Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
ACS Nano ; 18(34): 23757-23772, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39141816

RESUMEN

Toll-like receptors (TLRs) are a class of pattern recognition receptors (PRRs) crucial for the detection of infections and activation of downstream signaling pathways that lead to the production of pro-inflammatory cytokines and interferons. The TLR pathway is an attractive actively studied target pathway. Because of their strong immunostimulatory activity, TLRs are thought to be a "double-edged sword" for systemic treatment, even in the cancer field. To solve this, we have developed dextran-based TAM targeting activating conjugate (D-TAC) technology, which successfully uses tumor-associated macrophages (TAMs) to deliver the TLR7 agonist DSP-0509. We used low molecular weight dextran to target CD206 high M2-type macrophages, activate them, and induce a change in phenotype to antitumor M1-type macrophages with rapid clearance from the body and astonishing antitumor activity. We also demonstrated that the antitumor effect of our best drug candidate 5DEX-0509R is dependent on the abundance of TAMs, which is consistent with their mechanism of action. We believe that 5DEX-0509R generated by D-TAC technology can be a clinically applicable immunotherapy targeting the TLR signaling pathway.


Asunto(s)
Antineoplásicos , Nanomedicina , Receptor Toll-Like 7 , Macrófagos Asociados a Tumores , Animales , Ratones , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/metabolismo , Macrófagos Asociados a Tumores/efectos de los fármacos , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/química , Dextranos/química , Dextranos/farmacología , Ratones Endogámicos C57BL , Humanos , Línea Celular Tumoral , Femenino , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/inmunología
2.
BMC Immunol ; 25(1): 48, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39054418

RESUMEN

BACKGROUND: TLR7 is a key player in the antiviral immunity. TLR7 signaling activates antigen-presenting cells including DCs and macrophages. This activation results in the adaptive immunity including T cells and B cells. Therefore, TLR7 is an important molecule of the immune system. Based on these observations, TLR7 agonists considered to become a therapy weaponize the immune system against cancer. Radiation therapy (RT) is one of the standard cancer therapies and is reported to modulate the tumor immune response. In this study, we aimed to investigate the anti-tumor activity in combination of TLR7 agonist, DSP-0509, with RT and underlying mechanism. RESULT: We showed that anti-tumor activity is enhanced by combining RT with the TLR7 agonist DSP-0509 in the CT26, LM8, and 4T1 inoculated mice models. We found that once- weekly (q1w) dosing of DSP-0509 rather than biweekly (q2w) dosing is needed to achieve superior anti-tumor activities in CT26 model. Spleen cells from the mice in RT/DSP-0509 combination treatment group showed increased tumor lytic activity, inversely correlated with tumor volume, as measured by the chromium-release cytotoxicity assay. We also found the level of cytotoxic T lymphocytes (CTLs) increased in the spleens of completely cured mice. When the mice completely cured by combination therapy were re-challenged with CT26 cells, all mice rejected CT26 cells but accepted Renca cells. This rejection was not observed with CD8 depletion. Furthermore, levels of splenic effector memory CD8 T cells were increased in the combination therapy group. To explore the factors responsible for complete cure by combination therapy, we analyzed peripheral blood leukocytes (PBLs) mRNA from completely cured mice. We found that Havcr2low, Cd274low, Cd80high, and Il6low were a predictive signature for the complete response to combination therapy. An analysis of tumor-derived mRNA showed that combination of RT and DSP-0509 strongly increased the expression of anti-tumor effector molecules including Gzmb and Il12. CONCLUSION: These data suggest that TLR7 agonist, DSP-0509, can be a promising concomitant when used in combination with RT by upregulating CTLs activity and gene expression of effector molecules. This combination can be an expecting new radio-immunotherapeutic strategy in clinical trials.


Asunto(s)
Receptor Toll-Like 7 , Animales , Receptor Toll-Like 7/agonistas , Ratones , Línea Celular Tumoral , Femenino , Activación de Linfocitos/efectos de los fármacos , Ratones Endogámicos BALB C , Glicoproteínas de Membrana/agonistas , Terapia Combinada , Humanos , Ratones Endogámicos C57BL , Neoplasias/terapia , Neoplasias/inmunología , Neoplasias/tratamiento farmacológico , Modelos Animales de Enfermedad , Linfocitos T/inmunología , Linfocitos T/efectos de los fármacos
4.
Int J Mol Sci ; 24(7)2023 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-37047449

RESUMEN

Our laboratory has identified and developed a unique human-engineered domain (HED) structure that was obtained from the human Alpha-2-macroglobulin receptor-associated protein based on the three-dimensional structure of the Z-domain derived from Staphylococcal protein A. This HED retains µM binding activity to the human IgG1CH2-CH3 elbow region. We determined the crystal structure of HED in association with IgG1's Fc. This demonstrated that HED preserves the same three-bundle helix structure and Fc-interacting residues as the Z domain. HED was fused to the single chain variable fragment (scFv) of mAb 4D5 to produce an antibody-like protein capable of interacting with the p185Her2/neu ectodomain and the Fc of IgG. When further fused with murine IFN-γ (mIFN-γ) at the carboxy terminus, the novel species exhibited antitumor efficacy in vivo in a mouse model of human breast cancer. The HED is a novel platform for the therapeutic utilization of engineered proteins to alleviate human disease.


Asunto(s)
Neoplasias de la Mama , Anticuerpos de Cadena Única , Humanos , Animales , Ratones , Femenino , Anticuerpos de Cadena Única/genética , Proteína Estafilocócica A/química
5.
Front Immunol ; 14: 1055671, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36793737

RESUMEN

TLR7 is an innate immune receptor that recognizes single-stranded RNAs, and its activation leads to anti-tumor immune effects. Although it is the only approved TLR7 agonist in cancer therapy, imiquimod is allowed to be administered with topical formulation. Thus, systemic administrative TLR7 agonist is expected in terms of expanding applicable cancer types. Here, we demonstrated the identification and characterization of DSP-0509 as a novel small-molecule TLR7 agonist. DSP-0509 is designed to have unique physicochemical features that could be administered systemically with a short half-life. DSP-0509 activated bone marrow-derived dendritic cells (BMDCs) and induced inflammatory cytokines including type I interferons. In the LM8 tumor-bearing mouse model, DSP-0509 reduced tumor growth not only in subcutaneous primary lesions but also in lung metastatic lesions. DSP-0509 inhibited tumor growth in several syngeneic tumor-bearing mouse models. We found that the CD8+ T cell infiltration of tumor before treatment tended to be positively correlated with anti-tumor efficacy in several mouse tumor models. The combination of DSP-0509 with anti-PD-1 antibody significantly enhanced the tumor growth inhibition compared to each monotherapy in CT26 model mice. In addition, the effector memory T cells were expanded in both the peripheral blood and tumor, and rejection of tumor re-challenge occurred in the combination group. Moreover, synergistic anti-tumor efficacy and effector memory T cell upregulation were also observed for the combination with anti-CTLA-4 antibody. The analysis of the tumor-immune microenvironment by using the nCounter assay revealed that the combination of DSP-0509 with anti-PD-1 antibody enhanced infiltration by multiple immune cells including cytotoxic T cells. In addition, the T cell function pathway and antigen presentation pathway were activated in the combination group. We confirmed that DSP-0509 enhanced the anti-tumor immune effects of anti-PD-1 antibody by inducing type I interferons via activation of dendritic cells and even CTLs. In conclusion, we expect that DSP-0509, a new TLR7 agonist that synergistically induces anti-tumor effector memory T cells with immune checkpoint blockers (ICBs) and can be administered systemically, will be used in the treatment of multiple cancers.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Interferón Tipo I , Neoplasias , Receptor Toll-Like 7 , Animales , Ratones , Adyuvantes Inmunológicos/farmacología , Modelos Animales de Enfermedad , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/tratamiento farmacológico , Receptor Toll-Like 7/agonistas , Microambiente Tumoral
6.
Acta Radiol ; 64(1): 336-345, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35118883

RESUMEN

BACKGROUND: It is unclear whether deep-learning-based super-resolution technology (SR) or compressed sensing technology (CS) can accelerate magnetic resonance imaging (MRI) . PURPOSE: To compare SR accelerated images with CS images regarding the image similarity to reference 2D- and 3D gradient-echo sequence (GRE) brain MRI. MATERIAL AND METHODS: We prospectively acquired 1.3× and 2.0× faster 2D and 3D GRE images of 20 volunteers from the reference time by reducing the matrix size or increasing the CS factor. For SR, we trained the generative adversarial network (GAN), upscaling the low-resolution images to the reference images with twofold cross-validation. We compared the structural similarity (SSIM) index of accelerated images to the reference image. The rate of incorrect answers of a radiologist discriminating faster and reference image was used as a subjective image similarity (ISM) index. RESULTS: The SR demonstrated significantly higher SSIM than the CS (SSIM=0.9993-0.999 vs. 0.9947-0.9986; P < 0.001). In 2D GRE, it was challenging to discriminate the SR image from the reference image, compared to the CS (ISM index 40% vs. 17.5% in 1.3×; P = 0.039 and 17.5% vs. 2.5% in 2.0×; P = 0.034). In 3D GRE, the CS revealed a significantly higher ISM index than the SR (22.5% vs. 2.5%; P = 0.011) in 2.0 × faster images. However, the ISM index was identical for the 2.0× CS and 1.3× SR (22.5% vs. 27.5%; P = 0.62) with comparable time costs. CONCLUSION: The GAN-based SR outperformed CS in image similarity with 2D GRE for MRI acceleration. In addition, CS was more advantageous in 3D GRE than SR.


Asunto(s)
Imagenología Tridimensional , Imagen por Resonancia Magnética , Humanos , Presión , Encéfalo/diagnóstico por imagen , Procesamiento de Imagen Asistido por Computador/métodos
7.
Radiol Cardiothorac Imaging ; 4(6): e220111, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36601449

RESUMEN

Purpose: To evaluate the image quality of high-spatial-resolution two-dimensional (2D) late gadolinium enhancement (LGE) cardiac MRI compared with conventional normal-resolution LGE MRI. Materials and Methods: This prospective study included participants suspected of having cardiomyopathy who underwent cardiac MRI between March 2021 and December 2021. Normal-resolution and high-resolution 2D LGE sequences (inversion recovery [IR] and phase-sensitive inversion recovery [PSIR]) were performed at 3 T. Resolution was compared between normal-resolution and high-resolution images obtained in a quality assurance phantom. In vivo image quality and resolution were evaluated qualitatively using a five-point scoring system. Receiver operating characteristic curve analysis was used for LGE detection performance. Border sharpness was assessed with profile curve measurement. The contrast-to-noise ratio (CNR) between hyperenhancement and remote myocardium and LGE detection performance were calculated using normal-resolution IR images as the reference. Results: In total, 120 participants were evaluated (mean age, 56 years ± 17 [SD]; 72 men). Features smaller than 1 mm were detectable only on high-resolution images of the phantom. In vivo, the image resolution score with high-resolution LGE was 4.14-4.24, which was higher than the normal-resolution LGE reference score of 2.99 (P < .05). Border sharpness was higher in high-resolution images (P < .001). Receiver operating characteristic curve analysis revealed no evidence of a difference in LGE detection between normal-resolution and high-resolution images. There was also no evidence of a change in CNR of LGE in IR and PSIR magnitude compared with reference images. Conclusion: Comparison of image quality in 2D high-resolution and normal-resolution LGE cardiac MRI demonstrated the highest resolution for high-resolution IR and high-resolution PSIR magnitude sequences.Keywords: Cartilage Imaging, MRI, Cardiac, Heart, Imaging Sequences, Comparative Studies Supplemental material is available for this article. © RSNA, 2022.

8.
Front Immunol ; 11: 621, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32328070

RESUMEN

Arginine methylation is a post-translational modification that regulates many biological processes. However, the role of arginine methylation in immune cells is not well studied. Here we report an essential role of protein arginine methyltransferase 5 (PRMT5) in T cell homeostasis and activation-induced expansion. Using T cell-specific PRMT5 conditional knockout mice, we found that PRMT5 is required for natural killer T (NKT) cell but not for conventional or regulatory T (Treg) cell development after the double positive (DP) stage in the thymus. In contrast, PRMT5 was required for optimal peripheral T cell maintenance, for the transition of naïve T cells to effector/memory phenotype, and for early T cell development before the DP stage in a cell-intrinsic manner. Accordingly, PRMT5-deleted T cells showed impaired IL-7-mediated survival and TCR-induced proliferation in vitro. The latter was more pronounced and attributed to reduced responsiveness to IL-2. Acute deletion of PRMT5 revealed that not only naïve but also effector/memory T cells were impaired in TCR-induced proliferation in a development-independent manner. Reduced expression of common γ chain (γc), a shared receptor component for several cytokines including IL-7 and IL-2, on PRMT5-deleted T cells may be in part responsible for the defect. We further showed that PRMT5 was partially required for homeostatic T cell survival but absolutely required for lymphopenic T cell expansion in vivo. Thus, we propose that PRMT5 is required for T cell survival and proliferation by maintaining cytokine signaling, especially during proliferation. The inhibition of PRMT5 may provide a novel strategy for the treatment of diseases where uncontrolled T cell activation has a role, such as autoimmunity.


Asunto(s)
Células T Asesinas Naturales/fisiología , Proteína-Arginina N-Metiltransferasas/metabolismo , Animales , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Citocinas/metabolismo , Homeostasis , Memoria Inmunológica , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína-Arginina N-Metiltransferasas/genética , Transducción de Señal , Linfocitos T Reguladores/inmunología
9.
Front Immunol ; 10: 2486, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31681337

RESUMEN

Regulatory T cells (Tregs) are engaged in maintaining immune homeostasis and preventing autoimmunity. Treg cells include thymic Treg cells and peripheral Treg cells, both of which can suppress the immune response via multiple distinct mechanisms. The differentiation, proliferation, suppressive function and survival of Treg cells are affected by distinct energy metabolic programs. Tissue-resident Treg cells hold unique features in comparison with the lymphoid organ Treg cells. Foxp3 transcription factor is a lineage master regulator for Treg cell development and suppressive activity. Accumulating evidence indicates that the activity of Foxp3 protein is modulated by various post-translational modifications (PTMs), including phosphorylation, O-GlcNAcylation, acetylation, ubiquitylation and methylation. These modifications affect multiple aspects of Foxp3 function. In this review, we define features of Treg cells and roles of Foxp3 in Treg biology, and summarize current research in PTMs of Foxp3 protein involved in modulating Treg function. This review also attempts to define Foxp3 dimer modifications relevant to mediating Foxp3 activity and Treg suppression. Understanding Foxp3 protein features and modulation mechanisms may help in the design of rational therapies for immune diseases and cancer.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica/inmunología , Procesamiento Proteico-Postraduccional/inmunología , Linfocitos T Reguladores/inmunología , Acetilación , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Proliferación Celular/genética , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica/genética , Homeostasis/genética , Homeostasis/inmunología , Humanos , Procesamiento Proteico-Postraduccional/genética , Linfocitos T Reguladores/metabolismo
10.
Front Immunol ; 10: 174, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30800128

RESUMEN

Regulatory T cells (Tregs) are a subpopulation of T cells that are specialized in suppressing immune responses. Here we show that the arginine methyl transferase protein PRMT5 can complex with FOXP3 transcription factors in Tregs. Mice with conditional knock out (cKO) of PRMT5 expression in Tregs develop severe scurfy-like autoimmunity. In these PRMT5 cKO mice, the spleen has reduced numbers of Tregs, but normal numbers of Tregs are found in the peripheral lymph nodes. These peripheral Tregs that lack PRMT5, however, display a limited suppressive function. Mass spectrometric analysis showed that FOXP3 can be di-methylated at positions R27, R51, and R146. A point mutation of Arginine (R) 51 to Lysine (K) led to defective suppressive functions in human CD4 T cells. Pharmacological inhibition of PRMT5 by DS-437 also reduced human Treg functions and inhibited the methylation of FOXP3. In addition, DS-437 significantly enhanced the anti-tumor effects of anti-erbB2/neu monoclonal antibody targeted therapy in Balb/c mice bearing CT26Her2 tumors by inhibiting Treg function and induction of tumor immunity. Controlling PRMT5 activity is a promising strategy for cancer therapy in situations where host immunity against tumors is attenuated in a FOXP3 dependent manner.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Neoplasias de la Mama/terapia , Factores de Transcripción Forkhead/metabolismo , Técnicas de Inactivación de Genes , Inmunoterapia/métodos , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Receptor ErbB-2/inmunología , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Autoinmunidad/genética , Femenino , Factores de Transcripción Forkhead/genética , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Terapia Molecular Dirigida/métodos , Mutación Puntual , Receptor ErbB-2/antagonistas & inhibidores , Linfocitos T Reguladores/metabolismo , Transfección
11.
Oncoimmunology ; 7(4): e1300739, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29632709

RESUMEN

Despite substantial clinical progress with targeted therapies, current antibody-based approaches have limited efficacy at controlling HER2/neu-positive breast cancers, especially in the absence of chemotherapies. Previously, we showed that the combination of IFNγ and anti-HER2/neu antibody synergistically reduces tumor growth in an in vivo implanted mammary tumor model. Here, we report a recombinant approach to produce an anti-HER2/neu scFv and IFNγ fusion protein using an engineered effector domain (EED) scaffold. The new molecule induces in vitro apoptosis in an IFNγ receptor-dependent manner. At a very low dose in the in vivo xenografted tumor models, the new EED-IFNγ fusion protein demonstrates superior activity over the anti-HER2/neu antibody and is even active on tumors that are resistant to anti-HER2/neu antibody therapy. Examination of tumor infiltrated macrophages and lymphocytes reveals that the fusion protein can induce changes in tumor microenvironment to support immune reactivity against tumors. Our studies have defined a targeted immunotherapy approach for the treatment of cancers.

12.
Oncotarget ; 9(3): 3631-3640, 2018 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-29423071

RESUMEN

Prostate cancer is the most common cancer occurring in men in the United States. The monoclonal antibody F77 that was originally developed in our laboratory recognizes mainly glycolipids as well as O-linked glycosylation on proteins in prostate cancer cells. We have identified a spliced form of glycoprotein CD44 as one critical protein expressing the F77 antigen. The F77-specific glycosylation occurs on multiple potential glycosylation sites on the CD44 protein encoded by the fourteenth exon. CD44 is a tumor stem cell marker and is known to induce tumor stemness and metastasis. Knockdown of CD44 or FUT1 genes dramatically reduced F77-induced apoptosis in prostate cancer cell lines. We developed an ELISA using both a CD44 antibody and F77 to identify the special form of glycosylated CD44 from prostate cancer cells as well as from serum samples of prostate cancer patients. These results reveal a CD44-dependent mechanism for F77 to induce tumor cell apoptosis, and a new strategy for the detection of glycosylated CD44 proteins secreted by prostate cancer cells.

13.
Sci Immunol ; 2(12)2017 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-28783662

RESUMEN

CD4+FOXP3+ regulatory T (Treg) cells are critical mediators of immune tolerance, and their deficiency owing to FOXP3 mutations in immunodysregulation polyendocrinopathy enteropathy X-linked syndrome (IPEX) patients results in severe autoimmunity. Different FOXP3 mutations result in a wide range of disease severity, reflecting the relative importance of the affected residues in the integrity of the FOXP3 protein and its various molecular interactions. We characterized the cellular and molecular impact of the most common IPEX mutation, p.A384T, on patient-derived Treg cells. We found that the p.A384T mutation abrogated the suppressive capacity of Treg cells while preserving FOXP3's ability to repress inflammatory cytokine production. This selective functional impairment is partly due to a specific disruption of FOXP3A384T binding to the histone acetyltransferase Tat-interacting protein 60 (TIP60) (KAT5) and can be corrected using allosteric modifiers that enhance FOXP3-TIP60 interaction. These findings reveal the functional impact of TIP60 in FOXP3-driven Treg biology and provide a potential target for therapeutic manipulation of Treg activity.

14.
Exp Dermatol ; 25(6): 466-71, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26910392

RESUMEN

We previously reported that allergic responses to nickel (Ni) were minimal in mice deficient in the histamine-forming enzyme histidine decarboxylase (HDC-KO), suggesting an involvement of histamine in allergic responses to Ni. However, it remains unclear how histamine is involved in the process of Ni allergy. Here, we examined the role of histamine in Ni allergy using a murine model previously established by us. Mice were sensitized to Ni by intraperitoneal injection of a NiCl2 -lipopolysaccharide (LPS) mixture. Ten days later, allergic inflammation was elicited by challenging ear-pinnas intradermally with NiCl2 . Then, ear-swelling was measured. Pyrilamine (histamine H1-receptor antagonist) or cromoglicate (mast cell stabilizer) was intravenously injected 1 h before the sensitization or the challenge. In cell-transfer experiments, spleen cells from Ni-sensitized donor mice were intravenously transferred into non-sensitized recipient mice. In both sensitized and non-sensitized mice, 1 mm or more NiCl2 (injected into ear-pinnas) induced transient non-allergic inflammation (Ni-TI) with accompanying mast cell degranulation. LPS did not affect the magnitude of this Ni-TI. Pyrilamine and cromoglicate reduced either the Ni-TI or the ensuing allergic inflammation when administered before Ni-TI (at either the sensitization or elicitation step), but not if administered when the Ni-TI had subsided. Experiments on HDC-KO and H1-receptor-KO mice, and also cell-transfer experiments using these mice, demonstrated histamine's involvement in both the sensitization and elicitation steps. These results suggest that mast cell histamine-mediated Ni-TI promotes subsequent allergic inflammatory responses to Ni, raising the possibility that control of Ni-TI by drugs may be effective at preventing or reducing Ni allergy.


Asunto(s)
Dermatitis Alérgica por Contacto/inmunología , Histamina/metabolismo , Mastocitos/fisiología , Níquel/inmunología , Animales , Cromolin Sódico , Dermatitis Alérgica por Contacto/metabolismo , Femenino , Ratones Endogámicos C57BL , Ratones Noqueados
15.
Cell Rep ; 12(12): 2049-59, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26365188

RESUMEN

Reversion of the malignant phenotype of erbB2-transformed cells can be driven by anti-erbB2/neu monoclonal antibodies (mAbs), which disrupt the receptor's kinase activity. We examined the biologic effects of IFN-γ alone or after anti-erbB2/neu mAb treatment of erbB2-positive cells. IFN-γ had no effect on its own. Treatment of the tumors with anti-erbB2/neu mAbs followed by IFN-γ led to dramatic inhibition of tumor growth in vitro and in vivo with minimal mAb dosing. Sequential therapy enhanced the effects of chemotherapy. Moreover, IFN-γ with mAb treatment of mice with IFNγR knockdown tumors did not demonstrate marked synergistic eradication effects, indicating an unexpected role of IFN-γ on the tumor itself. Additionally, mAb and IFN-γ treatment also induced immune host responses that enhanced tumor eradication. Biochemical analyses identified loss of Snail expression in tumor cells, reflecting diminution of tumor-stem-cell-like properties as a consequence of altered activity of GSK3-ß and KLF molecules.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica , Interferón gamma/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Trastuzumab/farmacología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Fenotipo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Transducción de Señal , Factores de Transcripción de la Familia Snail , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Carga Tumoral/efectos de los fármacos , Receptor de Interferón gamma
16.
J Biol Chem ; 290(33): 20211-20, 2015 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25987564

RESUMEN

Regulation of the extent of immune responses is a requirement to maintain self-tolerance and limit inflammatory processes. CD4(+)CD25(+)Foxp3(+) regulatory T (Treg) cells play a role in regulation. The Foxp3 transcription factor is considered a dominant regulator for Treg cell development and function. Foxp3 function itself is directly regulated by multiple posttranslational modifications that occur in response to various external stimuli. The Foxp3 protein is a component of several dynamic macromolecular regulatory complexes. The complexes change constituents over time and through different signals to regulate the development and function of regulatory T cells. Here we identified a mechanism regulating Foxp3 level and activity that operates through discrete phosphorylation. The Pim-2 kinase can phosphorylate Foxp3, leading to decreased suppressive functions of Treg cells. The amino-terminal domain of Foxp3 is modified at several sites by Pim-2 kinase. This modification leads to altered expression of proteins related to Treg cell functions and increased Treg cell lineage stability. Treg cell suppressive function can be up-regulated by either pharmacologically inhibiting Pim-2 kinase activity or by genetically knocking out Pim-2 in rodent Treg cells. Deficiency of Pim-2 activity increases murine host resistance to dextran sodium sulfate-induced colitis in vivo, and a Pim-2 small molecule kinase inhibitor also modified Treg cell functions. Our studies define a pathway for limiting the regulation of Foxp3 function because the Pim-2 kinase represents a potential therapeutic target for modulating the Treg cell suppressive activities in controlling immune responses.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Linfocitos T Reguladores/inmunología , Secuencia de Aminoácidos , Animales , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Fosforilación , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/genética
17.
Cell Rep ; 7(5): 1471-1480, 2014 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-24835996

RESUMEN

The human FOXP3 molecule is an oligomeric transcriptional factor able to mediate activities that characterize T regulatory cells, a class of lymphocytes central to the regulation of immune responses. The activity of FOXP3 is regulated at the posttranslational level, in part by two histone acetyltransferases (HATs): TIP60 and p300. TIP60 and p300 work cooperatively to regulate FOXP3 activity. Initially, p300 and TIP60 interactions lead to the activation of TIP60 and facilitate acetylation of K327 of TIP60, which functions as a molecular switch to allow TIP60 to change binding partners. Subsequently, p300 is released from this complex, and TIP60 interacts with and acetylates FOXP3. Maximal induction of FOXP3 activities is observed when both p300 and TIP60 are able to undergo cooperative interactions. Conditional knockout of TIP60 in Treg cells significantly decreases the Treg population in the peripheral immune organs, leading to a scurfy-like fatal autoimmune disease.


Asunto(s)
Proteína p300 Asociada a E1A/metabolismo , Factores de Transcripción Forkhead/metabolismo , Histona Acetiltransferasas/metabolismo , Transactivadores/metabolismo , Secuencia de Aminoácidos , Animales , Enfermedades Autoinmunes/metabolismo , Proteína p300 Asociada a E1A/genética , Factores de Transcripción Forkhead/genética , Células HEK293 , Histona Acetiltransferasas/química , Histona Acetiltransferasas/genética , Humanos , Lisina/metabolismo , Lisina Acetiltransferasa 5 , Ratones , Datos de Secuencia Molecular , Mutación , Unión Proteica , Linfocitos T Reguladores/metabolismo , Transactivadores/química , Transactivadores/genética
18.
Exp Mol Pathol ; 95(1): 38-45, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23644046

RESUMEN

p300 is one of several acetyltransferases that regulate FOXP3 acetylation and functions. Our recent studies have defined a complex set of histone acetyltransferase interactions which can lead to enhanced or repressed changes in FOXP3 function. We have explored the use of a natural p300 inhibitor, Garcinol, as a tool to understand mechanisms by which p300 regulates FOXP3 acetylation. In the presence of Garcinol, p300 appears to become disassociated from the FOXP3 complex and undergoes lysosome-dependent degradation. As a consequence of p300's physical absence, FOXP3 becomes less acetylated and eventually degraded, a process that cannot be rescued by the proteasome inhibitor MG132. p300 plays a complex role in FOXP3 acetylation, as it could also acetylate a subset of four Lys residues that repressively regulate total FOXP3 acetylation. Garcinol acts as a degradation device to reduce the suppressive activity of regulatory T cells (Treg) and to enhance the in vivo anti-tumor activity of a targeted therapeutic anti-p185(her2/neu) (ERBB2) antibody in MMTV-neu transgenics implanted with neu transformed breast tumor cells. Our studies provide the rationale for molecules that disrupt p300 stability to limit Treg functions in targeted therapies for cancers.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Proteína p300 Asociada a E1A/metabolismo , Factores de Transcripción Forkhead/metabolismo , Lisosomas/metabolismo , Linfocitos T Reguladores/inmunología , Terpenos/farmacología , Acetilación/efectos de los fármacos , Animales , Línea Celular , Cloroquina/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Factores de Transcripción Forkhead/genética , Histona Acetiltransferasas/antagonistas & inhibidores , Humanos , Lisina Acetiltransferasa 5 , Lisosomas/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Terapia Molecular Dirigida/métodos , Mutación , Linfocitos T Reguladores/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Cancer Res ; 73(8): 2619-27, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23396586

RESUMEN

Recruitment of immune cells to tumor cells targeted by a therapeutic antibody can heighten the antitumor efficacy of the antibody. For example, p185(her2/neu)-targeting antibodies not only downregulate the p185(her2/neu) kinase (ERBB2) but also trigger complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC) through the antibody Fc region. Here, we describe a generalized strategy to improve immune cell recruitment to targeted cancer cells, using a modified scFv antibody we call a "Grababody" that binds the target protein and endogenous immunoglobulins. The model system we used to illustrate the use of this platform recognizes p185(her2/neu) and includes an IgG binding domain. The recombinant scFv Grababody that was created recruited circulating human IgGs and attracted immune cells carrying Fc receptors to tumor cells that expressed p185(her2/neu). The presence of the IgG binding domain significantly enhanced CDC and ADCC activity and improved antitumor activity in vivo. Our results illustrate a novel general approach to improve antibody-like proteins for therapeutic applications.


Asunto(s)
Anticuerpos Antineoplásicos/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Neoplasias/inmunología , Anticuerpos de Cadena Única/inmunología , Animales , Anticuerpos Antineoplásicos/administración & dosificación , Anticuerpos Antineoplásicos/metabolismo , Afinidad de Anticuerpos/inmunología , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Línea Celular , Activación de Complemento , Citotoxicidad Inmunológica , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Ratones , Neoplasias/terapia , Unión Proteica , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Anticuerpos de Cadena Única/administración & dosificación , Anticuerpos de Cadena Única/metabolismo , Trasplante Heterólogo
20.
Arch Oral Biol ; 58(6): 628-37, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23245859

RESUMEN

OBJECTIVE: Nitrogen-containing bisphosphonates (NBPs), the first-choice drugs for diseases that cause enhanced bone resorption, may injure jawbones and gastrointestinal tissues. In rodents, NBPs cause necrosis at injection sites. Bisphosphonates accumulate within bones, especially where there is inflammation. We hypothesized that if jawbone-accumulated NBPs are released, they may directly injure cells around the jawbones. To examine this hypothesis, we compared the direct effects of zoledronate (NBP) and/or etidronate (non-NBP) on various cells, including periodontal cells. DESIGN: Various human tumour cells (such as squamous carcinoma cells and prostate adenocarcinoma cells) and periodontal cells (such as gingival fibroblasts and periodontal ligament cells) were incubated with or without zoledronate and/or etidronate. Cell viability and cytotoxicity were determined by tetrazolium dye assay and by FITC-Annexin V/propidium iodide assay, respectively. RESULTS: Zoledronate, at 100µM, was toxic to all types of cells tested, while its toxicity varied among cells at both 1 and 10µM. There was no clear difference between tumour cells and non-tumour cells in sensitivity to the cytotoxicity of zoledronate. In contrast, etidronate was not toxic at 1-100µM in any of the cells tested. Interestingly, etidronate reduced the cytotoxicity of zoledronate in many cell-types, including gingival fibroblasts. CONCLUSIONS: These results, together with those reported by others and those from our previous in vivo experiments, suggest that NBPs, upon release from jawbones (e.g., during dental surgery or bone infection), may directly injure various cells located around the jawbones, and that etidronate may be protective against the cytotoxicity of NBPs in periodontal tissues.


Asunto(s)
Conservadores de la Densidad Ósea/toxicidad , Difosfonatos/toxicidad , Ácido Etidrónico/toxicidad , Imidazoles/toxicidad , Adenocarcinoma/patología , Anexina A5 , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/patología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Colorantes , Cemento Dental/efectos de los fármacos , Difosfonatos/antagonistas & inhibidores , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Ácido Etidrónico/farmacología , Fibroblastos/efectos de los fármacos , Fluoresceína-5-Isotiocianato/análogos & derivados , Colorantes Fluorescentes , Encía/citología , Encía/efectos de los fármacos , Humanos , Imidazoles/antagonistas & inhibidores , Leucemia Monocítica Aguda/patología , Células Madre Mesenquimatosas/efectos de los fármacos , Ligamento Periodontal/citología , Ligamento Periodontal/efectos de los fármacos , Propidio , Sales de Tetrazolio , Venas Umbilicales/citología , Ácido Zoledrónico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA