Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 93(15)2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31092578

RESUMEN

Respiratory syncytial virus (RSV) infection of seronegative children previously immunized with formalin-inactivated (FI) RSV has been associated with serious enhanced respiratory disease (ERD). The phenomenon was reproduced in the cotton rat and the mouse, and both preclinical models have been routinely used to evaluate the safety of new RSV vaccine candidates. More recently, we demonstrated that immunizations with suboptimal doses of the RSV fusion (F) antigen, in its post- or prefusion conformation, and in the presence of a Th1-biasing adjuvant, unexpectedly led to ERD in the cotton rat model. To assess if those observations are specific to the cotton rat and to elucidate the mechanism by which vaccination with low antigen doses can drive ERD post-RSV challenge, we evaluated RSV post-F antigen dose de-escalation in BALB/c mice in the presence of a Th1-biasing adjuvant. While decreasing antigen doses, we observed an increase in lung inflammation associated with an upregulation of proinflammatory cytokines. The amplitude of the lung histopathology was comparable to that of FI-RSV-induced ERD, confirming the observations made in the cotton rat. Importantly, depletion of CD4+ T cells prior to viral challenge completely abrogated ERD, preventing proinflammatory cytokine upregulation and the infiltration of T cells, neutrophils, eosinophils, and macrophages into the lung. Overall, low-antigen-dose-induced ERD resembles FI-RSV-induced ERD, except that the former appears in the absence of detectable levels of viral replication and in the context of a Th1-biased immune response. Taken together, our observations reinforce the recent concept that vaccines developed for RSV-naïve individuals should be systematically tested under suboptimal dosing conditions.IMPORTANCE RSV poses a significant health care burden and is the leading cause of serious lower-respiratory-tract infections in young children. A formalin-inactivated RSV vaccine developed in the 1960s not only showed a complete lack of efficacy against RSV infection but also induced severe lung disease enhancement in vaccinated children. Since then, establishing safety in preclinical models has been one of the major challenges to RSV vaccine development. We recently observed in the cotton rat model that suboptimal immunizations with RSV fusion protein could induce lung disease enhancement. In the present study, we extended suboptimal dosing evaluation to the mouse model. We confirmed the induction of lung disease enhancement by vaccinations with low antigen doses and dissected the associated immune mechanisms. Our results stress the need to evaluate suboptimal dosing for any new RSV vaccine candidate developed for seronegative infants.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Inmunización/métodos , Enfermedades Pulmonares/patología , Infecciones por Virus Sincitial Respiratorio/patología , Vacunas contra Virus Sincitial Respiratorio/efectos adversos , Proteínas Virales de Fusión/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Modelos Animales de Enfermedad , Inmunización/efectos adversos , Pulmón/patología , Enfermedades Pulmonares/fisiopatología , Ratones , Ratones Endogámicos BALB C , Infecciones por Virus Sincitial Respiratorio/fisiopatología , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/efectos adversos
2.
J Virol ; 91(8)2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28148790

RESUMEN

Respiratory syncytial virus (RSV) infection of children previously immunized with a nonlive, formalin-inactivated (FI)-RSV vaccine has been associated with serious enhanced respiratory disease (ERD). Consequently, detailed studies of potential ERD are a critical step in the development of nonlive RSV vaccines targeting RSV-naive children and infants. The fusion glycoprotein (F) of RSV in either its postfusion (post-F) or prefusion (pre-F) conformation is a target for neutralizing antibodies and therefore an attractive antigen candidate for a pediatric RSV subunit vaccine. Here, we report the evaluation of RSV post-F and pre-F in combination with glucopyranosyl lipid A (GLA) integrated into stable emulsion (SE) (GLA-SE) and alum adjuvants in the cotton rat model. Immunization with optimal doses of RSV F antigens in the presence of GLA-SE induced high titers of virus-neutralizing antibodies and conferred complete lung protection from virus challenge, with no ERD signs in the form of alveolitis. To mimic a waning immune response, and to assess priming for ERD under suboptimal conditions, an antigen dose de-escalation study was performed in the presence of either GLA-SE or alum. At low RSV F doses, alveolitis-associated histopathology was unexpectedly observed with either adjuvant at levels comparable to FI-RSV-immunized controls. This occurred despite neutralizing-antibody titers above the minimum levels required for protection and with no/low virus replication in the lungs. These results emphasize the need to investigate a pediatric RSV vaccine candidate carefully for priming of ERD over a wide dose range, even in the presence of strong neutralizing activity, Th1 bias-inducing adjuvant, and protection from virus replication in the lower respiratory tract.IMPORTANCE RSV disease is of great importance worldwide, with the highest burden of serious disease occurring upon primary infection in infants and children. FI-RSV-induced enhanced disease, observed in the 1960s, presented a major and ongoing obstacle for the development of nonlive RSV vaccine candidates. The findings presented here underscore the need to evaluate a nonlive RSV vaccine candidate during preclinical development over a wide dose range in the cotton rat RSV enhanced-disease model, as suboptimal dosing of several RSV F subunit vaccine candidates led to the priming for ERD. These observations are relevant to the validity of the cotton rat model itself and to safe development of nonlive RSV vaccines for seronegative infants and children.


Asunto(s)
Acrecentamiento Dependiente de Anticuerpo , Infecciones por Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Células TH1/inmunología , Células Th2/inmunología , Proteínas Virales de Fusión/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Compuestos de Alumbre/administración & dosificación , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Modelos Animales de Enfermedad , Lípido A/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/genética , Sigmodontinae , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas Virales de Fusión/genética
3.
Antimicrob Agents Chemother ; 60(10): 5640-8, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27401576

RESUMEN

Methicillin-resistant Staphylococcus aureus (MRSA) causes large-scale epidemics of acute bacterial skin and skin structure infections (ABSSSI) within communities across the United States. Animal models that reproduce ABSSSI as they occur in humans are urgently needed to test new therapeutic strategies. Alpha-toxin plays a critical role in a variety of staphylococcal infection models in mice, but its role in the pathogenesis of ABSSSI remains to be elucidated in rabbits, which are similar to humans in their susceptibility to S. aureus superantigens and certain bicomponent pore-forming leukocidins. We report here a new rabbit model of ABSSSI and show that those infected with a mutant deficient in expression of alpha-toxin (Δhla) developed a small dermonecrotic lesion, whereas those infected with isogenic USA300 MRSA wild-type or complemented Δhla strains developed ABSSSI that mimic the severe infections that occur in humans, including the large central dermonecrotic core surrounded by erythema, induration, and marked subcutaneous hemorrhage. More importantly, immunoprophylaxis with MEDI4893*, an anti-alpha-toxin human monoclonal antibody, significantly reduced the severity of disease caused by a USA300 wild-type strain to that caused by the Δhla mutant, indicating that this toxin could be completely neutralized during infection. Thus, this study illustrates a potential high standard for the development of new immunotherapeutic agents in which a toxin-neutralizing antibody provides protection to the same degree achieved with a toxin gene knockout. When MEDI4893* was administered as adjunctive therapy with a subtherapeutic dose of linezolid, the combination was significantly more efficacious than either agent alone in reducing the severity of ABSSSI.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Toxinas Bacterianas/inmunología , Proteínas Hemolisinas/inmunología , Enfermedades Cutáneas Bacterianas/microbiología , Infecciones Cutáneas Estafilocócicas/tratamiento farmacológico , Animales , Anticuerpos Monoclonales Humanizados , Toxinas Bacterianas/genética , Anticuerpos ampliamente neutralizantes , Modelos Animales de Enfermedad , Proteínas Hemolisinas/genética , Humanos , Linezolid/sangre , Linezolid/farmacología , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/inmunología , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Profilaxis Pre-Exposición/métodos , Conejos , Enfermedades Cutáneas Bacterianas/inmunología , Infecciones Cutáneas Estafilocócicas/inmunología , Infecciones Cutáneas Estafilocócicas/microbiología
4.
Antimicrob Agents Chemother ; 59(1): 299-309, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25348518

RESUMEN

Alpha-toxin (AT) is a major virulence determinant in Staphylococcus aureus skin and soft tissue infection models. We previously demonstrated that prophylactic administration of 2A3, an AT-neutralizing monoclonal antibody (MAb), prevents S. aureus disease in a mouse dermonecrosis model by neutralizing AT-mediated tissue necrosis and immune evasion. In the present study, MEDI4893*, an affinity-optimized version of 2A3, was characterized for therapeutic activity in the dermonecrosis model as a single agent and in combination with two frontline antibiotics, vancomycin and linezolid. MEDI4893* postinfection therapy was found to exhibit a therapeutic treatment window similar to that for linezolid but longer than that for vancomycin. Additionally, when combined with either vancomycin or linezolid, MEDI4893* resulted in reduced tissue damage, increased neutrophil and macrophage infiltration and abscess formation, and accelerated healing relative to those with the antibiotic monotherapies. These data suggest that AT neutralization with a potent MAb holds promise for both prophylaxis and adjunctive therapy with antibiotics and may be a valuable addition to currently available options for the treatment of S. aureus skin and soft tissue infections.


Asunto(s)
Antibacterianos/farmacología , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Toxinas Bacterianas/inmunología , Proteínas Hemolisinas/inmunología , Infecciones Cutáneas Estafilocócicas/tratamiento farmacológico , Animales , Antibacterianos/farmacocinética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Anticuerpos ampliamente neutralizantes , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Linezolid/farmacocinética , Linezolid/farmacología , Ratones Endogámicos BALB C , Necrosis/tratamiento farmacológico , Necrosis/microbiología , Infecciones Cutáneas Estafilocócicas/inmunología , Infecciones Cutáneas Estafilocócicas/patología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/patogenicidad , Vancomicina/farmacocinética , Vancomicina/farmacología
5.
Neurobiol Dis ; 43(3): 598-608, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21624467

RESUMEN

The activation of nuclear factor κB (NF-κB) contributes to muscle degeneration that results from dystrophin deficiency in human Duchenne muscular dystrophy (DMD) and in the mdx mouse. In dystrophic muscle, NF-κB participates in inflammation and failure of muscle regeneration. Peptides containing the NF-κB Essential Modulator (NEMO) binding domain (NBD) disrupt the IκB kinase complex, thus blocking NF-κB activation. The NBD peptide, which is linked to a protein transduction domain to achieve in vivo peptide delivery to muscle tissue, was systemically delivered to mdx mice for 4 or 7 weeks to study NF-κB activation, histological changes in hind limb and diaphragm muscle and ex vivo function of diaphragm muscle. Decreased NF-κB activation, decreased necrosis and increased regeneration were observed in hind limb and diaphragm muscle in mdx mice treated systemically with NBD peptide, as compared to control mdx mice. NBD peptide treatment resulted in improved generation of specific force and greater resistance to lengthening activations in diaphragm muscle ex vivo. Together these data support the potential of NBD peptides for the treatment of DMD by modulating dystrophic pathways in muscle that are downstream of dystrophin deficiency.


Asunto(s)
Quinasa I-kappa B/administración & dosificación , Quinasa I-kappa B/farmacocinética , Péptidos y Proteínas de Señalización Intracelular/administración & dosificación , Péptidos y Proteínas de Señalización Intracelular/farmacocinética , Músculo Esquelético/fisiología , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/metabolismo , Péptidos/uso terapéutico , Animales , Diafragma/patología , Diafragma/fisiología , Modelos Animales de Enfermedad , Distrofina/deficiencia , Distrofina/genética , Quinasa I-kappa B/uso terapéutico , Péptidos y Proteínas de Señalización Intracelular/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Músculo Esquelético/inervación , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/patología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/fisiología , Necrosis/prevención & control , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/genética , Regeneración Nerviosa/fisiología , Péptidos/administración & dosificación , Péptidos/farmacocinética , Estructura Terciaria de Proteína/genética
6.
Mol Cancer Ther ; 19(8): 1649-1659, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32404408

RESUMEN

Multiple myeloma is a hematologic cancer that disrupts normal bone marrow function and has multiple lines of therapeutic options, but is incurable as patients ultimately relapse. We developed a novel antibody-drug conjugate (ADC) targeting CS-1, a protein that is highly expressed on multiple myeloma tumor cells. The anti-CS-1 mAb specifically bound to cells expressing CS-1 and, when conjugated to a cytotoxic pyrrolobenzodiazepine payload, reduced the viability of multiple myeloma cell lines in vitro In mouse models of multiple myeloma, a single administration of the CS-1 ADC caused durable regressions in disseminated models and complete regression in a subcutaneous model. In an exploratory study in cynomolgus monkeys, the CS-1 ADC demonstrated a half-life of 3 to 6 days; however, no highest nonseverely toxic dose was achieved, as bone marrow toxicity was dose limiting. Bone marrow from dosed monkeys showed reductions in progenitor cells as compared with normal marrow. In vitro cell killing assays demonstrated that the CS-1 ADC substantially reduced the number of progenitor cells in healthy bone marrow, leading us to identify previously unreported CS-1 expression on a small population of progenitor cells in the myeloid-erythroid lineage. This finding suggests that bone marrow toxicity is the result of both on-target and off-target killing by the ADC.


Asunto(s)
Anticuerpos Monoclonales/química , Antineoplásicos/farmacología , Benzodiazepinas/química , Inmunoconjugados/farmacología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de Microfilamentos/antagonistas & inhibidores , Mieloma Múltiple/tratamiento farmacológico , Pirroles/química , Animales , Antineoplásicos/química , Apoptosis , Proliferación Celular , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Inmunoconjugados/química , Macaca fascicularis , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas de Microfilamentos/inmunología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Pediatr Crit Care Med ; 8(6): 570-5, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17906594

RESUMEN

OBJECTIVES: The passage of unconjugated bilirubin (UCB) across the blood-brain barrier into the central nervous system is a crucial first step in the development of kernicterus. The objective of the current study was to characterize the passage of UCB across primary bovine brain microvascular endothelial cell (BBMVEC) monolayers in vitro. DESIGN: Experimental study. SETTING: Research institute. SUBJECTS: BBMVECs. INTERVENTIONS: Tritiated UCB (H-UCB) transport at 60, 80, 100, 200, 300, and 400 nM concentrations was tested in both the apical to basolateral (A--> B) and basolateral to apical (B-->A) directions in BBMVEC monolayers in vitro with or without preincubation with pharmacologic active transport inhibitors cyclosporine A, indomethacin, or MK571. MEASUREMENTS AND MAIN RESULTS: The rate of H-UCB transport in the B-->A direction was 6.2- to 7.3-fold higher than in the A-->B direction, suggesting active efflux of UCB. Cyclosporine A (5 microM), a model inhibitor of P-glycoprotein, enhanced A-->B while decreasing B-->A UCB transport, resulting in an overall decrease in BBMVEC UCB efflux of between 46% and 54%. Indomethacin (10 microM) and MK-571 (50 microM), respectively a substrate and potent inhibitor of multidrug resistance-associated protein-1, had no effect. CONCLUSIONS: We conclude that 1) UCB is transported by BBMVEC monolayers in vitro in a net B-->A direction (i.e., active efflux); and 2) cyclosporine A partially inhibits such transport. We speculate that the blood-brain barrier limits the passage and central nervous system retention of UCB by active transport and that this may be accounted in part by P-glycoprotein.


Asunto(s)
Antioxidantes/metabolismo , Bilirrubina/metabolismo , Encéfalo/irrigación sanguínea , Células Endoteliales/metabolismo , Animales , Antioxidantes/administración & dosificación , Bilirrubina/administración & dosificación , Transporte Biológico/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Western Blotting , Bovinos , Células Endoteliales/efectos de los fármacos , Técnicas In Vitro , Kernicterus
8.
PLoS One ; 12(11): e0188708, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29182682

RESUMEN

Respiratory syncytial virus (RSV) is recognized as an important cause of lower and upper respiratory tract infections in older adults, and a successful vaccine would substantially lower morbidity and mortality in this age group. Recently, two vaccine candidates based on soluble purified glycoprotein F (RSV F), either alone or adjuvanted with glucopyranosyl lipid A formulated in a stable emulsion (GLA-SE), failed to reach their primary endpoints in clinical efficacy studies, despite demonstrating the desired immunogenicity profile and efficacy in young rodent models. Here, one of the RSV F vaccine candidates (post-fusion conformation, RSV post-F), and a stabilized pre-fusion form of RSV F (RSV pre-F, DS-Cav1) were evaluated in aged BALB/c mice. Humoral and cellular immunogenicity elicited after immunization of naïve, aged mice was generally lower compared to young animals. In aged mice, RSV post-F vaccination without adjuvant poorly protected the respiratory tract from virus replication, and addition of GLA-SE only improved protection in the lungs, but not in nasal turbinates. RSV pre-F induced higher neutralizing antibody titers compared to RSV post-F (as previously reported) but interestingly, RSV F-specific CD8 T cell responses were lower compared to RSV post-F responses regardless of age. The vaccines were also tested in RSV seropositive aged mice, in which both antigen forms similarly boosted neutralizing antibody titers, although GLA-SE addition boosted neutralizing activity only in RSV pre-F immunized animals. Cell-mediated immune responses in the aged mice were only slightly boosted and well below levels induced in seronegative young mice. Taken together, the findings suggest that the vaccine candidates were not able to induce a strong anti-RSV immune response in recipient mice with an aged immune system, in agreement with recent human clinical trial results. Therefore, the aged mouse model could be a useful tool to evaluate improved vaccine candidates, targeted to prevent RSV disease in older adults.


Asunto(s)
Factores de Edad , Proteínas Recombinantes de Fusión/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Humanos , Inmunidad Celular , Ratones , Ratones Endogámicos BALB C , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/metabolismo
9.
Diabetes ; 66(7): 2007-2018, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28408435

RESUMEN

Neurturin (NRTN), a member of the glial-derived neurotrophic factor family, was identified from an embryonic chicken pancreatic cDNA library in a screen for secreted factors. In this study, we assessed the potential antidiabetic activities of NRTN relative to liraglutide, a glucagon-like peptide 1 receptor agonist, in Zucker diabetic fatty (ZDF) rats. Subcutaneous administration of NRTN to 8-week-old male ZDF rats prevented the development of hyperglycemia and improved metabolic parameters similar to liraglutide. NRTN treatment increased pancreatic insulin content and ß-cell mass and prevented deterioration of islet organization. However, unlike liraglutide-treated rats, NRTN-mediated improvements were not associated with reduced body weight or food intake. Acute NRTN treatment did not activate c-Fos expression in key feeding behavior and metabolic centers in ZDF rat brain or directly enhance glucose-stimulated insulin secretion from pancreatic ß-cells. Treating 10-week-old ZDF rats with sustained hyperglycemia with liraglutide resulted in some alleviation of hyperglycemia, whereas NRTN was not as effective despite improving plasma lipids and fasting glucose levels. Interestingly, coadministration of NRTN and liraglutide normalized hyperglycemia and other metabolic parameters, demonstrating that combining therapies with distinct mechanism(s) can alleviate advanced diabetes. This emphasizes that therapeutic combinations can be more effective to manage diabetes in individuals with uncontrolled hyperglycemia.


Asunto(s)
Glucemia/efectos de los fármacos , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Hipoglucemiantes/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Liraglutida/farmacología , Neurturina/farmacología , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Modelos Animales de Enfermedad , Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/agonistas , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Masculino , Tamaño de los Órganos , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Zucker
10.
Sci Transl Med ; 8(329): 329ra31, 2016 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-26962155

RESUMEN

Broad-spectrum antibiotic use may adversely affect a patient's beneficial microbiome and fuel cross-species spread of drug resistance. Although alternative pathogen-specific approaches are rationally justified, a major concern for this precision medicine strategy is that co-colonizing or co-infecting opportunistic bacteria may still cause serious disease. In a mixed-pathogen lung infection model, we find that the Staphylococcus aureus virulence factor α toxin potentiates Gram-negative bacterial proliferation, systemic spread, and lethality by preventing acidification of bacteria-containing macrophage phagosomes, thereby reducing effective killing of both S. aureus and Gram-negative bacteria. Prophylaxis or early treatment with a single α toxin neutralizing monoclonal antibody prevented proliferation of co-infecting Gram-negative pathogens and lethality while also promoting S. aureus clearance. These studies suggest that some pathogen-specific, antibody-based approaches may also work to reduce infection risk in patients colonized or co-infected with S. aureus and disparate drug-resistant Gram-negative bacterial opportunists.


Asunto(s)
Toxinas Bacterianas/efectos adversos , Proteínas Hemolisinas/efectos adversos , Infecciones Oportunistas/microbiología , Infecciones del Sistema Respiratorio/microbiología , Infecciones Estafilocócicas/microbiología , Ácidos/metabolismo , Animales , Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Calpaína/metabolismo , Coinfección/microbiología , Activación Enzimática/efectos de los fármacos , Humanos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/patología , Lisosomas/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/patología , Ratones , Viabilidad Microbiana/efectos de los fármacos , Modelos Biológicos , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Infecciones Oportunistas/patología , Neumonía/microbiología , Neumonía/patología , Pseudomonas aeruginosa/efectos de los fármacos , Infecciones del Sistema Respiratorio/patología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/crecimiento & desarrollo
11.
J Appl Physiol (1985) ; 93(2): 407-17, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12133845

RESUMEN

Muscular dystrophies are a clinically and genetically heterogeneous group of disorders that show myofiber degeneration and regeneration. Identification of animal models of muscular dystrophy has been instrumental in research on the pathogenesis, pathophysiology, and treatment of these disorders. We review our understanding of the functional status of dystrophic skeletal muscle from selected animal models with a focus on 1) the mdx mouse model of Duchenne muscular dystrophy, 2) the Bio 14.6 delta-sarcoglycan-deficient hamster model of limb-girdle muscular dystrophy, and 3) transgenic null mutant murine lines of sarcoglycan (alpha, beta, delta, and gamma) deficiencies. Although biochemical data from these models suggest that the dystrophin-sarcoglycan-dystroglycan-laminin network is critical for structural integrity of the myofiber plasma membrane, emerging studies of muscle physiology suggest a more complex picture, with specific functional deficits varying considerably from muscle to muscle and model to model. It is likely that changes in muscle structure and function, downstream of the specific, primary biochemical deficiency, may alter muscle contractile properties.


Asunto(s)
Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/fisiopatología , Distrofia Muscular de Duchenne/fisiopatología , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos mdx , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/genética
12.
PDA J Pharm Sci Technol ; 67(1): 2-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23385559

RESUMEN

The primary purpose of an environmental monitoring program is to provide oversight for microbiological cleanliness of manufacturing operation and document the state of control of the facility. Key to the success of the program is the establishment of alert and action limits. In practice, several statistical methods including normal, Poisson, and negative binomial modeling have been routinely used to set these limits. However, data collected from clean rooms or controlled locations often display excess of zeros and overdispersion, caused by sampling population heterogeneity. Such data render it inappropriate to use the traditional methods to set alert and action levels. In this paper, a method based on a zero-inflated negative binomial model is proposed for the above instances. The method provides an enhanced alternative for trending environmental data of classified rooms, and it is demonstrated to show a clear improvement in terms of model fitting and parameter estimation. LAY ABSTRACT: The primary purpose of an environmental monitoring program is to provide oversight for microbiological cleanliness of manufacturing operation and document the state of control of the facility. Key to the success of the program is the establishment of alert and action limits. In practice, several statistical methods including normal, Poisson, and negative binomial modeling have been routinely used to set these limits. However, data collected from clean rooms or controlled locations often display excess of zeros and overdispersion, caused by sampling population heterogeneity. Such data render it inappropriate to use the traditional methods to set alert and action levels. In this paper, a method based on a zero-inflated negative binomial model is proposed for the above instances. The method provides an enhanced alternative for trending environmental data of classified rooms, and it is demonstrated to show a clear improvement in terms of model fitting and parameter estimation.


Asunto(s)
Modelos Estadísticos , Análisis de Regresión , Monitoreo del Ambiente , Humanos , Distribución de Poisson
13.
PLoS One ; 8(10): e75103, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24098366

RESUMEN

An optimal host response against Staphylococcus aureus skin and soft tissue infections (SSTI) is dependent on IL-1ß and IL-17 mediated abscess formation. Alpha toxin (AT), an essential virulence factor for SSTI, has been reported to damage tissue integrity; however its effect on the immune response has not been investigated. Here, we demonstrate that infection with USA300 AT isogenic mutant (Δhla), or passive immunization with an AT neutralizing mAb, 2A3, 24 h prior to infection with wild type USA300 (WT), resulted in dermonecrotic lesion size reduction, and robust neutrophil infiltration. Infiltration correlates with increase in proinflammatory cytokines and chemokines, as well as enhanced bacterial clearance relative to immunization with a negative control mAb. In addition, infection with Δhla, or with WT +2A3, resulted in an early influx of innate IL-17(+)γδT cells and a more rapid induction of an adaptive immune response as measured by Th1 and Th17 cell recruitment at the site of infection. These results are the first direct evidence of a role for AT in subverting the innate and adaptive immune responses during a S. aureus SSTI. Further, these effects of AT can be overcome with a high affinity anti-AT mAb resulting in a reduction in disease severity.


Asunto(s)
Inmunidad Adaptativa , Toxinas Bacterianas/metabolismo , Inmunidad Innata , Piel/microbiología , Piel/patología , Staphylococcus aureus/metabolismo , Animales , Anticuerpos Neutralizantes/inmunología , Toxinas Bacterianas/inmunología , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos BALB C , Necrosis/inmunología , Piel/inmunología , Infecciones de los Tejidos Blandos/inmunología , Infecciones de los Tejidos Blandos/microbiología , Staphylococcus aureus/fisiología , Células TH1/inmunología , Células TH1/metabolismo
14.
Biol Neonate ; 90(1): 40-5, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16534185

RESUMEN

BACKGROUND: The hyperbilirubinemic j/j Gunn rat is frequently used to study the effects of neonatal hyperbilirubinemia on the developing central nervous system (CNS). Despite evidence that the cerebellar region and males are predisposed to bilirubin-induced brain injury in this animal model, there are limited regional and no sex-specific brain bilirubin content data. OBJECTIVE/METHODS: To characterize and contrast the regional (cortex, brainstem, cerebellum) and sex-specific CNS bilirubin contents of hyperbilirubinemic j/j Gunn rat pups and their age-matched (15-19 days) nonjaundiced J/j counterparts. Pups were injected 24 h prior to sacrifice with sulfadimethoxine (200 mg/kg i.p.) to enhance the CNS bilirubin content. RESULTS: The CNS bilirubin contents in each region and total serum bilirubin levels were significantly greater in jaundiced j/j pups versus nonjaundiced J/j pups. Within the sulfadimethoxine-treated male j/j cohort, the mean brain bilirubin content was highest in the cerebellum (18.9 +/- 7.8 microg/g), intermediate in the brainstem (10.7 +/- 8.0 microg/g), and lowest in the cortex (4.7 +/- 3.0 microg/g) (F = 11.31, p < 0.001 by ANOVA), and the cerebellar bilirubin level was significantly higher than in the littermate-matched sulfadimthoxine-treated j/j female pups (p < 0.02). The serum albumin levels were not different between j/j male and j/j female pups. CONCLUSIONS: We conclude that the brain bilirubin content of hyperbilirubinemic j/j Gunn rat pups is greater than in nonjaundiced J/j pups and varies as a function of CNS region and sex. We speculate that the higher cerebellar bilirubin content may preferentially predispose male j/j Gunn rat pups to bilirubin-induced neurotoxicity.


Asunto(s)
Bilirrubina/metabolismo , Encéfalo/metabolismo , Hiperbilirrubinemia/metabolismo , Ratas Gunn/fisiología , Animales , Femenino , Masculino , Ratas , Caracteres Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA