Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Autophagy ; 10(10): 1787-800, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25136802

ABSTRACT

Autophagy, a "self-eating" cellular process, has dual roles in promoting and suppressing tumor growth, depending on cellular context. PTP4A3/PRL-3, a plasma membrane and endosomal phosphatase, promotes multiple oncogenic processes including cell proliferation, invasion, and cancer metastasis. In this study, we demonstrate that PTP4A3 accumulates in autophagosomes upon inhibition of autophagic degradation. Expression of PTP4A3 enhances PIK3C3-BECN1-dependent autophagosome formation and accelerates LC3-I to LC3-II conversion in an ATG5-dependent manner. PTP4A3 overexpression also enhances the degradation of SQSTM1, a key autophagy substrate. These functions of PTP4A3 are dependent on its catalytic activity and prenylation-dependent membrane association. These results suggest that PTP4A3 functions to promote canonical autophagy flux. Unexpectedly, following autophagy activation, PTP4A3 serves as a novel autophagic substrate, thereby establishing a negative feedback-loop that may be required to fine-tune autophagy activity. Functionally, PTP4A3 utilizes the autophagy pathway to promote cell growth, concomitant with the activation of AKT. Clinically, from the largest ovarian cancer data set (GSE 9899, n = 285) available in GEO, high levels of expression of both PTP4A3 and autophagy genes significantly predict poor prognosis of ovarian cancer patients. These studies reveal a critical role of autophagy in PTP4A3-driven cancer progression, suggesting that autophagy could be a potential Achilles heel to block PTP4A3-mediated tumor progression in stratified patients with high expression of both PTP4A3 and autophagy genes.


Subject(s)
Autophagy , Neoplasm Proteins/metabolism , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Protein Tyrosine Phosphatases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagy/drug effects , Autophagy/genetics , Biocatalysis/drug effects , CHO Cells , Cell Cycle Proteins/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Chloroquine/pharmacology , Cricetinae , Cricetulus , Disease Progression , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Models, Biological , Ovarian Neoplasms/genetics , Phagosomes/drug effects , Phagosomes/metabolism , Prenylation/drug effects , Sequestosome-1 Protein , Substrate Specificity/drug effects , Survival Analysis
2.
EMBO Mol Med ; 5(9): 1351-66, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23929599

ABSTRACT

FLT3-ITD mutations are prevalent mutations in acute myeloid leukaemia (AML). PRL-3, a metastasis-associated phosphatase, is a downstream target of FLT3-ITD. This study investigates the regulation and function of PRL-3 in leukaemia cell lines and AML patients associated with FLT3-ITD mutations. PRL-3 expression is upregulated by the FLT3-STAT5 signalling pathway in leukaemia cells, leading an activation of AP-1 transcription factors via ERK and JNK pathways. PRL-3-depleted AML cells showed a significant decrease in cell growth. Clinically, high PRL-3 mRNA expression was associated with FLT3-ITD mutations in four independent AML datasets with 1158 patients. Multivariable Cox-regression analysis on our Cohort 1 with 221 patients identified PRL-3 as a novel prognostic marker independent of other clinical parameters. Kaplan-Meier analysis showed high PRL-3 mRNA expression was significantly associated with poorer survival among 491 patients with normal karyotype. Targeting PRL-3 reversed the oncogenic effects in FLT3-ITD AML models in vitro and in vivo. Herein, we suggest that PRL-3 could serve as a prognostic marker to predict poorer survival and as a promising novel therapeutic target for AML patients.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Neoplasm Proteins/biosynthesis , Protein Tyrosine Phosphatases/biosynthesis , fms-Like Tyrosine Kinase 3/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression , Humans
3.
Oncotarget ; 3(2): 158-71, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22374986

ABSTRACT

Antibodies are considered as 'magic bullets' because of their high specificity. It is believed that antibodies are too large to routinely enter the cytosol, thus antibody therapeutic approach has been limited to extracellular or secreted proteins expressed by cancer cells. However, many oncogenic proteins are localized within the cell. To explore the possibility of antibody therapies against intracellular targets, we generated a chimeric antibody targeting the intracellular PRL-3 oncoprotein to assess its antitumor activities in mice. Remarkably, we observed that the PRL-3 chimeric antibody could efficiently and specifically reduce the formation of PRL-3 expressing metastatic tumors. We further found that natural killer (NK) cells were important in mediating the therapeutic effect, which was only observed in a nude mouse model (T-cell deficient), but not in a Severe Combined Immunodeficiency' (scid ) mouse model (B- and T-cell deficient), indicating the anticancer effect also depends on host B-cell activity. Our study involving 377 nude and scid mice suggest that antibodies targeting intracellular proteins can be developed to treat cancer.


Subject(s)
Antibodies, Monoclonal/immunology , Immediate-Early Proteins/immunology , Killer Cells, Natural/immunology , Molecular Targeted Therapy/methods , Protein Tyrosine Phosphatases/immunology , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Humans , Lymphocyte Activation/immunology , Melanoma/drug therapy , Mice , Mice, Nude , Mice, SCID , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use
4.
Sci Transl Med ; 3(99): 99ra85, 2011 Sep 07.
Article in English | MEDLINE | ID: mdl-21900592

ABSTRACT

Antibody-based therapies have better specificity and thus improved efficacy over standard chemotherapy regimens, which result in extended survival and improved quality of life for cancer patients. Because antibodies are viewed as too large to access intracellular locations, antibody therapy has traditionally targeted extracellular or secreted proteins expressed by cancer cells. However, many oncogenic proteins are found within the cell (such as intracellular phosphatases/kinases and transcription factors) and have therefore not been pursued for antibody therapies. Here, we explored the possibility of antibody therapy or vaccination against intracellular proteins. As proofs of concept, we selected three representative intracellular proteins as immunogens for tumor vaccine studies: PRL-3 (phosphatase of regenerating liver 3), a cancer-associated phosphatase; EGFP (enhanced green fluorescent protein), a general reporter; and mT (polyomavirus middle T), the polyomavirus middle T oncoprotein. A variety of tumors that expressed these intracellular proteins were clearly inhibited by their respective exogenous antibodies or by antigen-induced host antibodies (vaccination). These anticancer activities were reproducibly observed in hundreds of C57BL/6 tumor-bearing mice and MMTV-PymT transgenic breast tumor mice. Our in vivo data suggest that immunotherapies can target not only extracellular but also intracellular oncoproteins.


Subject(s)
Antineoplastic Agents/immunology , Cancer Vaccines/immunology , Immunotherapy/methods , Neoplasms/immunology , Oncogene Proteins/immunology , Vaccination , Animals , Antigens, Polyomavirus Transforming/immunology , Antineoplastic Agents/therapeutic use , B-Lymphocytes/immunology , Cell Line, Tumor , Female , Green Fluorescent Proteins/immunology , Humans , Immediate-Early Proteins/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Neoplasm Metastasis/pathology , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Protein Tyrosine Phosphatases/immunology , Survival Rate
5.
J Cell Biochem ; 111(5): 1087-98, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21053359

ABSTRACT

The deregulated expression of members of the phosphatase of regenerating liver (PRL) family has been implicated in the metastatic progression of multiple human cancers. Importantly, PRL-1 and PRL-3 both possess the capacity to drive key steps in metastatic progression. Yet, little is known about the regulation and oncogenic mechanisms of this emerging class of dual-specificity phosphatases. This prospect article details the involvement of PRLs in the metastatic cascade, the regulatory mechanisms controlling PRL expression, and recent efforts in the characterization of PRL-modulated pathways and substrates using biochemical and high-throughput approaches. Current advances and future prospects in anti-cancer therapy targeting this family are also discussed.


Subject(s)
Neoplasm Metastasis/pathology , Neoplasm Proteins/physiology , Protein Tyrosine Phosphatases/physiology , Dual-Specificity Phosphatases , Humans , Neoplasm Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...