Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
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
3.
J Med Chem ; 66(10): 6577-6590, 2023 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-37155693

RESUMEN

Highly functionalized skeletons of macrolide natural products gain access to rare spatial arrangements of atoms, where changes in stereochemistry can have a profound impact on the structure and function. Spliceosome modulators present a unique consensus motif, with the majority targeting a key interface within the SF3B spliceosome complex. Our recent preparative-scale synthetic campaign of 17S-FD-895 provided unique access to stereochemical analogues of this complex macrolide. Here, we report on the preparation and systematic activity evaluation of multiple FD-895 analogues. These studies examine the effects of modifications at specific stereocenters within the molecule and highlight future directions for medicinal chemical optimization of spliceosome modulators.


Asunto(s)
Macrólidos , Macrólidos/farmacología
4.
Blood Adv ; 7(16): 4302-4314, 2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37216228

RESUMEN

Human telomeres are tandem arrays that are predominantly composed of 5'-TTAGGG-3' nucleotide sequences at the terminal ends of chromosomes. These sequences serve 2 primary functions: they preserve genomic integrity by protecting the ends of chromosomes, preventing inappropriate degradation by DNA repair mechanisms, and they prevent loss of genetic information during cellular division. When telomeres shorten to reach a critical length, termed the Hayflick limit, cell senescence or death is triggered. Telomerase is a key enzyme involved in synthesizing and maintaining the length of telomeres within rapidly dividing cells and is upregulated across nearly all malignant cells. Accordingly, targeting telomerase to inhibit uncontrolled cell growth has been an area of great interest for decades. In this review, we summarize telomere and telomerase biology because it relates to both physiologic and malignant cells. We discuss the development of telomere- and telomerase-targeted therapeutic candidates within the realm of myeloid malignancies. We overview all mechanisms of targeting telomerase that are currently in development, with a particular focus on imetelstat, an oligonucleotide with direct telomerase inhibitory properties that has advanced the furthest in clinical development and has demonstrated promising data in multiple myeloid malignancies.


Asunto(s)
Trastornos Mieloproliferativos , Neoplasias , Telomerasa , Humanos , Telomerasa/metabolismo , Telómero/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Senescencia Celular , Secuencia de Bases , Trastornos Mieloproliferativos/genética
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.
Stem Cell Reports ; 18(8): 1599-1609, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-36563687

RESUMEN

Developing cellular therapies is not straightforward. This Perspective summarizes the experience of a group of academic stem cell investigators working in different clinical areas and aims to share insight into what we wished we knew before starting. These include (1) choosing the stem cell line and assessing the genome of both the starting and final product, (2) familiarity with GMP manufacturing, reagent validation, and supply chain management, (3) product delivery issues and the additional regulatory challenges, (4) the relationship between clinical trial design and preclinical studies, and (5) the market approval requirements, pathways, and partnerships needed.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Células Madre , Humanos , Línea Celular
8.
Int J Mol Sci ; 23(6)2022 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-35328625

RESUMEN

Advanced prostate cancer (PCa) patients with bone metastases are treated with androgen pathway directed therapy (APDT). However, this treatment invariably fails and the cancer becomes castration resistant. To elucidate resistance mechanisms and to provide a more predictive pre-clinical research platform reflecting tumor heterogeneity, we established organoids from a patient-derived xenograft (PDX) model of bone metastatic prostate cancer, PCSD1. APDT-resistant PDX-derived organoids (PDOs) emerged when cultured without androgen or with the anti-androgen, enzalutamide. Transcriptomics revealed up-regulation of neurogenic and steroidogenic genes and down-regulation of DNA repair, cell cycle, circadian pathways and the severe acute respiratory syndrome (SARS)-CoV-2 host viral entry factors, ACE2 and TMPRSS2. Time course analysis of the cell cycle in live cells revealed that enzalutamide induced a gradual transition into a reversible dormant state as shown here for the first time at the single cell level in the context of multi-cellular, 3D living organoids using the Fucci2BL fluorescent live cell cycle tracker system. We show here a new mechanism of castration resistance in which enzalutamide induced dormancy and novel basal-luminal-like cells in bone metastatic prostate cancer organoids. These PDX organoids can be used to develop therapies targeting dormant APDT-resistant cells and host factors required for SARS-CoV-2 viral entry.


Asunto(s)
Neoplasias Óseas/genética , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/genética , Organoides/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/genética , Andrógenos/farmacología , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Benzamidas/farmacología , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , COVID-19/genética , COVID-19/metabolismo , COVID-19/virología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Nitrilos/farmacología , Feniltiohidantoína/farmacología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Virales/genética , Receptores Virales/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiología , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Trasplante Heterólogo , Internalización del Virus
9.
Nat Commun ; 13(1): 659, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35115489

RESUMEN

Kinase signaling fuels growth of B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Yet its role in leukemia initiation is unclear and has not been shown in primary human hematopoietic cells. We previously described activating mutations in interleukin-7 receptor alpha (IL7RA) in poor-prognosis "ph-like" BCP-ALL. Here we show that expression of activated mutant IL7RA in human CD34+ hematopoietic stem and progenitor cells induces a preleukemic state in transplanted immunodeficient NOD/LtSz-scid IL2Rγnull mice, characterized by persistence of self-renewing Pro-B cells with non-productive V(D)J gene rearrangements. Preleukemic CD34+CD10highCD19+ cells evolve into BCP-ALL with spontaneously acquired Cyclin Dependent Kinase Inhibitor 2 A (CDKN2A) deletions, as commonly observed in primary human BCP-ALL. CRISPR mediated gene silencing of CDKN2A in primary human CD34+ cells transduced with activated IL7RA results in robust development of BCP-ALLs in-vivo. Thus, we demonstrate that constitutive activation of IL7RA can initiate preleukemia in primary human hematopoietic progenitors and cooperates with CDKN2A silencing in progression into BCP-ALL.


Asunto(s)
Subunidad alfa del Receptor de Interleucina-7/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Células Precursoras de Linfocitos B/inmunología , Transducción de Señal/inmunología , Animales , Antígenos CD34/genética , Antígenos CD34/inmunología , Antígenos CD34/metabolismo , Secuencia de Bases , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/inmunología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Expresión Génica/inmunología , Humanos , Subunidad alfa del Receptor de Interleucina-7/genética , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Células Precursoras de Linfocitos B/metabolismo , RNA-Seq/métodos , Receptores de Citocinas/genética , Receptores de Citocinas/inmunología , Receptores de Citocinas/metabolismo , Transducción de Señal/genética , Análisis de la Célula Individual/métodos , Trasplante Heterólogo
11.
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
12.
Blood Cancer Discov ; 1(1): 48-67, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32974613

RESUMEN

Persistence of drug-resistant quiescent leukemic stem cells (LSC) and impaired natural killer (NK) cell immune response account for relapse of chronic myelogenous leukemia (CML). Inactivation of protein phosphatase 2A (PP2A) is essential for CML-quiescent LSC survival and NK cell antitumor activity. Here we show that MIR300 has antiproliferative and PP2A-activating functions that are dose dependently differentially induced by CCND2/CDK6 and SET inhibition, respectively. MIR300 is upregulated in CML LSCs and NK cells by bone marrow microenvironment (BMM) signals to induce quiescence and impair immune response, respectively. Conversely, BCR-ABL1 downregulates MIR300 in CML progenitors to prevent growth arrest and PP2A-mediated apoptosis. Quiescent LSCs escape apoptosis by upregulating TUG1 long noncoding RNA that uncouples and limits MIR300 function to cytostasis. Genetic and pharmacologic MIR300 modulation and/or PP2A-activating drug treatment restore NK cell activity, inhibit BMM-induced growth arrest, and selectively trigger LSC apoptosis in vitro and in patient-derived xenografts; hence, the importance of MIR300 and PP2A activity for CML development and therapy.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva , MicroARNs , Humanos , Células Asesinas Naturales , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , MicroARNs/genética , Células Madre Neoplásicas , Inhibidores de Proteínas Quinasas/metabolismo , Proteína Fosfatasa 2/genética , Microambiente Tumoral/genética
13.
Cell Stem Cell ; 22(6): 801-805, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29859170

RESUMEN

The California Institute for Regenerative Medicine (CIRM) Alpha Stem Cell Clinic (ASCC) Network was launched in 2015 to address a compelling unmet medical need for rigorous, FDA-regulated, stem cell-related clinical trials for patients with challenging, incurable diseases. Here, we describe our multi-center experiences addressing current and future challenges.


Asunto(s)
Técnicas de Laboratorio Clínico , Medicina Regenerativa , Investigación con Células Madre , Células Madre/citología , California , Ensayos Clínicos como Asunto , Humanos , Trasplante de Células Madre
14.
Nat Commun ; 8(1): 1922, 2017 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-29203771

RESUMEN

Despite novel therapies, relapse of multiple myeloma (MM) is virtually inevitable. Amplification of chromosome 1q, which harbors the inflammation-responsive RNA editase adenosine deaminase acting on RNA (ADAR)1 gene, occurs in 30-50% of MM patients and portends a poor prognosis. Since adenosine-to-inosine RNA editing has recently emerged as a driver of cancer progression, genomic amplification combined with inflammatory cytokine activation of ADAR1 could stimulate MM progression and therapeutic resistance. Here, we report that high ADAR1 RNA expression correlates with reduced patient survival rates in the MMRF CoMMpass data set. Expression of wild-type, but not mutant, ADAR1 enhances Alu-dependent editing and transcriptional activity of GLI1, a Hedgehog (Hh) pathway transcriptional activator and self-renewal agonist, and promotes immunomodulatory drug resistance in vitro. Finally, ADAR1 knockdown reduces regeneration of high-risk MM in serially transplantable patient-derived xenografts. These data demonstrate that ADAR1 promotes malignant regeneration of MM and if selectively inhibited may obviate progression and relapse.


Asunto(s)
Adenosina Desaminasa/genética , Mieloma Múltiple/genética , Recurrencia Local de Neoplasia/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteína con Dedos de Zinc GLI1/metabolismo , Adenosina Desaminasa/metabolismo , Adulto , Anciano , Animales , Estudios de Casos y Controles , Progresión de la Enfermedad , Resistencia a Antineoplásicos/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Técnicas In Vitro , Masculino , Ratones , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Recurrencia Local de Neoplasia/metabolismo , Trasplante de Neoplasias , Pronóstico , Edición de ARN/genética , Proteínas de Unión al ARN/metabolismo
15.
Nat Rev Cancer ; 17(6): 381-392, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28416802

RESUMEN

Cancer stem cells (CSCs) can regenerate all facets of a tumour as a result of their stem cell-like capacity to self-renew, survive and become dormant in protective microenvironments. CSCs evolve during tumour progression in a manner that conforms to Charles Darwin's principle of natural selection. Although somatic DNA mutations and epigenetic alterations promote evolution, post-transcriptional RNA modifications together with RNA binding protein activity (the 'epitranscriptome') might also contribute to clonal evolution through dynamic determination of RNA function and gene expression diversity in response to environmental stimuli. Deregulation of these epitranscriptomic events contributes to CSC generation and maintenance, which governs cancer progression and drug resistance. In this Review, we discuss the role of malignant RNA processing in CSC generation and maintenance, including mechanisms of RNA methylation, RNA editing and RNA splicing, and the functional consequences of their aberrant regulation in human malignancies. Finally, we highlight the potential of these events as novel CSC biomarkers as well as therapeutic targets.


Asunto(s)
Neoplasias/genética , Neoplasias/patología , Células Madre Neoplásicas/fisiología , Edición de ARN/fisiología , Biomarcadores de Tumor , Humanos , Células Madre Neoplásicas/patología
16.
Cell Stem Cell ; 19(5): 599-612, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27570067

RESUMEN

Age-related human hematopoietic stem cell (HSC) exhaustion and myeloid-lineage skewing promote oncogenic transformation of hematopoietic progenitor cells into therapy-resistant leukemia stem cells (LSCs) in secondary acute myeloid leukemia (AML). While acquisition of clonal DNA mutations has been linked to increased rates of secondary AML for individuals older than 60 years, the contribution of RNA processing alterations to human hematopoietic stem and progenitor aging and LSC generation remains unclear. Comprehensive RNA sequencing and splice-isoform-specific PCR uncovered characteristic RNA splice isoform expression patterns that distinguished normal young and aged human stem and progenitor cells (HSPCs) from malignant myelodysplastic syndrome (MDS) and AML progenitors. In splicing reporter assays and pre-clinical patient-derived AML models, treatment with a pharmacologic splicing modulator, 17S-FD-895, reversed pro-survival splice isoform switching and significantly impaired LSC maintenance. Therapeutic splicing modulation, together with monitoring splice isoform biomarkers of healthy HSPC aging versus LSC generation, may be employed safely and effectively to prevent relapse, the leading cause of leukemia-related mortality.


Asunto(s)
Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Empalme del ARN/genética , Animales , Supervivencia Celular/genética , Senescencia Celular/genética , Técnicas de Cocultivo , Células HEK293 , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Síndromes Mielodisplásicos/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Empalmosomas/metabolismo , Células del Estroma/metabolismo
17.
Oncotarget ; 7(35): 56013-56029, 2016 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-27463014

RESUMEN

Multiple myeloma cell growth relies on intrinsic aggressiveness, due to a high karyotypic instability, or on the support from bone marrow (BM) niche.We and other groups have provided evidences that Notch signaling is related to tumor cell growth, pharmacological resistance, localization/recirculation in the BM and bone disease.This study indicates that high gene expression levels of Notch signaling members (JAG1, NOTCH2, HES5 and HES6) correlate with malignant progression or high-risk disease, and Notch signaling may participate in myeloma progression by increasing the BM levels of interleukin-6 (IL-6), a major player in myeloma cell growth and survival. Indeed, in vitro results, confirmed by correlation analysis on gene expression profiles of myeloma patients and immunohistochemical studies, demonstrated that Notch signaling controls IL-6 gene expression in those myeloma cells capable of IL-6 autonomous production as well as in surrounding BM stromal cells. In both cases Notch signaling activation may be triggered by myeloma cell-derived Jagged ligands. The evidence that Notch signaling positively controls IL-6 in the myeloma-associated BM makes this pathway a key mediator of tumor-directed reprogramming of the bone niche.This work strengthens the rationale for a novel Notch-directed therapy in multiple myeloma based on the inhibition of Jagged ligands.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Interleucina-6/metabolismo , Células Madre Mesenquimatosas/patología , Mieloma Múltiple/genética , Receptores Notch/metabolismo , Transducción de Señal/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Médula Ósea/patología , Línea Celular Tumoral , Técnicas de Cocultivo , Progresión de la Enfermedad , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Interleucina-6/genética , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Proteína Jagged-2/genética , Proteína Jagged-2/metabolismo , Ligandos , Células Madre Mesenquimatosas/metabolismo , Mieloma Múltiple/patología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores Notch/genética , Proteínas Represoras/metabolismo , Regulación hacia Arriba
18.
Cell Stem Cell ; 19(2): 177-191, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27292188

RESUMEN

Post-transcriptional adenosine-to-inosine RNA editing mediated by adenosine deaminase acting on RNA1 (ADAR1) promotes cancer progression and therapeutic resistance. However, ADAR1 editase-dependent mechanisms governing leukemia stem cell (LSC) generation have not been elucidated. In blast crisis chronic myeloid leukemia (BC CML), we show that increased JAK2 signaling and BCR-ABL1 amplification activate ADAR1. In a humanized BC CML mouse model, combined JAK2 and BCR-ABL1 inhibition prevents LSC self-renewal commensurate with ADAR1 downregulation. Lentiviral ADAR1 wild-type, but not an editing-defective ADAR1(E912A) mutant, induces self-renewal gene expression and impairs biogenesis of stem cell regulatory let-7 microRNAs. Combined RNA sequencing, qRT-PCR, CLIP-ADAR1, and pri-let-7 mutagenesis data suggest that ADAR1 promotes LSC generation via let-7 pri-microRNA editing and LIN28B upregulation. A small-molecule tool compound antagonizes ADAR1's effect on LSC self-renewal in stromal co-cultures and restores let-7 biogenesis. Thus, ADAR1 activation represents a unique therapeutic vulnerability in LSCs with active JAK2 signaling.


Asunto(s)
Adenosina Desaminasa/metabolismo , Autorrenovación de las Células , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , MicroARNs/metabolismo , Proteínas de Unión al ARN/metabolismo , Adenosina Desaminasa/genética , Animales , Secuencia de Bases , Autorrenovación de las Células/genética , Proteínas de Fusión bcr-abl/metabolismo , Regulación Leucémica de la Expresión Génica , Janus Quinasa 2/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Ratones , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Edición de ARN/genética , Proteínas de Unión al ARN/genética , Transducción de Señal/genética
19.
Sci Rep ; 6: 23885, 2016 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-27041210

RESUMEN

While implicated in therapeutic resistance, malignant progenitor cell cycle kinetics have been difficult to quantify in real-time. We developed an efficient lentiviral bicistronic fluorescent, ubiquitination-based cell cycle indicator reporter (Fucci2BL) to image live single progenitors on a defined niche coupled with cell cycle gene expression analysis. We have identified key differences in cell cycle regulatory gene expression and transit times between normal and chronic myeloid leukemia progenitors that may inform cancer stem cell eradication strategies.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Leucemia Mielógena Crónica BCR-ABL Positiva/diagnóstico por imagen , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Células Madre Neoplásicas/metabolismo , Análisis de Secuencia de ARN/métodos , Nicho de Células Madre , Ciclo Celular , Línea Celular , Técnicas de Cocultivo , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Humanos , Cinética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Microscopía Confocal , Imagen de Lapso de Tiempo
20.
Asian J Urol ; 3(4): 229-239, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29264191

RESUMEN

OBJECTIVE: Bone metastasis occurs in up to 90% of men with advanced prostate cancer and leads to fractures, severe pain and therapy-resistance. Bone metastases induce a spectrum of types of bone lesions which can respond differently to therapy even within individual prostate cancer patients. Thus, the special environment of the bone makes the disease more complicated and incurable. A model in which bone lesions are reproducibly induced that mirrors the complexity seen in patients would be invaluable for pre-clinical testing of novel treatments. The microstructural changes in the femurs of mice implanted with PCSD1, a new patient-derived xenograft from a surgical prostate cancer bone metastasis specimen, were determined. METHODS: Quantitative micro-computed tomography (micro-CT) and histological analyses were performed to evaluate the effects of direct injection of PCSD1 cells or media alone (Control) into the right femurs of Rag2-/-γc-/- male mice. RESULTS: Bone lesions formed only in femurs of mice injected with PCSD1 cells. Bone volume (BV) was significantly decreased at the proximal and distal ends of the femurs (p < 0.01) whereas BV (p < 0.05) and bone shaft diameter (p < 0.01) were significantly increased along the femur shaft. CONCLUSION: PCSD1 cells reproducibly induced bone loss leading to osteolytic lesions at the ends of the femur, and, in contrast, induced aberrant bone formation leading to osteoblastic lesions along the femur shaft. Therefore, the interaction of PCSD1 cells with different bone region-specific microenvironments specified the type of bone lesion. Our approach can be used to determine if different bone regions support more therapy resistant tumor growth, thus, requiring novel treatments.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...