Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Med Res Arch ; 11(8)2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37736242

RESUMEN

Immunotherapy, including immune cell therapy and targeted therapy, is gradually developed through the ongoing discovery of molecular compounds or immune cells. Choosing the best one or the best combination of target compounds and immune-cell therapy is a challenge for clinical scientists and clinicians. We have found variable efficacy individually after tumor-infiltrating lymphocyte (TIL) therapy, and now TILs have been discovered in a group of heterogeneous immune cells. To select the best immunotherapy for each patient, we started to study TIL genomics, including single-cell mRNA differential display from TIL published in 2007 and single-cell RNA-seq from TIL published in 2013, set up TIL quantitative network in 2015, researched machine-learning model for immune therapy in 2022. These manual reports single-cell RNA-seq data combined with machine learning to evaluate the optimal compounds and immune cells for individual patients. The machine-learning model, one of artificial intelligence, can estimate targeting genomic variance from single-cell RNA-seq so that they can cover thirteen kinds of immune cell therapies and ongoing FDA-approved targeted therapies such as PD1 inhibitors, PDL1 inhibitors, and CTLA4 inhibitors, as well as other different treatments such as HDACI or DNMT1 inhibitors, FDA-approved drugs. Moreover, also cover Phase-1, Phase-2, Phase-3, and Phase-4 of clinical trials, such as TIL, CAR T-cells, TCR T-cells. Single-cell RNA-seq with an Artificial intelligence estimation system is much better than our published models from microarrays or just cell therapy. The medical goal is to address three issues in clinical immunotherapy: the increase of efficacy; the decrease of adverse effects and the decrease of the cost in clinical applications.

3.
Front Immunol ; 13: 973881, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36341370

RESUMEN

Lymphocytes in tumor tissue are called tumor-infiltrating lymphocytes (TILs), and they play a key role in the control and treatment of tumor diseases. Since the discovery in 1987 that cultured TILs can kill tumor cells more than 100 times more effectively than T-cells cultured from peripheral blood in melanoma, it has been confirmed that cultured TILs can successfully cure clinical patients with melanoma. Since 1989, after we investigated TIL isolation performance from solid tumors, we modified some procedures to increase efficacy, and thus successfully established new TIL isolation and culture methods in 1994. Moreover, our laboratory and clinicians using our cultured TILs have published more than 30 papers. To improve the efficacy of TILs, we have been carrying out studies of TIL efficacy to treat solid tumor diseases for approximately 30 years. The three main questions of TIL study have been "How do TILs remain silent in solid tumor tissue?", "How do TILs attack homologous and heterologous antigens from tumor cells of solid tumors?", and "How do TILs infiltrate solid tumor tissue from a distance into tumor sites to kill tumor cells?". Research on these three issues has increasingly answered these questions. In this review I summarize the main issues surrounding TILs in treating solid tumors. This review aims to study the killing function of TILs from solid tumor tissues, thereby ultimately introducing the optimal strategy for patients suffering from solid tumors through personalized immunotherapy in the near future.


Asunto(s)
Linfocitos Infiltrantes de Tumor , Melanoma , Humanos , Inmunoterapia , Linfocitos/patología , Recuento de Linfocitos
5.
PLoS One ; 17(5): e0261799, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35639781

RESUMEN

Sickle cell disease (SCD) is an inherited blood disorder caused by a mutation in the HBB gene leading to hemoglobin S production and polymerization under hypoxia conditions leading to vaso-occlusion, chronic hemolysis, and progressive organ damage. This disease affects ~100,000 people in the United States and millions worldwide. An effective therapy for SCD is fetal hemoglobin (HbF) induction by pharmacologic agents such as hydroxyurea, the only Food and Drug Administration-approved drug for this purpose. Therefore, the goal of our study was to determine whether salubrinal (SAL), a selective protein phosphatase 1 inhibitor, induces HbF expression through the stress-signaling pathway by activation of p-eIF2α and ATF4 trans-activation in the γ-globin gene promoter. Sickle erythroid progenitors treated with 24µM SAL increased F-cells levels 1.4-fold (p = 0.021) and produced an 80% decrease in reactive oxygen species. Western blot analysis showed SAL enhanced HbF protein by 1.6-fold (p = 0.0441), along with dose-dependent increases of p-eIF2α and ATF4 levels. Subsequent treatment of SCD mice by a single intraperitoneal injection of SAL (5mg/kg) produced peak plasma concentrations at 6 hours. Chronic treatments of SCD mice with SAL mediated a 2.3-fold increase in F-cells (p = 0.0013) and decreased sickle erythrocytes supporting in vivo HbF induction.


Asunto(s)
Anemia de Células Falciformes , Hemoglobina Fetal , Animales , Cinamatos/farmacología , Cinamatos/uso terapéutico , Factor 2 Eucariótico de Iniciación/metabolismo , Hemoglobina Fetal/metabolismo , Humanos , Ratones , Transducción de Señal , Tiourea/análogos & derivados
6.
Blood Cells Mol Dis ; 93: 102626, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34856533

RESUMEN

Induction of fetal hemoglobin (HbF) expression ameliorates the clinical severity and prolong survival in persons with sickle cell disease (SCD). Hydroxyurea (HU) is the only FDA-approved HbF inducer however, additional therapeutics that produce an additive effect in SCD are needed. To this end, development of potent Class I histone deacetylase inhibitors (HDACi) for HbF induction represents a rational molecularly targeted approach. In studies here, we evaluated CT-101, a novel Class I-restricted HDACi, a Largazole derivative, for pharmacodynamics, cytotoxicity, and targeted epigenetic effects. In SCD-derived erythroid progenitors, CT-101 induced HbF expression with additive activity in combination with HU. CT-101 preferentially activated γ-globin gene transcription, increased acetylated histone H3 levels, and conferred an open chromatin conformation in the γ-globin promoter. These data indicate CT-101 represents a strong potential candidate as a molecularly targeted inducer of HbF.


Asunto(s)
Anemia de Células Falciformes , gamma-Globinas , Anemia de Células Falciformes/tratamiento farmacológico , Anemia de Células Falciformes/genética , Epigénesis Genética , Células Eritroides/metabolismo , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Expresión Génica , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Tomografía Computarizada por Rayos X , gamma-Globinas/genética
7.
Biomed J Sci Tech Res ; 47(2): 38211-38216, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-37817882

RESUMEN

Immune-cell therapy and targeting therapy are in rapid development to treat tumor diseases. However, current immune-cell therapy and targeting immunotherapy often face three challenges (three Ss): safety challenges such as cytokine releasing syndrome (C.R.S.); specificity targeting problems such as low efficacy caused by off-targeting tumor cells; unsatisfying payment are confounded to clinical patients and physicians. We have been studying immunotherapy for more than thirty years, and recently, personalized immunotherapy to treat tumor disease has been proposed. After we discovered quiescent genes from immune cells within the tumor microenvironment, we set up single-cell genomics analysis, studying heterogeneous immune responses from multiple tumor antigens (neo-antigen); here, we further introduce a new generation of immunotherapy module by using a machine-learning model to assess optimal immunotherapy. The machine-learning model combined with single-cell genomic analysis can predict optimal immune-cell (such as T-cells) and other optimal targeting drugs such as PD1 and CTLA4 inhibitors for the patient to use.

8.
Blood Cells Mol Dis ; 89: 102561, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33744514

RESUMEN

Increased expression of developmentally silenced fetal globin (HBG) reduces the clinical severity of ß-hemoglobinopathies. Benserazide has a relatively benign safety profile having been approved for 50 years in Europe and Canada for Parkinson's disease treatment. Benserazide was shown to activate HBG gene transcription in a high throughput screen, and subsequent studies confirmed fetal hemoglobin (HbF) induction in erythroid progenitors from hemoglobinopathy patients, transgenic mice containing the entire human ß-globin gene (ß-YAC) and anemic baboons. The goal of this study is to evaluate efficacies and plasma exposure profiles of benserazide racemate and its enantiomers to select the chemical form for clinical development. Intermittent treatment with all forms of benserazide in ß-YAC mice significantly increased proportions of red blood cells expressing HbF and HbF protein per cell with similar pharmacokinetic profiles and with no cytopenia. These data contribute to the regulatory justification for development of the benserazide racemate. Additionally, dose ranges and frequencies required for HbF induction using racemic benserazide were explored. Orally administered escalating doses of benserazide in an anemic baboon induced γ-globin mRNA up to 13-fold and establish an intermittent dose regimen for clinical studies as a therapeutic candidate for potential treatment of ß-hemoglobinopathies.


Asunto(s)
Anemia de Células Falciformes/tratamiento farmacológico , Benserazida/farmacología , Dopaminérgicos/farmacología , Hemoglobina Fetal/genética , Regulación hacia Arriba/efectos de los fármacos , Talasemia beta/tratamiento farmacológico , Anemia de Células Falciformes/genética , Animales , Expresión Génica/efectos de los fármacos , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Papio , Talasemia beta/genética , gamma-Globinas/genética
9.
Blood Cells Mol Dis ; 79: 102345, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31351219

RESUMEN

Pharmacologic induction of fetal hemoglobin (HbF) is an effective strategy for treating sickle cell disease (SCD) by ameliorating disease severity. Hydroxyurea is the only FDA-approved agent that induces HbF, but significant non-responders and requirement for frequent monitoring of blood counts for drug toxicity limit clinical usefulness. Therefore, we investigated a novel prodrug conjugate of butyric acid (BA) and δ-aminolevulinate (ALA) as a potential HbF inducing agent, using erythroid precursors and a preclinical ß-YAC mouse model. We observed significantly increased γ-globin gene transcription and HbF expression mediated by AN-233 in K562 cells. Moreover, AN-233 stimulated mild heme biosynthesis and inhibited expression of heme-regulated eIF2α kinase involved in silencing γ-globin expression. Studies using primary erythroid precursors generated from sickle peripheral blood mononuclear cells verified the ability of AN-233 to induce HbF, increase histone H3 and H4 acetylation levels at the γ-globin promoter and reduce erythroid precursor sickling by 50%. Subsequent drug treatment of ß-YAC transgenic mice confirmed HbF induction in vivo by AN-233 through an increase in the percentage of HbF positive red blood cells and HbF levels measured by flow cytometry. These data support the potential development of AN-233 for the treatment of SCD.


Asunto(s)
Anemia de Células Falciformes/terapia , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/efectos de los fármacos , Ácidos Levulínicos/farmacología , Profármacos/farmacología , Animales , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Humanos , Células K562 , Ácidos Levulínicos/uso terapéutico , Ratones , Ratones Transgénicos , Activación Transcripcional , gamma-Globinas/genética
10.
Br J Haematol ; 186(1): 91-100, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30891745

RESUMEN

Sickle cell disease (SCD) affects over 2 million people worldwide with high morbidity and mortality in underdeveloped countries. Therapeutic interventions aimed at reactivating fetal haemoglobin (HbF) is an effective approach for improving survival and ameliorating the clinical severity of SCD. A class of agents that inhibit DNA methyltransferase (DNMT) activity show promise as HbF inducers because off-target effects are not observed at low concentrations. However, these compounds are rapidly degraded by cytidine deaminase when taken by oral administration, creating a critical barrier to clinical development for SCD. We previously demonstrated that microRNA29B (MIR29B) inhibits de novo DNMT synthesis, therefore, the goal of our study was to determine if MIR29 mediates HbF induction. Overexpression of MIR29B in human KU812 cells and primary erythroid progenitors significantly increased the percentage of HbF positive cells, while decreasing the expression of DNMT3A and the HBG repressor MYB. Furthermore, HBG promoter methylation levels decreased significantly following MIR29B overexpression in human erythroid progenitors. We subsequently, observed higher MIR29B expression in SCD patients with higher HbF levels compared to those with low HbF. Our findings provide evidence for the ability of MIR29B to induce HbF and supports further investigation to expand treatment options for SCD.


Asunto(s)
Anemia de Células Falciformes/genética , Epigénesis Genética/efectos de los fármacos , Hemoglobina Fetal/genética , MicroARNs/fisiología , Activación Transcripcional/efectos de los fármacos , gamma-Globinas/genética , Línea Celular , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , ADN (Citosina-5-)-Metiltransferasas/efectos de los fármacos , ADN Metiltransferasa 3A , Metilasas de Modificación del ADN/biosíntesis , Metilasas de Modificación del ADN/efectos de los fármacos , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/metabolismo , Humanos , MicroARNs/antagonistas & inhibidores
11.
Exp Biol Med (Maywood) ; 244(2): 171-182, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30674214

RESUMEN

IMPACT STATEMENT: Sickle cell disease (SCD) is a group of inherited blood disorders caused by mutations in the human ß-globin gene, leading to the synthesis of abnormal hemoglobin S, chronic hemolysis, and oxidative stress. Inhibition of hemoglobin S polymerization by fetal hemoglobin holds the greatest promise for treating SCD. The transcription factor NRF2, is the master regulator of the cellular oxidative stress response and activator of fetal hemoglobin expression. In animal models, various small chemical molecules activate NRF2 and ameliorate the pathophysiology of SCD. This review discusses the mechanisms of NRF2 regulation and therapeutic strategies of NRF2 activation to design the treatment options for individuals with SCD.


Asunto(s)
Anemia de Células Falciformes/metabolismo , Hemoglobina Fetal/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Anemia de Células Falciformes/tratamiento farmacológico , Regulación de la Expresión Génica , Hemoglobina Falciforme/metabolismo , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/fisiología , Modelos Biológicos , Factor 2 Relacionado con NF-E2/fisiología , Estrés Oxidativo , Complejo de la Endopetidasa Proteasomal/metabolismo , Complejo de la Endopetidasa Proteasomal/fisiología , Transducción de Señal/efectos de los fármacos
12.
Exp Hematol ; 70: 85-96.e5, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30412705

RESUMEN

Inherited genetic modifiers and pharmacologic agents that enhance fetal hemoglobin (HbF) expression reverse the clinical severity of sickle cell disease (SCD). Recent efforts to develop novel strategies of HbF induction include discovery of molecular targets that regulate γ-globin gene transcription and translation. The purpose of this study was to perform genome-wide microRNA (miRNA) analysis to identify genes associated with HbF expression in patients with SCD. We isolated RNA from purified reticulocytes for microarray-based miRNA expression profiling. Using samples from patients with contrasting HbF levels, we observed an eightfold upregulation of miR-144-3p (miR-144) and miR-144-5p in the low-HbF group compared with those with high HbF. Additional analysis by reverse transcription quantitative polymerase chain reaction confirmed individual miR-144 expression levels of subjects in the two groups. Subsequent functional studies in normal and sickle erythroid progenitors showed NRF2 gene silencing by miR-144 and concomitant repression of γ-globin transcription; by contrast, treatment with miR-144 antagomir reversed its silencing effects in a dose-dependent manner. Because NRF2 regulates reactive oxygen species levels, additional studies investigated mechanisms of HbF regulation using a hemin-induced oxidative stress model. Treatment of KU812 cells with hemin produced an increase in NRF2 expression and HbF induction that reversed with miR-144 pretreatment. Chromatin immunoprecipitation assay confirmed NRF2 binding to the γ-globin antioxidant response element, which was inhibited by miR-144 mimic treatment. The genome-wide miRNA microarray and primary erythroid progenitor data support a miR-144/NRF2-mediated mechanism of γ-globin gene regulation in SCD.


Asunto(s)
Anemia de Células Falciformes/metabolismo , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/biosíntesis , Regulación de la Expresión Génica , MicroARNs/biosíntesis , Factor 2 Relacionado con NF-E2/metabolismo , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/patología , Línea Celular , Células Precursoras Eritroides/patología , Femenino , Hemoglobina Fetal/genética , Estudio de Asociación del Genoma Completo , Humanos , Masculino , MicroARNs/genética , Factor 2 Relacionado con NF-E2/genética , Especies Reactivas de Oxígeno/metabolismo
14.
JCI Insight ; 2(20)2017 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-29046485

RESUMEN

Sickle cell disease (SCD) results from a point mutation in the ß-globin gene forming hemoglobin S (HbS), which polymerizes in deoxygenated erythrocytes, triggering recurrent painful vaso-occlusive crises and chronic hemolytic anemia. Reactivation of fetal Hb (HbF) expression ameliorates these symptoms of SCD. Nuclear factor (erythroid derived-2)-like 2 (Nrf2) is a transcription factor that triggers cytoprotective and antioxidant pathways to limit oxidative damage and inflammation and increases HbF synthesis in CD34+ stem cell-derived erythroid progenitors. We investigated the ability of dimethyl fumarate (DMF), a small-molecule Nrf2 agonist, to activate γ-globin transcription and enhance HbF in tissue culture and in murine and primate models. DMF recruited Nrf2 to the γ-globin promoters and the locus control region of the ß-globin locus in erythroleukemia cells, elevated HbF in SCD donor-derived erythroid progenitors, and reduced hypoxia-induced sickling. Chronic DMF administration in SCD mice induced HbF and increased Nrf2-dependent genes to detoxify heme and limit inflammation. This improved hematological parameters, reduced plasma-free Hb, and attenuated inflammatory markers. Chronic DMF administration to nonanemic primates increased γ-globin mRNA in BM and HbF protein in rbc. DMF represents a potential therapy for SCD to induce HbF and augment vasoprotection and heme detoxification.


Asunto(s)
Anemia de Células Falciformes/tratamiento farmacológico , Anemia/tratamiento farmacológico , Dimetilfumarato/metabolismo , Dimetilfumarato/farmacología , Hemoglobina Fetal/metabolismo , Hemo/metabolismo , Animales , Antioxidantes/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Inflamación , Leucemia Eritroblástica Aguda/metabolismo , Ratones , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , ARN Mensajero/metabolismo , Bazo/metabolismo , gamma-Globinas/genética
16.
Exp Biol Med (Maywood) ; 241(7): 719-29, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26940952

RESUMEN

Sickle cell anemia is a common genetic disorder caused by a point mutation in the sixth codon of the ß-globin gene affecting people of African descent worldwide. A wide variety of clinical phenotypes ranging from mild to severe symptoms and complications occur due to hemoglobin S polymerization, red blood cell sickling, and vaso-occlusion. Research efforts are ongoing to develop strategies of fetal hemoglobin (HbF; α2γ2) induction to inhibit sickle hemoglobin polymerization and improve clinical outcomes. Insights have been gained from investigating mutations in the ß-globin locus or transcription factors involved in the mechanisms of hemoglobin switching. Recent efforts to expand molecular targets that modulate γ-globin expression involve microRNAs that work through posttranscriptional gene regulation. Therefore, the goal of our study was to identify novel microRNA genes involved in fetal hemoglobin expression. Using in silico analysis, we identified a miR-34a binding site in the γ-globin mRNA which was tested for functional relevance. Stable expression of the shMIMIC miR-34a lentivirus vector increased fetal hemoglobin levels in single cell K562 clones consistent with silencing of a γ-globin gene repressor. Furthermore, miR-34a promoted cell differentiation supported by increased expression of KLF1, glycophorin A, and the erythropoietin receptor. Western blot analysis of known negative regulators of γ-globin including YY1, histone deacetylase 1, and STAT3, which are regulated by miR-34a showed no change in YY1 and histone deacetylase 1 levels; however, total- and phosphorylated-STAT3 levels were decreased in single cell miR-34a K562 clones. These data support a mechanism of fetal hemoglobin activation by miR-34a involving STAT3 gene silencing.


Asunto(s)
Hemoglobina Fetal/metabolismo , Regulación de la Expresión Génica/genética , MicroARNs/fisiología , Regiones no Traducidas 3' , Silenciador del Gen , Glicoforinas/metabolismo , Células HEK293 , Humanos , Células K562/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Polimorfismo de Nucleótido Simple/genética , Receptores de Eritropoyetina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/fisiología , Humedales
17.
PLoS One ; 10(12): e0144660, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26713848

RESUMEN

High-level fetal (γ) globin expression ameliorates clinical severity of the beta (ß) hemoglobinopathies, and safe, orally-bioavailable γ-globin inducing agents would benefit many patients. We adapted a LCR-γ-globin promoter-GFP reporter assay to a high-throughput robotic system to evaluate five diverse chemical libraries for this activity. Multiple structurally- and functionally-diverse compounds were identified which activate the γ-globin gene promoter at nanomolar concentrations, including some therapeutics approved for other conditions. Three candidates with established safety profiles were further evaluated in erythroid progenitors, anemic baboons and transgenic mice, with significant induction of γ-globin expression observed in vivo. A lead candidate, Benserazide, emerged which demonstrated > 20-fold induction of γ-globin mRNA expression in anemic baboons and increased F-cell proportions by 3.5-fold in transgenic mice. Benserazide has been used chronically to inhibit amino acid decarboxylase to enhance plasma levels of L-dopa. These studies confirm the utility of high-throughput screening and identify previously unrecognized fetal globin inducing candidates which can be developed expediently for treatment of hemoglobinopathies.


Asunto(s)
Anemia/genética , Hemoglobina Fetal/genética , Ensayos Analíticos de Alto Rendimiento , Papio , Activación Transcripcional/efectos de los fármacos , gamma-Globinas/genética , Animales , Benserazida/efectos adversos , Benserazida/farmacología , Evaluación Preclínica de Medicamentos , Células Precursoras Eritroides/efectos de los fármacos , Loratadina/efectos adversos , Loratadina/análogos & derivados , Loratadina/farmacología , Ratones , Ratones Transgénicos , ARN Mensajero/genética , ARN Mensajero/metabolismo
18.
Exp Biol Med (Maywood) ; 240(8): 1050-64, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26283707

RESUMEN

The developmental regulation of globin gene expression has shaped research efforts to establish therapeutic modalities for individuals affected with sickle cell disease and ß-thalassemia. Fetal hemoglobin has been shown to block sickle hemoglobin S polymerization to improve symptoms of sickle cell disease; moreover, fetal hemoglobin functions to replace inadequate hemoglobin A synthesis in ß-thalassemia thus serving as an effective therapeutic target. In the perinatal period, fetal hemoglobin is synthesized at high levels followed by a decline to adult levels by one year of age. It is known that naturally occurring mutations in the γ-globin gene promoters and distant cis-acting transcription factors produce persistent fetal hemoglobin synthesis after birth to ameliorate clinical symptoms. Major repressor proteins that silence γ-globin during development have been targeted for gene therapy in ß-hemoglobinopathies patients. In parallel effort, several classes of pharmacological agents that induce fetal hemoglobin expression through molecular and cell signaling mechanisms have been identified. Herein, we reviewed the progress made in the discovery of signaling molecules targeted by pharmacologic agents that enhance γ-globin expression and have the potential for future drug development to treat the ß-hemoglobinopathies.


Asunto(s)
Envejecimiento , Anemia de Células Falciformes/tratamiento farmacológico , Anemia de Células Falciformes/metabolismo , Hemoglobina Fetal/biosíntesis , Transducción de Señal/efectos de los fármacos , Humanos , Talasemia beta/tratamiento farmacológico , Talasemia beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA