Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.306
Filtrar
1.
BMC Cancer ; 24(1): 326, 2024 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-38461240

RESUMEN

BACKGROUND: FLI1 is an oncogenic transcription factor that promotes diverse malignancies through mechanisms that are not fully understood. Herein, FLI1 is shown to regulate the expression of Ubiquitin Associated and SH3 Domain Containing A/B (UBASH3A/B) genes. UBASH3B and UBASH3A are found to act as an oncogene and tumor suppressor, respectively, and their combined effect determines erythroleukemia progression downstream of FLI1. METHODS: Promoter analysis combined with luciferase assays and chromatin immunoprecipitation (ChIP) analysis were applied on the UBASH3A/B promoters. RNAseq analysis combined with bioinformatic was used to determine the effect of knocking-down UBASH3A and UBASH3B in leukemic cells. Downstream targets of UBASH3A/B were inhibited in leukemic cells either via lentivirus-shRNAs or small molecule inhibitors. Western blotting and RT-qPCR were used to determine transcription levels, MTT assays to assess proliferation rate, and flow cytometry to examine apoptotic index. RESULTS: Knockdown of FLI1 in erythroleukemic cells identified the UBASH3A/B genes as potential downstream targets. Herein, we show that FLI1 directly binds to the UBASH3B promoter, leading to its activation and leukemic cell proliferation. In contrast, FLI1 indirectly inhibits UBASH3A transcription via GATA2, thereby antagonizing leukemic growth. These results suggest oncogenic and tumor suppressor roles for UBASH3B and UBASH3A in erythroleukemia, respectively. Mechanistically, we show that UBASH3B indirectly inhibits AP1 (FOS and JUN) expression, and that its loss leads to inhibition of apoptosis and acceleration of proliferation. UBASH3B also positively regulates the SYK gene expression and its inhibition suppresses leukemia progression. High expression of UBASH3B in diverse tumors was associated with worse prognosis. In contrast, UBASH3A knockdown in erythroleukemic cells increased proliferation; and this was associated with a dramatic induction of the HSP70 gene, HSPA1B. Accordingly, knockdown of HSPA1B in erythroleukemia cells significantly accelerated leukemic cell proliferation. Accordingly, overexpression of UBASH3A in different cancers was predominantly associated with good prognosis. These results suggest for the first time that UBASH3A plays a tumor suppressor role in part through activation of HSPA1B. CONCLUSIONS: FLI1 promotes erythroleukemia progression in part by modulating expression of the oncogenic UBASH3B and tumor suppressor UBASH3A.


Asunto(s)
Leucemia Eritroblástica Aguda , Proteína Proto-Oncogénica c-fli-1 , Humanos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Línea Celular Tumoral , Regulación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteína Proto-Oncogénica c-fli-1/genética , Proteína Proto-Oncogénica c-fli-1/metabolismo , ARN Interferente Pequeño/genética , Proteína EWS de Unión a ARN/genética , Proteínas Tirosina Fosfatasas/metabolismo
2.
Exp Cell Res ; 433(2): 113853, 2023 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-37944576

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is causative of the ongoing coronavirus disease 2019 (COVID-19) pandemic. The SARS-CoV-2 Spike protein (S-protein) plays an important role in the early phase of SARS-CoV-2 infection through efficient interaction with ACE2. The S-protein is produced by RNA-based COVID-19 vaccines, that were fundamental for the reduction of the viral spread within the population and the clinical severity of COVID-19. However, the S-protein has been hypothesized to be responsible for damaging cells of several tissues and for some important side effects of RNA-based COVID-19 vaccines. Considering the impact of COVID-19 and SARS-CoV-2 infection on the hematopoietic system, the aim of this study was to verify the effect of the BNT162b2 vaccine on erythroid differentiation of the human K562 cell line, that has been in the past intensively studied as a model system mimicking some steps of erythropoiesis. In this context, we focused on hemoglobin production and induced expression of embryo-fetal globin genes, that are among the most important features of K562 erythroid differentiation. We found that the BNT162b2 vaccine suppresses mithramycin-induced erythroid differentiation of K562 cells. Reverse-transcription-qPCR and Western blotting assays demonstrated that suppression of erythroid differentiation was associated with sharp inhibition of the expression of α-globin and γ-globin mRNA accumulation. Inhibition of accumulation of ζ-globin and ε-globin mRNAs was also observed. In addition, we provide in silico studies suggesting a direct interaction between SARS-CoV-2 Spike protein and Hb Portland, that is the major hemoglobin produced by K562 cells. This study thus provides information suggesting the need of great attention on possible alteration of hematopoietic parameters following SARS-CoV-2 infection and/or COVID-19 vaccination.


Asunto(s)
COVID-19 , Leucemia Eritroblástica Aguda , Humanos , Células K562 , Plicamicina/farmacología , Plicamicina/metabolismo , Vacunas contra la COVID-19/metabolismo , Vacuna BNT162 , Leucemia Eritroblástica Aguda/metabolismo , COVID-19/prevención & control , COVID-19/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Hemoglobinas/metabolismo , ARN Mensajero/genética , Células Eritroides/metabolismo
3.
Blood ; 141(18): 2245-2260, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-36735909

RESUMEN

The NFIA-ETO2 fusion is the product of a t(1;16)(p31;q24) chromosomal translocation, so far, exclusively found in pediatric patients with pure erythroid leukemia (PEL). To address the role for the pathogenesis of the disease, we facilitated the expression of the NFIA-ETO2 fusion in murine erythroblasts (EBs). We observed that NFIA-ETO2 significantly increased proliferation and impaired erythroid differentiation of murine erythroleukemia cells and of primary fetal liver-derived EBs. However, NFIA-ETO2-expressing EBs acquired neither aberrant in vitro clonogenic activity nor disease-inducing potential upon transplantation into irradiated syngenic mice. In contrast, in the presence of 1 of the most prevalent erythroleukemia-associated mutations, TP53R248Q, expression of NFIA-ETO2 resulted in aberrant clonogenic activity and induced a fully penetrant transplantable PEL-like disease in mice. Molecular studies support that NFIA-ETO2 interferes with erythroid differentiation by preferentially binding and repressing erythroid genes that contain NFI binding sites and/or are decorated by ETO2, resulting in a activity shift from GATA- to ETS-motif-containing target genes. In contrast, TP53R248Q does not affect erythroid differentiation but provides self-renewal and survival potential, mostly via downregulation of known TP53 targets. Collectively, our work indicates that NFIA-ETO2 initiates PEL by suppressing gene expression programs of terminal erythroid differentiation and cooperates with TP53 mutation to induce erythroleukemia.


Asunto(s)
Leucemia Eritroblástica Aguda , Proteínas Represoras , Animales , Ratones , Proteínas Represoras/genética , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/metabolismo , Diferenciación Celular/genética , Eritroblastos/metabolismo , Factores de Transcripción NFI/metabolismo
4.
Food Chem Toxicol ; 168: 113336, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35963475

RESUMEN

The novel di-and triphenyltin(IV) dithiocarbamate compounds represented as RnSnL2 (where R = C4H9, C6H5; n = 2,3; L = N,N-dithiocarbamate), Ph2Sn(N,N-diisopropyldithiocarbamate) (OC1), Ph3Sn(N,N-diisopropyldithiocarbamate) (OC2), Ph2Sn(N,N-diallyldithiocarbamate) (OC3), Ph3Sn(N,N-diallyldithiocarbamate) (OC4), and Ph2Sn(N,N-diethyldithiocarbamate) (OC5) were assessed for their cytotoxicity in K562 human erythroleukemia cells. All compounds inhibited the growth of cells at low micromolar concentrations (<10 µM), and the mechanism underlying their antiproliferative effects on K562 cells was apoptosis, as corroborated by the exposure of plasma membrane phosphatidylserine. OC2, which showed the most promising antiproliferative activity, was selected for further analyses. The results demonstrated that OC2 induced apoptosis in K562 cells via an intrinsic mitochondrial pathway triggered upon DNA damage, an early apoptotic signal. Subsequently, OC2 produced excessive intracellular reactive oxygen species. The role of oxidative stress was corroborated by the significant reduction in GSH levels and percentage of apoptosis in NAC-pretreated cells. OC2 could arrest the cell cycle progression in the S phase. These new findings elucidate the antiproliferative potential of OC2 in the K562 human erythroleukemia cells and warrant further investigation, specifically to determine the exact signaling pathway underlying its antileukemic efficacy.


Asunto(s)
Leucemia Eritroblástica Aguda , Apoptosis , Daño del ADN , Ditiocarba/análogos & derivados , Ditiocarba/metabolismo , Ditiocarba/farmacología , Humanos , Células K562 , Leucemia Eritroblástica Aguda/tratamiento farmacológico , Leucemia Eritroblástica Aguda/metabolismo , Mitocondrias , Compuestos Orgánicos de Estaño , Fosfatidilserinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
5.
Oxid Med Cell Longev ; 2022: 5885342, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35140839

RESUMEN

Acute erythroleukemia (AEL) is an infrequent subtype of acute myeloid leukemia (AML) with worse prognosis. Though the last decade has seen major advances in the novel features and genomic landscape in AEL, there is still a lack of specific therapeutic targets and effective treatment approaches for this disease. Here, we found a novel oncogene KEL that specifically and aberrantly expressed in patients with AEL. In this study, we demonstrated that KEL promoted cell proliferation and the downregulation of KEL reversed drug resistance in AEL cells to JQ1. Our findings suggested that KEL contributed to gain of H3K27 acetylation and promoted erythroid differentiation induced by GATA1. Additionally, GATA1 and TAL1 as cotranscription factors (TFs) modulated the expression of KEL. Maintaining cell viability and differentiation, KEL also played parts in the immune evasion of tumor cells. Our work expands the current knowledge regarding molecular mechanisms involved in cancer onset and progression, offering promising therapeutic target to broaden the treatment options.


Asunto(s)
Carcinogénesis/genética , Progresión de la Enfermedad , Leucemia Eritroblástica Aguda/genética , Glicoproteínas de Membrana/genética , Metaloendopeptidasas/genética , Oncogenes , Transducción de Señal/genética , Animales , Estudios de Casos y Controles , Diferenciación Celular/genética , Proliferación Celular/genética , Regulación hacia Abajo/genética , Factor de Transcripción GATA1/metabolismo , Regulación Leucémica de la Expresión Génica , Células HEK293 , Humanos , Células K562 , Leucemia Eritroblástica Aguda/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Metaloendopeptidasas/metabolismo , Ratones , Ratones Endogámicos NOD , Trasplante de Neoplasias/métodos , Transfección
6.
FASEB J ; 36(3): e22220, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35195928

RESUMEN

Erythroid differentiation (ED) is a complex cellular process entailing morphologically distinct maturation stages of erythroblasts during terminal differentiation. Studies of actin filament (F-actin) assembly and organization during terminal ED have revealed essential roles for the F-actin pointed-end capping proteins, tropomodulins (Tmod1 and Tmod3). Tmods bind tropomyosins (Tpms), which enhance Tmod capping and F-actin stabilization. Tmods can also nucleate F-actin assembly, independent of Tpms. Tmod1 is present in the red blood cell (RBC) membrane skeleton, and deletion of Tmod1 in mice leads to a mild compensated anemia due to mis-regulated F-actin lengths and membrane instability. Tmod3 is not present in RBCs, and global deletion of Tmod3 leads to embryonic lethality in mice with impaired ED. To further decipher Tmod3's function during ED, we generated a Tmod3 knockout in a mouse erythroleukemia cell line (Mel ds19). Tmod3 knockout cells appeared normal prior to ED, but showed defects during progression of ED, characterized by a marked failure to reduce cell and nuclear size, reduced viability, and increased apoptosis. Tmod3 does not assemble with Tmod1 and Tpms into the Triton X-100 insoluble membrane skeleton during ED, and loss of Tmod3 had no effect on α1,ß1-spectrin and protein 4.1R assembly into the membrane skeleton. However, F-actin, Tmod1 and Tpms failed to assemble into the membrane skeleton during ED in absence of Tmod3. We propose that Tmod3 nucleation of F-actin assembly promotes incorporation of Tmod1 and Tpms into membrane skeleton F-actin, and that this is integral to morphological maturation and cell survival during erythroid terminal differentiation.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Eritroblastos/citología , Eritropoyesis , Leucemia Eritroblástica Aguda/metabolismo , Tropomodulina/metabolismo , Animales , Línea Celular Tumoral , Eritroblastos/metabolismo , Leucemia Eritroblástica Aguda/sangre , Ratones , Multimerización de Proteína , Espectrina/metabolismo , Tropomodulina/genética
8.
Int J Mol Sci ; 22(21)2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34769424

RESUMEN

A series of heterocyclic compounds containing a spiro-fused pyrrolo[3,4-a]pyrrolizine and tryptanthrin framework have been synthesized and studied as potential antitumor agents. Cytotoxicity of products was screened against human erythroleukemia (K562) and human cervical carcinoma (HeLa) cell lines. Among the screened compounds. 4a, 4b and 5a were active against human erythroleukemia (K562) cell line, while 4a and 5a were active against cervical carcinoma (HeLa) cell line. In agreement with the DNA cytometry studies, the tested compounds have achieved significant cell-cycle perturbation with higher accumulation of cells in G2/M phase and induced apoptosis. Using confocal microscopy, we found that with 4a and 5a treatment of HeLa cells, actin filaments disappeared, and granular actin was distributed diffusely in the cytoplasm in 76-91% of cells. We discovered that HeLa cells after treatment with compounds 4a and 5a significantly reduced the number of cells with filopodium-like membrane protrusions (from 63 % in control cells to 29% after treatment) and a decrease in cell motility.


Asunto(s)
Antineoplásicos/farmacología , Leucemia Eritroblástica Aguda/tratamiento farmacológico , Pirrolidinas/síntesis química , Pirrolidinas/farmacología , Quinazolinas/farmacología , Compuestos de Espiro/farmacología , Neoplasias del Cuello Uterino/tratamiento farmacológico , Antineoplásicos/síntesis química , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/patología , Quinazolinas/síntesis química , Compuestos de Espiro/síntesis química , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología
9.
Nat Commun ; 12(1): 6241, 2021 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-34716321

RESUMEN

Precise control of gene expression during differentiation relies on the interplay of chromatin and nuclear structure. Despite an established contribution of nuclear membrane proteins to developmental gene regulation, little is known regarding the role of inner nuclear proteins. Here we demonstrate that loss of the nuclear scaffolding protein Matrin-3 (Matr3) in erythroid cells leads to morphological and gene expression changes characteristic of accelerated maturation, as well as broad alterations in chromatin organization similar to those accompanying differentiation. Matr3 protein interacts with CTCF and the cohesin complex, and its loss perturbs their occupancy at a subset of sites. Destabilization of CTCF and cohesin binding correlates with altered transcription and accelerated differentiation. This association is conserved in embryonic stem cells. Our findings indicate Matr3 negatively affects cell fate transitions and demonstrate that a critical inner nuclear protein impacts occupancy of architectural factors, culminating in broad effects on chromatin organization and cell differentiation.


Asunto(s)
Cromatina/química , Leucemia Eritroblástica Aguda/patología , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Factor de Unión a CCCTC , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/fisiología , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Células Madre Embrionarias/fisiología , Células Eritroides/patología , Leucemia Eritroblástica Aguda/metabolismo , Ratones Noqueados , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas de Unión al ARN/genética , Cohesinas
10.
Blood ; 138(9): 790-805, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34473231

RESUMEN

Therapy-related myeloid neoplasms (t-MNs) are high-risk late effects with poorly understood pathogenesis in cancer survivors. It has been postulated that, in some cases, hematopoietic stem and progenitor cells (HSPCs) harboring mutations are selected for by cytotoxic exposures and transform. Here, we evaluate this model in the context of deficiency of CUX1, a transcription factor encoded on chromosome 7q and deleted in half of t-MN cases. We report that CUX1 has a critical early role in the DNA repair process in HSPCs. Mechanistically, CUX1 recruits the histone methyltransferase EHMT2 to DNA breaks to promote downstream H3K9 and H3K27 methylation, phosphorylated ATM retention, subsequent γH2AX focus formation and propagation, and, ultimately, 53BP1 recruitment. Despite significant unrepaired DNA damage sustained in CUX1-deficient murine HSPCs after cytotoxic exposures, they continue to proliferate and expand, mimicking clonal hematopoiesis in patients postchemotherapy. As a consequence, preexisting CUX1 deficiency predisposes mice to highly penetrant and rapidly fatal therapy-related erythroleukemias. These findings establish the importance of epigenetic regulation of HSPC DNA repair and position CUX1 as a gatekeeper in myeloid transformation.


Asunto(s)
Cromosomas de los Mamíferos , Reparación del ADN , Epigénesis Genética , Regulación Leucémica de la Expresión Génica , Proteínas de Homeodominio , Leucemia Eritroblástica Aguda , Proteínas de Neoplasias , Neoplasias Primarias Secundarias , Proteínas Nucleares , Proteínas Represoras , Animales , Cromosomas de los Mamíferos/genética , Cromosomas de los Mamíferos/metabolismo , Hematopoyesis Clonal , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/metabolismo , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Primarias Secundarias/genética , Neoplasias Primarias Secundarias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
11.
Mol Med Rep ; 24(5)2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34458927

RESUMEN

Natural agents have been used to restart the process of differentiation that is inhibited during leukemic transformation of hematopoietic stem or progenitor cells. Autophagy is a housekeeping pathway that maintains cell homeostasis against stress by recycling macromolecules and organelles and plays an important role in cell differentiation. In the present study, an experimental model was established to investigate the involvement of autophagy in the megakaryocyte differentiation of human erythroleukemia (HEL) cells induced by diosgenin [also known as (25R)­Spirosten­5­en­3b­ol]. It was demonstrated that Atg7 expression was upregulated from day 1 of diosgenin­induced differentiation and was accompanied by a significant elevation in the conversion of light chain 3 A/B (LC3­A/B)­I to LC3­A/B­II. Autophagy was modulated before or after the induction of megakaryocyte differentiation using 3­methyladenine (3­MA, autophagy inhibitor) and metformin (Met, autophagy initiation activator). 3­MA induced a significant accumulation of the LC3 A/B­II form at day 8 of differentiation. It was revealed that 3­MA had a significant repressive effect on the nuclear (polyploidization) and membrane glycoprotein V [(GpV) expression] maturation. On the other hand, autophagy activation increased GpV genomic expression, but did not change the nuclear maturation profile after HEL cells treatment with Met. It was concluded that autophagy inhibition had a more prominent effect on the diosgenin­differentiated cells than autophagy activation.


Asunto(s)
Autofagia/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diosgenina/farmacología , Leucemia Eritroblástica Aguda/metabolismo , Megacariocitos/metabolismo , Apoptosis/efectos de los fármacos , Autofagia/genética , Línea Celular , Humanos , ARN Mensajero/metabolismo
12.
Int J Mol Sci ; 22(15)2021 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-34360557

RESUMEN

Among the eight human glutathione peroxidase isoforms, glutathione peroxidase 4 (GPX4) is the only enzyme capable of reducing complex lipid peroxides to the corresponding alcohols. In mice, corruption of the Gpx4 gene leads to embryonic lethality and more detailed expression silencing studies have implicated the enzyme in several physiological processes (e.g., embryonal cerebrogenesis, neuronal function, male fertility). Experiments with conditional knockout mice, in which expression of the Gpx4 gene was silenced in erythroid precursors, indicated a role of Gpx4 in erythropoiesis. To test this hypothesis in a cellular in vitro model we transfected mouse erythroleukemia cells with a Gpx4 siRNA construct and followed the expression kinetics of erythropoietic gene products. Our data indicate that Gpx4 is expressed at high levels in mouse erythroleukemia cells and that expression silencing of the Gpx4 gene delays in vitro erythropoiesis. However, heterozygous expression of a catalytically inactive Gpx4 mutant (Gpx4+/Sec46Ala) did not induce a defective erythropoietic phenotype in different in vivo and ex vivo models. These data suggest that Gpx4 plays a role in erythroid differentiation of mouse erythroleukemia cells but that heterozygous expression of a catalytically inactive Gpx4 is not sufficient to compromise in vivo and ex vivo erythropoiesis.


Asunto(s)
Eritropoyesis , Leucemia Eritroblástica Aguda/patología , Mitocondrias/patología , Estrés Oxidativo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Animales , Leucemia Eritroblástica Aguda/etiología , Leucemia Eritroblástica Aguda/metabolismo , Masculino , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo
13.
Proc Natl Acad Sci U S A ; 118(34)2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34404729

RESUMEN

Chimeric antigen receptor (CAR)-engineered T cell therapies have been recognized as powerful strategies in cancer immunotherapy; however, the clinical application of CAR-T is currently constrained by severe adverse effects in patients, caused by excessive cytotoxic activity and poor T cell control. Herein, we harnessed a dietary molecule resveratrol (RES)-responsive transactivator and a transrepressor to develop a repressible transgene expression (RESrep) device and an inducible transgene expression (RESind) device, respectively. After optimization, these tools enabled the control of CAR expression and CAR-mediated antitumor function in engineered human cells. We demonstrated that a resveratrol-repressible CAR expression (RESrep-CAR) device can effectively inhibit T cell activation upon resveratrol administration in primary T cells and a xenograft tumor mouse model. Additionally, we exhibit how a resveratrol-inducible CAR expression (RESind-CAR) device can achieve fine-tuned and reversible control over T cell activation via a resveratrol-titratable mechanism. Furthermore, our results revealed that the presence of RES can activate RESind-CAR T cells with strong anticancer cytotoxicity against cells in vitro and in vivo. Our study demonstrates the utility of RESrep and RESind devices as effective tools for transgene expression and illustrates the potential of RESrep-CAR and RESind-CAR devices to enhance patient safety in precision cancer immunotherapies.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Inmunoterapia Adoptiva/métodos , Leucemia Eritroblástica Aguda/inmunología , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Animales , Apoptosis , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/terapia , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Immunother Cancer ; 9(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33737343

RESUMEN

BACKGROUND: CAR-T cells immunotherapy is a breakthrough in the treatment of hematological malignancies such as acute lymphoblastic leukemia (ALL) and B-cell malignancies. However, CAR-T therapies face major hurdles such as the lack of tumor-specific antigen (TSA), and immunosuppressive tumor microenvironment sometimes caused by the tumorous expression of immune checkpoints (ICPs) such as HLA-G. Indeed, HLA-G is remarkable because it is both a potent ICP and a TSA. HLA-G tumor expression causes immune escape by impairing innate and adaptive immune responses and by inducing a suppressive microenvironment. Yet, to date, no immunotherapy targets it. METHODS: We have developed two anti-HLA-G third-generation CARs based on new anti-HLA-G monoclonal antibodies. RESULTS: Anti-HLA-G CAR-T cells were specific for immunosuppressive HLA-G isoforms. HLA-G-activated CAR-T cells polarized toward T helper 1, and became cytotoxic against HLA-G+ tumor cells. In vivo, anti-HLA-G CAR-T cells were able to control and eliminate HLA-G+ tumor cells. The interaction of tumor-HLA-G with interleukin (IL)T2-expressing T cells is known to result in effector T cell functional inhibition, but anti-HLA-G CAR-T cells were insensitive to this inhibition and still exerted their function even when expressing ILT2. Lastly, we show that anti-HLA-G CAR-T cells differentiated into long-term memory effector cells, and seemed not to lose function even after repeated stimulation by HLA-G-expressing tumor cells. CONCLUSION: We report for the first time that HLA-G, which is both a TSA and an ICP, constitutes a valid target for CAR-T cell therapy to specifically target and eliminate both tumor cells and HLA-G+ suppressive cells.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Antígenos HLA-G/metabolismo , Inmunoterapia Adoptiva , Leucemia Eritroblástica Aguda/terapia , Células T de Memoria/trasplante , Receptores Quiméricos de Antígenos/genética , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Antígenos CD/metabolismo , Diferenciación Celular , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Antígenos HLA-G/inmunología , Humanos , Memoria Inmunológica , Células K562 , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/inmunología , Leucemia Eritroblástica Aguda/metabolismo , Receptor Leucocitario Tipo Inmunoglobulina B1/metabolismo , Células T de Memoria/inmunología , Células T de Memoria/metabolismo , Ratones Endogámicos NOD , Ratones SCID , Fenotipo , Receptores Quiméricos de Antígenos/metabolismo , Factores de Tiempo , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Physiol Rep ; 9(3): e14745, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33587325

RESUMEN

Hematopoietic disorders, particularly hemolytic anemias, commonly lead to bone loss. We have previously reported that actively proliferating cancer cells stimulate osteoclastogenesis from late precursors in a RANKL-independent manner. We theorized that cancer cells exploit the physiological role of bone resorption to support expanding hematopoietic bone marrow and examined if hematopoietic cells can trigger osteoclastogenesis. Using phlebotomy-induced acute anemia in mice, we found strong correlation between augmented erythropoiesis and increased osteoclastogenesis. Conditioned medium (CM) from K562 erythroleukemia cells and primary mouse erythroblasts stimulated osteoclastogenesis when added to RANKL-primed precursors from mouse bone marrow or RAW264.7 cells. Using immunoblotting and mass spectrometry, PRDX2 was identified as a factor produced by erythroid cells in vitro and in vivo. PRDX2 was detected in K562-derived exosomes, and inhibiting exosomal release significantly decreased the osteoclastogenic capacity of K562 CM. Recombinant PRDX2 induced osteoclast formation from RANKL-primed primary or RAW 264.7 precursors to levels comparable to achieved with continuous RANKL treatment. Thus, increased bone marrow erythropoiesis secondary to anemia leads to upregulation of PRDX2, which is released in the exosomes and acts to induce osteoclast formation. Increased bone resorption by the osteoclasts expands bone marrow cavity, which likely plays a supporting role to increase blood cell production.


Asunto(s)
Anemia/metabolismo , Eritropoyesis , Exosomas/metabolismo , Osteoclastos/metabolismo , Osteogénesis , Comunicación Paracrina , Peroxirredoxinas/metabolismo , Anemia/sangre , Anemia/patología , Animales , Modelos Animales de Enfermedad , Eritroblastos/metabolismo , Femenino , Humanos , Células K562 , Leucemia Eritroblástica Aguda/metabolismo , Ratones , Ratones Endogámicos C57BL , Osteoclastos/patología , Peroxirredoxinas/sangre , Células RAW 264.7 , Transducción de Señal
18.
Curr Mol Med ; 21(10): 819-831, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33475071

RESUMEN

Acute erythroid leukemia (AEL) is a subtype of acute myeloid leukemia (AML) with features such as accumulation of maturation-arrested erythroblasts. Compared with AML, the progression of AEL is faster and the prognosis to available therapy is worse. However, its categorization is still being updated and the pathophysiology of AEL is still under research, making diagnosis and chemotherapy challenging for physicians. To achieve better outcomes, therapies should be optimized and new drugs should be developed. In this review, we summarize current strategies of diagnosis and therapies of AEL, and discuss prospective targets for chemotherapeutic agents based on the biological characteristics of AEL neoplastic cells as well as transcriptional factors and pathways related to erythroid differentiation.


Asunto(s)
Diferenciación Celular/genética , Leucemia Eritroblástica Aguda , Mutación , Proteínas de Neoplasias , Factores de Transcripción , Humanos , Leucemia Eritroblástica Aguda/tratamiento farmacológico , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Anticancer Agents Med Chem ; 21(4): 498-507, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32538736

RESUMEN

BACKGROUND AND PURPOSE: Leukemia is considered a top-listed ailment, according to WHO, which contributes to the death of a major population of the world every year. Paris Saponin VII (PS), a saponin which was isolated from the roots of Trillium kamtschaticum, from our group, was reported to provide hemostatic, cytotoxic and antimicrobial activities. However, its molecular mechanism underlying the anti-proliferative effects remains unclear. Thus, this study hypothesized to assess that mechanism in PS treated HEL cells. METHODS: The MTT assay was used to analyze the PS inhibited cell viability in the HEL cells. We further found that PS could induce S phase cell cycle arrest through flow cytometry as well as the western blot analysis of intrinsic and extrinsic apoptotic molecules. RESULTS: The MTT assay showed the IC50 concentration of PS as 0.667µM. The study revealed that PS treatment inhibits cell proliferation dose-dependently. It further caused mitochondrial membrane potential changes by PS treatment. Mechanistic protein expression revealed a dose-dependent upsurge for Bid and Bim molecules, while Bcl2 and PARP expression levels were significantly (P<0.05) down-regulated in PS treated HEL cells resulting in caspase -3 release and increased the Bim levels upon 24h of incubation. CONCLUSION: These findings indicate that PS possesses an excellent anti-leukemic activity via the regulation of the mitochondrial pathway, leading to S phase cell cycle arrest and caspase-dependent apoptosis, suggesting it as a potential alternative chemotherapeutic agent for leukemia patients.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Leucemia Eritroblástica Aguda/tratamiento farmacológico , Extractos Vegetales/farmacología , Saponinas/farmacología , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/aislamiento & purificación , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/patología , Membranas Mitocondriales/efectos de los fármacos , Estructura Molecular , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Saponinas/química , Saponinas/aislamiento & purificación , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Células Tumorales Cultivadas
20.
Syst Biol Reprod Med ; 66(6): 378-386, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32972244

RESUMEN

The Origin Replication Complex subunit 4 (ORC4) is one in six subunits of the Origin Replication Complexes (ORCs) which is essential for initiating licensing at DNA replication origins and recruiting adaptor molecules necessary for various cellular processes. Previously, we reported that ORC4 also plays a vital role in polar body extrusion (PBE) during oogenesis in which half the chromosomes are extruded from the oocyte. We hypothesized that ORC4 might play a broader role in chromatin elimination. We tested its role in enucleation during the development of erythrocytes. Murine erythroleukemia (MEL) cells can be propagated in culture indefinitely and can be induced to enucleate their DNA by treatment with Vacuolin-1, thereby mimicking normal erythrocyte enucleation. We found that ORC4 appeared around the nuclei of the MEL cells with Vacuolin-1 treatment, gradually increasing in thickness before enucleation. We then tested whether ORC4 was required for MEL enucleation by down regulating ORC4 with siRNA-ORC4 during Vacuolin-1 treatment and found that this prevented MEL enucleation. These data are consistent with the model that ORC4 is required for erythroblast enucleation just as it is for oocyte PBE. They suggest a new model in which ORC4 expression is a marker for the initiation to the enucleation pathway.


Asunto(s)
Núcleo Celular/metabolismo , Eritroblastos/metabolismo , Leucemia Eritroblástica Aguda/metabolismo , Oogénesis , Complejo de Reconocimiento del Origen/metabolismo , Cuerpos Polares/metabolismo , Animales , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/genética , Núcleo Celular/patología , Replicación del ADN , Eritroblastos/efectos de los fármacos , Eritroblastos/patología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/patología , Ratones , Complejo de Reconocimiento del Origen/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...