Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biol Blood Marrow Transplant ; 26(9): e227-e231, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32592856

RESUMEN

The emotional and physical toll on caregivers of cancer patients is well documented, but research evaluating the financial burdens and time commitments of caregivers is limited. We suspected that the rural location of our cancer center would intensify these burdens for caregivers. We conducted a prospective trial to assess the out-of-pocket expenses and time commitment of caregivers of hematopoietic stem cell transplantation recipients within the first 4 weeks after discharge from the hospital from a National Cancer Institute (NCI)-designated comprehensive cancer center. These results show that caregivers of autologous recipients paid out-of-pocket expenses of $196 over 4 weeks. If lost wages were included, the expenses increased to $736 during this period. Caregivers of allogeneic recipients had out-of-pocket expenses of $110 in 4 weeks, or a total of $610 when lost wages were included. In the month after discharge from the hospital, caregivers traveled a median distance of 450 miles or 560 miles, depending on whether the patient received an autologous transplant or an allogeneic transplant, respectively. These results demonstrate a compelling need to address caregiver support, given the significant financial out-of-pocket expenses and time commitment.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Neoplasias , Cuidadores , Gastos en Salud , Humanos , Neoplasias/terapia , Estudios Prospectivos , Trasplante Autólogo
2.
Blood Coagul Fibrinolysis ; 30(8): 419-422, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31483321

RESUMEN

: Primary myelofibrosis (PMF) is a clonal hematopoietic stem cell disorder characterized by fibrosis of the marrow cavity, marked megakaryocyte atypia and progressive cytopenias. Although thrombosis predominates, bleeding is the primary manifestation in up to 20% of patients and may be life-threatening. In this report, we document restoration of megakaryocyte and platelet structure and function in PMF after allogeneic hematopoietic cell transplantation (HCT). A 59-year-old man presented with recurrent episodes of postoperative bleeding preceding a diagnosis of primary myelofibrosis (PMF). Platelet aggregation and secretion studies showed abnormal responses to all agonists tested (epinephrine, ADP, arachidonic acid, U46619, collagen, ristocetin) despite the presence of thrombocytosis. After an allogeneic HCT, platelet morphology and function studies were all normal. The pathophysiology of platelet dysfunction in myeloid neoplasia is not well understood but, as highlighted in our report, restoration of platelet function by HCT supports a clonal process involving an early hematopoietic progenitor cell.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/etiología , Trasplante de Células Madre Hematopoyéticas , Mielofibrosis Primaria/etiología , Recuperación de la Función , Trastornos de las Plaquetas Sanguíneas/terapia , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Hemorragia/terapia , Humanos , Masculino , Persona de Mediana Edad , Agregación Plaquetaria/efectos de los fármacos , Mielofibrosis Primaria/complicaciones , Mielofibrosis Primaria/terapia , Trombocitosis , Trasplante Homólogo , Resultado del Tratamiento
3.
Cancer Immunol Res ; 7(7): 1079-1090, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31088847

RESUMEN

Tumor hypoxia is a negative prognostic factor that is implicated in oncogenic signal activation, immune escape, and resistance to treatment. Identifying the mechanistic role of hypoxia in immune escape and resistance to immune-checkpoint inhibitors may aid the identification of therapeutic targets. We and others have shown that V-domain Ig suppressor of T-cell activation (VISTA), a negative checkpoint regulator in the B7 family, is highly expressed in the tumor microenvironment in tumor models and primary human cancers. In this study, we show that VISTA and HIF1α activity are correlated in a cohort of colorectal cancer patients. High VISTA expression was associated with worse overall survival. We used the CT26 colon cancer model to investigate the regulation of VISTA by hypoxia. Compared with less hypoxic tumor regions or draining lymph nodes, regions of profound hypoxia in the tumor microenvironment were associated with increased VISTA expression on tumor-infiltrating myeloid-derived suppressor cells (MDSC). Using chromatin immunoprecipitation and genetic silencing, we show that hypoxia-inducible factor (HIF)-1α binding to a conserved hypoxia response element in the VISTA promoter upregulated VISTA on myeloid cells. Further, antibody targeting or genetic ablation of VISTA under hypoxia relieved MDSC-mediated T-cell suppression, revealing VISTA as a mediator of MDSC function. Collectively, these data suggest that targeting VISTA may mitigate the deleterious effects of hypoxia on antitumor immunity.


Asunto(s)
Adenocarcinoma/inmunología , Antígenos B7/metabolismo , Neoplasias Colorrectales/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/fisiopatología , Células Supresoras de Origen Mieloide/inmunología , Microambiente Tumoral/inmunología , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Apoptosis , Antígenos B7/genética , Estudios de Casos y Controles , Proliferación Celular , Estudios de Cohortes , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Pronóstico , Tasa de Supervivencia , Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Células Tumorales Cultivadas
4.
Blood ; 127(11): 1384-5, 2016 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-26989189

RESUMEN

One of the major goals of hemoglobinopathy research has been to devise improved pharmacologic strategies for the induction of fetal hemoglobin (HbF) in people with sickle cell disease and ß-thalassemia. In this issue of Blood, Dulmovits and colleagues report that pomalidomide, a drug approved by the US Food and Drug Administration (FDA) for treatment of multiple myeloma, induces HbF production by decreasing levels of several key transcriptional repressors of fetal globin gene expression. In addition, they show that pomalidomide induces HbF in differentiating erythroid cells from people with sickle cell disease and in myeloma patients.


Asunto(s)
Células Madre Hematopoyéticas/efectos de los fármacos , Talidomida/análogos & derivados , Transcripción Genética/efectos de los fármacos , gamma-Globinas/genética , Humanos
5.
J Community Support Oncol ; 13(9): 316-22, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26859751

RESUMEN

BACKGROUND: About 1 in 7 of all hospitalized patients is readmitted within 30 days of discharge. The cost of readmissions is significant, with Medicare readmissions alone costing the health care system an estimated $28 billion a year. OBJECTIVE: To identify the rates of and causes for readmission within 100 days of patients receiving a hematopoietic stem cell transplant. METHODS: We performed a retrospective review of 235 consecutive transplant recipients (autologous, n = 144; allogeneic, n = 91) to determine rates and causes for readmission within 100 days of patients receiving a transplant. Medical records and hospital readmissions were reviewed for each patient. RESULTS: 36 allogeneic patients accounted for 56 readmissions. 23 autologous patients accounted for 26 readmissions. Autologous transplant recipients were most commonly readmitted for the development of a fever (n = 15 patients) or cardiopulmonary issues (n = 4). The most prevalent reasons for readmission in the allogeneic recipients included a fever (n = 21) or the development or exacerbation of graft-versus-host disease (n = 5). The readmission length of stay was 6 days (median range, 1-91 days) for allogeneic patients and 4 days (median range, 1-22 days) for autologous patients. There was no difference in survival between the readmitted and the non-readmitted cohorts (𝑃 = .55 for allogeneic patients; 𝑃 = .24 for autologous patients). Although allogeneic graft recipients demonstrated a higher readmission rate (39.6%) compared with autologous recipients (16%), none of the variables examined, including age, gender, performance status, diagnosis, remission status at the time of transplant, comorbidities, type of preparative chemotherapy regimen or donor type, identified patients at increased risk for readmission. LIMITATIONS: Variations in clinical care, physician practices, and patient characteristics need to be considered when examining readmission rates. Most of the allogeneic patient population included unrelated donor recipients (65%) who received nonmyeloablative conditioning regimens (81% of allogeneic recipients). These features may not be characteristic of other centers. CONCLUSIONS: In these high-risk patients, readmissions following a transplant are common. Enhanced predischarge education by nurses and pharmacists, along with ongoing outpatient education and rigorous outpatient follow-up through phone calls or social media may decrease readmission rates.

6.
Blood ; 124(17): 2730-4, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25170120

RESUMEN

Fetal hemoglobin (HbF) induction can ameliorate the clinical severity of sickle cell disease and ß-thalassemia. We previously reported that activation of the eukaryotic initiation factor 2α (eIF2α) stress pathway increased HbF through a posttranscriptional mechanism. In this study, we explored the underlying means by which salubrinal, an activator of eIF2α signaling, enhances HbF production in primary human erythroid cells. Initial experiments eliminated changes in globin messenger RNA (mRNA) stability or cellular location and reduction of adult hemoglobin as possible salubrinal mechanisms. We then determined that salubrinal selectively increased the number of actively translating ribosomes on γ-globin mRNA. This enhanced translation efficiency occurred in the recovery phase of the stress response as phosphorylation of eIF2α and global protein synthesis returned toward baseline. These findings highlight γ-globin mRNA translation as a novel mechanism for regulating HbF production and as a pharmacologic target for induction of HbF.


Asunto(s)
Células Eritroides/metabolismo , Hemoglobina Fetal/genética , Biosíntesis de Proteínas , gamma-Globinas/genética , Western Blotting , Cinamatos/farmacología , Células Eritroides/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/farmacología , Hemoglobina Fetal/metabolismo , Humanos , Células K562 , Fosforilación/efectos de los fármacos , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Tiourea/análogos & derivados , Tiourea/farmacología , gamma-Globinas/metabolismo
7.
Blood Cells Mol Dis ; 52(4): 214-24, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24314748

RESUMEN

Increased fetal hemoglobin (HbF) expression is beneficial for ß-hemoglobinopathy patients; however, current inducing agents do not possess the ideal combination of efficacy, safety and ease of use. Better understanding the mechanisms involved in γ-globin gene induction is critical for designing improved therapies, as no complete mechanism for any inducing agent has been identified. Given the cytotoxic nature of most known inducing drugs, we hypothesized that γ-globin is a cell stress response gene, and that induction occurs via activation of cell stress signaling pathways. We tested this hypothesis by investigating the ability of physical stresses including heat-shock (HS), UV- and X-irradiation and osmotic shock to increase γ-globin gene expression in erythroid cells. Experiments in K562 and KU812 cells showed that each of these stresses increased steady-state γ-globin mRNA levels, but only after 3-5days of treatments. HS and UV also increased γ-globin mRNA and HbF levels in differentiating primary human erythroid cells. Mechanistic studies showed that HS affects γ-globin mRNA at multiple levels, including nascent transcription and transcript stability, and that induction is dependent on neither the master regulator of the canonical HS response, HSF1, nor p38 MAPK. Inhibitor panel testing identified PI3K inhibitor LY294002 as a novel inducing agent and revealed potential roles for NFκB and VEGFR/PDGFR/Raf kinases in HS-mediated γ-globin gene induction. These findings suggest that cell stress signaling pathways play an important role in γ-globin gene induction and may provide novel targets for the pharmacologic induction of fetal hemoglobin.


Asunto(s)
Células Eritroides/metabolismo , Hemoglobina Fetal/biosíntesis , Hemoglobina Fetal/genética , Regulación de la Expresión Génica , Estrés Fisiológico , gamma-Globinas/biosíntesis , gamma-Globinas/genética , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Eritropoyesis/genética , Regulación de la Expresión Génica/efectos de los fármacos , Factores de Transcripción del Choque Térmico , Hemoglobinopatías/genética , Calor , Humanos , Células K562 , Presión Osmótica , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Rayos Ultravioleta , Rayos X , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Am J Hematol ; 89(3): 339-41, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24127129

RESUMEN

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare hematological disorder that is often suspected in a patient presenting with non-immune hemolytic anemia associated with pancytopenia or venous thrombosis. This disorder is a consequence of acquired somatic mutations in the phosphatidylinositol glycan class A (PIG-A) gene in the hematopoietic stem cells (HSC) of patients. The presence of these mutations leads to production of blood cells with decreased glycosyl phosphatidylinositol-anchored cell surface proteins, making red blood cells derived from the clone more sensitive to complement mediated hemolysis. The diagnosis of PNH may be difficult in some cases due a low proportion of PNH cells in the blood and occasionally due to difficulties in selecting the most appropriate diagnostic studies. The latest generation of tests allow for detection of very small populations of PNH cells, for following the natural course and response to therapy of the disease, and for helping to decide when to initiate therapy with monoclonal antibody targeting the terminal complement protein C5 (Eculizumab), anticoagulation, and in some cases allogeneic HSC transplant. In this article, we review the different diagnostic tests available to clinicians for PNH diagnosis.


Asunto(s)
Examen de la Médula Ósea , Análisis Mutacional de ADN , Hemoglobinuria Paroxística/diagnóstico , Inmunofenotipificación/métodos , Antígeno CD24/sangre , Antígenos CD59/sangre , Citometría de Flujo/métodos , Glicosilfosfatidilinositoles/sangre , Hemoglobinuria Paroxística/sangre , Hemoglobinuria Paroxística/genética , Humanos , Receptores de Lipopolisacáridos/sangre , Proteínas de la Membrana/genética
9.
Mol Cancer Ther ; 12(8): 1504-14, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23723123

RESUMEN

Proteins of the BCL2 family provide a survival mechanism in many human malignancies, including chronic lymphocytic leukemia (CLL). The BCL2 inhibitor ABT-263 (navitoclax) is active in clinical trials for lymphoid malignancies, yet resistance is expected on the basis of preclinical models. We recently showed that vinblastine can dramatically sensitize several leukemia cell lines to ABT-737 (the experimental congener of ABT-263). The goal of these experiments was to determine the impact of vinblastine on ABT-737 sensitivity in CLL cells isolated from peripheral blood and to define the underlying mechanism. Freshly isolated CLL cells from 35 patients, as well as normal lymphocytes and platelets, were incubated with various microtubule-disrupting agents plus ABT-737 to assess sensitivity to the single agents and the combination. ABT-737 and vinblastine displayed a range of sensitivity as single agents, and vinblastine markedly sensitized all CLL samples to ABT-737 within six hours. Vinblastine potently induced the proapoptotic protein PMAIP1 (NOXA) in both time- and dose-dependent manner and this was required for the observed apoptosis. Combretastatin A4, which dissociates microtubules by binding to a different site, had the same effect, confirming that interaction of these agents with microtubules is the initial target. Similarly, vincristine and vinorelbine induced NOXA and enhanced CLL sensitivity to ABT-737. Furthermore, vinblastine plus ABT-737 overcame stroma-mediated resistance to ABT-737 alone. Apoptosis was induced with clinically achievable concentrations with no additional toxicity to normal lymphocytes or platelets. These results suggest that vinca alkaloids may improve the clinical efficacy of ABT-263 in patients with CLL.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Compuestos de Bifenilo/farmacología , Resistencia a Antineoplásicos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Leucemia Linfocítica Crónica de Células B/genética , Nitrofenoles/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Sulfonamidas/farmacología , Vinblastina/farmacología , Antineoplásicos Fitogénicos/toxicidad , Apoptosis/efectos de los fármacos , Apoptosis/genética , Compuestos de Bifenilo/toxicidad , Plaquetas/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/genética , Humanos , Leucemia Linfocítica Crónica de Células B/metabolismo , Linfocitos/efectos de los fármacos , Nitrofenoles/toxicidad , Piperazinas/farmacología , Piperazinas/toxicidad , Pronóstico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Células del Estroma/metabolismo , Sulfonamidas/toxicidad , Moduladores de Tubulina/farmacología , Moduladores de Tubulina/toxicidad , Vinblastina/toxicidad
10.
Blood ; 122(4): 477-85, 2013 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-23690448

RESUMEN

Strategies to increase fetal hemoglobin (HbF) levels can ameliorate symptoms and improve the lives of ß-hemoglobinopathy patients. Although most studies have focused on induction of γ-globin gene expression as an approach to induce HbF, we hypothesized that post-transcriptional regulation of HbF plays an underappreciated yet important role in controlling HbF levels. In the present study, we investigated whether increasing eukaryotic initiation factor 2α (eIF2α) phosphorylation, a key regulator of protein translation, could enhance HbF post-transcriptionally in human primary erythroid cells. Initial analysis using a known inhibitor of eIF2α dephosphorylation, salubrinal, revealed that elevated eIF2α phosphorylation enhanced HbF production without changing globin gene expression, proliferation, or cell differentiation. These results were further supported by the post-transcriptional induction of HbF by other pharmacologic activators of the eIF2α pathway and by genetic inactivation of the negative regulators, GADD34 and CReP. Additionally, we found that this novel mechanism of increasing HbF could be combined with clinically relevant transcriptional activators of γ-globin gene expression to additively enhance HbF. Taken together, these findings identify eIF2α phosphorylation as a post-transcriptional regulator of HbF induction that may be pharmacologically targeted, either alone or in combination, in ß-hemoglobinopathy patients.


Asunto(s)
Factor 2 Eucariótico de Iniciación/metabolismo , Hemoglobina Fetal/metabolismo , Cinamatos/farmacología , Inhibidores Enzimáticos/farmacología , Células Precursoras Eritroides/efectos de los fármacos , Células Precursoras Eritroides/metabolismo , Factor 2 Eucariótico de Iniciación/fisiología , Hemoglobinopatías/genética , Hemoglobinopatías/metabolismo , Hemoglobinopatías/terapia , Humanos , Células K562 , Modelos Biológicos , Terapia Molecular Dirigida/métodos , Fosforilación/fisiología , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Tiourea/análogos & derivados , Tiourea/farmacología , Transcripción Genética
11.
Blood ; 121(5): 830-9, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23223429

RESUMEN

UNLABELLED: Although increased fetal hemoglobin (HbF) levels have proven benefit for people with ß-hemoglobinopathies, all current HbF-inducing agents have limitations. We previously reported that drugs that activate the NRF2 antioxidant response signaling pathway increase HbF in primary human erythroid cells. In an attempt to increase HbF levels achieved with NRF2 activators, in the present study, we investigated potential complementary activity between these agents and HMG-CoA reductase inhibitors (statins) based on their ability to induce KLF2 protein levels. Experiments in K562 cells showed that simvastatin increased KLF2 mRNA and protein and KLF2 binding to HS2 of the ß-globin locus control region and enhanced -globin mRNA production by the NRF2 activator Tert-butylhydroquinone (tBHQ). When tested in differentiating primary human erythroid cells, simvastatin induced HbF alone and additively with tBHQ, but it did not increase KLF2 mRNA or locus control region binding above levels seen with normal differentiation. Investigating alternative mechanisms of action, we found that both simvastatin and tBHQ suppress ß-globin mRNA and KLF1 and BCL11A mRNA and protein, similar to what is seen in people with an HPFH phenotype because of KLF1 haploinsufficiency. These findings identify statins as a potential class of HbF-inducing agents and suggest a novel mechanism of action based on pharmacologic suppression of KLF1 and BCL11A gene expression. KEY POINTS: Simvastatin and tBHQ suppress KLF1 and BCL11 gene expression and additively increase fetal hemoglobin in primary human erythroid cells. Because both drugs are FDA-approved, these findings could lead to clinical trials in the relatively near future.


Asunto(s)
Proteínas Portadoras/biosíntesis , Células Eritroides/metabolismo , Hemoglobina Fetal/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Hidroquinonas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Factores de Transcripción de Tipo Kruppel/biosíntesis , Proteínas Nucleares/biosíntesis , Simvastatina/farmacología , Proteínas Portadoras/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Eritroides/citología , Femenino , Hemoglobina Fetal/genética , Regulación de la Expresión Génica/genética , Sitios Genéticos/fisiología , Humanos , Células K562 , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Nucleares/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras
12.
Cancer Biol Ther ; 12(4): 314-25, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21768777

RESUMEN

The efficacy of many chemotherapeutic agents can be attenuated by expression of the anti-apoptotic proteins Bcl-2, Bcl-X(L) and Mcl-1. Flavopiridol and dinaciclib are cyclin-dependent kinase 7 and 9 inhibitors that transcriptionally inhibit expression of Mcl-1. We have investigated the ability of flavopiridol and dinaciclib to sensitize a panel of leukemia cell lines to vinblastine and paclitaxel. Both drugs acutely sensitized most of the leukemia lines to vinblastine, with 100% apoptosis in 4 h. Furthermore, dinaciclib sensitized freshly isolated chronic lymphocytic leukemia cells to vinblastine. This rapid induction of apoptosis was attributed to vinblastine-mediated activation of JNK because (a) flavopiridol and dinaciclib failed to induce apoptosis when combined with non-JNK activating concentrations of vinblastine; (b) JNK inhibitors suppressed JNK activity and prevented apoptosis; (c) flavopiridol did not potentiate apoptosis induced by paclitaxel which does not activate JNK in these cells; and (d) Jurkat cells failed to activate JNK in response to vinblastine and were not sensitive to combinations of vinblastine and flavopiridol or dinaciclib. The rapid induction of apoptosis by this combination in multiple cell systems but not in normal lymphocytes provides justification for performing a clinical trial to assess the efficacy in patients.


Asunto(s)
Apoptosis/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Flavonoides/farmacología , Piperidinas/farmacología , Compuestos de Piridinio/farmacología , Vinblastina/farmacología , Antracenos/farmacología , Antineoplásicos Fitogénicos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Óxidos N-Cíclicos , Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 9 Dependiente de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/metabolismo , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Células HL-60 , Humanos , Immunoblotting , Indolizinas , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Células Jurkat , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Paclitaxel/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Células U937 , Quinasa Activadora de Quinasas Ciclina-Dependientes
13.
Blood Cells Mol Dis ; 47(2): 107-16, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21641240

RESUMEN

The ß-hemoglobinopathies and thalassemias are serious genetic blood disorders affecting the ß-globin chain of hemoglobin A (α(2)ß(Α)(2)). Their clinical severity can be reduced by enhancing expression of fetal hemoglobin (γ-globin), producing HbF (α(2)γ(2,)). In studies reported here, γ-globin induction by 23 novel, structurally-unrelated compounds, which had been predicted through molecular modeling and in silico screening of a 13,000 chemical library, was evaluated in vitro in erythroid progenitors cultured from normal subjects and ß-thalassemia patients, and in vivo in transgenic mice or anemic baboons. Four predicted candidates were found to have high potency, with 4- to 8-fold induction of HbF. Two of these compounds have pharmacokinetic profiles favorable for clinical application. These studies thus effectively identified high potency γ-globin inducing candidate therapeutics and validated the utility of in silico molecular modeling.


Asunto(s)
Anemia/tratamiento farmacológico , Productos Biológicos/administración & dosificación , Diseño de Fármacos , Células Precursoras Eritroides/efectos de los fármacos , Hemoglobina Fetal/biosíntesis , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Talasemia beta/tratamiento farmacológico , gamma-Globinas/biosíntesis , Administración Oral , Anemia/genética , Anemia/metabolismo , Animales , Productos Biológicos/química , Productos Biológicos/uso terapéutico , Células Cultivadas , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/genética , Expresión Génica , Humanos , Inyecciones Intravenosas , Ratones , Ratones Transgénicos , Modelos Moleculares , Papio , Flebotomía , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Globinas beta/deficiencia , Globinas beta/genética , Talasemia beta/genética , Talasemia beta/metabolismo , gamma-Globinas/genética
14.
Blood ; 117(22): 5987-97, 2011 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-21464371

RESUMEN

Although hematopoietic stem cell transplantation and gene therapy have the potential to cure ß-thalassemia and sickle cell disease, they are not currently available to most people with these diseases. In the near term, pharmacologic induction of fetal hemoglobin (HbF) may offer the best possibility for safe, effective, and widely available therapy. In an effort to define new pathways for targeted drug development for HbF induction, we evaluated the nuclear factor erythroid 2-related factor 2 (NRF2) antioxidant response element signaling pathway. We found that 3 well-known activators of this pathway increased γ-globin mRNA at nontoxic doses in K562 cells. Tert-butylhydroquinone (tBHQ), the most active of these compounds, increased cellular levels and nuclear translocation of NRF2 and binding of NRF2 to the γ-globin promoter. siRNA knockdown of NRF2 inhibited γ-globin induction by tBHQ. When tested in human primary erythroid cells, tBHQ induced NRF2 binding to the γ-globin promoter, increased γ-globin mRNA and HbF, and suppressed ß-globin mRNA and HbA, resulting in a > 3-fold increase in the percentage of HbF. These results suggest that drugs that activate the NRF2/antioxidant response element signaling pathway have the potential to induce therapeutic levels of HbF in people with ß-hemoglobinopathies.


Asunto(s)
Antioxidantes/uso terapéutico , Hemoglobina Fetal/metabolismo , Hidroquinonas/uso terapéutico , Factor 2 Relacionado con NF-E2/metabolismo , Transducción de Señal/efectos de los fármacos , Talasemia beta/prevención & control , gamma-Globinas/genética , Western Blotting , Diferenciación Celular , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Células Eritroides/metabolismo , Humanos , Células K562 , Luciferasas/metabolismo , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/genética , Regiones Promotoras Genéticas , Transporte de Proteínas , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Elementos de Respuesta , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Globinas beta , Talasemia beta/metabolismo , Talasemia beta/patología , gamma-Globinas/metabolismo
15.
Mol Cancer Ther ; 9(4): 791-802, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20371726

RESUMEN

Chemotherapeutic agents modify intracellular signaling that culminates in the inhibition of Bcl-2 family members and initiates apoptosis. Inhibition of the extracellular signal-regulated kinase by PD98059 dramatically accelerates vinblastine-mediated apoptosis in ML-1 leukemia with cells dying in 4 hours from all phases of the cell cycle. Inhibition of protein synthesis by cycloheximide also markedly accelerated vinblastine-induced apoptosis, showing that the proteins required for this acute apoptosis are constitutively expressed. Vinblastine induced the rapid induction of Mcl-1 that was inhibited by PD98059 and cycloheximide. No change in Bcl-2 or Bcl-X was observed. We hypothesize that ML-1 cells use Mcl-1 for protection from the rapid vinblastine-induced apoptosis. This was confirmed by targeting Mcl-1 with short hairpin RNA. We also investigated the response of 13 other leukemia and lymphoma cell lines and cells from seven chronic lymphocytic leukemia patients. Four cell lines and all chronic lymphocytic leukemia cells were killed in 6 hours by vinblastine alone. Two additional cell lines were sensitized to vinblastine by PD98059, which suppressed Mcl-1. This acute apoptosis either alone or in combination with PD98059 required vinblastine-mediated activation of c-Jun-NH(2)-terminal kinase. PD98059 did not suppress Mcl-1 in other cell lines whereas sorafenib did, but this did not sensitize the cells to vinblastine, suggesting that the acute apoptosis varies depending on which Bcl-2 protein mediates protection. Most of the cell lines were sensitized to vinblastine by cycloheximide, suggesting that inhibition of a short-lived protein in addition to Mcl-1 can acutely sensitize cells. These results suggest several clinical strategies that might provide an effective therapy for selected patients. Mol Cancer Ther; 9(4); 791-802. (c)2010 AACR.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Leucemia/patología , Linfoma/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Vinblastina/farmacología , Línea Celular Tumoral , Cicloheximida/farmacología , Citoprotección/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Flavonoides/farmacología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Leucemia/enzimología , Leucemia Linfocítica Crónica de Células B/enzimología , Leucemia Linfocítica Crónica de Células B/patología , Linfoma/enzimología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides
16.
Exp Hematol ; 37(7): 807-813.e2, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19460471

RESUMEN

OBJECTIVE: DNA methylation has long been implicated in developmental beta-globin gene regulation. However, the mechanism underlying this regulation is unclear, especially because these genes do not contain CpG islands. This has led us to propose and test the hypothesis that, just as for histone modifications, developmentally specific changes in human beta-like globin gene expression are associated with long-range changes in DNA methylation. MATERIALS AND METHODS: Bisulfite sequencing was used to determine the methylation state of individual CpG dinucleotides across the beta-globin locus in uncultured primary human erythroblasts from fetal liver and bone marrow, and in primitive-like erythroid cells derived from human embryonic stem cells. RESULTS: beta-globin locus CpGs are generally highly methylated, but domains of DNA hypomethylation spanning thousands of base pairs are established around the most highly expressed genes during each developmental stage. These large domains of DNA hypomethylation are found within domains of histone modifications associated with gene expression. We also find hypomethylation of a small proportion of gamma-globin promoters in adult erythroid cells, suggesting a mechanism by which adult erythroid cells produce fetal hemoglobin. CONCLUSION: This is one of the first reports to show that changes in DNA methylation patterns across large domains around non-CpG island genes correspond with changes in developmentally regulated histone modifications and gene expression. These data support a new model in which extended domains of DNA hypomethylation and active histone marks are coordinately established to achieve developmentally specific gene expression of non-CpG island genes.


Asunto(s)
Metilación de ADN , Regulación del Desarrollo de la Expresión Génica , Transcripción Genética , Globinas beta/genética , Islas de CpG , Humanos , Regiones Promotoras Genéticas
17.
Exp Hematol ; 37(7): 799-806.e4, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19460472

RESUMEN

OBJECTIVE: The regulation of the beta-globin switch remains undetermined, and understanding this mechanism has important benefits for clinical and basic science. Histone modifications regulate gene expression and this study determines the presence of three important histone modifications across the beta-globin locus in erythroblasts with different beta-like globin-expression profiles. Understanding the chromatin associated with weak gamma gene expression in bone marrow cells is an important objective, with the goal of ultimately inducing postnatal expression of weak gamma-globin to cure beta-hemoglobinopathies. MATERIALS AND METHODS: These studies use uncultured primary fetal and bone marrow erythroblasts and human embryonic stem cell-derived primitive-like erythroblasts. Chromatin immunoprecipitation with antibodies against modified histones reveals DNA associated with such histones. Precipitated DNA is quantitated by real-time polymerase chain reaction for 40 sites across the locus. RESULTS: Distribution of histone modifications differs at each developmental stage. The most highly expressed genes at each stage are embedded within large domains of modifications associated with expression (acetylated histone H3 [H3ac] and dimethyl lysine 4 of histone H3 [H3K4me2]). Moderately expressed genes have H3ac and H3K4me2 in the immediate area around the gene. Dimethyl lysine 9 of histone H3 (H3K9me2), a mark associated with gene suppression, is present at the epsilon and gamma genes in bone marrow cells, suggesting active suppression of these genes. CONCLUSION: This study reveals complex patterns of histone modifications associated with highly expressed, moderately expressed, and unexpressed genes. Activation of gamma postnatally will likely require extensive modification of the histones in a large domain around the gamma genes.


Asunto(s)
Histonas/metabolismo , Globinas beta/metabolismo , Células Cultivadas , Inmunoprecipitación de Cromatina , Humanos , Reacción en Cadena de la Polimerasa
18.
Exp Hematol ; 36(9): 1057-72, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18718415

RESUMEN

A major goal of hemoglobinopathy research is to develop treatments that correct the underlying molecular defects responsible for sickle cell disease and beta-thalassemia. One approach to achieving this goal is the pharmacologic induction of fetal hemoglobin (HbF). This strategy is capable of inhibiting the polymerization of sickle hemoglobin and correcting the globin chain imbalance of beta-thalassemia. Despite this promise, none of the currently available HbF-inducing agents exhibit the combination of efficacy, safety, and convenience of use that would make them applicable to most patients. The recent success of targeted drug therapies for malignant diseases suggests that this approach could be effective for developing optimal HbF-inducing agents. A first step in applying this approach is the identification of specific molecular targets. However, while >70 HbF-inducing agents have been described, neither molecular mechanisms nor target molecules have been definitively verified for any of these compounds. To help focus investigation in this area, we have reviewed known HbF-inducing agents and their proposed mechanisms of action. We find that in many cases, current models inadequately explain key experimental results. By integrating features of the erythropoietic stress model of HbF induction with data from recent intracellular signaling experiments, we have developed a new model that has the potential to explain several findings that are inconsistent with previous models and to unify most HbF-inducing agents under a common mechanism: cell stress signaling. If correct, this or related models could lead to new opportunities for development of targeted therapies for the beta-hemoglobinopathies.


Asunto(s)
Eritrocitos/metabolismo , Eritropoyesis/fisiología , Hemoglobina Fetal/biosíntesis , Regulación de la Expresión Génica/fisiología , Globinas/biosíntesis , Hemoglobinopatías/tratamiento farmacológico , Modelos Genéticos , Estrés Fisiológico/genética , Adolescente , Animales , Butiratos/farmacología , Butiratos/uso terapéutico , Ensayos Clínicos como Asunto/estadística & datos numéricos , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Sistemas de Liberación de Medicamentos , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/uso terapéutico , Eritrocitos/patología , Eritropoyesis/efectos de los fármacos , Hemoglobina Fetal/genética , Regulación de la Expresión Génica/efectos de los fármacos , Globinas/genética , Trasplante de Células Madre Hematopoyéticas , Hemoglobinopatías/sangre , Hemoglobinopatías/genética , Hemoglobinopatías/fisiopatología , Inhibidores de Histona Desacetilasas , Humanos , Hidroxiurea/farmacología , Hidroxiurea/uso terapéutico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Estrés Fisiológico/fisiopatología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
19.
Blood ; 111(1): 411-20, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17916742

RESUMEN

5-azacytidine (5-Aza) is a potent inducer of fetal hemoglobin (HbF) in people with beta-thalassemia and sickle cell disease. Two models have been proposed to explain this activity. The first is based on the drug's ability to inhibit global DNA methylation, including the fetal globin genes, resulting in their activation. The second is based on 5-Aza's cytotoxicity and observations that HbF production is enhanced during marrow recovery. We tested these models using human primary cells in an in vitro erythroid differentiation system. We found that doses of 5-Aza that produce near maximal induction of gamma-globin mRNA and HbF do not alter cell growth, differentiation kinetics, or cell cycle, but do cause a localized demethylation of the gamma promoter. However, when we reduced gamma promoter methylation to levels equivalent to those seen with 5-Aza or to the lower levels seen in primary fetal erythroid cells using DNMT1 siRNA and shRNA, we observed no induction of gamma-globin mRNA or HbF. These results suggest that 5-Aza induction of HbF is not the result of global DNA demethylation or of changes in differentiation kinetics, but involves an alternative, previously unrecognized mechanism. Other results suggest that posttranscriptional regulation plays an important role in the 5-Aza response.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Azacitidina/farmacología , Metilación de ADN/efectos de los fármacos , Células Eritroides/efectos de los fármacos , Hemoglobina Fetal/genética , Adulto , Diferenciación Celular/fisiología , Células Cultivadas , Células Eritroides/citología , Células Eritroides/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Globinas/genética , Humanos , Cinética , Regiones Promotoras Genéticas/fisiología , ARN Mensajero/metabolismo , ARN Interferente Pequeño
20.
Blood ; 110(4): 1343-52, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17456718

RESUMEN

The mechanisms underlying the human fetal-to-adult beta-globin gene switch remain to be determined. While there is substantial experimental evidence to suggest that promoter DNA methylation is involved in this process, most data come from studies in nonhuman systems. We have evaluated human gamma- and beta-globin promoter methylation in primary human fetal liver (FL) and adult bone marrow (ABM) erythroid cells. Our results show that, in general, promoter methylation and gene expression are inversely related. However, CpGs at -162 of the gamma promoter and -126 of the beta promoter are hypomethylated in ABM and FL, respectively. We also studied gamma-globin promoter methylation during in vitro differentiation of erythroid cells. The gamma promoters are initially hypermethylated in CD34(+) cells. The upstream gamma promoter CpGs become hypomethylated during the preerythroid phase of differentiation and are then remethylated later, during erythropoiesis. The period of promoter hypomethylation correlates with transient gamma-globin gene expression and may explain the previously observed fetal hemoglobin production that occurs during early adult erythropoiesis. These results provide the first comprehensive survey of developmental changes in human gamma- and beta-globin promoter methylation and support the hypothesis that promoter methylation plays a role in human beta-globin locus gene switching.


Asunto(s)
Diferenciación Celular/genética , Metilación de ADN , Células Eritroides/citología , Regulación del Desarrollo de la Expresión Génica , Globinas/genética , Regiones Promotoras Genéticas/genética , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Linaje de la Célula/genética , Células Cultivadas , Eritroblastos , Hemoglobina Fetal/genética , Feto/citología , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Humanos , Hígado/citología , Secuencias Reguladoras de Ácidos Nucleicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...