Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters

Publication year range
1.
Mol Cancer ; 23(1): 50, 2024 03 09.
Article in English | MEDLINE | ID: mdl-38461268

ABSTRACT

Despite advancements in treatment protocols, cancer is one of the leading cause of deaths worldwide. Therefore, there is a need to identify newer and personalized therapeutic targets along with screening technologies to combat cancer. With the advent of pan-omics technologies, such as genomics, transcriptomics, proteomics, metabolomics, and lipidomics, the scientific community has witnessed an improved molecular and metabolomic understanding of various diseases, including cancer. In addition, three-dimensional (3-D) disease models have been efficiently utilized for understanding disease pathophysiology and as screening tools in drug discovery. An integrated approach utilizing pan-omics technologies and 3-D in vitro tumor models has led to improved understanding of the intricate network encompassing various signalling pathways and molecular cross-talk in solid tumors. In the present review, we underscore the current trends in omics technologies and highlight their role in understanding genotypic-phenotypic co-relation in cancer with respect to 3-D in vitro tumor models. We further discuss the challenges associated with omics technologies and provide our outlook on the future applications of these technologies in drug discovery and precision medicine for improved management of cancer.


Subject(s)
Multiomics , Neoplasms , Humans , Precision Medicine/methods , Genomics/methods , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/diagnosis , Metabolomics/methods , Drug Discovery
2.
Cell ; 138(2): 377-88, 2009 Jul 23.
Article in English | MEDLINE | ID: mdl-19632185

ABSTRACT

Prostate apoptosis response-4 (Par-4) is a proapoptotic protein with intracellular functions in the cytoplasm and nucleus. Unexpectedly, we noted Par-4 protein is spontaneously secreted by normal and cancer cells in culture, and by Par-4 transgenic mice that are resistant to spontaneous tumors. Short exposure to endoplasmic reticulum (ER) stress-inducing agents further increased cellular secretion of Par-4 by a brefeldin A-sensitive pathway. Secretion occurred independently of caspase activation and apoptosis. Interestingly, extracellular Par-4 induced apoptosis by binding to the stress response protein, glucose-regulated protein-78 (GRP78), expressed at the surface of cancer cells. The interaction of extracellular Par-4 and cell surface GRP78 led to apoptosis via ER stress and activation of the FADD/caspase-8/caspase-3 pathway. Moreover, apoptosis inducible by TRAIL, which also exerts cancer cell-specific effects, is dependent on extracellular Par-4 signaling via cell surface GRP78. Thus, Par-4 activates an extrinsic pathway involving cell surface GRP78 receptor for induction of apoptosis.


Subject(s)
Apoptosis , Receptors, Thrombin/metabolism , Animals , Brefeldin A/pharmacology , Cell Line , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Transgenic , Protein Structure, Tertiary , Protein Transport/drug effects , Receptors, Thrombin/chemistry
3.
J Biol Chem ; 298(7): 102123, 2022 07.
Article in English | MEDLINE | ID: mdl-35697073

ABSTRACT

High levels of redox enzymes have been commonly observed in various types of human cancer, although whether and how the enzymes contribute to cancer malignancy and therapeutic resistance have yet to be understood. Peroxiredoxin IV (Prx4) is an antioxidant with bona fide peroxidase and molecular chaperone functions. Here, we report that Prx4 is highly expressed in prostate cancer patient specimens, as well as established prostate cancer cell lines, and that its levels can be further stimulated through the activation of androgen receptor signaling. We used lentivirus-mediated shRNA knockdown and CRISPR-Cas9 based KO techniques to establish Prx4-depleted prostate cancer cells, which showed delayed cell cycle progression, reduced rate of cell proliferation, migration, and invasion compared to control cells. In addition, we used proteome profiler phosphokinase arrays to identify signaling changes in Prx4-depleted cells; we found that loss of Prx4 results in insufficient phosphorylation of both Akt and its downstream kinase GSK3α/ß. Moreover, we demonstrate that Prx4-depleted cells are more sensitive to ionizing radiation as they display compromised ability to scavenge reactive oxygen species and increased accumulation of DNA damage. In mouse xenograft models, we show depletion of Prx4 leads to significant suppression of tumor growth, and tumors formed by Prx4-depleted cells respond more effectively to radiation therapy. Our findings suggest that increased levels of Prx4 contribute to the malignancy and radioresistance of prostate cancer through the activation of Akt/GSK3 signaling pathways. Therefore, strategies targeting Prx4 may be utilized to potentially inhibit tumor growth and overcome radioresistance in prostate cancer.


Subject(s)
Prostatic Neoplasms , Proto-Oncogene Proteins c-akt , Animals , Cell Line, Tumor , Glycogen Synthase Kinase 3/metabolism , Humans , Male , Mice , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/radiotherapy , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
4.
Mol Cancer ; 21(1): 61, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35209919

ABSTRACT

The advent of molecular profiling has revolutionized the treatment of lung cancer by comprehensively delineating the genomic landscape of the epidermal growth factor receptor (EGFR) gene. Drug resistance caused by EGFR mutations and genetic polymorphisms of drug metabolizing enzymes and transporters impedes effective treatment of EGFR mutant and resistant lung cancer. This review appraises current literature, opportunities, and challenges associated with liquid biopsy and pharmacogenomic (PGx) testing as precision therapy tools in the management of EGFR mutant and resistant lung cancers. Liquid biopsy could play a potential role in selection of precise tyrosine kinase inhibitor (TKI) therapies during different phases of lung cancer treatment. This selection will be based on the driver EGFR mutational status, as well as monitoring the development of potential EGFR mutations arising during or after TKIs treatment, since some of these new mutations may be druggable targets for alternative TKIs. Several studies have identified the utility of liquid biopsy in the identification of EGFR driver and acquired resistance with good sensitivities for various blood-based biomarkers. With a plethora of sequencing technologies and platforms available currently, further evaluations using randomized controlled trials (RCTs) in multicentric, multiethnic and larger patient cohorts could enable optimization of liquid-based assays for the detection of EGFR mutations, and support testing of CYP450 enzymes and drug transporter polymorphisms to guide precise dosing of EGFR TKIs.


Subject(s)
Liquid Biopsy , Lung Neoplasms , Drug Resistance, Neoplasm , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Pharmacogenetics , Precision Medicine , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Randomized Controlled Trials as Topic
5.
Blood ; 131(26): 2943-2954, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29695515

ABSTRACT

Prostate apoptosis response-4 (Par-4), a proapoptotic tumor suppressor protein, is downregulated in many cancers including renal cell carcinoma, glioblastoma, endometrial, and breast cancer. Par-4 induces apoptosis selectively in various types of cancer cells but not normal cells. We found that chronic lymphocytic leukemia (CLL) cells from human patients and from Eµ-Tcl1 mice constitutively express Par-4 in greater amounts than normal B-1 or B-2 cells. Interestingly, knockdown of Par-4 in human CLL-derived Mec-1 cells results in a robust increase in p21/WAF1 expression and decreased growth due to delayed G1-to-S cell-cycle transition. Lack of Par-4 also increased the expression of p21 and delayed CLL growth in Eµ-Tcl1 mice. Par-4 expression in CLL cells required constitutively active B-cell receptor (BCR) signaling, as inhibition of BCR signaling with US Food and Drug Administration (FDA)-approved drugs caused a decrease in Par-4 messenger RNA and protein, and an increase in apoptosis. In particular, activities of Lyn, a Src family kinase, spleen tyrosine kinase, and Bruton tyrosine kinase are required for Par-4 expression in CLL cells, suggesting a novel regulation of Par-4 through BCR signaling. Together, these results suggest that Par-4 may play a novel progrowth rather than proapoptotic role in CLL and could be targeted to enhance the therapeutic effects of BCR-signaling inhibitors.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Gene Expression Regulation, Leukemic , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Cell Cycle , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Deletion , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mice, Inbred C57BL , Mice, Inbred NOD , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Up-Regulation
6.
J Immunol ; 200(12): 4180-4189, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29712773

ABSTRACT

Chronic lymphocytic leukemia (CLL) patients progressively develop an immunosuppressive state. CLL patients have more plasma IL-10, an anti-inflammatory cytokine, than healthy controls. In vitro human CLL cells produce IL-10 in response to BCR cross-linking. We used the transgenic Eµ-T cell leukemia oncogene-1 (TCL1) mouse CLL model to study the role of IL-10 in CLL associated immunosuppression. Eµ-TCL mice spontaneously develop CLL because of a B cell-specific expression of the oncogene, TCL1. Eµ-TCL1 mouse CLL cells constitutively produce IL-10, which is further enhanced by BCR cross-linking, CLL-derived IL-10 did not directly affect survival of murine or human CLL cells in vitro. We tested the hypothesis that the CLL-derived IL-10 has a critical role in CLL disease in part by suppressing the host immune response to the CLL cells. In IL-10R-/- mice, wherein the host immune cells are unresponsive to IL-10-mediated suppressive effects, there was a significant reduction in CLL cell growth compared with wild type mice. IL-10 reduced the generation of effector CD4 and CD8 T cells. We also found that activation of BCR signaling regulated the production of IL-10 by both murine and human CLL cells. We identified the transcription factor, Sp1, as a novel regulator of IL-10 production by CLL cells and that it is regulated by BCR signaling via the Syk/MAPK pathway. Our results suggest that incorporation of IL-10 blocking agents may enhance current therapeutic regimens for CLL by potentiating host antitumor immune response.


Subject(s)
Interleukin-10/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Animals , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation/physiology , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins/immunology , Signal Transduction/immunology
7.
Int J Mol Sci ; 21(18)2020 Sep 10.
Article in English | MEDLINE | ID: mdl-32927660

ABSTRACT

Collagen prolyl 4-hydroxylase 1 (C-P4H1) is an α-ketoglutarate (α-KG)-dependent dioxygenase that catalyzes 4-hydroxylation of proline on collagen. C-P4H1-induced prolyl hydroxylation is required for proper collagen deposition and cancer metastasis. Therefore, targeting C-P4H1 is considered a potential therapeutic strategy for collagen-related cancer progression and metastasis. However, no C-P4H1 inhibitors are available for clinical testing, and the high content assay is currently not available for C-P4H1 inhibitor screening. In the present study, we developed a high-throughput screening assay by quantifying succinate, a byproduct of C-P4H-catalyzed hydroxylation. C-P4H1 is the major isoform of collagen prolyl 4-hydroxylases (CP4Hs) that contributes the majority prolyl 4-hydroxylase activity. Using C-P4H1 tetramer purified from the eukaryotic expression system, we showed that the Succinate-GloTM Hydroxylase assay was more sensitive for measuring C-P4H1 activity compared with the hydroxyproline colorimetric assay. Next, we performed high-throughput screening with the FDA-approved drug library and identified several new C-P4H1 inhibitors, including Silodosin and Ticlopidine. Silodosin and Ticlopidine inhibited C-P4H1 activity in a dose-dependent manner and suppressed collagen secretion and tumor invasion in 3D tissue culture. These C-P4H1 inhibitors provide new agents to test clinical potential of targeting C-P4H1 in suppressing cancer progression and metastasis.


Subject(s)
Antineoplastic Agents/analysis , High-Throughput Screening Assays/methods , Prolyl-Hydroxylase Inhibitors/analysis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Humans , Indoles/chemistry , Ticlopidine/chemistry
8.
Carcinogenesis ; 37(11): 1027-1040, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27543608

ABSTRACT

Mouse double minute 2 (MDM2) protein functionally inactivates the tumor suppressor p53 in human cancer. Conventional MDM2 inhibitors provide limited clinical application as they interfere only with the MDM2-p53 interaction to release p53 from MDM2 sequestration but do not prevent activated p53 from transcriptionally inducing MDM2 expression. Here, we report a rationally synthesized chalcone-based pyrido[ b ]indole, CPI-7c, as a unique small-molecule inhibitor of MDM2, which not only inhibited MDM2-p53 interaction but also promoted MDM2 degradation. CPI-7c bound to both RING and N-terminal domains of MDM2 to promote its ubiquitin-mediated degradation and p53 stabilization. CPI-7c-induced p53 directly recruited to the promoters of DR4 and DR5 genes and enhanced their expression, resulting in sensitization of TNF-related apoptosis-inducing ligand (TRAIL)-resistant cancer cells toward TRAIL-induced apoptosis. Collectively, we identified CPI-7c as a novel small-molecule inhibitor of MDM2 with a unique two-prong mechanism of action that sensitized TRAIL-resistant cancer cells to apoptosis by modulating the MDM2-p53-DR4/DR5 pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Carbolines/pharmacology , Drug Resistance, Neoplasm , Propiophenones/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Carbolines/chemistry , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Models, Molecular , Molecular Targeted Therapy , Promoter Regions, Genetic , Propiophenones/chemistry , Protein Binding , Protein Stability , Proteolysis , Proto-Oncogene Proteins c-mdm2/chemistry , Proto-Oncogene Proteins c-mdm2/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Transcription, Genetic/drug effects , Tumor Suppressor Protein p53/metabolism , Ubiquitination/drug effects , Up-Regulation
9.
Nat Chem Biol ; 10(11): 924-926, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25218743

ABSTRACT

The tumor suppressor protein prostate apoptosis response-4 (Par-4), which is secreted by normal cells, selectively induces apoptosis in cancer cells. We identified a 3-arylquinoline derivative, designated Arylquin 1, as a potent Par-4 secretagogue in cell cultures and mice. Mechanistically, Arylquin 1 binds vimentin, displaces Par-4 from vimentin for secretion and triggers the efficient paracrine apoptosis of diverse cancer cells. Thus, targeting vimentin with Par-4 secretagogues efficiently induces paracrine apoptosis of tumor cells.


Subject(s)
Aminoquinolines/pharmacology , Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Neoplasms/pathology , Vimentin/antagonists & inhibitors , Aminoquinolines/administration & dosage , Aminoquinolines/chemistry , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Molecular Structure , Paracrine Communication/drug effects , Vimentin/chemistry
10.
Org Biomol Chem ; 14(1): 74-84, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26548370

ABSTRACT

Advanced prostate tumors usually metastasize to the lung, bone, and other vital tissues and are resistant to conventional therapy. Prostate apoptosis response-4 protein (Par-4) is a tumor suppressor that causes apoptosis in therapy-resistant prostate cancer cells by binding specifically to a receptor, Glucose-regulated protein-78 (GRP78), found only on the surface of cancer cells. 3-Arylquinolines or "arylquins" induce normal cells to release Par-4 from the intermediate filament protein, vimentin and promote Par-4 secretion that targets cancer cells in a paracrine manner. A structure-activity study identified arylquins that promote Par-4 secretion, and an evaluation of arylquin binding to the hERG potassium ion channel using a [(3)H]-dofetilide binding assay permitted the identification of structural features that separated this undesired activity from the desired Par-4 secretory activity. A binding study that relied on the natural fluorescence of arylquins and that used the purified rod domain of vimentin (residues 99-411) suggested that the mechanism behind Par-4 release involved arylquin binding to multiple sites in the rod domain.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Quinolones/metabolism , Quinolones/pharmacology , Vimentin/metabolism , Binding Sites/drug effects , Endoplasmic Reticulum Chaperone BiP , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Molecular Structure , Quinolones/chemistry , Stereoisomerism , Structure-Activity Relationship , Vimentin/chemistry
11.
Tetrahedron Lett ; 56(23): 3382-3384, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-26236052

ABSTRACT

Nitrosation and cyclization of 4-(3-aminothieno[2,3-b]pyridine-2-yl)-2H-chromen-2-ones 1 afforded substituted 6H-chromeno[3,4-c]pyrido[3',2':4,5]thieno[2,3-e]pyridazin-6-ones 2 that inhibited the intermediary filament protein, vimentin, at low micromolar concentrations. This inhibition promoted the secretion of Prostate Apoptosis Response-4 protein (Par-4), which selectively triggered apoptosis in prostate cancer cells such as CWR22Rv1, LNCaP-derivative C4-2B, PC-3 and its aggressive analog, PC-3 MM2.

12.
Acta Neuropathol ; 128(5): 723-32, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25135281

ABSTRACT

Prostate apoptosis response-4 (Par-4) is an endogenous tumor suppressor that selectively induces apoptosis in a variety of cancers. Although it has been the subject of intensive research in other cancers, less is known about its significance in gliomas, including whether it is regulated by key driver mutations, has therapeutic potential against glioma stem cells (GSCs), and/or is a prognostic marker. We found that patient-derived gliomas with mutant isocitrate dehydrogenase 1 have markedly lower Par-4 expression (P < 0.0001), which was validated by The Cancer Genome Atlas dataset (P = 2.0 E-13). The metabolic product of mutant IDH1, D-2-hydroxyglutarate (2-HG), can suppress Par-4 transcription in vitro via inhibition of promoter activity as well as enhanced mRNA degradation, but interestingly not by direct DNA promoter hypermethylation. The Selective for Apoptosis induction in Cancer cells (SAC) domain within Par-4 is highly active against glioma cells, including orthotopic xenografts of patient-derived primary GSCs (P < 0.0001). Among high-grade gliomas that are IDH1 wild type, those that express more Par-4 have significantly longer median survival (18.4 vs. 8.0 months, P = 0.002), a finding confirmed in two external GBM cohorts. Together, these data suggest that Par-4 is a significant component of the mutant IDH1 phenotype, that the activity of 2-HG is complex and can extend beyond direct DNA hypermethylation, and that Par-4 is a promising therapeutic strategy against GSCs. Furthermore, not every effect of mutant IDH1 necessarily contributes to the overall favorable prognosis seen in such tumors; inhibition of Par-4 may be one such effect.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics , Isocitrate Dehydrogenase/genetics , Mutation/genetics , Neoplastic Stem Cells/pathology , AC133 Antigen , Analysis of Variance , Animals , Antigens, CD/metabolism , Apoptosis Regulatory Proteins/genetics , Brain Neoplasms , Cell Line, Tumor , DNA Methylation , Disease Models, Animal , Flow Cytometry , Glioma/pathology , Glycoproteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Peptides/metabolism , RNA, Messenger , Tissue Array Analysis , Transplantation, Heterologous
13.
Adv Exp Med Biol ; 818: 155-66, 2014.
Article in English | MEDLINE | ID: mdl-25001535

ABSTRACT

Tumor suppressor genes play an important role in preventing neoplastic transformation and maintaining normal tissue homeostasis. Par-4 is one such tumor suppressor which is unique in its ability to selectively induce apoptosis in cancer cells while leaving the normal cells unaffected. The cancer cell specific activity of Par-4 is elicited through intracellular as well as extracellular mechanisms. Intracellularly Par-4 acts through the inhibition of pro-survival pathways and activation of Fas mediated apoptosis whereas extracellular (secreted Par-4) acts by binding to cell surface GRP78 leading to activation of the extrinsic apoptotic pathway. Many studies have highlighted the importance of Par-4 not only in preventing cancer development/recurrence but also as a promising anticancer therapeutic agent.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Protein Binding , Tumor Suppressor Proteins/genetics , fas Receptor/genetics , fas Receptor/metabolism
14.
Commun Biol ; 7(1): 732, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38886572

ABSTRACT

Ferroptosis is a unique iron-dependent form of non-apoptotic cell death characterized by devastating lipid peroxidation. Whilst growing evidence suggests that ferroptosis is a type of autophagy-dependent cell death, the underlying molecular mechanisms regulating ferroptosis are largely unknown. In this study, through an unbiased RNA-sequencing screening, we demonstrate the activation of a multi-faceted tumor-suppressor protein Par-4/PAWR during ferroptosis. Functional studies reveal that genetic depletion of Par-4 effectively blocks ferroptosis, whereas Par-4 overexpression sensitizes cells to undergo ferroptosis. More importantly, we have determined that Par-4-triggered ferroptosis is mechanistically driven by the autophagic machinery. Upregulation of Par-4 promotes activation of ferritinophagy (autophagic degradation of ferritin) via the nuclear receptor co-activator 4 (NCOA4), resulting in excessive release of free labile iron and, hence, enhanced lipid peroxidation and ferroptosis. Inhibition of Par-4 dramatically suppresses the NCOA4-mediated ferritinophagy signaling axis. Our results also establish that Par-4 activation positively correlates with reactive oxygen species (ROS) production, which is critical for ferritinophagy-mediated ferroptosis. Furthermore, Par-4 knockdown effectively blocked ferroptosis-mediated tumor suppression in the mouse xenograft models. Collectively, these findings reveal that Par-4 has a crucial role in ferroptosis, which could be further exploited for cancer therapy.


Subject(s)
Autophagy , Ferroptosis , Nuclear Receptor Coactivators , Reactive Oxygen Species , Ferroptosis/genetics , Humans , Animals , Nuclear Receptor Coactivators/metabolism , Nuclear Receptor Coactivators/genetics , Mice , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Lipid Peroxidation , Iron/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Signal Transduction
15.
Breast Cancer Res ; 15(5): 314, 2013.
Article in English | MEDLINE | ID: mdl-24164776

ABSTRACT

Therapy resistance and disease recurrence are two of the most challenging aspects in breast cancer treatment. A recent article in Cancer Cell makes a significant contribution toward a better understanding of this therapeutic problem by establishing downregulation of the tumor suppressor Par-4 as the primary determinant of breast cancer recurrence. This viewpoint brings forth the importance of their findings and its implications on future research and therapy.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Breast Neoplasms/etiology , Neoplasm Recurrence, Local/etiology , Animals , Female , Humans
16.
Am J Cancer Res ; 13(3): 976-991, 2023.
Article in English | MEDLINE | ID: mdl-37034206

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths. Lung cancer cells develop resistance to apoptosis by suppressing the secretion of the tumor suppressor Par-4 protein (also known as PAWR) and/or down-modulating the Par-4 receptor GRP78 on the cell surface (csGRP78). We sought to identify FDA-approved drugs that elevate csGRP78 on the surface of lung cancer cells and induce Par-4 secretion from the cancer cells and/or normal cells in order to inhibit cancer growth in an autocrine or paracrine manner. In an unbiased screen, we identified crizotinib (CZT), an inhibitor of activated ALK/MET/ROS1 receptor tyrosine kinase, as an inducer of csGRP78 expression in ALK-negative, KRAS or EGFR mutant lung cancer cells. Elevation of csGRP78 in the lung cancer cells was dependent on activation of the non-receptor tyrosine kinase SRC by CZT. Inhibition of SRC activation in the cancer cells prevented csGRP78 translocation but promoted Par-4 secretion by CZT, implying that activated SRC prevented Par-4 secretion. In normal cells, CZT did not activate SRC and csGRP78 elevation but induced Par-4 secretion. Consequently, CZT induced Par-4 secretion from normal cells and elevated csGRP78 in the ALK-negative tumor cells to cause paracrine apoptosis in cancer cell cultures and growth inhibition of tumor xenografts in mice. Thus, CZT induces differential activation of SRC in normal and cancer cells to trigger the pro-apoptotic Par-4-GRP78 axis. As csGRP78 is a targetable receptor, CZT can be repurposed to elevate csGRP78 for inhibition of ALK-negative lung tumors.

17.
Cells ; 12(21)2023 10 27.
Article in English | MEDLINE | ID: mdl-37947608

ABSTRACT

C1orf74, also known as URCL4, has been reported to have higher expression and be associated with poor prognosis in lung adenocarcinoma patients, and its role in regulation of the EGFR/AKT/mTORC1 pathway has been recently elucidated. In the current study, we used publicly available data and experimental validation of C1orf74 gene expression and its association with prognosis in cervical cancer patients. qRT-PCR was performed using RNA from cervical cancer cell lines and twenty-five cervical cancer patients. Data from TNMplot revealed that mRNA expression of the C1orf74 gene in primary tumor tissues, as well as metastatic tissues from cervical cancer patients, was significantly higher compared to normal cervical tissues. HPV-positive tumors had higher expression of this gene compared to HPV-negative tumors. qPCR analysis also demonstrated higher expression of C1orf74 in HPV-positive cervical cancer cell lines and most cervical cancer patients. The promoter methylation levels of the C1orf74 gene in cervical cancer tissues were lower compared to normal cervical tissues (p < 0.05). Collectively, our study indicates that higher expression of the C1orf74 gene caused by hypomethylation of its promoter is associated with poor overall survival in cervical cancer patients. Thus, C1orf74 is a novel prognostic marker in cervical cancer.


Subject(s)
Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , Uterine Cervical Neoplasms/metabolism , Cell Line, Tumor , Papillomavirus Infections/pathology , Cervix Uteri/metabolism , Gene Expression
18.
J Cell Physiol ; 227(12): 3715-21, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22552839

ABSTRACT

Par-4 is a pro-apoptotic, tumor suppressor protein that induces apoptosis selectively in cancer cells. Endoplasmic reticulum-stress and higher levels of protein kinase A in tumor cells confer the coveted feature of cancer selective response to extracellular and intracellular Par-4, respectively. Recent studies have shown that systemic Par-4 confers resistance to tumor growth in mice, and that tumor-resistance is transferable by bone-marrow transplantation. Moreover, recombinant Par-4 inhibits the growth of tumors in mice. As systemic Par-4 induces apoptosis via cell surface GRP78, strategies that promote GRP78 trafficking to the cell surface are expected sensitize cancer cells to circulating levels of Par-4. This review illustrates the domains and mechanisms by which Par-4 orchestrates the apoptotic process in both cell culture models and in physiological settings.


Subject(s)
Apoptosis/physiology , Gene Expression Regulation, Neoplastic/physiology , Receptors, Thrombin/metabolism , Animals , Endoplasmic Reticulum Chaperone BiP , Genetic Predisposition to Disease , Humans , Neoplasms/metabolism , Receptors, Thrombin/genetics
19.
Int J Cancer ; 131(11): 2519-27, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22447412

ABSTRACT

Tobacco use significantly increases the risk of developing cancer. Moreover, there is growing evidence that tobacco use decreases survival in cancer patients. Nicotine, a systemically available component of tobacco, is associated with tumor promotion and decreased apoptosis in cell culture; however, the role of nicotine on response to radiotherapy (RT) or chemoradiotherapy (CRT) in vivo has not been evaluated. Our study evaluated the effects of nicotine administration on cancer cell survival in cell culture and mouse models. Nicotine increased survival in two cell lines following RT in vitro. Nicotine administration in mice during fractionated RT or CRT increased xenograft regrowth as compared to RT or CRT alone. Nicotine increased hypoxia-inducible factor 1-alpha (HIF-1α) expression in tumor xenografts without altering expression of carbonic-anhydrase, a clinical marker of tumor hypoxia. The effects of nicotine on HIF-1α expression were transient, returning to baseline levels within 2-3 days after nicotine removal. Further mechanistic studies indicated that inhibition of phosphoinositide-3-kinase (PI3K) prevented nicotine-mediated increases in HIF-1α expression as well as the prosurvival effects of nicotine on RT. These findings imply that during tobacco use, nicotine may function as a systemic agent through acute and reversible regulation of HIF-1α expression and a decreased therapeutic response.


Subject(s)
Lung Neoplasms/pathology , Lung Neoplasms/therapy , Nicotine/toxicity , Animals , Carbonic Anhydrases/genetics , Carbonic Anhydrases/metabolism , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Humans , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Nude , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Random Allocation , Xenograft Model Antitumor Assays
20.
Leuk Lymphoma ; 63(8): 1810-1822, 2022 08.
Article in English | MEDLINE | ID: mdl-35258388

ABSTRACT

The chronic lymphocytic leukemia (CLL) microenvironment has been receiving an increasing amount of attention, but there is currently limited data surrounding how the microenvironment affects initial development of CLL. We determined that the spleen is the initial site of CLL growth through monitoring of transgenic Eµ-TCL1 mice that develop CLL. Subsequently, we isolated stromal cells from the spleens of Eµ-TCL1 mice (EMST cells) that induce CLL cell division in vitro. Both cell-cell contact and soluble factors were involved in EMST-induced CLL cell division. These stromal cells are present in significantly larger numbers in the spleen than other lymphoid organs. We also noted that splenectomy delayed CLL development in Eµ-TCL1 mice and completely prevented CLL development in adoptive transfer mice. Our findings will allow future studies surrounding the CLL microenvironment to focus upon the splenic stromal cells.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Animals , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mice , Mice, Transgenic , Proto-Oncogene Proteins , Spleen , Stromal Cells , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL