Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Mol Cell ; 81(6): 1216-1230.e9, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33606996

ABSTRACT

Interferon-γ (IFN-γ)-mediated adaptive resistance is one major barrier to improving immunotherapy in solid tumors. However, the mechanisms are not completely understood. Here, we report that IFN-γ promotes nuclear translocation and phase separation of YAP after anti-PD-1 therapy in tumor cells. Hydrophobic interactions of the YAP coiled-coil domain mediate droplet initiation, and weak interactions of the intrinsically disordered region in the C terminus promote droplet formation. YAP partitions with the transcription factor TEAD4, the histone acetyltransferase EP300, and Mediator1 and forms transcriptional hubs for maximizing target gene transcriptions, independent of the canonical STAT1-IRF1 transcription program. Disruption of YAP phase separation reduced tumor growth, enhanced immune response, and sensitized tumor cells to anti-PD-1 therapy. YAP activity is negatively correlated with patient outcome. Our study indicates that YAP mediates the IFN-γ pro-tumor effect through its nuclear phase separation and suggests that YAP can be used as a predictive biomarker and target of anti-PD-1 combination therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Drug Resistance, Neoplasm , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy , Interferon-gamma/metabolism , Neoplasms, Experimental , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Transcription Factors/metabolism , A549 Cells , Adaptor Proteins, Signal Transducing/genetics , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , HEK293 Cells , Humans , Interferon-gamma/genetics , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Transcription Factors/genetics , YAP-Signaling Proteins
2.
Prep Biochem Biotechnol ; 45(8): 743-53, 2015.
Article in English | MEDLINE | ID: mdl-25207990

ABSTRACT

Human transferrin receptor, referred as hTfR1, is ubiquitously expressed at low levels in most normal human tissues; however, the expression level of hTfR1 at the blood-brain barrier (BBB) and in tumor tissues is relatively higher. hTfR1 is a type II homodimeric transmembrane protein. The extracellular domain of hTfR1 consists of three domains: helical domain, apical, and protease-like domain. In order to prepare hTfR1 antibody, which can be utilized to deliver drugs across BBB through receptor-mediated endocytosis, we began to express the nonligand binding domain of hTfR1 in Escherichia coli BL21 Transetta (DE3). The TfR1 gene was first obtained from HepG2 cells by reverse-transcription polymerase chain reaction (RT-PCR) and then inserted into pET 32a(c+) vector. The protein was expressed in the form of inclusion body with extremely high purity by the E. coli BL21 Transetta (DE3), and the purity was further improved by size-exclusion chromatography. The Western blot test indicated that the recombinant protein was TfR1 as expected. Above all, this report provided a convenient protocol that could be fulfilled in order to prepare hTfR1 inclusion body, which failed to be purified by an Ni(2+) affinity column.


Subject(s)
Antigens, CD/isolation & purification , Escherichia coli/chemistry , Inclusion Bodies/chemistry , Receptors, Transferrin/isolation & purification , Antigens, CD/biosynthesis , Antigens, CD/chemistry , Antigens, CD/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Inclusion Bodies/genetics , Inclusion Bodies/metabolism , Receptors, Transferrin/biosynthesis , Receptors, Transferrin/chemistry , Receptors, Transferrin/genetics , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
3.
STAR Protoc ; 2(2): 100595, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34169289

ABSTRACT

Anti-PD-1/PD-L1 therapy shows long-term effects in many cancer types, but resistance and relapse remain the main limitations of this therapy. Here, we describe a protocol to evaluate the tumor response to immunotherapy in a mouse lung cancer model. The protocol includes the establishment of the lung cancer mouse model, anti-PD-1 treatment, tumor-infiltrating lymphocyte isolation, immunofluorescence, and flow cytometry analysis. This protocol can also be applied to other cancer types and immunotherapies. For complete details on the use and execution of this protocol, please refer to Yu et al. (2021).


Subject(s)
Adenocarcinoma of Lung/therapy , Lung Neoplasms/therapy , Adenocarcinoma of Lung/immunology , Animals , Cell Separation , Disease Models, Animal , Flow Cytometry , Heterografts , Lung Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Treatment Outcome
4.
Cancer Lett ; 386: 24-34, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27832973

ABSTRACT

Metastatic disease is the major cause of death from cancer, and immunotherapy and chemotherapy have had limited success in reversing its progression. Researchers have suggested that inflammatory factors in the tumor environment can promote cancer invasion and metastasis, stimulating cancer progression. Thus, novel strategies that target cytokines and modulate the tumor microenvironment may emerge as important approaches for treating metastatic breast cancer. Specific neutralization of pathogenic TNF signaling using a TNFα antibody has gained increasing attention. Considering this, a selective human TNFα neutralized antibody was generated based on nanobody technology. A TNFα-specific nanobody was produced in Pichia pastoris with a molecular mass of 15 kDa and affinity constant of 2.05 nM. In the proliferation experiment, the TNFα nanobody could inhibit the proliferation of the breast cancer cell line MCF-7 induced by hTNFα in a dose-dependent manner. In the microinvasion model, the TNFα nanobody could inhibit the migration of the breast cancer cell lines MCF-7, MDA-MB-231 and the invasiveness of MDA-MB-231 induced by hTNFα in a dose-dependent manner. Drug administration of the combination of paclitaxel with the TNFα nanobody in vivo significantly enhanced the efficacy against 4T-1 breast tumor proliferation and lung metastasis; meanwhile, E-cadherin tumor epithelial marker expression was upregulated, supporting the anti-tumor therapeutic relevance of paclitaxel and the TNFα nanobody on EMT. This study highlights the importance of neutralizing low TNFα levels in the tumor microenvironment to sensitize the chemotherapeutic response, which has attractive potential for clinical applications.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Cell Movement/drug effects , Lung Neoplasms/prevention & control , Paclitaxel/pharmacology , Single-Domain Antibodies/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Antibodies, Neutralizing/administration & dosage , Antibody Affinity , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , MCF-7 Cells , Male , Mice, Inbred BALB C , Rats, Sprague-Dawley , Signal Transduction/drug effects , Single-Domain Antibodies/administration & dosage , Tumor Microenvironment , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL