Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Ther Nucleic Acids ; 30: 479-492, 2022 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-36420217

RESUMEN

Duchenne muscular dystrophy (DMD) is a devastating muscle-wasting disease that arises due to the loss of dystrophin expression, leading to progressive loss of motor and cardiorespiratory function. Four exon-skipping approaches using antisense phosphorodiamidate morpholino oligomers (PMOs) have been approved by the FDA to restore a DMD open reading frame, resulting in expression of a functional but internally deleted dystrophin protein, but in patients with single-exon duplications, exon skipping has the potential to restore full-length dystrophin expression. Cell-penetrating peptide-conjugated PMOs (PPMOs) have demonstrated enhanced cellular uptake and more efficient dystrophin restoration than unconjugated PMOs. In the present study, we demonstrate widespread PPMO-mediated dystrophin restoration in the Dup2 mouse model of exon 2 duplication, representing the most common single-exon duplication among patients with DMD. In this proof-of-concept study, a single intravenous injection of PPMO targeting the exon 2 splice acceptor site induced 45% to 68% exon 2-skipped Dmd transcripts in Dup2 skeletal muscles 15 days post-injection. Muscle dystrophin restoration peaked at 77% to 87% average dystrophin-positive fibers and 41% to 51% of normal signal intensity by immunofluorescence, and 15.7% to 56.8% of normal by western blotting 15 to 30 days after treatment. These findings indicate that PPMO-mediated exon skipping is a promising therapeutic strategy for muscle dystrophin restoration in the context of exon 2 duplications.

2.
Sci Rep ; 10(1): 6702, 2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32317649

RESUMEN

Pompe disease is caused by mutations in the GAA gene, resulting in deficient lysosomal acid-α-glucosidase activity in patients, and a progressive decline in mobility and respiratory function. Enzyme replacement therapy is one therapeutic option, but since not all patients respond to this treatment, alternative interventions should be considered. One GAA mutation, c.-32-13T > G, impacts upon normal exon 2 splicing and is found in two-thirds of late-onset cases. We and others have explored a therapeutic strategy using splice modulating phosphorodiamidate morpholino oligomers to enhance GAA exon 2 inclusion in the mature mRNA of patients with one c.-32-13T > G allele. We designed 20 oligomers and treated fibroblasts derived from five patients to identify an oligomer sequence that maximally increased enzyme activity in all fibroblasts. The most effective splice correcting oligomer was chosen to treat forced-myogenic cells, derived from fibroblasts from nine patients carrying the c.-32-13T > G mutation. After transfection, we show increased levels of the full-length GAA transcript, acid-α-glucosidase protein, and enzyme activity in all patients' myogenic cells, regardless of the nature of the mutation in the other allele. This data encourages the initiation of clinical trials to assess the therapeutic efficacy of this oligomer for those patients carrying the c.-32-13T > G mutation.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Oligonucleótidos Antisentido/farmacología , Empalme del ARN/genética , alfa-Glucosidasas/metabolismo , Edad de Inicio , Estudios de Casos y Controles , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Humanos , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , alfa-Glucosidasas/genética
3.
Neurology ; 94(21): e2270-e2282, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32139505

RESUMEN

OBJECTIVE: To report safety, pharmacokinetics, exon 53 skipping, and dystrophin expression in golodirsen-treated patients with Duchenne muscular dystrophy (DMD) amenable to exon 53 skipping. METHODS: Part 1 was a randomized, double-blind, placebo-controlled, 12-week dose titration of once-weekly golodirsen; part 2 is an ongoing, open-label evaluation. Safety and pharmacokinetics were primary and secondary objectives of part 1. Primary biological outcome measures of part 2 were blinded exon skipping and dystrophin protein production on muscle biopsies (baseline, week 48) evaluated, respectively, using reverse transcription PCR and Western blot and immunohistochemistry. RESULTS: Twelve patients were randomized to receive golodirsen (n = 8) or placebo (n = 4) in part 1. All from part 1 plus 13 additional patients received 30 mg/kg golodirsen in part 2. Safety findings were consistent with those previously observed in pediatric patients with DMD. Most of the study drug was excreted within 4 hours following administration. A significant increase in exon 53 skipping was associated with ∼16-fold increase over baseline in dystrophin protein expression at week 48, with a mean percent normal dystrophin protein standard of 1.019% (range, 0.09%-4.30%). Sarcolemmal localization of dystrophin was demonstrated by significantly increased dystrophin-positive fibers (week 48, p < 0.001) and a positive correlation (Spearman r = 0.663; p < 0.001) with dystrophin protein change from baseline, measured by Western blot and immunohistochemistry. CONCLUSION: Golodirsen was well-tolerated; muscle biopsies from golodirsen-treated patients showed increased exon 53 skipping, dystrophin production, and correct dystrophin sarcolemmal localization. CLINICALTRIALSGOV IDENTIFIER: NCT02310906. CLASSIFICATION OF EVIDENCE: This study provides Class I evidence that golodirsen is safe and Class IV evidence that it induces exon skipping and novel dystrophin as confirmed by 3 different assays.


Asunto(s)
Distrofina/biosíntesis , Distrofia Muscular de Duchenne/tratamiento farmacológico , Oligonucleótidos/uso terapéutico , Administración Intravenosa , Adolescente , Niño , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Distrofina/genética , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/sangre , Distrofia Muscular de Duchenne/genética , Eliminación de Secuencia/efectos de los fármacos
4.
Medicine (Baltimore) ; 98(26): e15858, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31261494

RESUMEN

This analysis aims to describe the outcomes of two nonambulatory patients with Duchenne muscular dystrophy (DMD) who participated in two clinical studies. The two consecutive trials of eteplirsen (studies 201 and 202) were conducted in patients with DMD (N = 12) and confirmed genetic mutations amenable to exon 51 skipping.In study 201, 12 patients were randomized to receive once-weekly, double-blind intravenous infusions of eteplirsen 30 or 50 mg/kg or placebo for 24 weeks; patients then received open-label eteplirsen during weeks 25 through 28. All 12 patients continued onto open-label extension study 202 and received long-term treatment with eteplirsen. We compared cardiac, pulmonary, and upper limb function and dystrophin production in the nonambulatory twin patients versus the 10 ambulatory patients through 240 combined treatment weeks.Ten study patients remained ambulatory through both studies, while the identical twin patients both experienced early, rapid loss of ambulation. The twin patients had greater disease severity at baseline (6-minute walk test [6MWT], 330 and 256 m) versus the other patients (n = 10; 6MWT range, 341-418 m). They maintained cardiac and upper limb function through combined week 240, with outcomes similar to those of the patients who remained ambulatory. Dystrophin production was confirmed following eteplirsen treatment.Despite the loss of ambulation, other markers of disease progression remained relatively stable in the eteplirsen-treated twin patients and were similar to those of the ambulatory patients.


Asunto(s)
Morfolinos/uso terapéutico , Distrofia Muscular de Duchenne/tratamiento farmacológico , Niño , Progresión de la Enfermedad , Enfermedades en Gemelos , Método Doble Ciego , Distrofina/genética , Distrofina/metabolismo , Humanos , Masculino , Morfolinos/efectos adversos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatología , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Prueba de Paso , Caminata
5.
Hum Mol Genet ; 28(18): 3091-3100, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31179493

RESUMEN

Duchenne muscular dystrophy (DMD) is a rare genetic disease affecting 1 in 3500-5000 newborn boys. It is due to mutations in the DMD gene with a consequent lack of dystrophin protein that leads to deterioration of myofibres and their replacement with fibro-adipogenic tissue. Out-of-frame mutations in the DMD gene can be modified by using antisense oligonucleotides (AONs) to promote skipping of specific exons such that the reading frame is restored and the resulting protein produced, though truncated, is functional. We have shown that AONs can also be used to knock down myostatin, a negative regulator of muscle growth and differentiation, through disruption of the transcript reading frame, and thereby enhance muscle strength. In young mdx mice, combined dystrophin and myostatin exon skipping therapy greatly improved DMD pathology, compared to the single dystrophin skipping approach. Here we show that in aged (>15-month-old) mdx mice, when the pathology is significantly more severe and more similar to the one observed in DMD patients, the effect of the combined therapy is slightly attenuated but still beneficial in improving the disease phenotype. These results confirm the beneficial outcome of the combination approach and support its translation into DMD clinical trials.


Asunto(s)
Distrofina/genética , Distrofina/metabolismo , Exones , Regulación de la Expresión Génica , Músculos/metabolismo , Miostatina/genética , Miostatina/metabolismo , Empalme del ARN , Animales , Modelos Animales de Enfermedad , Inmunohistoquímica , Ratones , Ratones Endogámicos mdx , Músculos/patología , Músculos/fisiopatología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , ARN Mensajero/genética
6.
J Neuromuscul Dis ; 5(4): 407-417, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30198876

RESUMEN

A new line of dystrophic mdx mice on the DBA/2J (D2) background has emerged as a candidate to study the efficacy of therapeutic approaches for Duchenne muscular dystrophy (DMD). These mice harbor genetic polymorphisms that appear to increase the severity of the dystropathology, with disease modifiers that also occur in DMD patients, making them attractive for efficacy studies and drug development. This workshop aimed at collecting and consolidating available data on the pathological features and the natural history of these new D2/mdx mice, for comparison with classic mdx mice and controls, and to identify gaps in information and their potential value. The overall aim is to establish guidance on how to best use the D2/mdx mouse model in preclinical studies.


Asunto(s)
Modelos Animales de Enfermedad , Distrofia Muscular Animal , Distrofia Muscular de Duchenne , Animales , Ratones , Ratones Endogámicos DBA , Ratones Endogámicos mdx
7.
Neurology ; 90(24): e2146-e2154, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29752304

RESUMEN

OBJECTIVE: To describe the quantification of novel dystrophin production in patients with Duchenne muscular dystrophy (DMD) after long-term treatment with eteplirsen. METHODS: Clinical study 202 was an observational, open-label extension of the randomized, controlled study 201 assessing the safety and efficacy of eteplirsen in patients with DMD with a confirmed mutation in the DMD gene amenable to correction by skipping of exon 51. Patients received once-weekly IV doses of eteplirsen 30 or 50 mg/kg. Upper extremity muscle biopsy samples were collected at combined study week 180, blinded, and assessed for dystrophin-related content by Western blot, Bioquant software measurement of dystrophin-associated immunofluorescence intensity, and percent dystrophin-positive fibers (PDPF). Results were contrasted with matched untreated biopsies from patients with DMD. Reverse transcription PCR followed by Sanger sequencing of newly formed slice junctions was used to confirm the mechanism of action of eteplirsen. RESULTS: Reverse transcription PCR analysis and sequencing of the newly formed splice junction confirmed that 100% of treated patients displayed the expected skipped exon 51 sequence. In treated patients vs untreated controls, Western blot analysis of dystrophin content demonstrated an 11.6-fold increase (p = 0.007), and PDPF analysis demonstrated a 7.4-fold increase (p < 0.001). The PDPF findings were confirmed in a re-examination of the sample (15.5-fold increase, p < 0.001). Dystrophin immunofluorescence intensity was 2.4-fold greater in treated patients than in untreated controls (p < 0.001). CONCLUSION: Taken together, the 4 assays, each based on unique evaluation mechanisms, provided evidence of eteplirsen muscle cell penetration, exon skipping, and induction of novel dystrophin expression. CLASSIFICATION OF EVIDENCE: This study provides Class II evidence of the muscle cell penetration, exon skipping, and induction of novel dystrophin expression by eteplirsen, as confirmed by 4 assays.


Asunto(s)
Distrofina/biosíntesis , Exones/genética , Morfolinos/uso terapéutico , Distrofia Muscular de Duchenne/tratamiento farmacológico , Biopsia , Niño , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Resultado del Tratamiento
8.
PLoS One ; 11(2): e0149582, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26915099

RESUMEN

T cells have the remarkable ability to recognize antigen with great specificity and in turn mount an appropriate and robust immune response. Critical to this process is the initial T cell antigen recognition and subsequent signal transduction events. This antigen recognition can be modulated at the site of TCR interaction with peptide:major histocompatibility (pMHC) or peptide interaction with the MHC molecule. Both events could have a range of effects on T cell fate. Though responses to antigens that bind sub-optimally to TCR, known as altered peptide ligands (APL), have been studied extensively, the impact of disrupting antigen binding to MHC has been highlighted to a lesser extent and is usually considered to result in complete loss of epitope recognition. Here we present a model of viral evasion from CD8 T cell immuno-surveillance by a lymphocytic choriomeningitis virus (LCMV) escape mutant with an epitope for which TCR affinity for pMHC remains high but where the antigenic peptide binds sub optimally to MHC. Despite high TCR affinity for variant epitope, levels of interferon regulatory factor-4 (IRF4) are not sustained in response to the variant indicating differences in perceived TCR signal strength. The CD8+ T cell response to the variant epitope is characterized by early proliferation and up-regulation of activation markers. Interestingly, this response is not maintained and is characterized by a lack in IL-2 and IFNγ production, increased apoptosis and an abrogated glycolytic response. We show that disrupting the stability of peptide in MHC can effectively disrupt TCR signal strength despite unchanged affinity for TCR and can significantly impact the CD8+ T cell response to a viral escape mutant.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Mutación , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Linfocitos T CD8-positivos/citología , Proliferación Celular , Supervivencia Celular , Regulación de la Expresión Génica , Antígenos de Histocompatibilidad/metabolismo , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Activación de Linfocitos , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL , Monitorización Inmunológica , Transducción de Señal
9.
Nucleic Acid Ther ; 24(2): 114-26, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24494586

RESUMEN

Activated and regulatory T cells express the negative co-stimulatory molecule cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) that binds B7 on antigen-presenting cells to mediate cellular responses. Single nucleotide polymorphisms in the CTLA-4 gene have been found to affect alternative splicing and are linked to autoimmune disease susceptibility or resistance. Increased expression of a soluble splice form (sCTLA-4), lacking the transmembrane domain encoded by exon 3, has been shown to accelerate autoimmune pathology. In contrast, an exon 2-deficient form lacking the B7 ligand binding domain (liCTLA-4), expressed by diabetes resistant mouse strains has been shown to be protective when expressed as a transgene in diabetes susceptible non-obese diabetic (NOD) mice. We sought to employ an antisense-targeted splice-switching approach to independently produce these CTLA-4 splice forms in NOD mouse T cells and observe their relative impact on spontaneous autoimmune diabetes susceptibility. In vitro antisense targeting of the splice acceptor site for exon 2 produced liCTLA-4 while targeting exon 3 produced the sCTLA-4 form in NOD T cells. The liCTLA-4 expressing T cells exhibited reduced activation, proliferation and increased adhesion to intercellular adhesion molecule-1 (ICAM-1) similar to treatment with agonist α-CTLA-4. Mice treated to produce liCTLA-4 at the time of elevated blood glucose levels exhibited a significant reduction in the incidence of insulitis and diabetes, whereas a marked increase in the incidence of both was observed in animals treated to produce sCTLA-4. These findings provide further support that alternative splice forms of CTLA-4 affects diabetes susceptibility in NOD mice and demonstrates the therapeutic utility of antisense mediated splice-switching for modulating immune responses.


Asunto(s)
Autoinmunidad/genética , Antígeno CTLA-4/genética , Diabetes Mellitus Tipo 1/genética , Susceptibilidad a Enfermedades/inmunología , Oligonucleótidos Antisentido/genética , Abatacept , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/inmunología , Adhesión Celular , Proliferación Celular , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Exones , Regulación de la Expresión Génica , Inmunoconjugados/farmacología , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos NOD , Datos de Secuencia Molecular , Oligonucleótidos Antisentido/inmunología , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Índice de Severidad de la Enfermedad , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
10.
J Infect Dis ; 208(10): 1553-60, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24130069

RESUMEN

BACKGROUND: Peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs) are synthetic DNA/RNA analogues that silence expression of specific genes. We studied whether PPMOs targeted to essential genes in Acinetobacter lwoffii and Acinetobacter baumannii are active in vitro and in vivo. METHODS: PPMOs were evaluated in vitro using minimum inhibitory concentration (MIC) and viability assays, and in vivo using murine pulmonary infection models with intranasal PPMO treatment. RESULTS: MICs of PPMOs ranged from 0.1 to 64 µM (approximately 0.6-38 µg/mL). The most effective PPMO tested was (RXR)4-AcpP, which is targeted to acpP. (RXR)4-AcpP reduced viability of A. lwoffii and A. baumannii by >10(3) colony-forming units/mL at 5-8 times MIC. Mice treated with ≥0.25 mg/kg of (RXR)4-AcpP survived longer and had less inflammation and bacterial lung burden than mice treated with a scrambled-sequence PPMO or phosphate-buffered saline. Treatment could be delayed after infection and still increase survival. CONCLUSIONS: PPMOs targeted to essential genes of A. lwoffii and A. baumannii were bactericidal and had MICs in a clinically relevant range. (RXR)4-AcpP increased survival of mice infected with A. lwoffii or A. baumannii, even when initial treatment was delayed after infection. PPMOs could be a viable therapeutic approach in dealing with multidrug-resistant Acinetobacter species.


Asunto(s)
Acinetobacter/efectos de los fármacos , Acinetobacter/genética , Silenciador del Gen , Morfolinos/farmacología , Oligonucleótidos Antisentido/genética , Acinetobacter/crecimiento & desarrollo , Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/mortalidad , Infecciones por Acinetobacter/terapia , Animales , Antibacterianos/farmacología , Modelos Animales de Enfermedad , Femenino , Ratones , Pruebas de Sensibilidad Microbiana , Morfolinos/administración & dosificación , Morfolinos/química , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos Antisentido/química , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/mortalidad , Neumonía Bacteriana/terapia
11.
Biores Open Access ; 2(1): 61-6, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23515175

RESUMEN

Phosphorodiamidate morpholino oligomers (PMO) are neutrally charged, sequence-specific antisense agents that interfere with targeted gene expression. PMO have been shown to be highly specific and potent therapies after cellular uptake, yet methods to detect PMO in tissue and inside the cell are limited. We offer in this report novel methods for the detection of cellular resident PMO using flow cytometry-fluorescence in situ hybridization (flow FISH) and a sandwich hybridization technique to quickly and sensitively quantify tissue resident PMO. These methods rely on oligonucleotide probes complementary to a PMO to specifically detect and quantify cell-associated and tissue resident PMO after in vitro or in vivo administration. Using the sandwich hybridization technique, we show that intranasally delivered PMO demonstrates zero-order clearance kinetics from the lung. Furthermore, PMO was detected in nonhematopoietic and hematopoietic cells of the lung regardless of influenza virus infection, although an increase in PMO uptake in infected hematopoietic cells was observed. Coincident measurement of target knock-down to cell-associated influenza A PMO concentration allowed for the calculation of an EC50.

12.
PLoS Pathog ; 8(12): e1003073, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23300439

RESUMEN

The viral family Arenaviridae includes a number of viruses that can cause hemorrhagic fever in humans. Arenavirus infection often involves multiple organs and can lead to capillary instability, impaired hemostasis, and death. Preclinical testing for development of antiviral or therapeutics is in part hampered due to a lack of an immunologically well-defined rodent model that exhibits similar acute hemorrhagic illness or sequelae compared to the human disease. We have identified the FVB mouse strain, which succumbs to a hemorrhagic fever-like illness when infected with lymphocytic choriomeningitis virus (LCMV). FVB mice infected with LCMV demonstrate high mortality associated with thrombocytopenia, hepatocellular and splenic necrosis, and cutaneous hemorrhage. Investigation of inflammatory mediators revealed increased IFN-γ, IL-6 and IL-17, along with increased chemokine production, at early times after LCMV infection, which suggests that a viral-induced host immune response is the cause of the pathology. Depletion of T cells at time of infection prevented mortality in all treated animals. Antisense-targeted reduction of IL-17 cytokine responsiveness provided significant protection from hemorrhagic pathology. F1 mice derived from FVB×C57BL/6 mating exhibit disease signs and mortality concomitant with the FVB challenged mice, extending this model to more widely available immunological tools. This report offers a novel animal model for arenavirus research and pre-clinical therapeutic testing.


Asunto(s)
Antivirales/uso terapéutico , Fiebres Hemorrágicas Virales/tratamiento farmacológico , Coriomeningitis Linfocítica/tratamiento farmacológico , Virus de la Coriomeningitis Linfocítica/efectos de los fármacos , Morfolinos/uso terapéutico , Animales , Antivirales/farmacología , Modelos Animales de Enfermedad , Interferón gamma/sangre , Interleucina-17/sangre , Interleucina-6/sangre , Hígado/patología , Hígado/virología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Ratones , Ratones Endogámicos C57BL , Morfolinos/farmacología , Bazo/patología , Bazo/virología , Trombocitopenia/virología , Replicación Viral
13.
J Immunol ; 182(4): 1829-35, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19201834

RESUMEN

One mechanism viruses use to subvert immune surveillance is through mutation of MHC contact residues of antigenic epitopes that weaken T cell recognition to the point that the immune system is ignorant of the infection. However, in contrast to ignorance, results presented herein demonstrate that intracellular signaling does occur upon stimulation with a lymphocytic choriomeningitis virus-derived escape mutant as demonstrated by the sustained activation of Src homology 2 domain-containing protein tyrosine phosphatase (SHP-1). In addition to the increased SHP-1 activity, we found that the mutated epitope failed to induce oxidation of SHP-1, further enhancing enzymatic activity. Sustained activation of SHP-1 in a reduced form correlated with ERK and early growth response gene 1 activation and failure of T cells to commit to the effector lineage. Thus, instead of immune ignorance, these studies demonstrate the activation of a negative signaling pathway that actively suppresses T cell responses and limits recognition of viral escape mutants.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Activación de Linfocitos/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Animales , Antígenos Virales/inmunología , Ratones , Reacción en Cadena de la Polimerasa
14.
Immunology ; 121(2): 207-15, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17250584

RESUMEN

During mammalian pregnancy the maternal thymus undergoes significant involution, and then recovers in size after birth. The mechanism behind this involution is not known, but it has been suggested that elevated levels of hormones during pregnancy induce the involution. We have recently shown that injection of 17beta-oestradiol into mice causes loss of early thymocyte precursors and inhibits proliferation of developing thymocytes. This suggests that elevated oestrogen in pregnancy may contribute to thymic involution. We have investigated this idea by examining the fate of thymocytes during mouse pregnancy in much greater detail than has been previously reported. Looking over a broad time-course, we find that pregnancy does not affect thymocyte precursor populations in the bone marrow, but induces a profound loss of early thymic progenitors in the thymus as early as day 12 x 5 of pregnancy. This loss is accompanied by decreased thymocyte proliferation, which returns to normal 2-4 days postpartum. No enhancement of apoptosis is detectable at any stage of pregnancy. We also find that there is a reduction in recent thymic emigrants after oestrogen treatment and at day 17 x 5 of pregnancy, suggesting that thymic involution during pregnancy influences the peripheral T-cell repertoire. The similarities between oestrogen-mediated involution and pregnancy-mediated involution suggest that oestrogen is a significant contributor to loss of thymocyte cellularity during pregnancy, and probably functions primarily by reducing thymocyte proliferation.


Asunto(s)
Embarazo/inmunología , Linfocitos T/inmunología , Timo/inmunología , Animales , Apoptosis , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Estradiol/sangre , Estradiol/farmacología , Femenino , Citometría de Flujo , Subunidad alfa del Receptor de Interleucina-2/análisis , Masculino , Ratones , Ratones Endogámicos C57BL , Células Madre/inmunología , Timo/efectos de los fármacos
15.
J Immunol ; 176(8): 4740-7, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16585567

RESUMEN

The size of the thymus can be greatly influenced by changes in the small number of early progenitors in the thymus. However, it is not known whether thymic cellularity feeds back to regulate the recruitment, survival, and expansion of progenitors. The transcription factor early growth response gene 1 (Egr1) has been implicated in controlling proliferation and survival in many cell types. We have previously shown that mice deficient in Egr1 have increased thymic cellularity. We now show that Egr1 regulates a negative feedback signal that controls the entry of cells into the thymus. Egr1-deficient mice have higher percentages of early T lineage progenitors in the thymus, yet Egr1-deficient mice have normal numbers of myelolymphoid progenitors in the bone marrow, and Egr1-deficient thymocytes show normal rates of apoptosis and proliferation at all stages of development. Evidence from mixed bone marrow chimeras shows that the ability of Egr1 to control progenitor recruitment is mediated by bone marrow-derived cells, but is not cell autonomous. Furthermore, Egr1-deficient thymuses have increased P-selectin expression. The data suggest that Egr1 mediates a feedback mechanism whereby the number of resident double negative thymocytes controls the entry of new progenitors into the thymus by regulating P-selectin expression on thymic endothelial cells.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Linfocitos T/citología , Linfocitos T/inmunología , Timo/citología , Timo/inmunología , Animales , Movimiento Celular , Proteína 1 de la Respuesta de Crecimiento Precoz/deficiencia , Proteína 1 de la Respuesta de Crecimiento Precoz/inmunología , Retroalimentación , Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Selectina-P/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
16.
Proc Natl Acad Sci U S A ; 102(46): 16735-40, 2005 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16267136

RESUMEN

The bone wasting induced by estrogen deficiency is, in part, a consequence of increased T cell production of the osteoclastogenic cytokine TNF-alpha. This phenomenon is due to an expansion of T cells, but the responsible mechanism is unknown. We now show that ovariectomy (ovx) disregulates T lymphopoiesis and induces bone loss by stimulating, through a rise in IL-7 levels, both thymic-dependent differentiation of bone marrow-derived progenitors and thymic-independent, peripheral expansion of mature T cells. Attesting to the relevance of the thymic effects, thymectomy decreases by approximately 50% the bone loss and the stimulation of T lymphopoiesis induced by ovx. In contrast, in vivo attenuation of the elevated IL-7 completely prevents the stimulation of T lymphopoiesis and the bone loss that follow ovx. Thus, the disruption of both T cell and bone homeostasis induced by ovx is mediated by IL-7 and due to both the thymic and extrathymic mechanisms. We conclude that IL-7 is a pivotal upstream target through which estrogen regulates hematopoietic and immune functions that are critical for bone homeostasis.


Asunto(s)
Estrógenos/deficiencia , Interleucina-7/fisiología , Osteoporosis/etiología , Linfocitos T/citología , Timo/citología , Animales , Secuencia de Bases , Cartilla de ADN , Femenino , Homeostasis , Interleucina-7/biosíntesis , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Osteoporosis/inmunología , Ovariectomía , Linfocitos T/inmunología , Timo/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis
17.
J Immunol ; 175(4): 2270-7, 2005 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16081795

RESUMEN

Early growth response gene 1 (Egr1) is a transcriptional regulator whose expression can be induced by multiple signals including the TCR. Egr1 has been shown to promote positive selection, but an investigation of its role in T cell homeostasis has not been reported. The possibility that similar signals control both positive selection and peripheral T cell homeostasis led us to investigate the role of Egr1 in the maintenance of peripheral T cells. We have found that on TCR transgenic backgrounds, Egr1-deficient mice have a reduction in their number of naive T cells. Although Egr1-deficient animals have a low percentage of mature thymocytes due to inefficient positive selection, the absolute number of mature thymocytes is only slightly reduced due to increased thymus size in Egr1-deficient mice. Despite possessing near normal numbers of mature thymocytes, we find that Egr1-deficient mice have poor accumulation of recent thymic emigrants (RTE) in the periphery. The poor accumulation of RTE in Egr1-deficient mice appears to originate from decreased survival of mature thymocytes and RTE, which we have observed both in vitro and in vivo. These findings suggest that an Egr1-mediated signal during positive selection promotes not only the production of single positive thymocytes, but also the survival of selected thymocytes until they can become established in the periphery.


Asunto(s)
Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/citología , Timo/citología , Animales , Agregación Celular/genética , Agregación Celular/inmunología , Diferenciación Celular/genética , Movimiento Celular/genética , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Proteína 1 de la Respuesta de Crecimiento Precoz/deficiencia , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Homeostasis/genética , Homeostasis/inmunología , Recuento de Linfocitos , Linfopenia/genética , Linfopenia/inmunología , Linfopenia/patología , Ratones , Ratones Endogámicos AKR , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética , Fase de Descanso del Ciclo Celular/genética , Fase de Descanso del Ciclo Celular/inmunología , Transducción de Señal/genética , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Timo/inmunología , Timo/metabolismo , Timo/patología
18.
J Urol ; 170(5): 1864-7, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14532794

RESUMEN

PURPOSE: We report on the relationship of patient age to prostate specific antigen (PSA) bounce after brachytherapy for prostate cancer. MATERIALS AND METHODS: From 1992 to 1997, 1,011 consecutive men with stage T1-T2NxM0 prostate cancer were treated with simultaneous irradiation, transperineal prostate I-125 implant followed by external beam irradiation. No patients received neoadjuvant hormones. There were 251, 491 and 269 men 60 years old or younger, 61 to 70 and 71 or older, respectively, at implant. PSA bounce is defined by a postirradiation PSA increase of 0.1 ng/ml or more above the level before bounce followed by a decrease to or below that level. Disease freedom is defined by a PSA cut point of 0.2 ng/ml. Median followup is 6 years (range 3 months to 10 years). RESULTS: The frequency of PSA bounce according to age 60 or younger, 61 to 70 and 71 or older is 57%, 41% and 26%, respectively, a significant difference (p <0.000), average time to bounce onset is 19, 20 and 25 months for these 3 age groups, respectively, a significant difference (p = 0.002), and average bounce duration is 11, 8 and 8 months, respectively, a significant difference (p <0.000). On multivariate analysis of age, pretreatment PSA, Gleason score, stage, implant dose, prostate volume, diabetes and race, only age is significantly associated with PSA bounce (p <0.0001). After adjusting for the inherent bias caused by PSA bounce, there is no significant difference in disease-free survival between men with and without bounce. CONCLUSIONS: Young men with prostate cancer treated with brachytherapy have a significantly higher frequency, earlier onset and longer duration of PSA bounce than older men.


Asunto(s)
Biomarcadores de Tumor/sangre , Braquiterapia , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/radioterapia , Radioterapia , Factores de Edad , Anciano , Terapia Combinada , Diagnóstico Diferencial , Supervivencia sin Enfermedad , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/sangre , Recurrencia Local de Neoplasia/diagnóstico , Estadificación de Neoplasias , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/patología , Dosificación Radioterapéutica , Estudios Retrospectivos
19.
J Biol Chem ; 277(12): 10028-36, 2002 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-11792697

RESUMEN

Recent studies have demonstrated that CD47 plays an important role in regulating human neutrophil (PMN) chemotaxis. Two ligands for CD47, thrombospondin and SIRPalpha, have been described. However, it is not known if SIRP-CD47 interactions play a role in regulating PMN migration. In this study, we show that SIRPalpha1 directly binds to the immunoglobulin variable domain loop of purified human CD47 and that such SIRP-CD47 interactions regulate PMN transmigration. Specifically, PMN migration across both human epithelial monolayers and collagen-coated filters was partially inhibited by anti-SIRP monoclonal antibodies. Similar kinetics of inhibition were observed for PMN transmigration in the presence of soluble, recombinant CD47 consisting of the SIRP-binding loop. In contrast, anti-CD47 monoclonal antibodies inhibited PMN transmigration by markedly different kinetics. Results of signal transduction experiments suggested differential regulation of PMN migration by SIRP versus CD47 by phosphatidylinositol 3-kinase and tyrosine kinases, respectively. Immunoprecipitation followed by Western blotting after SDS-PAGE under nonreducing conditions suggested that several SIRP protein species may be present in PMN. Stimulation of PMN with fMLP resulted in increased surface expression of these SIRP proteins, consistent with the existence of intracellular pools. Taken together, these results demonstrate that PMN migration is regulated by CD47 through SIRPalpha-dependent and SIRPalpha-independent mechanisms.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación , Proteínas Portadoras/metabolismo , Glicoproteínas de Membrana/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neutrófilos/citología , Neutrófilos/metabolismo , Receptores Inmunológicos , Fosfatasa Alcalina/metabolismo , Androstadienos/farmacología , Anticuerpos Monoclonales/metabolismo , Western Blotting , Antígenos CD18/metabolismo , Antígeno CD47 , Adhesión Celular , Movimiento Celular , Células Cultivadas , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Genisteína/farmacología , Humanos , Cinética , Ligandos , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/metabolismo , Factores de Tiempo , Wortmanina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...