Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
1.
Nat Commun ; 15(1): 1009, 2024 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-38307859

RESUMEN

Tumor-secreted factors contribute to the development of a microenvironment that facilitates the escape of cancer cells from immunotherapy. In this study, we conduct a retrospective comparison of the proteins secreted by hepatocellular carcinoma (HCC) cells in responders and non-responders among a cohort of ten patients who received Nivolumab (anti-PD-1 antibody). Our findings indicate that non-responders have a high abundance of secreted RNase1, which is associated with a poor prognosis in various cancer types. Furthermore, mice implanted with HCC cells that overexpress RNase1 exhibit immunosuppressive tumor microenvironments and diminished response to anti-PD-1 therapy. RNase1 induces the polarization of macrophages towards a tumor growth-promoting phenotype through activation of the anaplastic lymphoma kinase (ALK) signaling pathway. Targeting the RNase1/ALK axis reprograms the macrophage polarization, with increased CD8+ T- and Th1- cell recruitment. Moreover, simultaneous targeting of the checkpoint protein PD-1 unleashes cytotoxic CD8+ T-cell responses. Treatment utilizing both an ALK inhibitor and an anti-PD-1 antibody exhibits enhanced tumor regression and facilitates long-term immunity. Our study elucidates the role of RNase1 in mediating tumor resistance to immunotherapy and reveals an RNase1-mediated immunosuppressive tumor microenvironment, highlighting the potential of targeting RNase1 as a promising strategy for cancer immunotherapy in HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Humanos , Ratones , Quinasa de Linfoma Anaplásico , Carcinoma Hepatocelular/metabolismo , Linfocitos T CD8-positivos , Terapia de Inmunosupresión , Neoplasias Hepáticas/metabolismo , Estudios Retrospectivos , Ribonucleasas , Microambiente Tumoral
2.
Int J Biol Sci ; 19(14): 4644-4656, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37781042

RESUMEN

Anthracyclines are a class of conventionally and routinely used first-line chemotherapy drugs for cancer treatment. In addition to the direct cytotoxic effects, increasing evidence indicates that the efficacy of the drugs also depends on immunomodulatory effects with unknown mechanisms. Galectin-9 (Gal-9), a member of the ß-galactoside-binding protein family, has been demonstrated to induce T-cell death and promote immunosuppression in the tumor microenvironment. Here, we asked whether anthracycline-mediated immunomodulatory activity might be related to Gal-9. We found that combining doxorubicin with anti-Gal-9 therapy significantly inhibited tumor growth and prolonged overall survival in immune-competent syngeneic mouse models. Moreover, Gal-9 expression was increased in response to doxorubicin in various human and murine cancer cell lines. Mechanistically, doxorubicin induced tumoral Gal-9 by activating the STING/interferon ß pathway. Clinically, Gal-9 and p-STING levels were elevated in the tumor tissues of breast cancer patients treated with anthracyclines. Our study demonstrates Gal-9 upregulation in response to anthracyclines as a novel mechanism mediating immune escape and suggests targeting Gal-9 in combination with anthracyclines as a promising therapeutic strategy for cancer treatment.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Ratones , Animales , Antraciclinas/farmacología , Antraciclinas/uso terapéutico , Galectinas , Neoplasias/tratamiento farmacológico , Antibióticos Antineoplásicos/uso terapéutico , Doxorrubicina/uso terapéutico , Microambiente Tumoral
3.
J Oral Pathol Med ; 52(6): 554-563, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36807323

RESUMEN

BACKGROUND: Venous malformation (VM) is a kind of congenital vascular anomaly with a high incidence of recurrence, detailed pathogenesis and standard treatment of VM still lack now. Increasing evidence showed exosomal RNA plays a pivotal role in various diseases. However, the underlying mechanism of VM based on the potential differentially exosomal RNAs remains unclear. METHODS: Comparative high-throughput sequencing with serum exosomes from three VM patients and three healthy donors was used to explore differentially expressed (DE) circRNAs, DE lncRNAs, and DE mRNAs involving the formation of VM. We identified and verified DE circRNAs, DE lncRNAs, and DE mRNAs via qRT-PCR assay. We explored the potential functions of these exosomal DE non-coding RNAs via performing further Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis. Besides, circRNA/lncRNA-miRNA-mRNA linkages were also constructed to find their potential relationships in VM. RESULTS: A total of 121 circRNAs, 53 lncRNAs, and 42 mRNAs (|log2 FC| ≥ 2.0, FDR <0.05, n = 3) were determined to be differentially expressed. QRT-PCR validated that these top-changed DE circRNAs, lncRNAs, and mRNAs had significant expression changes. Functional studies demonstrated that DE circRNAs play a pivotal role in thyroid hormone signaling pathway, DE lncRNAs function as a key regulator in MAPK signaling pathway and DE miRNAs participate in the process of hepatocellular carcinoma mostly. CONCLUSION: Our study comprehensively depicted exosomal DE non-coding RNAs networks related to the pathogenesis of VM which can provide new insight into, a novel target for treating VM.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Circular/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Perfilación de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo
4.
Nat Cancer ; 3(10): 1211-1227, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36253486

RESUMEN

Poly(ADP-ribose) polymerase (PARP) inhibitors have demonstrated promising clinical activity in multiple cancers. However, resistance to PARP inhibitors remains a substantial clinical challenge. In the present study, we report that anaplastic lymphoma kinase (ALK) directly phosphorylates CDK9 at tyrosine-19 to promote homologous recombination (HR) repair and PARP inhibitor resistance. Phospho-CDK9-Tyr19 increases its kinase activity and nuclear localization to stabilize positive transcriptional elongation factor b and activate polymerase II-dependent transcription of HR-repair genes. Conversely, ALK inhibition increases ubiquitination and degradation of CDK9 by Skp2, an E3 ligase. Notably, combination of US Food and Drug Administration-approved ALK and PARP inhibitors markedly reduce tumor growth and improve survival of mice in PARP inhibitor-/platinum-resistant tumor xenograft models. Using human tumor biospecimens, we further demonstrate that phosphorylated ALK (p-ALK) expression is associated with resistance to PARP inhibitors and positively correlated with p-Tyr19-CDK9 expression. Together, our findings support a biomarker-driven, combinatorial treatment strategy involving ALK and PARP inhibitors to induce synthetic lethality in PARP inhibitor-/platinum-resistant tumors with high p-ALK-p-Tyr19-CDK9 expression.


Asunto(s)
Quinasa de Linfoma Anaplásico , Antineoplásicos , Neoplasias de la Mama , Quinasa 9 Dependiente de la Ciclina , Animales , Femenino , Humanos , Ratones , Quinasa de Linfoma Anaplásico/metabolismo , Antineoplásicos/farmacología , Biomarcadores , Neoplasias de la Mama/tratamiento farmacológico , Quinasa 9 Dependiente de la Ciclina/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Factor B de Elongación Transcripcional Positiva , Tirosina/química , Tirosina/metabolismo , Ubiquitina-Proteína Ligasas/efectos de los fármacos , Ubiquitina-Proteína Ligasas/metabolismo , Estados Unidos
5.
J Oral Pathol Med ; 51(10): 911-919, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35854627

RESUMEN

BACKGROUNDS: Head and neck vascular malformation (HNVM) is a highly complex congenital condition that is difficult to diagnose, monitor and treat. Therefore, it is critical to explore serum cytokines that may be related to its pathology and prognosis. METHODS: An antibody-based microarray was used to examine the expression of 31 angiogenic cytokines in 11 HNVM patients relative to 11 healthy subjects. ELISA was used to verify the results. We performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses of the differentially expressed cytokines (DECs). Additionally, we explored the function of DECs in human umbilical vein endothelial cells (HUVECs) in vitro via CCK-8, wound healing, transwell and tube formation assays. RESULTS: Expression of interleukin (IL)-10, matrix metallopeptidase-9 (MMP-9) and vascular endothelial growth factor receptor 2 (VEGF-R2) in HNVM patients was significantly higher, whereas levels of IL-12p40 and angiostatin were significantly lower in HNVM patients relative to healthy controls (p < 0.05). However, ELISA only verified that IL-10, MMP-9, VEGF-R2 and IL-12p40 had significant expression changes. Functional enrichment analysis revealed DECs mainly participated in the RAS signalling pathway. Functional studies demonstrated that IL-10, MMP-9 and VEGF-R2 promote cell proliferation, migration, invasion and tube formation, while IL-12p40 inhibited these processes in HUVECs. CONCLUSIONS: The present study not only indicates that IL-10, MMP-9, VEGF-R2 and IL-12p40 may participate in the development of HNVMs but also provides a theoretical basis for the discovery of new targeted molecules in the treatment of HNVMs.


Asunto(s)
Factor A de Crecimiento Endotelial Vascular , Malformaciones Vasculares , Humanos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Interleucina-10/metabolismo , Movimiento Celular , Metaloproteinasa 9 de la Matriz/metabolismo , Subunidad p40 de la Interleucina-12/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Malformaciones Vasculares/metabolismo , Citocinas/metabolismo
7.
J Biol Chem ; 298(4): 101817, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35278434

RESUMEN

Expression of the receptor tyrosine kinase ephrin receptor A10 (EphA10), which is undetectable in most normal tissues except for the male testis, has been shown to correlate with tumor progression and poor prognosis in several malignancies, including triple-negative breast cancer (TNBC). Therefore, EphA10 could be a potential therapeutic target, likely with minimal adverse effects. However, no effective clinical drugs against EphA10 are currently available. Here, we report high expression levels of EphA10 in tumor regions of breast, lung, and ovarian cancers as well as in immunosuppressive myeloid cells in the tumor microenvironment. Furthermore, we developed anti-EphA10 monoclonal antibodies (mAbs) that specifically recognize cell surface EphA10, but not other EphA family isoforms, and target tumor regions precisely in vivo with no apparent accumulation in other organs. In syngeneic TNBC mouse models, we found that anti-EphA10 mAb clone #4 enhanced tumor regression, therapeutic response rate, and T cell-mediated antitumor immunity. Notably, the chimeric antigen receptor T cells derived from clone #4 significantly inhibited TNBC cell viability in vitro and tumor growth in vivo. Together, our findings suggest that targeting EphA10 via EphA10 mAbs and EphA10-specific chimeric antigen receptor-T cell therapy may represent a promising strategy for patients with EphA10-positive tumors.


Asunto(s)
Anticuerpos Monoclonales , Receptores Quiméricos de Antígenos , Receptores de la Familia Eph , Linfocitos T , Neoplasias de la Mama Triple Negativas , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Ratones , Receptores de la Familia Eph/inmunología , Linfocitos T/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Biol Chem ; 298(4): 101821, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35283189

RESUMEN

Antibodies that target immune checkpoint proteins such as programmed cell death protein 1, programmed death ligand 1, and cytotoxic T-lymphocyte-associated antigen 4 in human cancers have achieved impressive clinical success; however, a significant proportion of patients fail to respond to these treatments. Galectin-9 (Gal-9), a ß-galactoside-binding protein, has been shown to induce T-cell death and facilitate immunosuppression in the tumor microenvironment by binding to immunomodulatory receptors such as T-cell immunoglobulin and mucin domain-containing molecule 3 and the innate immune receptor dectin-1, suggesting that it may have potential as a target for cancer immunotherapy. Here, we report the development of two novel Gal-9-neutralizing antibodies that specifically react with the N-carbohydrate-recognition domain of human Gal-9 with high affinity. We also show using cell-based functional assays that these antibodies efficiently protected human T cells from Gal-9-induced cell death. Notably, in a T-cell/tumor cell coculture assay of cytotoxicity, these antibodies significantly promoted T cell-mediated killing of tumor cells. Taken together, our findings demonstrate potent inhibition of human Gal-9 by neutralizing antibodies, which may open new avenues for cancer immunotherapy.


Asunto(s)
Anticuerpos Neutralizantes , Muerte Celular , Galectinas , Linfocitos T , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Neutralizantes/farmacología , Muerte Celular/efectos de los fármacos , Galectinas/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/terapia , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Microambiente Tumoral
9.
STAR Protoc ; 3(1): 101198, 2022 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-35243381

RESUMEN

Immunotherapy via PD-1/PD-L1 blockade is a promising strategy to eradicate cancer cells. However, the PD-L1 pathological level is inconsistent with the therapeutic response and is not a reliable biomarker to stratify patients for anti-PD-1/PD-L1 therapy. Here, we describe patient sample deglycosylation in an immunohistochemistry (IHC) assay to resolve this challenge. This protocol facilitates antigen retrieval by removing N-glycans from surface antigens on formalin-fixed paraffin-embedded (FFPE) tissue slides and can be applied in medical pathology for multiple cancer types. For complete details on the use and execution of this profile, please refer to Lee et al. (2019).


Asunto(s)
Antígeno B7-H1 , Neoplasias , Humanos , Inmunohistoquímica , Inmunoterapia , Neoplasias/terapia
10.
Am J Cancer Res ; 12(1): 123-137, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35141008

RESUMEN

The atezolizumab (Tecentriq), a humanized antibody against human programmed death ligand 1 (PD-L1), combined with nab-paclitaxel was granted with accelerated approval to treat unresectable locally advanced or metastatic triple-negative breast cancer (TNBC) due to the encouraging positive results of the phase 3 IMpassion130 trial using PD-L1 biomarker from immune cells to stratify patients. However, the post-market study IMpassion131 did not support the original observation, resulting in the voluntary withdrawal of atezolizumab from the indication in breast cancer by Genentech in 2021. Emerging evidence has revealed a high frequency of false negative result using the standard immunohistochemical (IHC) staining due to heavy glycosylation of PD-L1. The removal of glycosylation prevents from the false negative staining, enabling more accurate assessment of PD-L1 levels and improving prediction for response to immune checkpoint therapy. In the present study, the natural and de-glycosylated PD-L1 expression in tumor and immune cells from nine TNBC patients were analyzed by using clone 28-8 monoclonal antibody to correlate with treatment outcome. Our results demonstrate that: (1) Removal of the glycosylation indeed enhances the detection of PD-L1 by IHC staining, (2) The PD-L1 levels on tumor cell surface after removal of the glycosylation correlates well with clinical responses for atezolizumab treatment; (3) The criteria used in the IMpassion130 and IMpassion131 trials which scored the natural PD-L1 in the immune cells failed to correlate with the clinical response. Taken together, tumor cell surface staining of PD-L1 with de-glycosylation has a significant correlation with the clinical response for atezolizumab treatment, suggesting that treatment of atezolizumab may be worthy of further consideration with de-glycosylation procedure as a patient stratification strategy. A larger cohort to validate this important issue is warranted to ensure right patient population who could benefit from the existing FDA-approved drugs.

11.
Nat Commun ; 12(1): 7041, 2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34873175

RESUMEN

Despite unprecedented responses of some cancers to immune checkpoint blockade (ICB) therapies, the application of checkpoint inhibitors in pancreatic cancer has been unsuccessful. Glucocorticoids and glucocorticoid receptor (GR) signaling are long thought to suppress immunity by acting on immune cells. Here we demonstrate a previously undescribed tumor cell-intrinsic role for GR in activating PD-L1 expression and repressing the major histocompatibility complex class I (MHC-I) expression in pancreatic ductal adenocarcinoma (PDAC) cells through transcriptional regulation. In mouse models of PDAC, either tumor cell-specific depletion or pharmacologic inhibition of GR leads to PD-L1 downregulation and MHC-I upregulation in tumor cells, which in turn promotes the infiltration and activity of cytotoxic T cells, enhances anti-tumor immunity, and overcomes resistance to ICB therapy. In patients with PDAC, GR expression correlates with high PD-L1 expression, low MHC-I expression, and poor survival. Our results reveal GR signaling in cancer cells as a tumor-intrinsic mechanism of immunosuppression and suggest that therapeutic targeting of GR is a promising way to sensitize pancreatic cancer to immunotherapy.


Asunto(s)
Antígeno B7-H1/inmunología , Carcinoma Ductal Pancreático/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Neoplasias Pancreáticas/inmunología , Receptores de Glucocorticoides/inmunología , Escape del Tumor/inmunología , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/terapia , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Immunoblotting , Inmunoterapia/métodos , Estimación de Kaplan-Meier , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Escape del Tumor/genética
12.
Am J Transl Res ; 13(10): 11271-11286, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34786057

RESUMEN

Venous malformation (VM) is a kind of congenital vascular anomaly with high recurrence, and screening for VM lacks an efficient, inexpensive and noninvasive approach now. Serum miRNAs with stable structures are expected to become new postoperative and postablative monitoring biomarkers. Thus, we identified a prognostic serum miR-18a-5p and validated its function in VM. Notably, higher expression level of miR-18a-5p was detected in VM patients than in healthy individuals. We found that miR-18a-5p plays a promotive role in human umbilical vein endothelial cells in vitro. In addition, immunohistochemistry (IHC) results showed a distinct increase of vessels in miR-18a-5p mimics group and a decrease of vessels in inhibitors group compared to the control group in a murine VM model. Furthermore, thrombospondin-1 (TSP1), a potential miR-18a-5p-binding protein, was identified via RNA-seq, luciferase reporter and RNA immunoprecipitation (RIP) assays. Moreover, miR-18a-5p regulated the activation of P53 signaling pathway constituents and consequently led to the regulation of proliferation, migration, invasion and angiogenesis. These results provide a strong theoretical basis for further investigations into pathological mechanism of VM and may provide novel and noninvasive biomarker for VM diagnosis and monitoring.

13.
Cell Rep ; 36(8): 109537, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34433039

RESUMEN

Increased DNA replication and metastasis are hallmarks of cancer progression, while deregulated proliferation often triggers sustained replication stresses in cancer cells. How cancer cells overcome the growth stress and proceed to metastasis remains largely elusive. Proliferating cell nuclear antigen (PCNA) is an indispensable component of the DNA replication machinery. Here, we show that phosphorylation of PCNA on tyrosine 211 (pY211-PCNA) regulates DNA metabolism and tumor microenvironment. Abrogation of pY211-PCNA blocks fork processivity, resulting in biogenesis of single-stranded DNA (ssDNA) through a MRE11-dependent mechanism. The cytosolic ssDNA subsequently induces inflammatory cytokines through a cyclic GMP-AMP synthetase (cGAS)-dependent cascade, triggering an anti-tumor immunity by natural killer (NK) cells to suppress distant metastasis. Expression of pY211-PCNA is inversely correlated with cytosolic ssDNA and associated with poor survival in patients with cancer. Our results pave the way to biomarkers and therapies exploiting immune responsiveness to target metastatic cancer.


Asunto(s)
Neoplasias Experimentales/inmunología , Antígeno Nuclear de Célula en Proliferación/inmunología , Escape del Tumor , Microambiente Tumoral/inmunología , Animales , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Transgénicos , Neoplasias Experimentales/genética , Neoplasias Experimentales/mortalidad , Fosforilación , Antígeno Nuclear de Célula en Proliferación/genética , Microambiente Tumoral/genética , Tirosina/genética , Tirosina/inmunología
14.
Nat Commun ; 12(1): 2788, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33986289

RESUMEN

Human ribonuclease 1 (hRNase 1) is critical to extracellular RNA clearance and innate immunity to achieve homeostasis and host defense; however, whether it plays a role in cancer remains elusive. Here, we demonstrate that hRNase 1, independently of its ribonucleolytic activity, enriches the stem-like cell population and enhances the tumor-initiating ability of breast cancer cells. Specifically, secretory hRNase 1 binds to and activates the tyrosine kinase receptor ephrin A4 (EphA4) signaling to promote breast tumor initiation in an autocrine/paracrine manner, which is distinct from the classical EphA4-ephrin juxtacrine signaling through contact-dependent cell-cell communication. In addition, analysis of human breast tumor tissue microarrays reveals a positive correlation between hRNase 1, EphA4 activation, and stem cell marker CD133. Notably, high hRNase 1 level in plasma samples is positively associated with EphA4 activation in tumor tissues from breast cancer patients, highlighting the pathological relevance of the hRNase 1-EphA4 axis in breast cancer. The discovery of hRNase 1 as a secretory ligand of EphA4 that enhances breast cancer stemness suggests a potential treatment strategy by inactivating the hRNase 1-EphA4 axis.


Asunto(s)
Neoplasias de la Mama/patología , Carcinogénesis/patología , Efrina-A4/metabolismo , Células Madre Neoplásicas/patología , Ribonucleasa Pancreática/metabolismo , Antígeno AC133/metabolismo , Animales , Neoplasias de la Mama/genética , Carcinogénesis/genética , Línea Celular , Femenino , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Unión Proteica/genética , Ribonucleasa Pancreática/sangre , Ribonucleasa Pancreática/genética , Resultado del Tratamiento
15.
J Hepatol ; 74(4): 907-918, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33031845

RESUMEN

BACKGROUND & AIMS: There are currently limited therapeutic options for hepatocellular carcinoma (HCC), particularly when it is diagnosed at advanced stages. Herein, we examined the pathophysiological role of ROS1 and assessed the utility of ROS1-targeted therapy for the treatment of HCC. METHODS: Recombinant ribonucleases (RNases) were purified, and the ligand-receptor relationship between RNase7 and ROS1 was validated in HCC cell lines by Duolink, immunofluorescence, and immunoprecipitation assays. Potential interacting residues between ROS1 and RNase7 were predicted using a protein-protein docking approach. The oncogenic function of RNase7 was analyzed by cell proliferation, migration and invasion assays, and a xenograft mouse model. The efficacy of anti-ROS1 inhibitor treatment was evaluated in patient-derived xenograft (PDX) and orthotopic models. Two independent patient cohorts were analyzed to evaluate the pathological relevance of RNase7/ROS1. RESULTS: RNase7 associated with ROS1's N3-P2 domain and promoted ROS1-mediated oncogenic transformation. Patients with HCC exhibited elevated plasma RNase7 levels compared with healthy individuals. High ROS1 and RNase7 expression were strongly associated with poor prognosis in patients with HCC. In both HCC PDX and orthotopic mouse models, ROS1 inhibitor treatment markedly suppressed RNase7-induced tumorigenesis, leading to decreased plasma RNase7 levels and tumor shrinkage in mice. CONCLUSIONS: RNase7 serves as a high-affinity ligand for ROS1. Plasma RNase7 could be used as a biomarker to identify patients with HCC who may benefit from anti-ROS1 treatment. LAY SUMMARY: Receptor tyrosine kinases are known to be involved in tumorigenesis and have been targeted therapeutically for a number of cancers, including hepatocellular carcinoma. ROS1 is the only such receptor with kinase activity whose ligand has not been identified. Herein, we show that RNase7 acts as a ligand to activate ROS1 signaling. This has important pathophysiological and therapeutic implications. Anti-ROS1 inhibitors could be used to treatment patients with hepatocellular carcinoma and high RNase7 levels.


Asunto(s)
Carcinogénesis , Carcinoma Hepatocelular , Crizotinib/farmacología , Neoplasias Hepáticas , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Ribonucleasas/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Carcinogénesis/efectos de los fármacos , Carcinogénesis/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Ensayos de Migración Celular/métodos , Proliferación Celular/efectos de los fármacos , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ligandos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Ratones , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Exp Clin Cancer Res ; 39(1): 273, 2020 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-33278894

RESUMEN

BACKGROUND: MUC18 is a glycoprotein highly expressed on the surface of melanoma and other cancers which promotes tumor progression and metastasis. However, its mechanism of action and suitability as a therapeutic target are unknown. METHODS: A monoclonal antibody (mAb) (JM1-24-3) was generated from metastatic melanoma tumor live cell immunization, and high-throughput screening identified MUC18 as the target. RESULTS: Analysis of molecular interactions between MUC18 and JM1-24-3 revealed that the downstream signaling events depended on binding of the mAb to a conformational epitope on the extracellular domain of MUC18. JM1-24-3 inhibited melanoma cell proliferation, migration and invasion in vitro and reduced tumor growth and metastasis in vivo. CONCLUSION: These results confirm that MUC18 is mechanistically important in melanoma growth and metastasis, suggest that the MUC18 epitope identified is a promising therapeutic target, and that the JM1-24-3 mAb may serve as the basis for a potential therapeutic agent.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Melanoma/terapia , Animales , Anticuerpos Monoclonales/inmunología , Antígeno CD146/inmunología , Línea Celular Tumoral , Humanos , Masculino , Melanoma/inmunología , Ratones , Ratones Endogámicos A , Ratones Desnudos , Distribución Aleatoria , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Nat Cell Biol ; 22(10): 1264-1275, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32929201

RESUMEN

Although pyroptosis is critical for macrophages against pathogen infection, its role and mechanism in cancer cells remains unclear. PD-L1 has been detected in the nucleus, with unknown function. Here we show that PD-L1 switches TNFα-induced apoptosis to pyroptosis in cancer cells, resulting in tumour necrosis. Under hypoxia, p-Stat3 physically interacts with PD-L1 and facilitates its nuclear translocation, enhancing the transcription of the gasdermin C (GSDMC) gene. GSDMC is specifically cleaved by caspase-8 with TNFα treatment, generating a GSDMC N-terminal domain that forms pores on the cell membrane and induces pyroptosis. Nuclear PD-L1, caspase-8 and GSDMC are required for macrophage-derived TNFα-induced tumour necrosis in vivo. Moreover, high expression of GSDMC correlates with poor survival. Antibiotic chemotherapy drugs induce pyroptosis in breast cancer. These findings identify a non-immune checkpoint function of PD-L1 and provide an unexpected concept that GSDMC/caspase-8 mediates a non-canonical pyroptosis pathway in cancer cells, causing tumour necrosis.


Asunto(s)
Apoptosis , Antígeno B7-H1/metabolismo , Biomarcadores de Tumor/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias/patología , Piroptosis , Animales , Antígeno B7-H1/genética , Biomarcadores de Tumor/genética , Proliferación Celular , Proteínas de Unión al ADN/genética , Femenino , Humanos , Hipoxia/fisiopatología , Inflamasomas , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Necrosis , Neoplasias/genética , Neoplasias/metabolismo , Células Tumorales Cultivadas , Macrófagos Asociados a Tumores , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Theranostics ; 10(23): 10589-10605, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32929368

RESUMEN

Rationale: The oncogenesis of head and neck squamous cell carcinoma (HNSCC) is believed to result from oncogene activation and tumor suppressor inactivation. Here, we identified a new oncogenic role for the EREG gene in HNSCC. Methods: The TCGA database and immunohistochemistry assay were used to analyze expression of EREG in HNSCC tissues. Immunoblotting was performed to identify the EGFR-mediated pathways altered by EREG. The role of EREG in oncogenesis was investigated in vivo and in vitro. Results: Upregulated EREG expression predicted a poor prognosis and triggered HNSCC oncogenic transformation by activating the epidermal growth factor receptor (EGFR) signaling pathway. We also demonstrated the direct association of EREG with EGFR and that this binding required EGFR domains I and III and the N57 residue of EREG. Moreover, EREG overexpression was shown to promote HNSCC oncogenesis by inducing C-Myc expression, and the pharmacological inhibition of C-Myc rescued EREG-promoted HNSCC oncogenesis. Unlike other EGFR ligands, EREG could mimic EGFR mutations by sustaining the activation of the EGFR-Erk pathway, and high EREG expression was positively associated with the response to treatment with the EGFR inhibitor erlotinib. Furthermore, knockdown of EREG decreased sensitivity to erlotinib treatment in vitro and in vivo. Conclusions: These results identify the EREG-EGFR-C-Myc pathway as a crucial axis that drives HNSCC oncogenesis and show that EREG expression could be a predictive functional marker of sensitivity to erlotinib therapy in HNSCC.


Asunto(s)
Resistencia a Antineoplásicos/genética , Epirregulina/metabolismo , Clorhidrato de Erlotinib/farmacología , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Animales , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Línea Celular Tumoral , Epirregulina/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Clorhidrato de Erlotinib/uso terapéutico , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Masculino , Ratones , Persona de Mediana Edad , Mutación , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Nature ; 580(7804): 530-535, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32322062

RESUMEN

Cancer cells increase lipogenesis for their proliferation and the activation of sterol regulatory element-binding proteins (SREBPs) has a central role in this process. SREBPs are inhibited by a complex composed of INSIG proteins, SREBP cleavage-activating protein (SCAP) and sterols in the endoplasmic reticulum. Regulation of the interaction between INSIG proteins and SCAP by sterol levels is critical for the dissociation of the SCAP-SREBP complex from the endoplasmic reticulum and the activation of SREBPs1,2. However, whether this protein interaction is regulated by a mechanism other than the abundance of sterol-and in particular, whether oncogenic signalling has a role-is unclear. Here we show that activated AKT in human hepatocellular carcinoma (HCC) cells phosphorylates cytosolic phosphoenolpyruvate carboxykinase 1 (PCK1), the rate-limiting enzyme in gluconeogenesis, at Ser90. Phosphorylated PCK1 translocates to the endoplasmic reticulum, where it uses GTP as a phosphate donor to phosphorylate INSIG1 at Ser207 and INSIG2 at Ser151. This phosphorylation reduces the binding of sterols to INSIG1 and INSIG2 and disrupts the interaction between INSIG proteins and SCAP, leading to the translocation of the SCAP-SREBP complex to the Golgi apparatus, the activation of SREBP proteins (SREBP1 or SREBP2) and the transcription of downstream lipogenesis-related genes, proliferation of tumour cells, and tumorigenesis in mice. In addition, phosphorylation of PCK1 at Ser90, INSIG1 at Ser207 and INSIG2 at Ser151 is not only positively correlated with the nuclear accumulation of SREBP1 in samples from patients with HCC, but also associated with poor HCC prognosis. Our findings highlight the importance of the protein kinase activity of PCK1 in the activation of SREBPs, lipogenesis and the development of HCC.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Gluconeogénesis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipogénesis , Neoplasias Hepáticas/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Animales , Carcinogénesis , Carcinoma Hepatocelular/patología , Proliferación Celular , Modelos Animales de Enfermedad , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Neoplasias Hepáticas/patología , Masculino , Proteínas de la Membrana/química , Ratones , Ratones Desnudos , Oxiesteroles/metabolismo , Fosforilación , Pronóstico , Unión Proteica , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...