Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(19)2023 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-37834003

RESUMEN

The NOTCH ligands JAG1 and JAG2 have been correlated in vitro with multiple myeloma (MM) cell proliferation, drug resistance, self-renewal and a pathological crosstalk with the tumor microenvironment resulting in angiogenesis and osteoclastogenesis. These findings suggest that a therapeutic approach targeting JAG ligands might be helpful for the care of MM patients and lead us to explore the role of JAG1 and JAG2 in a MM in vivo model and primary patient samples. JAG1 and JAG2 protein expression represents a common feature in MM cell lines; therefore, we assessed their function through JAG1/2 conditional silencing in a MM xenograft model. We observed that JAG1 and JAG2 showed potential as therapeutic targets in MM, as their silencing resulted in a reduction in the tumor burden. Moreover, JAG1 and JAG2 protein expression in MM patients was positively correlated with the presence of MM cells in patients' bone marrow biopsies. Finally, taking advantage of the Multiple Myeloma Research Foundation (MMRF) CoMMpass global dataset, we showed that JAG2 gene expression level was a predictive biomarker associated with patients' overall survival and progression-free survival, independently from other main molecular or clinical features. Overall, these results strengthened the rationale for the development of a JAG1/2-tailored approach and the use of JAG2 as a predictive biomarker in MM.


Asunto(s)
Mieloma Múltiple , Humanos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Receptores Notch/metabolismo , Biomarcadores , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Ligandos , Microambiente Tumoral
2.
Proc Natl Acad Sci U S A ; 110(3): 1041-6, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23275297

RESUMEN

The molecular etiology of human progenitor reprogramming into self-renewing leukemia stem cells (LSC) has remained elusive. Although DNA sequencing has uncovered spliceosome gene mutations that promote alternative splicing and portend leukemic transformation, isoform diversity also may be generated by RNA editing mediated by adenosine deaminase acting on RNA (ADAR) enzymes that regulate stem cell maintenance. In this study, whole-transcriptome sequencing of normal, chronic phase, and serially transplantable blast crisis chronic myeloid leukemia (CML) progenitors revealed increased IFN-γ pathway gene expression in concert with BCR-ABL amplification, enhanced expression of the IFN-responsive ADAR1 p150 isoform, and a propensity for increased adenosine-to-inosine RNA editing during CML progression. Lentiviral overexpression experiments demonstrate that ADAR1 p150 promotes expression of the myeloid transcription factor PU.1 and induces malignant reprogramming of myeloid progenitors. Moreover, enforced ADAR1 p150 expression was associated with production of a misspliced form of GSK3ß implicated in LSC self-renewal. Finally, functional serial transplantation and shRNA studies demonstrate that ADAR1 knockdown impaired in vivo self-renewal capacity of blast crisis CML progenitors. Together these data provide a compelling rationale for developing ADAR1-based LSC detection and eradication strategies.


Asunto(s)
Adenosina Desaminasa/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Adenosina Desaminasa/genética , Empalme Alternativo , Animales , Crisis Blástica/etiología , Crisis Blástica/genética , Crisis Blástica/metabolismo , Crisis Blástica/patología , Transformación Celular Neoplásica , Progresión de la Enfermedad , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Mediadores de Inflamación/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mieloide de Fase Crónica/genética , Leucemia Mieloide de Fase Crónica/metabolismo , Leucemia Mieloide de Fase Crónica/patología , Ratones , Edición de ARN , Proteínas de Unión al ARN , Transcriptoma , Trasplante Heterólogo , Ensayo de Tumor de Célula Madre
3.
J Transl Med ; 13: 52, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25889244

RESUMEN

BACKGROUND: Deregulation of RNA editing by adenosine deaminases acting on dsRNA (ADARs) has been implicated in the progression of diverse human cancers including hematopoietic malignancies such as chronic myeloid leukemia (CML). Inflammation-associated activation of ADAR1 occurs in leukemia stem cells specifically in the advanced, often drug-resistant stage of CML known as blast crisis. However, detection of cancer stem cell-associated RNA editing by RNA sequencing in these rare cell populations can be technically challenging, costly and requires PCR validation. The objectives of this study were to validate RNA editing of a subset of cancer stem cell-associated transcripts, and to develop a quantitative RNA editing fingerprint assay for rapid detection of aberrant RNA editing in human malignancies. METHODS: To facilitate quantification of cancer stem cell-associated RNA editing in exons and intronic or 3'UTR primate-specific Alu sequences using a sensitive, cost-effective method, we established an in vitro RNA editing model and developed a sensitive RNA editing fingerprint assay that employs a site-specific quantitative PCR (RESSq-PCR) strategy. This assay was validated in a stably-transduced human leukemia cell line, lentiviral-ADAR1 transduced primary hematopoietic stem and progenitor cells, and in primary human chronic myeloid leukemia stem cells. RESULTS: In lentiviral ADAR1-expressing cells, increased RNA editing of MDM2, APOBEC3D, GLI1 and AZIN1 transcripts was detected by RESSq-PCR with improved sensitivity over sequencing chromatogram analysis. This method accurately detected cancer stem cell-associated RNA editing in primary chronic myeloid leukemia samples, establishing a cancer stem cell-specific RNA editing fingerprint of leukemic transformation that will support clinical development of novel diagnostic tools to predict and prevent cancer progression. CONCLUSIONS: RNA editing quantification enables rapid detection of malignant progenitors signifying cancer progression and therapeutic resistance, and will aid future RNA editing inhibitor development efforts.


Asunto(s)
Reprogramación Celular , Células Madre Neoplásicas/patología , Edición de ARN/genética , Adenosina Desaminasa/metabolismo , Biomarcadores de Tumor/metabolismo , Crisis Blástica/patología , Técnicas de Cocultivo , Progresión de la Enfermedad , Humanos , Células K562 , Lentivirus/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Modelos Biológicos , Reproducibilidad de los Resultados
4.
J Biol Chem ; 287(38): 31691-702, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22833673

RESUMEN

Parkinson disease is characterized by the loss of dopaminergic neurons mainly in the substantia nigra. Accumulation of α-synuclein and cell loss has been also reported in many other brain regions including the hippocampus, where it might impair adult neurogenesis, contributing to nonmotor symptoms. However, the molecular mechanisms of these alterations are still unknown. In this report we show that α-synuclein-accumulating adult rat hippocampus neural progenitors present aberrant neuronal differentiation, with reduction of Notch1 expression and downstream signaling targets. We characterized a Notch1 proximal promoter that contains p53 canonical response elements. In vivo binding of p53 represses the transcription of Notch1 in neurons. Moreover, we demonstrated that α-synuclein directly binds to the DNA at Notch1 promoter vicinity and also interacts with p53 protein, facilitating or increasing Notch1 signaling repression, which interferes with maturation and survival of neural progenitors cells. This study provides a molecular basis for α-synuclein-mediated disruption of adult neurogenesis in Parkinson disease.


Asunto(s)
Regulación de la Expresión Génica , Neurogénesis , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Receptor Notch1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , alfa-Sinucleína/metabolismo , Animales , Apoptosis , Linaje de la Célula , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Lentivirus/genética , Regiones Promotoras Genéticas , Ratas , Transducción de Señal
5.
Cell Rep Med ; 4(3): 100962, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36889320

RESUMEN

Pediatric acute myeloid leukemia (pAML) is typified by high relapse rates and a relative paucity of somatic DNA mutations. Although seminal studies show that splicing factor mutations and mis-splicing fuel therapy-resistant leukemia stem cell (LSC) generation in adults, splicing deregulation has not been extensively studied in pAML. Herein, we describe single-cell proteogenomics analyses, transcriptome-wide analyses of FACS-purified hematopoietic stem and progenitor cells followed by differential splicing analyses, dual-fluorescence lentiviral splicing reporter assays, and the potential of a selective splicing modulator, Rebecsinib, in pAML. Using these methods, we discover transcriptomic splicing deregulation typified by differential exon usage. In addition, we discover downregulation of splicing regulator RBFOX2 and CD47 splice isoform upregulation. Importantly, splicing deregulation in pAML induces a therapeutic vulnerability to Rebecsinib in survival, self-renewal, and lentiviral splicing reporter assays. Taken together, the detection and targeting of splicing deregulation represent a potentially clinically tractable strategy for pAML therapy.


Asunto(s)
Leucemia Mieloide Aguda , Células Madre , Adulto , Niño , Humanos , Empalme del ARN/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Isoformas de Proteínas/genética , Mutación , Factores de Empalme de ARN/genética , Proteínas Represoras/genética
6.
Cell Stem Cell ; 30(3): 250-263.e6, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36803553

RESUMEN

Adenosine deaminase acting on RNA1 (ADAR1) preserves genomic integrity by preventing retroviral integration and retrotransposition during stress responses. However, inflammatory-microenvironment-induced ADAR1p110 to p150 splice isoform switching drives cancer stem cell (CSC) generation and therapeutic resistance in 20 malignancies. Previously, predicting and preventing ADAR1p150-mediated malignant RNA editing represented a significant challenge. Thus, we developed lentiviral ADAR1 and splicing reporters for non-invasive detection of splicing-mediated ADAR1 adenosine-to-inosine (A-to-I) RNA editing activation; a quantitative ADAR1p150 intracellular flow cytometric assay; a selective small-molecule inhibitor of splicing-mediated ADAR1 activation, Rebecsinib, which inhibits leukemia stem cell (LSC) self-renewal and prolongs humanized LSC mouse model survival at doses that spare normal hematopoietic stem and progenitor cells (HSPCs); and pre-IND studies showing favorable Rebecsinib toxicokinetic and pharmacodynamic (TK/PD) properties. Together, these results lay the foundation for developing Rebecsinib as a clinical ADAR1p150 antagonist aimed at obviating malignant microenvironment-driven LSC generation.


Asunto(s)
Adenosina Desaminasa , Células Madre Hematopoyéticas , Ratones , Animales , Isoformas de Proteínas , Adenosina Desaminasa/genética
7.
Hum Mol Genet ; 19(R1): R12-20, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20413653

RESUMEN

Alzheimer's disease (AD) is characterized by cognitive impairment, progressive neurodegeneration and formation of amyloid-beta (Abeta)-containing plaques and neurofibrillary tangles composed of hyperphosphorylated tau. The neurodegenerative process in AD is initially characterized by synaptic damage accompanied by neuronal loss. In addition, recent evidence suggests that alterations in adult neurogenesis in the hippocampus might play a role. Synaptic loss is one of the strongest correlates to the cognitive impairment in patients with AD. Several lines of investigation support the notion that the synaptic pathology and defective neurogenesis in AD are related to progressive accumulation of Abeta oligomers rather than fibrils. Abnormal accumulation of Abeta resulting in the formation of toxic oligomers is the result of an imbalance between the levels of Abeta production, aggregation and clearance. Abeta oligomers might lead to synaptic damage by forming pore-like structures with channel activity; alterations in glutamate receptors; circuitry hyper-excitability; mitochondrial dysfunction; lysosomal failure and alterations in signaling pathways related to synaptic plasticity, neuronal cell and neurogenesis. A number of signaling proteins, including fyn kinase; glycogen synthase kinase-3beta (GSK3beta) and cyclin-dependent kinase-5 (CDK5), are involved in the neurodegenerative progression of AD. Therapies for AD might require the development of anti-aggregation compounds, pro-clearance pathways and blockers of hyperactive signaling pathways.


Asunto(s)
Enfermedad de Alzheimer/genética , Neurogénesis , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Humanos
8.
Am J Pathol ; 178(4): 1646-61, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21435449

RESUMEN

Recent treatments with highly active antiretroviral therapy (HAART) regimens have been shown to improve general clinical status in patients with human immunodeficiency virus (HIV) infection; however, the prevalence of cognitive alterations and neurodegeneration has remained the same or has increased. These deficits are more pronounced in the subset of HIV patients with the inflammatory condition known as HIV encephalitis (HIVE). Activation of signaling pathways such as GSK3ß and CDK5 has been implicated in the mechanisms of HIV neurotoxicity; however, the downstream mediators of these effects are unclear. The present study investigated the involvement of CDK5 and tau phosphorylation in the mechanisms of neurodegeneration in HIVE. In the frontal cortex of patients with HIVE, increased levels of CDK5 and p35 expression were associated with abnormal tau phosphorylation. Similarly, transgenic mice engineered to express the HIV protein gp120 exhibited increased brain levels of CDK5 and p35, alterations in tau phosphorylation, and dendritic degeneration. In contrast, genetic knockdown of CDK5 or treatment with the CDK5 inhibitor roscovitine improved behavioral performance in the water maze test and reduced neurodegeneration, abnormal tau phosphorylation, and astrogliosis in gp120 transgenic mice. These findings indicate that abnormal CDK5 activation contributes to the neurodegenerative process in HIVE via abnormal tau phosphorylation; thus, reducing CDK5 might ameliorate the cognitive impairments associated with HIVE.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Regulación de la Expresión Génica , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Proteínas tau/metabolismo , Adulto , Animales , Encéfalo/metabolismo , Encéfalo/virología , Femenino , Lóbulo Frontal/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Masculino , Ratones , Persona de Mediana Edad , Enfermedades Neurodegenerativas/virología , Fosfotransferasas/metabolismo , Purinas/farmacología , Roscovitina , Transducción de Señal , Proteínas tau/química
9.
Proc Natl Acad Sci U S A ; 106(31): 13010-5, 2009 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-19651612

RESUMEN

Neuronal accumulation of alpha-synuclein and Lewy body formation are characteristic to many neurodegenerative diseases, including Parkinson's disease (PD). This Lewy pathology appears to spread throughout the brain as the disease progresses. Furthermore, recent studies showed the occurrence of Lewy pathology in neurons grafted into the brains of PD patients, suggesting the spread of pathology from the host tissues to the grafts. The mechanism underlying this propagation is unknown. Here, we show that alpha-synuclein is transmitted via endocytosis to neighboring neurons and neuronal precursor cells, forming Lewy-like inclusions. Moreover, alpha-synuclein was transmitted from the affected neurons to engrafted neuronal precursor cells in a transgenic model of PD-like pathology. Failure of the protein quality control systems, especially lysosomes, promoted the accumulation of transmitted alpha-synuclein and inclusion formation. Cells exposed to neuron-derived alpha-synuclein showed signs of apoptosis, such as nuclear fragmentation and caspase 3 activation, both in vitro and in vivo. These findings demonstrate the cell-to-cell transmission of alpha-synuclein aggregates and provide critical insights into the mechanism of pathological progression in PD and other proteinopathies.


Asunto(s)
Apoptosis , Cuerpos de Lewy/fisiología , Neuronas/fisiología , Transmisión Sináptica , alfa-Sinucleína/metabolismo , Animales , Transporte Biológico , Línea Celular , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ratas , Trasplante de Células Madre , alfa-Sinucleína/toxicidad
10.
Transplant Cell Ther ; 28(8): 446-454, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35605882

RESUMEN

The Blood and Marrow Transplant Clinical Trials Network (BMT CTN) Myeloma Intergroup conducted a workshop on Immune and Cellular Therapy in Multiple Myeloma on January 7, 2022. This workshop included presentations by basic, translational, and clinical researchers with expertise in plasma cell dyscrasias. Four main topics were discussed: platforms for myeloma disease evaluation, insights into pathophysiology, therapeutic target and resistance mechanisms, and cellular therapy for multiple myeloma. Here we provide a comprehensive summary of these workshop presentations.


Asunto(s)
Mieloma Múltiple , Médula Ósea , Tratamiento Basado en Trasplante de Células y Tejidos , Ensayos Clínicos como Asunto , Humanos , Mieloma Múltiple/terapia
11.
J Neurosci ; 30(37): 12252-62, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20844121

RESUMEN

During aging and in the progression of Alzheimer's disease (AD), synaptic plasticity and neuronal integrity are disturbed. In addition to the alterations in plasticity in mature neurons, the neurodegenerative process in AD has been shown to be accompanied by alterations in neurogenesis. Members of the bone morphogenetic protein (BMP) family of growth factors have been implicated as important regulators of neurogenesis and neuronal cell fate determination during development; however, their role in adult neurogenesis and in AD is less clear. We show here by qRT-PCR analysis that BMP6 mRNA levels were significantly increased in the hippocampus of human patients with AD and in APP transgenic mice compared to controls. Immunoblot and immunohistochemical analyses confirmed that BMP6 protein levels were increased in human AD brains and APP transgenic mouse brains compared to controls and accumulated around hippocampal plaques. The increased levels of BMP6 were accompanied by defects in hippocampal neurogenesis in AD patients and APP transgenic mice. In support of a role for BMP6 in defective neurogenesis in AD, we show in an in vitro model of adult neurogenesis that treatment with amyloid-ß(1-42) protein (Aß) resulted in increased expression of BMP6, and that exposure to recombinant BMP6 resulted in reduced proliferation with no toxic effects. Together, these results suggest that Aß-associated increases in BMP6 expression in AD may have deleterious effects on neurogenesis in the hippocampus, and therapeutic approaches could focus on normalization of BMP6 levels to protect against AD-related neurogenic deficits.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Proteína Morfogenética Ósea 6/biosíntesis , Proteína Morfogenética Ósea 6/genética , Química Encefálica , Inhibición Neural/genética , Neurogénesis , Regulación hacia Arriba/genética , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/fisiología , Animales , Proteína Morfogenética Ósea 6/fisiología , Química Encefálica/genética , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Neurogénesis/genética
12.
J Neurovirol ; 17(4): 327-40, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21660601

RESUMEN

Hepatitis C virus (HCV) infection is a serious problem among those co-infected with human immunodeficiency virus; however, its impact in the central nervous system (CNS) remains unclear. This study aimed to investigate the mechanisms underlying HCV core protein-mediated neurodegeneration. Analysis of human HCV seropositive cases demonstrated widespread damage to neuronal dendritic processes and sustained activation of extracellular signal-related kinase (ERK); analogous pathologies were observed in wild type injected with HCV core protein into the hippocampus. In vitro analysis in neuronal cells exposed to HCV core demonstrated retraction of the neuronal processes in an ERK/Signal Transducer and Activator of Transcription 3 (STAT3)-dependent manner dependent on toll-like receptor 2 (TLR2) signaling activation. These results indicate that HCV core protein neurotoxicity may be mediated by the sustained activation of ERK/STAT3 via TLR2-IRAK1 signaling pathway. These pathways provide novel targets for development of neuroprotective treatments for HCV involvement of the CNS.


Asunto(s)
Hepacivirus , Hepatitis C/virología , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Receptor Toll-Like 2/metabolismo , Adulto , Animales , Western Blotting , Coinfección , Femenino , Silenciador del Gen/efectos de los fármacos , VIH/fisiología , Infecciones por VIH/complicaciones , Infecciones por VIH/metabolismo , Infecciones por VIH/patología , Infecciones por VIH/virología , Hepacivirus/patogenicidad , Hepacivirus/fisiología , Hepatitis C/complicaciones , Hepatitis C/metabolismo , Hepatitis C/patología , Hipocampo/patología , Hipocampo/virología , Humanos , Inmunohistoquímica , Inyecciones , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Quinasa Quinasa PAM/genética , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Persona de Mediana Edad , Neuronas/efectos de los fármacos , Neuronas/patología , ARN Interferente Pequeño/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT3/genética , Receptor Toll-Like 2/genética , Proteínas del Núcleo Viral/efectos adversos , Proteínas del Núcleo Viral/farmacología
13.
STAR Protoc ; 2(2): 100565, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-34136833

RESUMEN

Interferon regulatory factor 4 (IRF4) is a transcription factor that regulates normal and malignant immune cell development and is implicated in multiple myeloma pathogenesis. This protocol describes the use of combined cell surface and intranuclear staining with fluorescent antibodies to measure IRF4 protein expression within myeloma and normal immune cells. IRF4 protein quantification may provide a valuable prognostic tool to predict disease severity and sensitivity to IRF4-targeted therapies. This flow-cytometry-based procedure could also be rapidly translated into a clinically compatible assay. For complete details on the use and execution of this protocol, please refer to Mondala et al. (2021).


Asunto(s)
Células de la Médula Ósea/metabolismo , Citometría de Flujo/métodos , Factores Reguladores del Interferón/metabolismo , Mieloma Múltiple/metabolismo , Humanos , Límite de Detección , Mieloma Múltiple/patología
14.
Cell Stem Cell ; 28(4): 623-636.e9, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33476575

RESUMEN

In multiple myeloma, inflammatory and anti-viral pathways promote disease progression and cancer stem cell generation. Using diverse pre-clinical models, we investigated the role of interferon regulatory factor 4 (IRF4) in myeloma progenitor regeneration. In a patient-derived xenograft model that recapitulates IRF4 pathway activation in human myeloma, we test the effects of IRF4 antisense oligonucleotides (ASOs) and identify a lead agent for clinical development (ION251). IRF4 overexpression expands myeloma progenitors, while IRF4 ASOs impair myeloma cell survival and reduce IRF4 and c-MYC expression. IRF4 ASO monotherapy impedes tumor formation and myeloma dissemination in xenograft models, improving animal survival. Moreover, IRF4 ASOs eradicate myeloma progenitors and malignant plasma cells while sparing normal human hematopoietic stem cell development. Mechanistically, IRF4 inhibition disrupts cell cycle progression, downregulates stem cell and cell adhesion transcript expression, and promotes sensitivity to myeloma drugs. These findings will enable rapid clinical development of selective IRF4 inhibitors to prevent myeloma progenitor-driven relapse.


Asunto(s)
Mieloma Múltiple , Preparaciones Farmacéuticas , Animales , Ciclo Celular , Línea Celular Tumoral , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Recurrencia Local de Neoplasia , Oligonucleótidos Antisentido
15.
Cell Rep ; 34(4): 108670, 2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33503434

RESUMEN

Inflammation-dependent base deaminases promote therapeutic resistance in many malignancies. However, their roles in human pre-leukemia stem cell (pre-LSC) evolution to acute myeloid leukemia stem cells (LSCs) had not been elucidated. Comparative whole-genome and whole-transcriptome sequencing analyses of FACS-purified pre-LSCs from myeloproliferative neoplasm (MPN) patients reveal APOBEC3C upregulation, an increased C-to-T mutational burden, and hematopoietic stem and progenitor cell (HSPC) proliferation during progression, which can be recapitulated by lentiviral APOBEC3C overexpression. In pre-LSCs, inflammatory splice isoform overexpression coincides with APOBEC3C upregulation and ADAR1p150-induced A-to-I RNA hyper-editing. Pre-LSC evolution to LSCs is marked by STAT3 editing, STAT3ß isoform switching, elevated phospho-STAT3, and increased ADAR1p150 expression, which can be prevented by JAK2/STAT3 inhibition with ruxolitinib or fedratinib or lentiviral ADAR1 shRNA knockdown. Conversely, lentiviral ADAR1p150 expression enhances pre-LSC replating and STAT3 splice isoform switching. Thus, pre-LSC evolution to LSCs is fueled by primate-specific APOBEC3C-induced pre-LSC proliferation and ADAR1-mediated splicing deregulation.


Asunto(s)
Inflamación/inmunología , Leucemia Mieloide Aguda/fisiopatología , Proliferación Celular , Humanos , Células Madre Neoplásicas/metabolismo
16.
J Neurosci ; 29(4): 1115-25, 2009 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-19176820

RESUMEN

The endopeptidase neprilysin (NEP) is a major amyloid-beta (Abeta) degrading enzyme and has been implicated in the pathogenesis of Alzheimer's disease. Because NEP cleaves substrates other than Abeta, we investigated the potential role of NEP-mediated processing of neuropeptides in the mechanisms of neuroprotection in vivo. Overexpression of NEP at low levels in transgenic (tg) mice affected primarily the levels of neuropeptide Y (NPY) compared with other neuropeptides. Ex vivo and in vivo studies in tg mice and in mice that received lentiviral vector injections showed that NEP cleaved NPY into C-terminal fragments (CTFs), whereas silencing NEP reduced NPY processing. Immunoblot and mass spectrometry analysis showed that NPY 21-36 and 31-36 were the most abundant fragments generated by NEP activity in vivo. Infusion of these NPY CTFs into the brains of APP (amyloid precursor protein) tg mice ameliorated the neurodegenerative pathology in this model. Moreover, the amidated NPY CTFs protected human neuronal cultures from the neurotoxic effects of Abeta. This study supports the possibility that the NPY CTFs generated during NEP-mediated proteolysis might exert neuroprotective effects in vivo. This function of NEP represents a unique example of a proteolytic enzyme with dual action, namely, degradation of Abeta as well as processing of NPY.


Asunto(s)
Neprilisina/química , Neprilisina/metabolismo , Degeneración Nerviosa/prevención & control , Neuropéptido Y/uso terapéutico , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Arginina/análogos & derivados , Arginina/farmacología , Benzazepinas/farmacología , Células Cultivadas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Regulación de la Expresión Génica/genética , Vectores Genéticos/fisiología , Humanos , Ratones , Ratones Transgénicos , Neprilisina/genética , Degeneración Nerviosa/etiología , Factores de Crecimiento Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuropéptido Y/química , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/uso terapéutico , Receptores de Neuropéptido Y/antagonistas & inhibidores
17.
Neuron ; 46(6): 857-68, 2005 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-15953415

RESUMEN

Abnormal folding of alpha-synuclein (alpha-syn) is thought to lead to neurodegeneration and the characteristic symptoms of Lewy body disease (LBD). Since previous studies suggest that immunization might be a potential therapy for Alzheimer's disease, we hypothesized that immunization with human (h)alpha-syn might have therapeutic effects in LBD. For this purpose, halpha-syn transgenic (tg) mice were vaccinated with halpha-syn. In mice that produced high relative affinity antibodies, there was decreased accumulation of aggregated halpha-syn in neuronal cell bodies and synapses that was associated with reduced neurodegeneration. Furthermore, antibodies produced by immunized mice recognized abnormal halpha-syn associated with the neuronal membrane and promoted the degradation of halpha-syn aggregates, probably via lysosomal pathways. Similar effects were observed with an exogenously applied FITC-tagged halpha-syn antibody. These results suggest that vaccination is effective in reducing neuronal accumulation of halpha-syn aggregates and that further development of this approach might have a potential role in the treatment of LBD.


Asunto(s)
Modelos Animales de Enfermedad , Inmunización/métodos , Proteínas del Tejido Nervioso/inmunología , Enfermedad de Parkinson/inmunología , Enfermedad de Parkinson/terapia , Animales , Anticuerpos/metabolismo , Anticuerpos/uso terapéutico , Western Blotting/métodos , Catepsina D/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Diagnóstico por Imagen/métodos , Mapeo Epitopo/métodos , Humanos , Inmunohistoquímica/métodos , Cuerpos de Inclusión/metabolismo , Lisosomas/metabolismo , Ratones , Ratones Transgénicos , Modelos Inmunológicos , Proteínas del Tejido Nervioso/genética , Neuroglía/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Terminales Presinápticos/metabolismo , Fracciones Subcelulares/metabolismo , Sinaptofisina/metabolismo , Sinucleínas , alfa-Sinucleína
18.
J Neurosci ; 28(16): 4250-60, 2008 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-18417705

RESUMEN

Altered expression and mutations in alpha-synuclein (alpha-syn) have been linked to Parkinson's disease (PD) and related disorders. The neurological alterations in PD patients have been associated with degeneration of dopaminergic cells and other neuronal populations. Moreover, recent studies in murine models have shown that alterations in neurogenesis might also contribute to the neurodegenerative phenotype. However, the mechanisms involved and the effects of alpha-syn expression on neurogenesis are not yet clear. To this end, murine embryonic stem (mES) cells were infected with lentiviral (LV) vectors expressing wild-type (WT) and mutant alpha-syn. Compared with mES cells infected with LV-green fluorescent protein (GFP), cells expressing WT and mutant alpha-syn showed reduced proliferation as indicated by lower 5-bromo-2'-deoxyuridine uptake, increased apoptosis, and reduced expression of neuronal markers such as neuron specific enolase and beta-III tubulin. The alterations in neurogenesis in alpha-syn-expressing mES cells were accompanied by a reduction in Notch-1 and Hairy and enhancer of split-5 (Hes-5) mRNA and protein levels. Moreover, levels of total Notch-1 and Notch intracellular domain (NICD) were lower in mES cells expressing WT and mutant alpha-syn compared with GFP controls. The reduced survival of alpha-syn-expressing mES cells was reverted by overexpressing constitutively active NICD. Similarly, in alpha-syn transgenic mice, the alterations in neurogenesis in the hippocampal subgranular zone were accompanied by decreased Notch-1, NICD, and Hes-5 expression. Together, these results suggest that accumulation of alpha-syn might impair survival of NPCs by interfering with the Notch signaling pathway. Similar mechanisms could be at play in PD and Lewy body disease.


Asunto(s)
Diferenciación Celular/fisiología , Regulación hacia Abajo/fisiología , Células Madre Embrionarias/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Receptor Notch1/antagonistas & inhibidores , Receptor Notch1/biosíntesis , alfa-Sinucleína/fisiología , Animales , Proliferación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica/fisiología , Hipocampo/citología , Hipocampo/embriología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Neuronas/citología , Receptor Notch1/genética , Células Madre/citología , Células Madre/metabolismo , alfa-Sinucleína/genética
19.
J Neurochem ; 108(6): 1561-74, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19166511

RESUMEN

Parkinson's disease (PD) is characterized by accumulation of alpha-synuclein (alpha-syn) and degeneration of neuronal populations in cortical and subcortical regions. Mitochondrial dysfunction has been considered a potential unifying factor in the pathogenesis of the disease. Mutations in genes linked to familial forms of PD, including SNCA encoding alpha-syn and Pten-induced putative kinase 1 (PINK1), have been shown to disrupt mitochondrial activity. We investigated the mechanisms through which mutant Pink1 might disrupt mitochondrial function in neuronal cells with alpha-syn accumulation. For this purpose, a neuronal cell model of PD was infected with virally-delivered Pink1, and was analyzed for cell survival, mitochondrial activity and calcium flux. Mitochondrial morphology was analyzed by confocal and electron microscopy. These studies showed that mutant (W437X) but not wildtype Pink1 exacerbated the alterations in mitochondrial function promoted by mutant (A53T) alpha-syn. This effect was associated with increased intracellular calcium levels. Co-expression of both mutant Pink1 and alpha-syn led to alterations in mitochondrial structure and neurite outgrowth that were partially ameliorated by treatment with cyclosporine A, and completely restored by treatment with the mitochondrial calcium influx blocker Ruthenium Red, but not with other cellular calcium flux blockers. Our data suggest a role for mitochondrial calcium influx in the mechanisms of mitochondrial and neuronal dysfunction in PD. Moreover, these studies support an important function for Pink1 in regulating mitochondrial activity under stress conditions.


Asunto(s)
Calcio/metabolismo , Mitocondrias/fisiología , Mutación/genética , Proteínas Quinasas/genética , Adenosina Trifosfato/metabolismo , Alanina/genética , Análisis de Varianza , Animales , Línea Celular Tumoral , Cobalto/farmacología , Relación Dosis-Respuesta a Droga , Ácido Flufenámico/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Microscopía Electrónica de Transmisión/métodos , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Neuroblastoma/ultraestructura , Neuronas/metabolismo , Neuronas/ultraestructura , Ratas , Treonina/genética , Transfección , Triptófano/genética , alfa-Sinucleína/genética
20.
J Clin Invest ; 116(11): 3060-9, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17080199

RESUMEN

Alzheimer's disease (AD) is characterized by progressive neurodegeneration and cerebral accumulation of the beta-amyloid peptide (Abeta), but it is unknown what makes neurons susceptible to degeneration. We report that the TGF-beta type II receptor (TbetaRII) is mainly expressed by neurons, and that TbetaRII levels are reduced in human AD brain and correlate with pathological hallmarks of the disease. Reducing neuronal TGF-beta signaling in mice resulted in age-dependent neurodegeneration and promoted Abeta accumulation and dendritic loss in a mouse model of AD. In cultured cells, reduced TGF-beta signaling caused neuronal degeneration and resulted in increased levels of secreted Abeta and beta-secretase-cleaved soluble amyloid precursor protein. These results show that reduced neuronal TGF-beta signaling increases age-dependent neurodegeneration and AD-like disease in vivo. Increasing neuronal TGF-beta signaling may thus reduce neurodegeneration and be beneficial in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Envejecimiento/fisiología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Células Cultivadas , Dendritas/metabolismo , Dendritas/patología , Gliosis/metabolismo , Gliosis/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Degeneración Nerviosa/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proteínas Serina-Treonina Quinasas , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA