mediate the membrane association of certain GTPases. Blockade of this enzyme by atorvastatin (AT) inhibits the destructive proinflammatory T helper cell (Th)1 response during experimental autoimmune encephalomyelitis and may be beneficial in the treatment of multiple sclerosis and other Th1-mediated autoimmune diseases. Here we present evidence linking specific isoprenoid intermediates of the mevalonate pathway to signaling pathways that regulate T cell autoimmunity. We demonstrate that the isoprenoid geranylgeranyl-pyrophosphate (GGPP) mediates proliferation, whereas both GGPP and its precursor, farnesyl-PP, regulate the Th1 differentiation of myelin-reactive T cells. Depletion of these isoprenoid intermediates in vivo via oral AT administration hindered these T cell responses by decreasing geranylgeranylated RhoA and farnesylated Ras at the plasma membrane. This was associated with reduced extracellular signal-regulated kinase (ERK) and p38 phosphorylation and DNA binding of their cotarget c-fos in response to T cell receptor activation. Inhibition of ERK and p38 mimicked the effects of AT and induced a Th2 cytokine shift. Thus, by connecting isoprenoid availability to regulation of Th1/Th2 fate, we have elucidated a mechanism by which AT may suppress Th1-mediated central nervous system autoimmune disease.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/inmunología , Ácidos Heptanoicos/farmacología , Pirroles/farmacología , Terpenos/farmacología , Células TH1/inmunología , Células Th2/inmunología , Animales , Atorvastatina , Diferenciación Celular/inmunología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colesterol/sangre , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Ácidos Heptanoicos/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Ratones , Ratones Endogámicos , Ratones Transgénicos , Prenilación de Proteína/efectos de los fármacos , Pirroles/administración & dosificación , Células TH1/citología , Células TH1/efectos de los fármacos , Células Th2/citología , Células Th2/efectos de los fármacos , Proteínas ras/antagonistas & inhibidores , Proteínas ras/metabolismo , Proteínas ras/fisiología , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/metabolismo
10.
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
11.
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.

12.
Nat Med ; 8(2): 143-9, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11821898

RESUMEN

An expanded polyglutamine domain in huntingtin underlies the pathogenic events in Huntington disease (HD), characterized by chorea, dementia and severe weight loss, culminating in death. Transglutaminase (TGase) may be critical in the pathogenesis, via cross-linking huntingtin. Administration of the TGase competitive inhibitor, cystamine, to transgenic mice expressing exon 1 of huntingtin containing an expanded polyglutamine repeat, altered the course of their HD-like disease. Cystamine given intraperitoneally entered brain where it inhibited TGase activity. When treatment began after the appearance of abnormal movements, cystamine extended survival, reduced associated tremor and abnormal movements and ameliorated weight loss. Treatment did not influence the appearance or frequency of neuronal nuclear inclusions. Unexpectedly, cystamine treatment increased transcription of one of the two genes shown to be neuroprotective for polyglutamine toxicity in Drosophila, dnaj (also known as HDJ1 and Hsp40 in humans and mice, respectively). Inhibition of TGase provides a new treatment strategy for HD and other polyglutamine diseases.


Asunto(s)
Cistamina/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Enfermedad de Huntington/tratamiento farmacológico , Trastornos del Movimiento/prevención & control , Transglutaminasas/antagonistas & inhibidores , Animales , Encéfalo/enzimología , Humanos , Ratones , Ratones Transgénicos , Sobrevida , Transglutaminasas/genética , Pérdida de Peso/efectos de los fármacos
13.
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
14.
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
15.
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
16.
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
17.
J Clin Invest ; 116(4): 1037-44, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16543951

RESUMEN

One approach to improving efficacy in MS therapy is to identify medications that provide additive or synergistic benefit in combination. Orally administered cholesterol-lowering HMG-CoA reductase inhibitors (known as statins), which exhibit immunomodulatory properties and are effective in treatment of the MS model EAE, are being tested in MS. As atorvastatin can enhance protective Th2 responses and has a different mechanism of action than glatiramer acetate (GA), a parenterally administered immunomodulatory agent approved for MS treatment, we tested whether the combination of these agents could be beneficial in EAE. Combination therapy using suboptimal doses of atorvastatin and GA prevented or reversed clinical and histologic EAE. Secretion of proinflammatory Th1 cytokines was reduced--and conversely Th2 cytokine secretion was increased--in these mice, but not in mice treated with each drug alone at the same doses. Monocytes treated with the combination of suboptimal doses of atorvastatin and GA secreted an antiinflammatory type II cytokine pattern and, when used as APCs, promoted Th2 differentiation of naive myelin-specific T cells. Our results demonstrate that agents with different mechanisms of immune modulation can combine in a synergistic manner for the treatment of CNS autoimmunity and provide rationale for testing the combination of atorvastatin and GA in MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Ácidos Heptanoicos/uso terapéutico , Factores Inmunológicos/farmacología , Péptidos/uso terapéutico , Pirroles/uso terapéutico , Animales , Atorvastatina , Células Cultivadas , Citocinas/metabolismo , Quimioterapia Combinada , Femenino , Acetato de Glatiramer , Ácidos Heptanoicos/farmacología , Factores Inmunológicos/uso terapéutico , Ratones , Ratones Endogámicos , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/metabolismo , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Péptidos/farmacología , Pirroles/farmacología , Linfocitos T Reguladores/inmunología , Células TH1/inmunología
18.
J Neuroimmunol ; 178(1-2): 140-8, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16860400

RESUMEN

Statins, inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductase, are widely prescribed for their cholesterol-lowering properties to reduce atherogenesis and cardiovascular morbidity. Over recent years, statins have also been shown to exert pleiotropic immunomodulatory effects that might be of therapeutic benefit in autoimmune disorders. The primary mechanism by which statins alter immune function appears to be mediated through the inhibition of post-translational protein prenylation of small GTP-binding proteins and is largely independent of lipid-lowering. In experimental autoimmune encephalomyelitis (EAE), the mouse model for multiple sclerosis (MS), statins prevent or reverse paralysis and were recently shown to exert synergistic benefit when combined with agents approved for MS therapy. Based primarily upon the beneficial effects in EAE, statins are now being tested in patients in MS clinical trials.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso/tratamiento farmacológico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Transducción de Señal/inmunología , Animales , Presentación de Antígeno/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Ensayos Clínicos como Asunto , Humanos , Médula Espinal/efectos de los fármacos , Médula Espinal/inmunología , Linfocitos T/efectos de los fármacos
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA