Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Cell Mol Life Sci ; 78(19-20): 6533-6540, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34424346

RESUMEN

Chemotherapy-induced cognitive impairment (CICI) has been observed in a large fraction of cancer survivors. Although many of the chemotherapeutic drugs do not cross the blood-brain barrier, following treatment, the structure and function of the brain are altered and cognitive dysfunction occurs in a significant number of cancer survivors. The means by which CICI occurs is becoming better understood, but there still remain unsolved questions of the mechanisms involved. The hypotheses to explain CICI are numerous. More than 50% of FDA-approved cancer chemotherapy agents are associated with reactive oxygen species (ROS) that lead to oxidative stress and activate a myriad of pathways as well as inhibit pathways necessary for proper brain function. Oxidative stress triggers the activation of different proteins, one in particular is tumor necrosis factor alpha (TNFα). Following treatment with various chemotherapy agents, this pro-inflammatory cytokine binds to its receptors at the blood-brain barrier and translocates to the parenchyma via receptor-mediated endocytosis. Once in brain, TNFα initiates pathways that may eventually lead to neuronal death and ultimately cognitive impairment. TNFα activation of the c-jun N-terminal kinases (JNK) and Janus kinase-signal transducer and activator of transcription (JAK/STAT) pathways may contribute to both memory decline and loss of higher executive functions reported in patients after chemotherapy treatment. Chemotherapy also affects the brain's antioxidant capacity, allowing for accumulation of ROS. This review expands on these topics to provide insights into the possible mechanisms by which the intersection of oxidative stress and TNFΑ are involved in chemotherapy-induced cognitive impairment.


Asunto(s)
Antineoplásicos/efectos adversos , Deterioro Cognitivo Relacionado con la Quimioterapia/metabolismo , Estrés Oxidativo/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Humanos , Transducción de Señal/efectos de los fármacos
2.
Int J Mol Sci ; 23(1)2021 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-35008785

RESUMEN

Microglia/astrocyte and B cell neuroimmune responses are major contributors to the neurological deficits after traumatic spinal cord injury (SCI). Bruton tyrosine kinase (BTK) activation mechanistically links these neuroimmune mechanisms. Our objective is to use Ibrutinib, an FDA-approved BTK inhibitor, to inhibit the neuroimmune cascade thereby improving locomotor recovery after SCI. Rat models of contusive SCI, Western blot, immunofluorescence staining imaging, flow cytometry analysis, histological staining, and behavioral assessment were used to evaluate BTK activity, neuroimmune cascades, and functional outcomes. Both BTK expression and phosphorylation were increased at the lesion site at 2, 7, 14, and 28 days after SCI. Ibrutinib treatment (6 mg/kg/day, IP, starting 3 h post-injury for 7 or 14 days) reduced BTK activation and total BTK levels, attenuated the injury-induced elevations in Iba1, GFAP, CD138, and IgG at 7 or 14 days post-injury without reduction in CD45RA B cells, improved locomotor function (BBB scores), and resulted in a significant reduction in lesion volume and significant improvement in tissue-sparing 11 weeks post-injury. These results indicate that Ibrutinib exhibits neuroprotective effects by blocking excessive neuroimmune responses through BTK-mediated microglia/astroglial activation and B cell/antibody response in rat models of SCI. These data identify BTK as a potential therapeutic target for SCI.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Neuroinmunomodulación , Recuperación de la Función , Traumatismos de la Médula Espinal/inmunología , Adenina/análogos & derivados , Adenina/farmacología , Adenina/uso terapéutico , Agammaglobulinemia Tirosina Quinasa/metabolismo , Animales , Formación de Anticuerpos/efectos de los fármacos , Astrocitos/patología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Peso Corporal/efectos de los fármacos , Proteínas de Unión al Calcio/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunoglobulina G/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/patología , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Microglía/patología , Actividad Motora/efectos de los fármacos , Neuroinmunomodulación/efectos de los fármacos , Fosforilación/efectos de los fármacos , Piperidinas/farmacología , Piperidinas/uso terapéutico , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/metabolismo , Ratas , Recuperación de la Función/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Bazo/patología , Sindecano-1/metabolismo , Resultado del Tratamiento , Regulación hacia Arriba/efectos de los fármacos
3.
Blood ; 131(26): 2943-2954, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29695515

RESUMEN

Prostate apoptosis response-4 (Par-4), a proapoptotic tumor suppressor protein, is downregulated in many cancers including renal cell carcinoma, glioblastoma, endometrial, and breast cancer. Par-4 induces apoptosis selectively in various types of cancer cells but not normal cells. We found that chronic lymphocytic leukemia (CLL) cells from human patients and from Eµ-Tcl1 mice constitutively express Par-4 in greater amounts than normal B-1 or B-2 cells. Interestingly, knockdown of Par-4 in human CLL-derived Mec-1 cells results in a robust increase in p21/WAF1 expression and decreased growth due to delayed G1-to-S cell-cycle transition. Lack of Par-4 also increased the expression of p21 and delayed CLL growth in Eµ-Tcl1 mice. Par-4 expression in CLL cells required constitutively active B-cell receptor (BCR) signaling, as inhibition of BCR signaling with US Food and Drug Administration (FDA)-approved drugs caused a decrease in Par-4 messenger RNA and protein, and an increase in apoptosis. In particular, activities of Lyn, a Src family kinase, spleen tyrosine kinase, and Bruton tyrosine kinase are required for Par-4 expression in CLL cells, suggesting a novel regulation of Par-4 through BCR signaling. Together, these results suggest that Par-4 may play a novel progrowth rather than proapoptotic role in CLL and could be targeted to enhance the therapeutic effects of BCR-signaling inhibitors.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Regulación Leucémica de la Expresión Génica , Leucemia Linfocítica Crónica de Células B/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Ciclo Celular , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Eliminación de Gen , Humanos , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/patología , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal , Regulación hacia Arriba
4.
J Immunol ; 200(12): 4180-4189, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29712773

RESUMEN

Chronic lymphocytic leukemia (CLL) patients progressively develop an immunosuppressive state. CLL patients have more plasma IL-10, an anti-inflammatory cytokine, than healthy controls. In vitro human CLL cells produce IL-10 in response to BCR cross-linking. We used the transgenic Eµ-T cell leukemia oncogene-1 (TCL1) mouse CLL model to study the role of IL-10 in CLL associated immunosuppression. Eµ-TCL mice spontaneously develop CLL because of a B cell-specific expression of the oncogene, TCL1. Eµ-TCL1 mouse CLL cells constitutively produce IL-10, which is further enhanced by BCR cross-linking, CLL-derived IL-10 did not directly affect survival of murine or human CLL cells in vitro. We tested the hypothesis that the CLL-derived IL-10 has a critical role in CLL disease in part by suppressing the host immune response to the CLL cells. In IL-10R-/- mice, wherein the host immune cells are unresponsive to IL-10-mediated suppressive effects, there was a significant reduction in CLL cell growth compared with wild type mice. IL-10 reduced the generation of effector CD4 and CD8 T cells. We also found that activation of BCR signaling regulated the production of IL-10 by both murine and human CLL cells. We identified the transcription factor, Sp1, as a novel regulator of IL-10 production by CLL cells and that it is regulated by BCR signaling via the Syk/MAPK pathway. Our results suggest that incorporation of IL-10 blocking agents may enhance current therapeutic regimens for CLL by potentiating host antitumor immune response.


Asunto(s)
Interleucina-10/inmunología , Leucemia Linfocítica Crónica de Células B/inmunología , Animales , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos/inmunología , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas/inmunología , Transducción de Señal/inmunología
5.
Arterioscler Thromb Vasc Biol ; 38(11): 2706-2717, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30354229

RESUMEN

Objective- Continuous T-cell production from thymus is essential in replenishing naïve T-cell pool and maintaining optimal T-cell functions. However, the underlying mechanisms regulating the T-cell development in thymus remains largely unknown. Approach and Results- We identified SR-BI (scavenger receptor class B type 1), an HDL (high-density lipoprotein) receptor, as a novel modulator in T-cell development. We found that SR-BI deficiency in mice led to reduced thymus size and decreased T-cell production, which was accompanied by narrowed peripheral naïve T-cell pool. Further investigation revealed that SR-BI deficiency impaired progenitor thymic homing, causing a dramatic reduction in the percentage of earliest thymic progenitors, but did not affect other downstream T-cell developmental steps inside the thymus. As a result of the impaired progenitor thymic homing, SR-BI-deficient mice displayed delayed thymic regeneration postirradiation. Using a variety of experimental approaches, we revealed that the impaired T-cell development in SR-BI-deficient mice was not caused by hematopoietic SR-BI deficiency or SR-BI deficiency-induced hypercholesterolemia, but mainly attributed to the SR-BI deficiency in adrenal glands, as adrenal-specific SR-BI-deficient mice exhibited similar defects in T-cell development and thymic regeneration with SR-BI-deficient mice. Conclusions- This study demonstrates that SR-BI deficiency impaired T-cell development and delayed thymic regeneration by affecting progenitor thymic homing in mice, elucidating a previously unrecognized link between SR-BI and adaptive immunity.


Asunto(s)
Glándulas Suprarrenales/metabolismo , Proliferación Celular , Activación de Linfocitos , Regeneración , Receptores Depuradores de Clase B/metabolismo , Linfocitos T/metabolismo , Timocitos/metabolismo , Timo/metabolismo , Inmunidad Adaptativa , Glándulas Suprarrenales/inmunología , Animales , Células Cultivadas , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Antígenos Comunes de Leucocito/deficiencia , Antígenos Comunes de Leucocito/genética , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Receptores de LDL/deficiencia , Receptores de LDL/genética , Receptores Depuradores de Clase B/deficiencia , Receptores Depuradores de Clase B/genética , Transducción de Señal , Bazo/inmunología , Bazo/metabolismo , Linfocitos T/inmunología , Timocitos/inmunología , Timo/inmunología
6.
Nanomedicine ; 13(5): 1627-1636, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28300659

RESUMEN

In this report milk-derived exosomes have been investigated for oral delivery of the chemotherapeutic drug paclitaxel (PAC) as an alternative to conventional i.v. therapy for improved efficacy and reduced toxicity. PAC-loaded exosomes (ExoPAC) were found to have a particle size of ~108 nm, a narrow particle size distribution (PDI ~0.190), zeta potential (~ -7 mV) and a practical loading efficiency of ~8%. Exosomes and ExoPAC exhibited excellent stability in the presence of simulated-gastrointestinal fluids, and during the storage at -80 °C. A sustained release of PAC was also observed up to 48 h in vitro using PBS (pH 6.8). Importantly, ExoPAC delivered orally showed significant tumor growth inhibition (60%; P<0.001) against human lung tumor xenografts in nude mice. Treatment with i.p. PAC at the same dose as ExoPAC, however, showed modest but statistically insignificant inhibition (31%). Moreover, ExoPAC demonstrated remarkably lower systemic and immunologic toxicities as compared to i.v. PAC.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Exosomas , Paclitaxel/administración & dosificación , Animales , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Ratones Desnudos , Leche
7.
J Immunol ; 189(7): 3355-67, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22942427

RESUMEN

Dendritic cells (DCs) play an essential role in regulation of immune responses. In the periphery, Ag presentation by DCs is critical for adaptive responses; for this reason, DCs are often targets of adjuvants that enhance vaccine responses. Activated mature DCs enhance B cell activation and differentiation by providing cytokines like BAFF and a proliferation-inducing ligand. However, the role of immature DCs in B cell tolerance is not well studied. Recently, mouse immature bone marrow-derived DCs (iBMDCs) have been shown to suppress anti-IgM-induced B cell activation. In this study, we tested the ability of mouse DCs to modulate B cell functions during TLR activation. We found that iBMDCs potently suppressed proliferation and differentiation of various B cell subsets on TLR stimulation. However, iBMDCs did not affect CD40-mediated B cell activation. Optimal suppression of B cell activation by iBMDCs required cell contact via the CD22 receptor on B cells. The B cell suppression was a property of iBMDCs or DCs resident in the bone marrow (BM), but not mature BM-derived DCs or DCs resident in the spleen. Presence of iBMDCs also enhanced the Ag-induced apoptotic response of BM B cells, suggesting that the suppressive effects of iBMDCs may have a role in B cell tolerance.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Células de la Médula Ósea/inmunología , Células Dendríticas/inmunología , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal/inmunología , Animales , Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/inmunología , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Antígenos de Linfocitos B/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos B/fisiología , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Bazo/citología , Bazo/inmunología , Bazo/metabolismo
8.
Blood ; 118(26): 6849-59, 2011 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-22039254

RESUMEN

The link between EBV infection and Burkitt lymphoma (BL) is strong, but the mechanism underlying that link has been elusive. We have developed a mouse model for EBV-associated BL in which LMP2A, an EBV latency protein, and MYC are expressed in B cells. Our model has demonstrated the ability of LMP2A to accelerate tumor onset, increase spleen size, and bypass p53 inactivation. Here we describe the results of total gene expression analysis of tumor and pretumor B cells from our transgenic mouse model. Although we see many phenotypic differences and changes in gene expression in pretumor B cells, the transcriptional profiles of tumor cells from LMP2A/λ-MYC and λ-MYC mice are strikingly similar, with fewer than 20 genes differentially expressed. We evaluated the functional significance of one of the most interesting differentially expressed genes, Egr1, and found that it was not required for acceleration of tumor onset by LMP2A. Our studies demonstrate the remarkable ability of LMP2A to affect the pretumor B-cell phenotype and tumorigenesis without substantially altering gene expression in tumor cells.


Asunto(s)
Linfoma de Burkitt/genética , Linfoma de Burkitt/virología , Perfilación de la Expresión Génica , Herpesvirus Humano 4/fisiología , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Linfocitos B/virología , Linfoma de Burkitt/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Genes myc/genética , Herpesvirus Humano 4/genética , Interacciones Huésped-Patógeno , Humanos , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/metabolismo , Leucosialina/genética , Leucosialina/metabolismo , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/metabolismo , Bazo/patología , Bazo/virología , Proteínas de la Matriz Viral/genética
9.
Leuk Lymphoma ; 63(8): 1810-1822, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35258388

RESUMEN

The chronic lymphocytic leukemia (CLL) microenvironment has been receiving an increasing amount of attention, but there is currently limited data surrounding how the microenvironment affects initial development of CLL. We determined that the spleen is the initial site of CLL growth through monitoring of transgenic Eµ-TCL1 mice that develop CLL. Subsequently, we isolated stromal cells from the spleens of Eµ-TCL1 mice (EMST cells) that induce CLL cell division in vitro. Both cell-cell contact and soluble factors were involved in EMST-induced CLL cell division. These stromal cells are present in significantly larger numbers in the spleen than other lymphoid organs. We also noted that splenectomy delayed CLL development in Eµ-TCL1 mice and completely prevented CLL development in adoptive transfer mice. Our findings will allow future studies surrounding the CLL microenvironment to focus upon the splenic stromal cells.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , Animales , Leucemia Linfocítica Crónica de Células B/genética , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas , Bazo , Células del Estroma , Microambiente Tumoral
10.
Brain Res ; 1782: 147840, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35183524

RESUMEN

Cranial radiation is important for treating both primary brain tumors and brain metastases. A potential delayed side effect of cranial radiation is neurocognitive function decline. Early detection of CNS injury might prevent further neuronal damage. Extracellular vesicles (EVs) have emerged as a potential diagnostic tool because of their unique membranous characteristics and cargos. We investigated whether EVs can be an early indicator of CNS injury by giving C57BJ/6 mice 10 Gy cranial IR. EVs were isolated from sera to quantify: 1) number of EVs using nanoparticle tracking analysis (NTA); 2) Glial fibrillary acidic protein (GFAP), an astrocyte marker; and 3) protein-bound 4-hydroxy-2-nonenal (HNE) adducts, an oxidative damage marker. Brain tissues were prepared for immunohistochemistry staining and protein immunoblotting. The results demonstrate: 1) increased GFAP levels (p < 0.05) in EVs, but not brain tissue, in the IR group; and 2) increased HNE-bound protein adduction levels (p < 0.05). The results support using EVs as an early indicator of cancer therapy-induced neuronal injury.


Asunto(s)
Lesiones Encefálicas , Vesículas Extracelulares , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Lesiones Encefálicas/etiología , Lesiones Encefálicas/metabolismo , Vesículas Extracelulares/metabolismo , Ratones , Neuronas/metabolismo , Proteínas/metabolismo
11.
Cancers (Basel) ; 13(15)2021 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-34359601

RESUMEN

Lung cancer is the leading cause of cancer-related deaths worldwide. Non-small-cell lung cancer (NSCLC) is the most common type accounting for 84% of all lung cancers. Paclitaxel (PAC) is a widely used drug in the treatment of a broad spectrum of human cancers, including lung. While efficacious, PAC generally is not well tolerated and its limitations include low aqueous solubility, and significant toxicity. To overcome the dose-related toxicity of solvent-based PAC, we utilized bovine colostrum-derived exosomes as a delivery vehicle for PAC for the treatment of lung cancer. Colostrum provided higher yield of exosomes and could be loaded with higher amount of PAC compared to mature milk. Exosomal formulation of PAC (ExoPAC) showed higher antiproliferative activity and inhibition of colony formation against A549 cells compared with PAC alone, and also showed antiproliferative activity against a drug-resistant variant of A549. To further enhance its efficacy, exosomes were attached with a tumor-targeting ligand, folic acid (FA). FA-ExoPAC given orally showed significant inhibition (>50%) of subcutaneous tumor xenograft while similar doses of PAC showed insignificant inhibition. In the orthotopic lung cancer model, oral dosing of FA-ExoPAC achieved greater efficacy (55% growth inhibition) than traditional i.v. PAC (24-32% growth inhibition) and similar efficacy as i.v. Abraxane (59% growth inhibition). The FA-ExoPAC given i.v. exceeded the therapeutic efficacy of Abraxane (76% growth inhibition). Finally, wild-type animals treated with p.o. ExoPAC did not show gross, systemic or immunotoxicity. Solvent-based PAC caused immunotoxicity which was either reduced or completely mitigated by its exosomal formulations. These studies show that a tumor-targeted oral formulation of PAC (FA-ExoPAC) significantly improved the overall efficacy and safety profile while providing a user-friendly, cost-effective alternative to bolus i.v. PAC and i.v. Abraxane.

12.
J Proteome Res ; 9(12): 6232-41, 2010 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-20945937

RESUMEN

Doxorubicin (DOX) is an anticancer drug used for the treatment of solid tumors. The ability of DOX to treat cancer is not specific to cancer cells; some of the cells that are normal may also become targets of DOX, thereby altering the normal cellular functions and eventual cell loss. DOX effects have been studied in detail in heart because of its ability to cause cardiomyopathy. The exact mechanism of DOX-induced cardiomyopathy is not completely understood. One of organs that can be affected by DOX is thymus. DOX treatment leads to degeneration of thymus; however, since thymus undergoes age-dependent degeneration, researchers have understudied the effect of DOX on thymus. In the present investigation, we studied the effects of DOX on thymus, an organ that is important for the T-cell maturation. DOX treatment led to loss of cortical cells, decrease lymphopoiesis and increased the number of Hassells corpuscles, a marker of thymus aging. Proteomics analysis led to identification of a number of thymic proteins whose expression are altered by in vivo DOX treatment. Taken together, these results are consistent with the notion that DOX-treatment leads to thymic senescence.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Doxorrubicina/farmacología , Linfocitos T/efectos de los fármacos , Timo/efectos de los fármacos , Adenosina Trifosfatasas/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Apolipoproteína A-I/metabolismo , Electroforesis en Gel Bidimensional , Linfopoyesis/efectos de los fármacos , Masculino , Ratones , Tamaño de los Órganos/efectos de los fármacos , Proteínas/análisis , Proteínas/metabolismo , Proteómica , Linfocitos T/metabolismo , Linfocitos T/patología , Timo/metabolismo , Timo/patología
13.
J Immunol ; 181(7): 4590-602, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18802061

RESUMEN

Egr-1 (early growth response gene-1) is an immediate early gene encoding a zinc finger motif-containing transcription factor. Upon cross-linking of BCR, mature B cells undergo proliferation with an increase in Egr-1 message. Immature B lymphoma cells that express Egr-1 message and protein constitutively are growth inhibited when Egr-1 is down-regulated by negative signals from BCR or by antisense oligonucleotides. To test the hypothesis that Egr-1 is important for B cell development, we examined B cells from primary and secondary lymphoid organs in Egr-1(-/-) mice. Marginal zone B cell development was arrested in these mice, whereas the B cells in all other compartments were increased. To test the hypothesis that Egr-1 function may be partially compensated by other Egr family members, we developed transgenic mice expressing a dominant negative form of Egr-1, which lacks the trans activation domain but retains the DNA-binding domain, in a B cell-specific manner. There was a decrease in B lymphopoiesis in the bone marrow accompanied by a reduction in splenic immature and mature B cells as well as marginal zone B cells in the transgenic mice. Moreover, transgenic mice respond poorly to BCR cross-linking in vitro and T-independent and T-dependent Ags in vivo.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Diferenciación Celular/inmunología , Proliferación Celular , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Animales , Antígenos T-Independientes/fisiología , Linfocitos B/citología , Diferenciación Celular/genética , Línea Celular , Línea Celular Tumoral , Proteína 1 de la Respuesta de Crecimiento Precoz/deficiencia , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Femenino , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Linfopoyesis/genética , Linfopoyesis/inmunología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Transgénicos , Receptores de Antígenos de Linfocitos B/fisiología
14.
Mol Cancer ; 8: 132, 2009 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-20043832

RESUMEN

BACKGROUND: Previously we have shown that B cell receptor (BCR) expression and B cell receptor signaling pathways are important for the basal growth of B lymphoma cells. In particular we have shown that the activation of Syk, a non-src family protein tyrosine kinase and the mitogen activated protein kinases (MAPK), ERK and JNK that mediate BCR signals are required for the constitutive growth of B lymphoma cells. Since src family protein tyrosine kinases (SFKs) like Lyn are known to be needed for the phosphorylation of BCR co-receptors, Ig-alpha and Ig-beta, we hypothesized that one or more SFKs will be constitutively activated in B lymphoma cells and may be necessary for B lymphoma growth. RESULTS: Src kinase activity was found to be constitutively high in many murine and human B lymphoma cell lines and primary lymphoma samples. The specific pharmacological inhibitors of SFKs, PP1 and PP2 inhibited the proliferation of a number of both murine and human B lymphomas in a dose-dependent manner. Importantly, dasatinib (BMS-354825), an oral dual BCR-ABL and SFK specific inhibitor inhibited the growth of B lymphomas in the nanomolar range in vitro and strongly inhibited a mouse lymphoma growth in vivo. Among the SFKs, Lyn is predominantly phosphorylated and Lyn-specific small interfering RNA inhibited the growth of B lymphomas, supporting an important role for Lyn in B lymphoma growth. Suppression of SFK activity blocks BCR mediated signaling pathways. PMA or CpG can partially reverse the growth inhibition induced by SFK inhibition. Although blocking SFK activity inhibited the growth of a number of B lymphomas, some lymphomas such as SudHL-4, SudHL-6, OCI-Ly3 and OCI-Ly10 are more resistant due to an increased expression of the anti-apoptotic proteins Bcl-2 and Bcl-xL. CONCLUSIONS: These studies further support our concept that BCR signaling pathways are important for the continued growth of established B lymphoma cells. Some of the intermediates in this BCR pathway are potential immunotherapeutic targets. In particular, inhibition of SFK activity alone or in synergy with inhibition of the prosurvival Bcl-2 proteins holds promise in developing more effective treatments for B lymphoma patients.


Asunto(s)
Linfoma de Células B/enzimología , Familia-src Quinasas/metabolismo , Animales , Supervivencia Celular , Dasatinib , Humanos , Linfoma de Células B/patología , Ratones , Pirimidinas/metabolismo , Pirimidinas/farmacología , ARN Interferente Pequeño/metabolismo , Tiazoles/metabolismo , Tiazoles/farmacología
15.
J Clin Immunol ; 29(1): 12-21, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19067131

RESUMEN

INTRODUCTION: B lymphocytes remain in a resting state until activated by antigenic stimuli through interaction with the B cell receptor (BCR). Coreceptors on B cells can modulate the thresholds for signaling through the BCR for growth and differentiation. CD72 is a B cell coreceptor that has been shown to interact with CD100, a semaphorin, and to enhance BCR signaling. DISCUSSION: CD72 ligation induces a variety of early signaling events such as activation of the Src kinases Blk and Lyn and the non-src kinase Btk leading to activation of the mitogen-activated protein (MAP) kinases, events usually associated with positive signaling. CD72 signals can enable Btk-deficient B cells to overcome their unresponsiveness to BCR signaling. On the other hand, BCR-mediated signals are enhanced in CD72-deficient cells but are reduced in CD100 null cells. The dual effects of CD72 on B cells can be explained by its association with positive and negative signaling molecules. Thus, CD72 interacts with SHP-1, an SH2-domain containing protein tyrosine phosphatase, a negative regulator of signaling, and Grb2, an adaptor protein associated with the Ras/MAPK pathway. Ligation of CD72 also triggered its association with CD19, a positive modulator of B cell receptor signaling. We propose a dual signaling hypothesis to explain the growth and differentiation promoting properties of CD72. Deficiency in either CD72 or CD100 leads to autoimmunity in mouse models. CD72 expression and polymorphisms exhibit some association with autoimmune diseases such as lupus, Sjogren's syndrome, and type 1 diabetes.


Asunto(s)
Antígenos CD/inmunología , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos B/metabolismo , Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Semaforinas/inmunología , Agammaglobulinemia Tirosina Quinasa , Animales , Antígenos CD/genética , Antígenos CD19/inmunología , Antígenos CD19/metabolismo , Antígenos de Diferenciación de Linfocitos B/genética , Autoinmunidad/inmunología , Linfocitos B/metabolismo , Proteína Adaptadora GRB2/inmunología , Proteína Adaptadora GRB2/metabolismo , Humanos , Ratones , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteínas Tirosina Quinasas/inmunología , Proteínas Tirosina Quinasas/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Semaforinas/metabolismo , Transducción de Señal/inmunología
16.
Cancer Res ; 67(19): 9276-85, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17909035

RESUMEN

Prostate apoptosis response-4 (Par-4) is a tumor-suppressor protein that induces apoptosis in cancer cells, but not in normal/immortalized cells. The cancer-specific proapoptotic action of Par-4 is encoded in its centrally located SAC domain. We report here the characterization of a novel mouse model with ubiquitous expression of the SAC domain. Although SAC transgenic mice displayed normal development and life span, they were resistant to the growth of spontaneous, as well as oncogene-induced, autochthonous tumors. Resistance to tumorigenesis was linked to inhibition of nuclear factor-kappaB activity and induction of apoptosis by the SAC domain. Collectively, our findings provide genetic evidence that the SAC domain of Par-4 confers cancer resistance in transgenic mice without compromising normal viability or aging, and may have therapeutic significance.


Asunto(s)
Neoplasias Experimentales/genética , Neoplasias Experimentales/prevención & control , Receptores Proteinasa-Activados/biosíntesis , Animales , Apoptosis/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Estructura Terciaria de Proteína , Receptores Proteinasa-Activados/genética , Receptores Proteinasa-Activados/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética
17.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1088-1097, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30759363

RESUMEN

Increasing numbers of cancer patients survive and live longer than five years after therapy, but very often side effects of cancer treatment arise at same time. One of the side effects, chemotherapy-induced cognitive impairment (CICI), also called "chemobrain" or "chemofog" by patients, brings enormous challenges to cancer survivors following successful chemotherapeutic treatment. Decreased abilities of learning, memory, attention, executive function and processing speed in cancer survivors with CICI, are some of the challenges that greatly impair survivors' quality of life. The molecular mechanisms of CICI involve very complicated processes, which have been the subject of investigation over the past decades. Many mechanistic candidates have been studied including disruption of the blood-brain barrier (BBB), DNA damage, telomere shortening, oxidative stress and associated inflammatory response, gene polymorphism of neural repair, altered neurotransmission, and hormone changes. Oxidative stress is considered as a vital mechanism, since over 50% of FDA-approved anti-cancer drugs can generate reactive oxygen species (ROS) or reactive nitrogen species (RNS), which lead to neuronal death. In this review paper, we discuss these important candidate mechanisms, in particular oxidative stress and the cytokine, TNF-alpha and their potential roles in CICI.


Asunto(s)
Antineoplásicos/efectos adversos , Encéfalo/efectos de los fármacos , Supervivientes de Cáncer/estadística & datos numéricos , Disfunción Cognitiva/fisiopatología , Neoplasias/fisiopatología , Calidad de Vida , Antineoplásicos/uso terapéutico , Encéfalo/patología , Encéfalo/fisiopatología , Supervivientes de Cáncer/psicología , Disfunción Cognitiva/inducido químicamente , Humanos , Memoria/efectos de los fármacos , Modelos Biológicos , Neoplasias/tratamiento farmacológico , Neoplasias/psicología , Estrés Oxidativo/efectos de los fármacos
18.
Free Radic Biol Med ; 134: 1-8, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30593843

RESUMEN

Cancer treatments are developing fast and the number of cancer survivors could arise to 20 million in United State by 2025. However, a large fraction of cancer survivors demonstrate cognitive dysfunction and associated decreased quality of life both shortly, and often long-term, after chemotherapy treatment. The etiologies of chemotherapy induced cognitive impairment (CICI) are complicated, made more so by the fact that many anti-cancer drugs cannot cross the blood-brain barrier (BBB). Multiple related factors and confounders lead to difficulties in determining the underlying mechanisms. Chemotherapy induced, oxidative stress-mediated tumor necrosis factor-alpha (TNF-α) elevation was considered as one of the main candidate mechanisms underlying CICI. Doxorubicin (Dox) is a prototypical reactive oxygen species (ROS)-generating chemotherapeutic agent used to treat solid tumors and lymphomas as part of multi-drug chemotherapeutic regimens. We previously reported that peripheral Dox-administration leads to plasma protein damage and elevation of TNF-α in plasma and brain of mice. In the present study, we used TNF-α null (TNFKO) mice to investigate the role of TNF-α in Dox-induced, oxidative stress-mediated alterations in brain. We report that Dox-induced oxidative stress in brain is ameliorated and brain mitochondrial function assessed by the Seahorse-determined oxygen consumption rate (OCR) is preserved in brains of TNFKO mice. Further, we show that Dox-decreased the level of hippocampal choline-containing compounds and brain phospholipases activity are partially protected in TNFKO group in MRS study. Our results provide strong evidence that Dox-targeted mitochondrial damage and levels of brain choline-containing metabolites, as well as phospholipases changes decreased in the CNS are associated with oxidative stress mediated by TNF-α. These results are consistent with the notion that oxidative stress and elevated TNF-α in brain underlie the damage to mitochondria and other pathological changes that lead to CICI. The results are discussed with reference to our identifying a potential therapeutic target to protect against cognitive problems after chemotherapy.


Asunto(s)
Encéfalo/patología , Colina/metabolismo , Disfunción Cognitiva/inducido químicamente , Doxorrubicina/farmacología , Mitocondrias/patología , Neuronas/patología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Antibióticos Antineoplásicos/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos
19.
Redox Biol ; 26: 101237, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31276937

RESUMEN

Extracellular vesicles (EVs) generated from redox active anticancer drugs are released into the extracellular environment. These EVs contain oxidized molecules and trigger inflammatory responses by macrophages. Using a mouse model of doxorubicin (DOX)-induced tissue injury, we previously found that the major sources of circulating EVs are from heart and liver, organs that are differentially affected by DOX. Here, we investigated the effects of EVs from cardiomyocytes and those from hepatocytes on macrophage activation. EVs from H9c2 rat cardiomyocytes (H9c2 EVs) and EVs from FL83b mouse hepatocytes (FL83 b EVs) have different levels of protein-bound 4-hydroxynonenal and thus different immunostimulatory effects on mouse RAW264.7 macrophages. H9c2 EVs but not FL83 b EVs induced both pro-inflammatory and anti-inflammatory macrophage activation, mediated by NFκB and Nrf-2 pathways, respectively. DOX enhanced the effects of H9c2 EVs but not FL83 b EVs. While EVs from DOX-treated H9c2 cells (H9c2 DOXEVs) suppressed mitochondrial respiration and increased glycolysis of macrophages, EVs from DOX-treated FL83b cells (FL83b DOXEVs) enhanced mitochondrial reserve capacity. Mechanistically, the different immunostimulatory functions of H9c2 EVs and FL83 b EVs are regulated, in part, by the redox status of the cytoplasmic thioredoxin 1 (Trx1) of macrophages. H9c2 DOXEVs lowered the level of reduced Trx1 in cytoplasm while FL83b DOXEVs did the opposite. Trx1 overexpression alleviated the effect of H9c2 DOXEVs on NFκB and Nrf-2 activation and prevented the upregulation of their target genes. Our findings identify EVs as a novel Trx1-mediated redox mediator of immune response, which greatly enhances our understanding of innate immune responses during cancer therapy.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Vesículas Extracelulares/inmunología , Hepatocitos/química , Miocitos Cardíacos/química , Tiorredoxinas/inmunología , Aldehídos/inmunología , Aldehídos/metabolismo , Aldehídos/farmacología , Animales , Línea Celular , Medios de Cultivo Condicionados/química , Vesículas Extracelulares/química , Regulación de la Expresión Génica , Glucólisis/efectos de los fármacos , Hepatocitos/metabolismo , Activación de Macrófagos/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/inmunología , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Oxidación-Reducción , Células RAW 264.7 , Ratas , Tiorredoxinas/genética
20.
Blood Adv ; 3(8): 1255-1266, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30987970

RESUMEN

Prostate apoptosis response 4 (Par-4) is a tumor suppressor that prevents proliferation and induces cell death in several solid tumors. However, its role in B-cell malignancies has not been elucidated. To describe the role of Par-4 in chronic lymphocytic leukemia (CLL) pathogenesis, we developed a B-cell-specific human Par-4-overexpressing mouse model of CLL using the TCL1 leukemia model. While Par-4 transgenic mice did not display any obvious defects in B-cell development or function, disease burden as evidenced by abundance of CD19+CD5+ B cells in the peripheral blood was significantly reduced in Par-4 × TCL1 mice compared with TCL1 littermates. This conferred a survival advantage on the Par-4-overexpressing mice. In addition, a B-cell-specific knockout model displayed the opposite effect, where lack of Par-4 expression resulted in accelerated disease progression and abbreviated survival in the TCL1 model. Histological and flow cytometry-based analysis of spleen and bone marrow upon euthanasia revealed comparable levels of malignant B-cell infiltration in Par-4 × TCL1 and TCL1 individuals, indicating delayed but pathologically normal disease progression in Par-4 × TCL1 mice. In vivo analysis of splenic B-cell proliferation by 5-ethynyl-2-deoxyuridine incorporation indicated >50% decreased expansion of CD19+CD5+ cells in Par-4 × TCL1 mice compared with TCL1 littermates. Moreover, reduced nuclear p65 levels were observed in Par-4 × TCL1 splenic B cells compared with TCL1, suggesting suppressed NF-κB signaling. These findings have identified an in vivo antileukemic role for Par-4 through an NF-κB-dependent mechanism in TCL1-mediated CLL-like disease progression.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/biosíntesis , Carcinogénesis/metabolismo , Leucemia Linfocítica Crónica de Células B/metabolismo , Neoplasias Experimentales/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Animales , Carcinogénesis/genética , Humanos , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/patología , Ratones , Ratones Transgénicos , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Proteínas Proto-Oncogénicas/genética , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA