Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Hematol Oncol ; 15(1): 177, 2022 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-36581954

RESUMO

Anti-CD38 monoclonal antibodies (mAbs), daratumumab, and isatuximab have represented a breakthrough in the treatment of multiple myeloma (MM). Recently, CD38-based mAbs were expected to achieve increasing potential beyond MM, which encouraged us to develop new anti-CD38 mAbs to meet clinical needs. In this study, we developed a novel humanized anti-CD38 antibody, FTL004, which exhibited enhanced pro-apoptotic ability and negligible binding to red blood cells (RBCs). FTL004 presented a better ability to induce direct apoptosis independent of Fc-mediated cross-linking against lymphoma and MM cell lines as well as primary myeloma cells derived from MM patients. For instance, FTL004 induced RPMI 8226 cells with 55% early apoptosis cells compared with 20% in the isatuximab-treated group. Of interest, FTL004 showed ignorable binding to CD38 on human RBCs in contrast to tumor cells, even at concentrations up to 30 µg/mL. Furthermore, with an engineered Fc domain, FTL004 displayed stronger antibody-dependent cellular cytotoxicity (ADCC) against CD38+ malignant cells. In vivo MM and non-Hodgkin lymphoma tumor xenograft models showed that FTL004 possessed an effective anti-tumor effect. Cryo-electron microscopy structure resolved two epitope centers of FTL004 on CD38: one of which was unique while the other partly overlapped with that of isatuximab. Taken together, FTL004 distinguishes it from other CD38 targeting mAbs and represents a potential candidate for the treatment of MM and non-Hodgkin lymphoma.


Assuntos
Antineoplásicos , Linfoma não Hodgkin , Mieloma Múltiplo , Humanos , Mieloma Múltiplo/patologia , Microscopia Crioeletrônica , ADP-Ribosil Ciclase 1 , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linfoma não Hodgkin/tratamento farmacológico , Eritrócitos/patologia
2.
MedComm (2020) ; 1(3): 338-350, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34766126

RESUMO

Ovarian cancer is considered to be the most lethal gynecologic malignancy, and despite the development of conventional therapies and new therapeutic approaches, the patient's survival time remains short because of tumor recurrence and metastasis. Therefore, effective methods to control tumor progression are urgently needed. The oncofetal tumor-associated antigen 5T4 (trophoblast glycoprotein, TPBG) represents an appealing target for adoptive T-cell immunotherapy as it is highly expressed on the surface of various tumor cells, has very limited expression in normal tissues, and spreads widely in malignant tumors throughout their development. In this study, we generated second-generation human chimeric antigen receptor (CAR) T cells with redirected specificity to 5T4 (5T4 CAR-T) and demonstrated that these CAR-T cells can elicit lytic cytotoxicity in targeted tumor cells, in addition to the secretion of cytotoxic cytokines, including IFN-γ, IL-2, and GM-CSF. Furthermore, adoptive transfer of 5T4 CAR-T cells significantly delayed tumor formation in xenografts of peritoneal and subcutaneous animal models. These results demonstrate the potential efficacy and feasibility of 5T4 CAR-T cell immunotherapy and provide a theoretical basis for the clinical study of future immunotherapies targeting 5T4 for ovarian cancer.

3.
Int J Mol Sci ; 13(4): 4920-4936, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22606020

RESUMO

Endothelial cells (ECs) that line the lumen of blood vessels are important players in blood vessel formation, and EC migration is a key component of the angiogenic process. Thus, identification of genes that are specifically or preferentially expressed in vascular ECs and in-depth understanding of their biological functions may lead to discovery of new therapeutic targets. We have previously reported molecular characterization of human endothelial cell-specific molecule 2 (ECSM2)/endothelial cell-specific chemotaxis regulator (ECSCR). In the present study, we cloned two mouse full-length cDNAs by RT-PCR, which encode two putative ECSCR isoform precursors with considerable homology to the human ECSCR. Nucleotide sequence and exon-intron junction analyses suggested that they are alternative splicing variants (ECSCR isoform-1 and -2), differing from each other in the first and second exons. Quantitative RT-PCR results revealed that isoform-2 is the predominant form, which was most abundant in heart, lung, and muscles, and moderately abundant in uterus and testis. In contrast, the expression of isoform-1 seemed to be more enriched in testis. To further explore their potential cellular functions, we expressed GFP- and FLAG-tagged ECSCR isoforms, respectively, in an ECSCR deficient cell line (HEK293). Interestingly, the actual sizes of either ECSCR-GFP or -FLAG fusion proteins detected by immunoblotting are much larger than their predicted sizes, suggesting that both isoforms are glycoproteins. Fluorescence microscopy revealed that both ECSCR isoforms are localized at the cell surface, which is consistent with the structural prediction. Finally, we performed cell migration assays using mouse endothelial MS1 cells overexpressing GFP alone, isoform-1-GFP, and isoform-2-GFP, respectively. Our results showed that both isoforms significantly inhibited vascular epidermal growth factor (VEGF)-induced cell migration. Taken together, we have provided several lines of experimental evidence that two mouse ECSCR splicing variants/isoform precursors exist. They are differentially expressed in a variety of tissue types and likely involved in modulation of vascular EC migration. We have also defined the gene structure of mouse ECSCR using bioinformatics tools, which provides new information towards a better understanding of alternative splicing of ECSCR.


Assuntos
Processamento Alternativo/genética , Proteínas Reguladoras de Apoptose/genética , Movimento Celular/genética , Células Endoteliais/citologia , Neovascularização Fisiológica/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Clonagem Molecular , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Proteínas de Membrana , Camundongos , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Alinhamento de Sequência , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Mol Endocrinol ; 25(12): 2119-33, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22034225

RESUMO

Dysfunction and destruction of pancreatic islet ß-cells is a hallmark of diabetes. Better understanding of cell signals regulating ß-cell growth and antiapoptosis will allow development of therapeutic strategies for diabetes by preservation and expansion of ß-cell mass. GH and IGF-I share a complicated physiological relationship and have both been implicated in ß-cell function. GH and IGF-I exert their biological effects through binding to respective receptors (GHR and IGF-IR) and subsequently engaging downstream signaling pathways. However, their collaborative roles in modulation of ß-cell mass and the underlying molecular mechanisms remain poorly understood. In this study, we demonstrate that cultured ß-cells are appealing systems for investigating potential GH-IGF-I signaling cross talk. We uncover that GH specifically promotes formation of a protein complex containing GHR, Janus kinase 2 (a nonreceptor kinase coupled to GH/GHR signaling), and IGF-IR. More importantly, GH and IGF-I synergistically activate both signal transducer and activator of transcription 5 and Akt pathways. Concomitantly, ß-cells proliferate more robustly and are better protected from serum deprivation-induced apoptosis when exposed to GH and IGF-I in combination vs. GH or IGF-I alone. The augmented proliferative effects by GH and IGF-I are confirmed in isolated islets. Taken together, our findings strongly suggest that there exists a novel signaling relationship between GH/GHR and IGF-I/IGF-IR systems in ß-cells, i.e. IGF-IR may serve as a proximal component of GH/GHR signaling, contributing to enhancement of ß-cell mass and function. In support of this, IGF-IR knockdown in ß-cells resulted in the desensitization of acute GH-induced signal transducer and activator of transcription 5 activation.


Assuntos
Hormônio do Crescimento/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Células Secretoras de Insulina/fisiologia , Receptor Cross-Talk , Receptor IGF Tipo 1/metabolismo , Receptores da Somatotropina/metabolismo , Animais , Apoptose , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Ativação Enzimática , Técnicas de Silenciamento de Genes , Hormônio do Crescimento/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Células Secretoras de Insulina/metabolismo , Janus Quinase 2/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Multimerização Proteica , Interferência de RNA , Ratos , Receptor IGF Tipo 1/genética , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais
5.
PLoS One ; 6(6): e21482, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21720547

RESUMO

BACKGROUND: Despite its first discovery by in silico cloning of novel endothelial cell-specific genes a decade ago, the biological functions of endothelial cell-specific molecule 2 (ECSM2) have only recently begun to be understood. Limited data suggest its involvement in cell migration and apoptosis. However, the underlying signaling mechanisms and novel functions of ECSM2 remain to be explored. METHODOLOGY/PRINCIPAL FINDINGS: A rabbit anti-ECSM2 monoclonal antibody (RabMAb) was generated and used to characterize the endogenous ECSM2 protein. Immunoblotting, immunoprecipitation, deglycosylation, immunostaining and confocal microscopy validated that endogenous ECSM2 is a plasma membrane glycoprotein preferentially expressed in vascular endothelial cells (ECs). Expression patterns of heterologously expressed and endogenous ECSM2 identified that ECSM2 was particularly concentrated at cell-cell contacts. Cell aggregation and transwell assays showed that ECSM2 promoted cell-cell adhesion and attenuated basic fibroblast growth factor (bFGF)-driven EC migration. Gain or loss of function assays by overexpression or knockdown of ECSM2 in ECs demonstrated that ECSM2 modulated bFGF-directed EC motility via the FGF receptor (FGFR)-extracellular regulated kinase (ERK)-focal adhesion kinase (FAK) pathway. The counterbalance between FAK tyrosine phosphorylation (activation) and ERK-dependent serine phosphorylation of FAK was critically involved. A model of how ECSM2 signals to impact bFGF/FGFR-driven EC migration was proposed. CONCLUSIONS/SIGNIFICANCE: ECSM2 is likely a novel EC junctional protein. It can promote cell-cell adhesion and inhibit bFGF-mediated cell migration. Mechanistically, ECSM2 attenuates EC motility through the FGFR-ERK-FAK pathway. The findings suggest that ECSM2 could be a key player in coordinating receptor tyrosine kinase (RTK)-, integrin-, and EC junctional component-mediated signaling and may have important implications in disorders related to endothelial dysfunction and impaired EC junction signaling.


Assuntos
Movimento Celular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Junções Intercelulares/metabolismo , Proteínas de Membrana/metabolismo , Transdução de Sinais/efeitos dos fármacos , Anticorpos Monoclonais/imunologia , Proteínas Reguladoras de Apoptose , Adesão Celular/efeitos dos fármacos , Agregação Celular/efeitos dos fármacos , Linhagem Celular , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Técnicas de Silenciamento de Genes , Glicosilação/efeitos dos fármacos , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Junções Intercelulares/efeitos dos fármacos , Modelos Biológicos , Transporte Proteico/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo , beta Catenina/metabolismo
6.
J Biomed Biotechnol ; 2010: 134764, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20454583

RESUMO

Apoptosis plays an important role in embryonic development. PNAS-4 has been demonstrated to induce apoptosis in several cancer cells. In this study, we cloned Xenopus laevis PNAS-4 (xPNAS-4), which is homologous to the human PNAS-4 gene. Bioinformatics analysis for PNAS-4 indicated that xPNAS-4 shared 87.6% identity with human PNAS-4 and 85.5% with mouse PNAS-4. The phylogenetic tree of PNAS-4 protein was also summarized. An analysis of cellular localization using an EGFP-fused protein demonstrated that xPNAS-4 was localized in the perinuclear region of the cytoplasm. RT-PCR analysis revealed that xPNAS-4, as a maternally expressed gene, was present in all stages of early embryo development. Whole-mount in situ hybridization showed that xPNAS-4 was mainly expressed in ectoderm and mesoderm. Furthermore, microinjection of xPNAS-4 mRNA in vivo caused developmental defects manifesting as a small eye phenotype in the Xenopous embryos, and as a small eye or one-eye phenotype in developing zebrafish embryos. In addition, embryos microinjected with xPNAS-4 antisense morpholino oligonucleotides (MOs) exhibited a failure of head development and shortened axis.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Embrião não Mamífero/embriologia , Desenvolvimento Embrionário/genética , Proteínas de Xenopus/genética , Xenopus laevis/embriologia , Xenopus laevis/genética , Sequência de Aminoácidos , Animais , Apoptose , Proteínas Reguladoras de Apoptose/química , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular , Biologia Computacional , Embrião não Mamífero/anormalidades , Embrião não Mamífero/metabolismo , Anormalidades do Olho/patologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Microinjeções , Dados de Sequência Molecular , Filogenia , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Tempo , Proteínas de Xenopus/química , Proteínas de Xenopus/deficiência , Proteínas de Xenopus/metabolismo
7.
Acta Pharmacol Sin ; 31(4): 493-500, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20228829

RESUMO

AIM: The adverse effects of local anesthetics (LAs) on wound healing at surgical sites have been suggested, and may be related to their cytotoxicity. This study was aimed to compare the cellular toxicity of bupivacaine and lidocaine (two well-known LAs), and to explore the molecular mechanism(s). METHODS: Toxicity of bupivacaine and lidocaine was assessed in cultured mouse C2C12 myoblasts by cell viability and apoptosis assays. Effects of LAs on extracellular signal-regulated kinase (ERK) and protein kinase B (Akt) activation, which are essential for cell proliferation and survival, were evaluated by immunoblotting. RESULTS: Both LAs, especially bupivacaine, prevented cell growth and caused cell death in a dose-dependent manner. The half maximal inhibitory concentrations (IC(50)) for bupivacaine and lidocaine were 0.49+/-0.04 and 3.37+/-0.53 mmol/L, respectively. When applied at the same dilutions of commercially available preparations, the apoptotic effect induced by bupivacaine was more severe than that of lidocaine in C2C12 cells. Furthermore, bupivacaine significantly diminished the ERK activation, which may underlie its anti-proliferative actions. Both LAs suppressed Akt activation, which correlated with their effects on apoptosis. CONCLUSION: Our study demonstrated that, when used at the same dilutions from clinically relevant concentrations, bupivacaine is more cytotoxic than lidocaine in vitro. Anti-proliferation and cell death with concomitant apoptosis mediated by bupivacaine may offer an explanation for its adverse effects in vivo (eg slowing wound healing at the surgical sites). A less toxic, long-acting anesthetic may be needed.


Assuntos
Anestésicos Locais/efeitos adversos , Bupivacaína/efeitos adversos , Lidocaína/efeitos adversos , Mioblastos/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Camundongos , Mioblastos/citologia , Mioblastos/enzimologia , Proteínas Proto-Oncogênicas c-akt/metabolismo
8.
Genes Cells ; 14(3): 281-93, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19267780

RESUMO

Endothelial cell-specific molecules (ECSMs) play a pivotal role in the pathogenesis of many angiogenesis-related diseases. Since its initial discovery, the exact function of human ECSM2 has not been defined. In this study, by database mining, we identified a number of hypothetical proteins across species exhibiting substantial sequence homology to the human ECSM2. We showed that ECSM2 is preferentially expressed in endothelial cells and blood vessels. Their characteristic structures and unique expression patterns suggest that ECSM2 is an evolutionarily conserved gene and may have important functions. We further explored the potential roles of human ECSM2 at the molecular and cellular level. Using a reconstitution mammalian cell system, we demonstrated that ECSM2 mainly resides at the cell membrane, is critically involved in cell-shape changes and actin cytoskeletal rearrangement, and suppresses tyrosine phosphorylation signaling. More importantly, we uncovered that ECSM2 can cross-talk with epidermal growth factor receptor (EGFR) to attenuate the EGF-induced cell migration, possibly via inhibiting the Shc-Ras-ERK (MAP kinase) pathway. Given the importance of growth factor and receptor tyrosine kinase-mediated signaling and cell migration in angiogenesis-related diseases, our findings regarding the inhibitory effects of ECSM2 on EGF-mediated signaling and cell motility may have important therapeutic implications.


Assuntos
Actinas/metabolismo , Movimento Celular , Receptores ErbB/metabolismo , Proteínas de Membrana/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Linhagem Celular , Citoesqueleto/metabolismo , Células Endoteliais/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Humanos , Proteínas de Membrana/química , Modelos Biológicos , Fosforilação , Ratos , Transdução de Sinais , Tirosina/metabolismo , Peixe-Zebra
9.
Proteins ; 72(1): 323-32, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18214972

RESUMO

The stem cell factor receptor (c-Kit) plays critical roles in initiating cell growth and proliferation. Its kinase functional abnormality has been thought to associate with several human cancers. The regulation of c-Kit kinase activity is achieved by phosphorylation on the residues Tyr568 and Tyr570 within juxtamembrane region (JMR) and subsequent structural transition of JMR and activation loop (A-loop). However, the detailed conformational dynamics of JMR and A-loop are far from clear, especially whether their conformational changes are coupled or not during the kinase activation transition. In this investigation, the complete conformational transition pathway was determined using a series of nanosecond conventional molecular dynamics (MD) and targeted molecular dynamics (TMD) simulations in explicit water systems. The results of the MD simulations show that the phosphorylation of residues Tyr568 and Tyr570 within JMR induces the detachment of JMR from the kinase C-lobe and increases the fluctuation in the structure of JMR, thus appearing to initiate the kinase activation process. During the course of the TMD simulation, which characterizes the conformational transition of c-Kit from autoinhibitory to activated state, the JMR undergoes a rapid departure from the allosteric binding site and drifts into solvent, followed by the conformational flip of A-loop from inactive (fold) state to active (extended) state. A change in the orientation of helix alphaC in response to the motion of JMR and A-loop has also been observed. The computational results presented here indicate that the dissociation of JMR from the kinase domain is prerequisite to c-Kit activation, which is consistent with previous experiments.


Assuntos
Membrana Celular/enzimologia , Proteínas Proto-Oncogênicas c-kit/química , Proteínas Proto-Oncogênicas c-kit/metabolismo , Simulação por Computador , Cristalografia por Raios X , Ativação Enzimática , Humanos , Modelos Moleculares , Fosforilação , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Temperatura , Fatores de Tempo
10.
Biochem Biophys Res Commun ; 361(1): 74-8, 2007 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-17637457

RESUMO

Xenopus laevis has recently been determined as a novel study platform of gene function. In this study, we cloned Xenopus DRR1 (xDRR1), which is homologous to human down-regulated in renal carcinoma (DRR1) gene. Bioinformatics analysis for DRR1 indicated that xDRR1 shared 74% identity with human DRR1 and 66% with mouse DRR1, and the phlogenetic tree of DRR1 protein was summarized. The xDRR1 gene locates in nuclei determined by transfecting A549 cells with the recombinant plasmid pEGFP-N1/xDRR1. RT-PCR analysis revealed that xDRR1 gene was expressed in all stages of early embryo development and all kinds of detected tissues, and whole-mount in situ hybridization showed xDRR1 was mainly present along ectoderm and mesoderm. Furthermore, xDRR1 expression could suppress A549 cell growth by transfecting with plasmid pcDNA3.1(+)/xDRR1. xDRR1 probably plays important roles involving in cell growth regulation and Xenopus embryo development.


Assuntos
Desenvolvimento Embrionário , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Proteínas de Xenopus/genética , Proteínas de Xenopus/fisiologia , Sequência de Aminoácidos , Animais , Clonagem Molecular , Genes Supressores de Tumor , Inibidores do Crescimento/química , Inibidores do Crescimento/genética , Inibidores do Crescimento/fisiologia , Humanos , Dados de Sequência Molecular , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Homologia de Sequência de Aminoácidos , Proteínas de Xenopus/química , Xenopus laevis
11.
Biochem Biophys Res Commun ; 357(4): 924-30, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17466267

RESUMO

The VEGF family comprises seven members that are designated VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E, placental growth factor (PlGF), and VEGF-F. Of these factors, VEGF-D plays important roles for angiogenesis and lymphangiogenesis, and could promote tumor growth and lymphatic metastasis. In this study, we identified a zebrafish VEGF-D homolog that encodes a 272 amino acid protein including a PDGF (platelet-derived growth factor) domain characteristic to VEGF family. Expression profile demonstrated that the VEGF-D began expressed from 13 somite stage. Microinjecting zVEGF-D mRNA into zebrafish 1-cell stage embryos resulted in severe misguidance of intersegmental vessels (ISV) and abnormal connection between dorsal aorta and caudal vein. Microangiography indicated that these abnormal ISVs were not functional. Our studies therefore identified the first non-mammalian VEGF-D and established its in vivo role for vascular system development during vertebrate embryogenesis and provided an alternative animal model to further reveal functions of VEGF-D.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Desenvolvimento Embrionário/fisiologia , Neovascularização Fisiológica/fisiologia , Fator D de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/fisiologia , Animais , Regulação da Expressão Gênica no Desenvolvimento/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...