Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Sci Rep ; 14(1): 18293, 2024 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-39112658

RESUMO

The cytokine interleukin-6 (IL-6) plays a crucial role in autoimmune and inflammatory diseases. Understanding the precise mechanism of IL-6 interaction at the amino acid level is essential to develop IL-6-inhibiting compounds. In this study, we employed computer-guided drug design tools to predict the key residues that are involved in the interaction between IL-6 and its receptor IL-6R. Subsequently, we generated IL-6 mutants and evaluated their binding affinity to IL-6R and the IL-6R - gp130 complex, as well as monitoring their biological activities. Our findings revealed that the R167A mutant exhibited increased affinity for IL-6R, leading to enhanced binding to IL-6R - gp130 complex and subsequently elevated intracellular phosphorylation of STAT3 in effector cells. On the other hand, although E171A reduced its affinity for IL-6R, it displayed stronger binding to the IL-6R - gp130 complex, thereby enhancing its biological activity. Furthermore, we identified the importance of R178 and R181 for the precise recognition of IL-6 by IL-6R. Mutants R181A/V failed to bind to IL-6R, while maintaining an affinity for the IL-6 - gp130 complex. Additionally, deletion of the D helix resulted in complete loss of IL-6 binding affinity for IL-6R. Overall, this study provides valuable insights into the binding mechanism of IL-6 and establishes a solid foundation for future design of novel IL-6 inhibitors.


Assuntos
Interleucina-6 , Simulação de Acoplamento Molecular , Ligação Proteica , Receptores de Interleucina-6 , Interleucina-6/metabolismo , Interleucina-6/genética , Humanos , Receptores de Interleucina-6/metabolismo , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/química , Receptor gp130 de Citocina/metabolismo , Receptor gp130 de Citocina/genética , Receptor gp130 de Citocina/química , Mutagênese Sítio-Dirigida , Sítios de Ligação , Fator de Transcrição STAT3/metabolismo , Fosforilação , Mutação
2.
Heliyon ; 10(9): e30551, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38756565

RESUMO

Natural killer (NK) cells represent key player in immune surveillance to eliminate transformed or malignant cells. One of mechanisms of action of NK cells is antibody-dependent cell-mediated cytotoxicity (ADCC) by recognizing tumor antigens on the surface of cancer cells. However, the heterogeneity of tumor antigens and the scarcity of membrane surface targets significantly restrict this strategy. Recently, we constructed a new cargo by tethering a low pH insertion peptide (pHLIP) to the C terminus of the ectodomain of programed death ligand-1 (PD-L1) and demonstrated its ability to modulate immune responses. Herein, the potential application of PD-L1-pHLIP in cancer therapy was determined. pHLIP tethering had no effect on the binding capacity of PD-L1 protein to an anti-PD-L1 antibody (i.e. avelumab). Association of pHLIP rendered PD-L1 segment display on the surface of cellular membrane in the acidic buffer instead of the neutral solution. Importantly, plate-coated or beads-coupled PD-L1-pHLIP enable robust activation and expression of cytotoxic mediators of NK cells via engaging avelumab. Overall, this work provides proof of concept that recombinant PD-L1 protein decorated on the cellular membrane driven by pHLIP in combination with appropriate monoclonal antibody has potentials to elicit NK cytotoxicity, which may represent a novel and promising therapeutic avenue in cancer.

3.
Biomed Pharmacother ; 162: 114618, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37011485

RESUMO

CD47, as an innate immune checkpoint molecule, is an important target of cancer immunotherapy. We previously reported that a high-affinity SIRPα variant FD164 fused with IgG1 subtype Fc showed a better antitumor effect than wild-type SIRPα in an immunodeficient tumor-bearing model. However, CD47 is widely expressed in blood cells, and the drugs targeting CD47 may cause potential hematological toxicity. Herein, we modified the FD164 molecule by Fc mutation (N297A) to inactivate the Fc-related effector function and named it nFD164. Moreover, we further studied the potential of nFD164 as a candidate drug targeting CD47, including the stability, in vitro activity, antitumor activity of single or combined drugs in vivo, and hematological toxicity in humanized CD47/SIRPα transgenic mouse model. The results show that nFD164 maintains strong binding activity to CD47 on tumor cells, but has weak binding activity with red blood cells or white blood cells, and nFD164 has good drug stability under accelerated conditions (high temperature, bright light and freeze-thaw cycles). More importantly, in the immunodeficient or humanized CD47/SIRPα transgenic mice bearing tumor model, the combination of nFD164 and anti-CD20 antibody or anti-mPD-1 antibody had a synergistic antitumor effect. Especially in transgenic mouse models, nFD164 combined with anti-mPD-1 significantly enhanced tumor suppressive activity compared with anti-mPD-1 (P < 0.01) or nFD164 (P < 0.01) as a single drug and had fewer hematology-related side effects than FD164 or Hu5F9-G4. When these factors are taken together, nFD164 is a promising high-affinity CD47-targeting drug candidate with better stability, potential antitumor activity, and improved safety profile.


Assuntos
Antígeno CD47 , Neoplasias , Camundongos , Animais , Antígeno CD47/metabolismo , Imunoterapia/métodos , Modelos Animais de Doenças , Camundongos Transgênicos , Sistemas de Liberação de Medicamentos , Neoplasias/tratamento farmacológico , Fagocitose
4.
Front Immunol ; 14: 1090311, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36936961

RESUMO

The Interleukin-2 Family contains six kinds of cytokines, namely IL-2, IL-15, IL-4, IL-7, IL-9, and IL-21, all of which share a common γ chain. Many cytokines of the IL-2 family have been reported to be a driving force in immune cells activation. Therefore, researchers have tried various methods to study the anti-tumor effect of cytokines for a long time. However, due to the short half-life, poor stability, easy to lead to inflammatory storms and narrow safety treatment window of cytokines, this field has been tepid. In recent years, with the rapid development of protein engineering technology, some engineered cytokines have a significant effect in tumor immunotherapy, showing an irresistible trend of development. In this review, we will discuss the current researches of the IL-2 family and mainly focus on the application and achievements of engineered cytokines in tumor immunotherapy.


Assuntos
Citocinas , Neoplasias , Humanos , Citocinas/metabolismo , Interleucina-2/uso terapêutico , Imunoterapia/métodos , Neoplasias/terapia
5.
Int J Biol Sci ; 19(3): 916-935, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36778110

RESUMO

The mechanisms of self-renewal and pluripotency maintenance of human pluripotent stem cells (hPSCs) have not been fully elucidated, especially for the role of those poorly characterized long noncoding RNAs (lncRNAs). ESRG is a lncRNA highly expressed in hPSCs, and its functional roles are being extensively explored in the field. Here, we identified that the transcription of ESRG can be directly regulated by OCT4, a key self-renewal factor in hPSCs. Knockdown of ESRG induces hPSC differentiation, cell cycle arrest, and apoptosis. ESRG binds to MCM2, a replication-licensing factor, to sustain its steady-state level and nuclear location, safeguarding error-free DNA replication. Further study showed that ESRG knockdown leads to MCM2 abnormalities, resulting in DNA damage and activation of the p53 pathway, ultimately impairs hPSC self-renewal and pluripotency, and induces cell apoptosis. In summary, our study suggests that ESRG, as a novel target of OCT4, plays an essential role in maintaining the cell survival and self-renewal/pluripotency of hPSCs in collaboration with MCM2 to suppress p53 signaling. These findings provide critical insights into the mechanisms underlying the maintenance of self-renewal and pluripotency in hPSCs by lncRNAs.


Assuntos
Componente 2 do Complexo de Manutenção de Minicromossomo , Células-Tronco Pluripotentes , RNA Longo não Codificante , Proteína Supressora de Tumor p53 , Humanos , Diferenciação Celular/genética , Sobrevivência Celular/genética , Componente 2 do Complexo de Manutenção de Minicromossomo/genética , Componente 2 do Complexo de Manutenção de Minicromossomo/metabolismo , Células-Tronco Pluripotentes/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
6.
Int Immunopharmacol ; 115: 109634, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36584573

RESUMO

Antibody or antibody-like protein drugs related to tumor immunotherapy are now widely used. Here, we describe an antibody-fusion protein drug IMAB362-mIL-21 with mouse IL-21 (mIL-21) fused into the C-terminal domain of IMAB362 (a clinical antibody drug against Claudin18.2), that we expect can achieve tumor targeting and activate local anti-tumor immune response more effectively, while reducing the systemic side effects of individual cytokines. In vitro assays comparing the fusion protein IMAB362-mIL-21 to IMAB362 and mIL-21, IMAB362-mIL-21 was able to recognize its cognate antigen Claudin18.2 and natural receptor mIL-21R with similar binding affinities, mediate equivalent ADCC activity and activate IL-21R-mediated downstream signal pathway. In in vivo assays, IMAB362-mIL-21 produced stronger anti-tumor effects compared with IMAB362 or mIL-21 or their combination at equimolar concentrations. Moreover, according to routine blood indicators, mIL-21-Fc and the combined treatment group had significant decreases (P < 0.01) in red blood cells (RBC), hemoglobin (HGB) and hematocrit (HCT), while the IMAB362-mIL-21 group did not. The above results have shown that IMAB362-mIL-21 can produce better anti-tumor effects without obvious hematological toxicity, which is sufficient to show that this kind of antibody-cytokine protein has better application value than IMAB362 or IL-21 as single drugs or in combination. Therefore, this bifunctional molecule combined tumor-targeting and immune activation effectively and has good application prospects.


Assuntos
Neoplasias , Camundongos , Animais , Neoplasias/tratamento farmacológico , Interleucinas , Imunoterapia , Transdução de Sinais , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/uso terapêutico
7.
Front Immunol ; 13: 1047610, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36518768

RESUMO

Several clinical studies demonstrate that there exist other immune checkpoints overexpressed in some PD-1 inhibitor-resistant tumor patients. Among them, Lymphocyte-activation gene 3 (LAG-3) is one of the important immune checkpoint molecules and has been clinically demonstrated to have synergistic anti-tumor effects in combination with PD-1 antibody. In this study, we designed a novel 'knob-in-hole' PD-1/LAG-3 bispecific antibody (BsAb) YG-003D3. In conclusion, the BsAb maintained the similar affinity and thermal stability to the parental antibody, and the BsAb structure can be independent of each other in the process of double-target recognition, and the recognition activity will not be affected. Moreover, the BsAb can not only target PD-1 and LAG-3 on single cell simultaneously, but also bridge the two kinds of cells expressing PD-1 and LAG-3, so as to release the 'brake system of immune checkpoints' and activate immune cells to exert anti-tumor effects more effectively. Especially in the PBMCs activation assay, YG-003D3 induced stronger IFN-γ, IL-6, and TNF-α secretion compared to anti-PD-1 or anti-LAG-3 single drug group or even combined drug group. In the tumor killing experiment of PBMC in vitro, YG-003D3 has a better ability to activate PBMC to kill tumor cells than anti-PD-1 or anti-LAG-3 single drug group or even combined drug group, and the killing rate is as high as 20%. In a humanized PD-1/LAG-3 transgenic mouse subcutaneous tumor-bearing model, YG-003D3 showed good anti-tumor activity, even better than that of the combination group at the same molar concentration. Further studies have shown that YG-003D3 could significantly alter the proportion of immune cells in the tumor microenvironment. In particular, the proportion of CD45+, CD3+ T, CD8+ T cells in tumor tissue and the proportion of CD3+ T, CD8+ T, CD4+ T cells in peripheral blood were significantly increased. These results suggest that YG-003D3 exerts a potent antitumor effect by activating the body 's immune system. In summary, the BsAb YG-003D3 has good anti-tumor activity, which is expected to become a novel drug candidate for cancer immunotherapy.


Assuntos
Anticorpos Biespecíficos , Neoplasias , Camundongos , Animais , Linfócitos T CD8-Positivos , Leucócitos Mononucleares , Anticorpos Biespecíficos/farmacologia , Anticorpos Biespecíficos/uso terapêutico , Ativação Linfocitária , Imunoterapia , Neoplasias/tratamento farmacológico , Microambiente Tumoral
8.
Mol Pharmacol ; 102(3): 161-171, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35764384

RESUMO

Sialic acid-binding Ig-like lectin-15 is an important immunosuppressive molecule considered to be a key target in next-generation tumor immunotherapy. In this study, we screened 22 high-affinity antibodies that specifically recognize human Siglec-15 by using a large human phage antibody library, and five representative sequences were selected for further study. The results showed the binding activity of five antibodies to Siglec-15 (EC50 ranged from 0.02368 µg/mL to 0.07949 µg/mL), and in two Siglec-15-overexpressed cell lines, three antibodies had the strongest binding activity, so the two clones were discarded for further study. Subsequently, the affinity of three antibodies were measured by bio-layer interferometry technology (5-9 × 10E-09M). As the reported ligands of Siglec-15, the binding activity of Siglec-15 and sialyl-Tn, cluster of differentiation 44, myelin-associated glycoprotein, and leucine-rich repeat-containing protein 4C can be blocked by three of the antibodies. Among these, 3F1 had a competitive advantage. Then, the antibody 3F1 showed an obvious antibody-dependent cell-mediated cytotoxicity effect (EC50 was 0.85 µg/mL). Further, antibody 3F1 can reverse the inhibitory effect of Siglec-15 on lymphocyte proliferation (especially CD4+T and CD8+T) and cytokine release Interferon-γ. Given the above results, 3F1 was selected as a candidate for the in vivo pharmacodynamics study. In the tumor model of Balb/c Nude mice, 3F1 (10 mg/kg) showed certain antitumor effects [tumor growth inhibition (TGI) was 31.5%], while the combination of 3F1 (5 mg/kg) and Erbitux (5 mg/kg) showed significant antitumor effects (TGI was 48.7%) compared with the PBS group. In conclusion, novel human antibody 3F1 has antitumor activity and is expected to be an innovative candidate drug targeting Siglec-15 for tumor immunotherapy. SIGNIFICANCE STATEMENT: Siglec-15 is considered as an important target in the next generation of tumor immunotherapy. 3F1 is expected to be the most promising potential candidate for targeting Siglec-15 for cancer treatment and could provide a reference for the development of antitumor drugs.


Assuntos
Antígenos CD , Neoplasias , Animais , Antígenos CD/metabolismo , Humanos , Imunoglobulinas , Lectinas/química , Lectinas/metabolismo , Ligantes , Proteínas de Membrana , Camundongos , Camundongos Nus , Neoplasias/tratamento farmacológico
9.
Microbiol Spectr ; 10(3): e0221221, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35384693

RESUMO

Ebola virus, a member of the Filoviridae family, utilizes the attachment factors on host cells to support its entry and cause severe tissue damage. TIM-1 has been identified as a predominant attachment factor via interaction with phosphatidylserine (PS) localized on the viral envelope and glycoprotein (GP). In this study, we give the first demonstration that TIM-1 enhances the cellular entry of three species of Ebola virus, as well as those harboring GP mutations (A82V, T544I, and A82V T544I). Furthermore, two TIM-1 variants (i.e., TIM-1-359aa and TIM-1-364aa) had comparable effects on promoting Zaire Ebola virus (EBOV) attachment, internalization, and infection. Importantly, recombinant TIM-1 ectodomain (ECD) protein could decrease the infectivity of Ebola virus and display synergistic inhibitory effects with ADI-15946, a monoclonal antibody with broad neutralizing activity to Ebola virus. Of note, EBOV strains harboring GP mutations (K510E and D552N), which were refractory to antibody treatment, were still sensitive to TIM-1 protein-mediated impairment of infectivity, indicating that TIM-1 protein may represent an alternative therapeutic regimen when antibody evasion occurs. IMPORTANCE The viral genome has acquired numerous mutations with the potential to increase transmission during the 2013-to-2016 outbreak of Ebola virus. EBOV strains harboring GP mutations (A82V, T544I, and A82V T544I), which have been identified to increase viral infectivity in humans, have attracted our attention. Herein, we give the first report that polymorphic TIM-1 enhances the infectivity of three species of Ebola virus, as well as those harboring GP mutations (A82V, T544I, and A82V T544I). We show that recombinant TIM-1 ECD protein could decrease the infectivity of Ebola virus with or without a point mutation and displays synergistic inhibitory effects with ADI-15946. Furthermore, TIM-1 protein potently blocked cell entry of antibody-evading Ebola virus species. These findings highlight the role of TIM-1 in Ebola virus infection and indicate that TIM-1 protein represents a potential therapeutic avenue for Ebola virus and its mutated species.


Assuntos
Ebolavirus , Doença pelo Vírus Ebola , Ebolavirus/genética , Genoma Viral , Doença pelo Vírus Ebola/epidemiologia , Humanos , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus
10.
Nat Commun ; 12(1): 6943, 2021 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-34836962

RESUMO

The pathological role of reactive gliosis in CNS repair remains controversial. In this study, using murine ischemic and hemorrhagic stroke models, we demonstrated that microglia/macrophages and astrocytes are differentially involved in engulfing synapses in the reactive gliosis region. By specifically deleting MEGF10 and MERTK phagocytic receptors, we determined that inhibiting phagocytosis of microglia/macrophages or astrocytes in ischemic stroke improved neurobehavioral outcomes and attenuated brain damage. In hemorrhagic stroke, inhibiting phagocytosis of microglia/macrophages but not astrocytes improved neurobehavioral outcomes. Single-cell RNA sequencing revealed that phagocytosis related biological processes and pathways were downregulated in astrocytes of the hemorrhagic brain compared to the ischemic brain. Together, these findings suggest that reactive microgliosis and astrogliosis play individual roles in mediating synapse engulfment in pathologically distinct murine stroke models and preventing this process could rescue synapse loss.


Assuntos
Encéfalo/patologia , Gliose/imunologia , Infarto da Artéria Cerebral Média/complicações , Sinapses/patologia , Animais , Astrócitos/metabolismo , Encéfalo/citologia , Encéfalo/imunologia , Modelos Animais de Doenças , Regulação para Baixo/imunologia , Feminino , Gliose/patologia , Humanos , Infarto da Artéria Cerebral Média/imunologia , Infarto da Artéria Cerebral Média/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Fagocitose/genética , Fagocitose/imunologia , RNA-Seq , Análise de Célula Única , Sinapses/imunologia , c-Mer Tirosina Quinase/genética , c-Mer Tirosina Quinase/metabolismo
11.
Bioorg Chem ; 116: 105366, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34560561

RESUMO

In recent years, tumor immunotherapy, especially the combination of PD1/PD-L1 inhibitors and chemotherapy, has developed rapidly. However, the systemic side effects induced by chemotherapy remain a crucial problem that needs to be addressed. Antibody drug conjugates (ADCs) are exceptional target-specific prodrugs that greatly improve the therapeutic window of chemotherapy drugs. Therefore, designing PD-L1-targeting ADCs is an interesting research project. In this study, we confirmed for the first time that the commercial anti-PD-L1 antibody Atezolizumab has better endocytosis efficiencies than Avelumab, and was more suitable for ADC design. Then, the most popular cytotoxic payload MMAE was conjugated to Atezolizumab via a classical dipeptide (valine-alanine) linker to generate a bifunctional PD-L1 ADC (ADC 3). An in vitro cytotoxicity test indicated the potent tumor cell inhibitory activity of ADC 3, with EC50 values of 9.75 nM to 11.94 nM. In addition, a co-culture of PBMCs in vitro proved that ADC 3 retained the immune activation effect of the Atezolizumab antibody. Moreover, ADC 3 exhibited a higher tumor inhibition rate and tumor regression rate in humanized immune system mice. To the best of our knowledge, this is the most active PD-L1-ADC reported thus far, which may promote the development of immunotherapy and novel ADCs.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Desenvolvimento de Medicamentos , Imunoconjugados/farmacologia , Imunoterapia , Oligopeptídeos/farmacologia , Anticorpos Monoclonais Humanizados/química , Antineoplásicos/química , Antígeno B7-H1/imunologia , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Imunoconjugados/química , Estrutura Molecular , Oligopeptídeos/química , Relação Estrutura-Atividade , Células Tumorais Cultivadas
12.
Mol Pharmacol ; 100(3): 193-202, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34315811

RESUMO

Phagocytic resistance plays a key role in tumor-mediated immune escape, so phagocytosis immune checkpoints are a potential target for cancer immunotherapy. CD47 is one of the important phagocytosis immune checkpoints; thus, blocking the interaction between CD47 and signal regulatory protein α (SIRPα) may provide new options for cancer treatment. Using computer-aided targeted epitope mammalian cell-displayed antibody library, we screened and obtained an engineered SIRPα variant fragment crystallizable fusion protein, FD164, with higher CD47-binding activity than wild-type SIRPα Compared with wild-type SIRPα, FD164 has approximately 3-fold higher affinity for binding to CD47, which further enhanced its phagocytic effect in vitro and tumor suppressor activity in vivo. FD164 maintains the similar antitumor activity of the clinical research drug Hu5F9 in the mouse xenograft model. Furthermore, FD164 combined with rituximab can significantly improve the effect of single-agent therapy. On the other hand, compared with Hu5F9, FD164 does not cause hemagglutination, and its ability to bind to red blood cells or white blood cells is weaker at the same concentration. Finally, it was confirmed by computer structure prediction and alanine scanning experiments that the N45, E47, 52TEVYVK58, K60, 115EVTELTRE122, and E124 residues of CD47 are important for SIRPα or FD164 recognition. Briefly, we obtained a high-affinity SIRPα variant FD164 with balanced safety and effectiveness. SIGNIFICANCE STATEMENT: Up to now, few clinically marketed drugs targeting CD47 have been determined to be effective and safe. FD164, a potential signal regulatory protein α variant fragment crystallizable protein with balanced safety and effectiveness, could provide a reference for the development of antitumor drugs.


Assuntos
Antígenos de Diferenciação/genética , Antígenos de Diferenciação/imunologia , Antígeno CD47/imunologia , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Animais , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/uso terapêutico , Antígenos de Diferenciação/efeitos adversos , Antígenos de Diferenciação/química , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linfoma de Burkitt/tratamento farmacológico , Linfoma de Burkitt/imunologia , Linfoma de Burkitt/patologia , Antígeno CD47/química , Células CHO , Linhagem Celular , Cricetulus , Desenho de Fármacos , Epitopos/química , Epitopos/genética , Epitopos/imunologia , Hemaglutinação/efeitos dos fármacos , Imunoterapia , Camundongos SCID , Modelos Moleculares , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Receptores Imunológicos/química , Proteínas Recombinantes de Fusão/efeitos adversos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Rituximab/uso terapêutico , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Mater Chem B ; 9(32): 6347-6356, 2021 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-34251002

RESUMO

Gene therapy provides a promising treatment for glioblastoma multiforme, which mainly depends on two key aspects, crossing the blood brain barrier (BBB) effectively and transfecting target cells selectively. In this work, we reported a series of peptide-based vectors for transfecting glioma cells specifically consisting of several functional segments including a cell-penetrating peptide, targeting segment substance P (SP), an endosomal escape segment, a PEG linker and a stearyl moiety. The conformations and DNA-loading capacities of peptide vectors and the self-assembly behaviors of peptide/pGL3 complexes were characterized. The in vitro gene transfection was evaluated in U87, 293T-NK1R, and normal 293T cell lines. The transfection efficiency ratio of P-02 (SP-PEG4-K(C18)-(LLHH)3-R9) to Lipo2000 in the U87 cell line was about 36% higher than that in the 293T cell line. The neurokinin-1 receptor (NK1R) in U87 cells mediated the transfection process via interactions with the ligand SP in peptide vectors. The mechanism of NK1R mediated transfection was demonstrated by the use of gene-modified 293T cells expressing NK1R, as well as the gene transfection in the presence of free SP. Besides, P-02 could promote the pGL3 plasmids to cross the BBB model in vitro and achieved the EGFP gene transfection in the brain of zebrafish successfully. The designed peptide vectors, owing to their specific transfection capacity in glioma cells, provide a potential approach for glioblastoma multiforme gene therapy.


Assuntos
Técnicas de Transferência de Genes , Glioma/tratamento farmacológico , Receptores da Neurocinina-1/metabolismo , Substância P/uso terapêutico , Animais , Barreira Hematoencefálica , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neurotransmissores/química , Neurotransmissores/uso terapêutico , Receptores da Neurocinina-1/genética , Substância P/química , Peixe-Zebra
14.
J Allergy Clin Immunol ; 147(6): 2225-2235, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33705829

RESUMO

BACKGROUND: Psoriasis is a chronic inflammatory skin disease with disturbed interplay between immune cells and keratinocytes. A strong IFN-γ signature is characteristic for psoriasis skin, but the role of IFN-γ has been elusive. MicroRNAs are short RNAs regulating gene expression. OBJECTIVE: Our aim was to investigate the role of miR-149 in psoriasis and in the inflammatory responses of keratinocytes. METHODS: miR-149 expression was measured by quantitative RT-PCR in keratinocytes isolated from healthy skin and lesional and nonlesional psoriasis skin. Synthetic miR-149 was injected intradermally into the back skin of mice, and imiquimod was applied to induce psoriasis-like skin inflammation, which was then evaluated at the morphologic, histologic, and molecular levels. miR-149 was transiently overexpressed or inhibited in keratinocytes in combination with IFN-γ- and/or TNF-related weak inducer of apoptosis (TWEAK)-treatment. RESULTS: Here we report a microRNA-mediated mechanism by which IFN-γ primes keratinocytes to inflammatory stimuli. Treatment with IFN-γ results in a rapid and long-lasting suppression of miR-149 in keratinocytes. Depletion of miR-149 in keratinocytes leads to widespread transcriptomic changes and induction of inflammatory mediators with enrichment of the TWEAK pathway. We show that IFN-γ-mediated suppression of miR-149 leads to amplified inflammatory responses to TWEAK. TWEAK receptor (TWEAKR/Fn14) is identified as a novel direct target of miR-149. The in vivo relevance of this pathway is supported by decreased miR-149 expression in psoriasis keratinocytes, as well as by the protective effect of synthetic miR-149 in the imiquimod-induced mouse model of psoriasis. CONCLUSION: Our data define a new mechanism, in which IFN-γ primes keratinocytes for TWEAK-induced inflammatory responses through suppression of miR-149, promoting skin inflammation.


Assuntos
Citocina TWEAK/metabolismo , Regulação da Expressão Gênica , Interferon gama/metabolismo , MicroRNAs/genética , Psoríase/etiologia , Psoríase/metabolismo , Transdução de Sinais , Animais , Apoptose/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Queratinócitos/metabolismo , Camundongos , Psoríase/patologia
15.
J Med Chem ; 63(22): 13825-13850, 2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33186040

RESUMO

A series of novel linear aliphatic amine-linked triaryl derivatives as inhibitors of PD-1/PD-L1 were designed, synthesized, and evaluated in vitro and in vivo. In this chemical series, compound 58 showed the most potent inhibitory activity and binding affinity with hPD-L1, with an IC50 value of 12 nM and a KD value of 16.2 pM, showing a binding potency approximately 2000-fold that of hPD-1. Compound 58 could bind with hPD-L1 on the cellular surface and competitively block the interaction of hPD-1 with hPD-L1. In a T cell function assay, 58 restored the T cell function, leading to increased IFN-γ secretion. Moreover, in a humanized mouse model, compound 58 significantly inhibited tumor growth without obvious toxicity and showed moderate PK properties after intravenous injection. These results indicated that 58 is a promising lead for further development of small-molecule PD-1/PD-L1 inhibitors for cancer therapy.


Assuntos
Antineoplásicos/síntese química , Antígeno B7-H1/antagonistas & inibidores , Desenho de Fármacos , Ácidos Graxos/síntese química , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Antígeno B7-H1/química , Antígeno B7-H1/metabolismo , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Ácidos Graxos/metabolismo , Ácidos Graxos/farmacologia , Feminino , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor de Morte Celular Programada 1/química , Receptor de Morte Celular Programada 1/metabolismo , Células RAW 264.7 , Ratos Sprague-Dawley , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
16.
Mucosal Immunol ; 12(5): 1164-1173, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31278373

RESUMO

Alternative splicing (AS) of mRNA is known to be involved in regulation of immune cell differentiation and activation. Elongation factor Tu GTP binding domain containing 2 (Eftud2) is an AS factor to potentially modulate innate immune response in macrophages. In this study, we investigate its involvement in the pathogenesis of colitis-associated cancer (CAC). Using an established mouse model of CAC, we show that Eftud2 is constantly overexpressed in the colonic tissues as well as infiltrating macrophages. Myeloid-specific knockout of Eftud2 remarkably suppresses chronic intestinal inflammation and tumorigenesis, which is associated with decreased production of inflammatory cytokines and tumorigenic factors. Repression of colonic inflammation and colorectal tumor development in Eftud2-deficient mice is due to the impaired activation of NF-κB signaling in LPS-challenged macrophages. Furthermore, the alteration of Eftud2-mediated AS involving the components of TLR4-NF-κB cascades underlies the impairment of NF-κB activation. Overall, these findings provide new insights into the tight link between inflammation and cancer and modulation of AS in innate immune signals may be a potentially therapeutic avenue for CAC treatment.


Assuntos
Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/metabolismo , Colite/complicações , Colite/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Ribonucleoproteína Nuclear Pequena U5/metabolismo , Animais , Biomarcadores , Transformação Celular Neoplásica/genética , Colite/patologia , Neoplasias do Colo/etiologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Deleção de Genes , Imunidade Inata , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Ribonucleoproteína Nuclear Pequena U5/genética , Transdução de Sinais , Spliceossomos/metabolismo , Receptor 4 Toll-Like/metabolismo
17.
Oncol Lett ; 17(6): 5784-5792, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31186805

RESUMO

AXL receptor tyrosine kinase ligand (AXL), a tyrosine kinase receptor that is commonly overexpressed in numerous types of cancer, significantly promotes drug resistance and metastasis in tumor cells. Inhibition of the AXL/growth arrest-specific 6 (Gas6) signaling pathway is emerging as a potential anticancer therapeutic strategy. In the present study, on the basis of the three-dimensional complex structure of AXL/Gas6, the critical residues (E56, E59 and T77) in AXL binding to Gas6 were determined using computer graphics analysis and the distance geometry method. Subsequently, four-variant AXL-ECD-Fc-M1 (G32S, D87G, V92A and G127R) and AXL-ECD-Fc-M2 (G32A, D87A, V92A and G127A) were predicted as high-affinity mutants; AXL-ECD-Fc-M3 (E56R and T77R) and AXL-ECD-Fc-M4 (E59R and T77R) were predicted as low-affinity mutants. The results of the present study revealed that the half-maximal effect concentrations of AXL-ECD-Fc-M1 and AXL-ECD-Fc-M2 were ~0.141 and 0.375 µg/ml, respectively, whereas that of the wild-type protein (AXL-ECD-Fc-WT) was 0.514 µg/ml. Furthermore, adding the high-affinity mutants into culture medium to capture free Gas6 significantly inhibited AXL/Gas6 binding and thus blocked the downstream signaling pathway. In addition, the high-affinity mutants effectively suppressed the migration and metastasis of SKOV3 and A549 cells. Conversely, compared with AXL-ECD-Fc-WT, the low-affinity AXL mutants AXL-ECD-Fc-M3 and AXL-ECD-Fc-M4 lost all inhibitory activities. These findings highlight AXL as a potential therapeutic target and demonstrated that the key residues E56, E59 and T77 may be crucial sites for abolishing the activity of the AXL/Gas6 pathway in cancer therapy.

18.
Scand J Immunol ; 90(2): e12777, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31075180

RESUMO

TAM family members (TYRO3, AXL and MERTK) play essential roles in the resolution of inflammation and in infectious diseases and cancer. AXL, a tyrosine kinase receptor, is commonly overexpressed in several solid tumours and numerous hematopoietic malignancies including acute myeloid leukaemia, acute lymphocytic leukaemia, chronic myeloid leukaemia, chronic lymphocytic leukaemia and multiple myeloma. AXL significantly promotes tumour cell migration, invasion and metastasis, as well as angiogenesis. AXL also plays an important role in inflammation and macrophage ontogeny. Recent studies have revealed that AXL contributes to leukaemic phenotypes through activation of oncogenic signalling pathways that lead to increased cell migration and proliferation. To evaluate the mechanisms underlying the role of AXL signalling in tumour metastasis, we screened a phage display library to generate a novel human monoclonal antibody, named DAXL-88, that recognizes both human and murine AXL. The concentrations of DAXL-88 required for 50% maximal binding to human and murine AXL were 0.118 and 0.164 µg/mL, respectively. Furthermore, DAXL-88 bound to human AXL with high affinity (KD  ~ 370 pM). DAXL-88 blocked the interaction between AXL and its ligand, growth arrest-specific gene 6 (GAS6), with a half maximal inhibitory concentration of 2.16 µg/mL. Moreover, DAXL-88 inhibited AXL/GAS6-dependent cell signalling, which is implicated in cell migration and invasion. In conclusion, the novel anti-AXL DAXL-88 high-affinity antibody blocks the interaction between AXL and GAS6 and inhibits tumour cell migration and invasion induced by GAS6. Thus, DAXL-88 offers promise for the development of targeted therapeutic strategies in solid tumours, leukaemias and other lymphoid neoplasms.


Assuntos
Anticorpos Monoclonais/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neoplasias Ovarianas/patologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Células A549 , Anticorpos Monoclonais/imunologia , Linhagem Celular Tumoral , Movimento Celular , Feminino , Humanos , Simulação de Acoplamento Molecular , Invasividade Neoplásica/patologia , Invasividade Neoplásica/prevenção & controle , Metástase Neoplásica/prevenção & controle , Ligação Proteica , Proteínas Proto-Oncogênicas/imunologia , Receptores Proteína Tirosina Quinases/imunologia , Transdução de Sinais , Receptor Tirosina Quinase Axl
19.
Transl Stroke Res ; 10(6): 695-704, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-30680639

RESUMO

A rodent model of embolic middle cerebral artery occlusion is used to mimic cerebral embolism in clinical patients. Thrombolytic therapy is the effective treatment for this ischemic injury. However, it is difficult to detect thrombolysis dynamically in living animals. Synchrotron radiation angiography may provide a novel approach to directly monitor the thrombolytic process and assess collateral circulation after embolic stroke. Thirty-six adult Sprague-Dawley rats underwent the embolic stroke model procedure and were then treated with tissue plasminogen activator. The angiographic images were obtained in vivo by synchrotron radiation angiography. Synchrotron radiation angiography confirmed the successful establishment of occlusion and detected the thrombolysis process after the thrombolytic treatment. The time of thrombolytic recanalization was unstable during embolic stroke. The infarct volume increased as the recanalization time was delayed from 2 to 6 h (p < 0.05). The collateral circulation of the internal carotid artery to the ophthalmic artery, the olfactory artery to the ophthalmic artery, and the posterior cerebral artery to the middle cerebral artery opened after embolic stroke and manifested different opening rates (59%, 24%, and 75%, respectively) in the rats. The opening of the collateral circulation from the posterior cerebral artery to the middle cerebral artery alleviated infarction in rats with successful thrombolysis (p < 0.05). The cerebral vessels of the circle of Willis narrowed after thrombolysis (p < 0.05). Synchrotron radiation angiography provided a unique tool to dynamically detect and assess the thrombolysis process and the collateral circulation during thrombolytic therapy.


Assuntos
Fibrinolíticos/uso terapêutico , Acidente Vascular Cerebral/diagnóstico por imagem , Acidente Vascular Cerebral/tratamento farmacológico , Terapia Trombolítica , Ativador de Plasminogênio Tecidual/uso terapêutico , Animais , Isquemia Encefálica/diagnóstico por imagem , Isquemia Encefálica/tratamento farmacológico , Angiografia Cerebral/métodos , Circulação Colateral/efeitos dos fármacos , Infarto da Artéria Cerebral Média/diagnóstico por imagem , Infarto da Artéria Cerebral Média/tratamento farmacológico , Masculino , Ratos , Ratos Sprague-Dawley , Síncrotrons
20.
Oncol Rep ; 38(5): 2677-2684, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29048687

RESUMO

Ovarian cancer is one of the most common gynecologic cancers and the leading cause of mortality in women worldwide. HER2/neu is overexpressed in various types of cancers and is most commonly associated with decreased survival. Trastuzumab is a humanized anti-HER2 monoclonal antibody for the treatment of HER2-positive breast cancers. However, primary and/or acquired resistance occurs in up to 62% patients during the first year of treatment. Vascular endothelial growth factor (VEGF) is a well­known angiogenesis factor involved in many physiological and pathological processes. Its significance has been implicated in promoting tumor growth and metastasis via angiogenesis. In the present study, we demonstrated that the upregulation of SP1 enhanced expression of VEGF promoting the angiogenesis and migration of trastuzumab-resistant ovarian cancer cell line SKOV3-T. Our in vitro and in vivo results both gave evidence that the SP1-VEGF axis was responsible for the enhanced malignancy, angiogenesis and migration in the acquired trastuzumab-resistant ovarian cancer cell model.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Neovascularização Patológica/patologia , Neoplasias Ovarianas/patologia , Fator de Transcrição Sp1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Humanos , Camundongos , Invasividade Neoplásica , Transplante de Neoplasias , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Fator de Transcrição Sp1/genética , Trastuzumab , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA