Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Cancer Causes Control ; 32(6): 567-575, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33909208

ABSTRACT

PURPOSE: Incidence of breast cancer (BC), particularly in young women, are rising in India. Without population-based mammography screening, rising rates cannot be attributed to screening. Investigations are needed to understand the potential drivers of this trend. METHODS: An international team of experts convened to discuss the trends, environmental exposures, and clinical implications associated with BC in India and outlined recommendations for its management. RESULTS: Panels were structured across three major BC themes (n = 10 presentations). The symposium concluded with a semi-structured Think Tank designed to elicit short-term and long-term goals that could address the challenges of BC in India. CONCLUSION: There was consensus that the prevalence of late-stage BC and the high BC mortality rates are associated with the practice of detection, which is primarily through clinical and self-breast exams, as opposed to mammography. Triple-Negative BC (TNBC) was extensively discussed, including TNBC etiology and potential risk factors, the limited treatment options, and if reported TNBC rates are supported by rigorous scientific evidence. The Think Tank session yielded long-term and short-term goals to further BC reduction in India and included more regional etiological studies on environmental exposures using existing India-based cohorts and case-control studies, standardization for molecular subtyping of BC cases, and improving the public's awareness of breast health.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/epidemiology , Breast Neoplasms/etiology , Early Detection of Cancer/standards , Environmental Exposure , Humans , India , Mammography , Reference Standards , Risk Factors
2.
Sci Adv ; 6(34)2020 08.
Article in English | MEDLINE | ID: mdl-32937365

ABSTRACT

Identification of targeted therapies for TNBC is an urgent medical need. Using a drug combination screen reliant on synthetic lethal interactions, we identified clinically relevant combination therapies for different TNBC subtypes. Two drug combinations targeting the BET family were further explored. The first, targeting BET and CXCR2, is specific for mesenchymal TNBC and induces apoptosis, whereas the second, targeting BET and the proteasome, is effective for major TNBC subtypes and triggers ferroptosis. Ferroptosis was induced at low drug doses and was associated with increased cellular iron and decreased glutathione levels, concomitant with reduced levels of GPX4 and key glutathione biosynthesis genes. Further functional studies, analysis of clinical datasets and breast cancer specimens revealed a unique vulnerability of TNBC to ferroptosis inducers, enrichment of ferroptosis gene signature, and differential expression of key proteins that increase labile iron and decrease glutathione levels. This study identified potent combination therapies for TNBC and unveiled ferroptosis as a promising therapeutic strategy.

3.
Adv Cancer Res ; 147: 109-160, 2020.
Article in English | MEDLINE | ID: mdl-32593399

ABSTRACT

The human epidermal growth factor receptor (HER) family of receptor tyrosine kinases (RTKs) are among the first layer of molecules that receive, interpret, and transduce signals leading to distinct cancer cell phenotypes. Since the discovery of the tooth-lid factor-later characterized as the epidermal growth factor (EGF)-and its high-affinity binding EGF receptor, HER kinases have emerged as one of the commonly upregulated or hyperactivated or mutated kinases in epithelial tumors, thus allowing HER1-3 family members to regulate several hallmarks of cancer development and progression. Each member of the HER family exhibits shared and unique structural features to engage multiple receptor activation modes, leading to a range of overlapping and distinct phenotypes. EGFR, the founding HER family member, provided the roadmap for the development of the cell surface RTK-directed targeted cancer therapy by serving as a prototype/precursor for the currently used HER-directed cancer drugs. We herein provide a brief account of the discoveries, defining moments, and historical context of the HER family and guidepost advances in basic, translational, and clinical research that solidified a prominent position of the HER family in cancer research and treatment. We also discuss the significance of HER3 pseudokinase in cancer biology; its unique structural features that drive transregulation among HER1-3, leading to a superior proximal signaling response; and potential role of HER3 as a shared effector of acquired therapeutic resistance against diverse oncology drugs. Finally, we also narrate some of the current drawbacks of HER-directed therapies and provide insights into postulated advances in HER biology with extensive implications of these therapies in cancer research and treatment.


Subject(s)
ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Neoplasms/drug therapy , Neoplasms/enzymology , Protein Kinase Inhibitors/therapeutic use , Animals , Disease Progression , Drug Resistance, Neoplasm , Humans , Molecular Targeted Therapy , Mutation , Neoplasms/genetics , Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Signal Transduction
5.
Cell Death Dis ; 9(10): 985, 2018 09 24.
Article in English | MEDLINE | ID: mdl-30250159

ABSTRACT

The tumor suppressor Hippo pathway negatively regulates the transcriptional coactivators Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) to inhibit cell growth and control organ size, whereas activation of YAP and TAZ is implicated in tumorigenesis and cancer metastasis. Here, we report that the nonreceptor tyrosine kinase PYK2 positively regulates TAZ and YAP transcriptional activity in triple-negative breast cancer (TNBC). We found that inhibition of PYK2 expression or its kinase activity substantially affects the steady-state level of TAZ and markedly facilitates its proteasomal degradation. This effect was specific to PYK2 inhibition and was not obtained by inhibition of FAK. Destabilization of TAZ was associated with profound effect of PYK2 inhibition on cell growth at low-density concomitant with reduced expression of TAZ-target genes and induction of cell apoptosis. We further show that PYK2 enhances the tyrosine phosphorylation of both TAZ and LATS1/2 and concomitantly TAZ stability, and that PYK2 protein level correlates with the level of TAZ protein in primary breast tumors. Together these observations suggest that PYK2 is an important regulator of the Hippo pathway, and its tyrosine kinase activity has a striking effect on TAZ stabilization and activation in TNBC.


Subject(s)
Focal Adhesion Kinase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Cell Line, Tumor , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 2/antagonists & inhibitors , Focal Adhesion Kinase 2/genetics , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , HEK293 Cells , Humans , Lithium Chloride/pharmacology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proteolysis , Quinolones/pharmacology , Sulfones/pharmacology , Trans-Activators , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transfection , Triple Negative Breast Neoplasms/pathology , YAP-Signaling Proteins
6.
PLoS Comput Biol ; 14(6): e1006192, 2018 06.
Article in English | MEDLINE | ID: mdl-29920512

ABSTRACT

Prediction of drug combinations that effectively target cancer cells is a critical challenge for cancer therapy, in particular for triple-negative breast cancer (TNBC), a highly aggressive breast cancer subtype with no effective targeted treatment. As signalling pathway networks critically control cancer cell behaviour, analysis of signalling network activity and crosstalk can help predict potent drug combinations and rational stratification of patients, thus bringing therapeutic and prognostic values. We have previously showed that the non-receptor tyrosine kinase PYK2 is a downstream effector of EGFR and c-Met and demonstrated their crosstalk signalling in basal-like TNBC. Here we applied a systems modelling approach and developed a mechanistic model of the integrated EGFR-PYK2-c-Met signalling network to identify and prioritize potent drug combinations for TNBC. Model predictions validated by experimental data revealed that among six potential combinations of drug pairs targeting the central nodes of the network, including EGFR, c-Met, PYK2 and STAT3, co-targeting of EGFR and PYK2 and to a lesser extent of EGFR and c-Met yielded strongest synergistic effect. Importantly, the synergy in co-targeting EGFR and PYK2 was linked to switch-like cell proliferation-associated responses. Moreover, simulations of patient-specific models using public gene expression data of TNBC patients led to predictive stratification of patients into subgroups displaying distinct susceptibility to specific drug combinations. These results suggest that mechanistic systems modelling is a powerful approach for the rational design, prediction and prioritization of potent combination therapies for individual patients, thus providing a concrete step towards personalized treatment for TNBC and other tumour types.


Subject(s)
Antineoplastic Agents , ErbB Receptors/metabolism , Focal Adhesion Kinase 2/metabolism , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , Triple Negative Breast Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Computational Biology , Databases, Genetic , Drug Synergism , Female , Gene Expression Profiling , Humans , Signal Transduction/drug effects , Signal Transduction/physiology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
7.
Cancer Res ; 77(1): 86-99, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27793840

ABSTRACT

Triple-negative breast cancer (TNBC) is a highly aggressive, heterogeneous disease with poor prognosis and no effective targeted therapies. EGFR is highly expressed in basal-like TNBC and is considered as a potential therapeutic target. However, EGFR targeting exerts only marginal clinical benefits, possibly due to activation of compensatory signaling pathways, which are frequently associated with HER3 upregulation. Here we show that concomitant targeting of EGFR and the nonreceptor tyrosine kinases PYK2/FAK synergistically inhibits the proliferation of basal-like TNBC cells in vitro and attenuates tumor growth in a mouse xenograft model. Dual targeting of EGFR and PYK2/FAK inhibited complementary key growth and survival pathways mediated by AKT, S6K, STAT3, and ERK1/2 activation. PYK2 inhibition also abrogated HER3 upregulation in response to EGFR antagonists, thereby circumventing HER3-associated drug resistance. Mechanistically, PYK2 inhibition facilitated the proteasomal degradation of HER3 while inducing upregulation of NDRG1 (N-myc downstream regulated 1 gene). NDRG1 enhanced the interaction of HER3 with the ubiquitin ligase NEDD4, while PYK2, which interacts with NEDD4 and HER3, interfered with NEDD4-HER3 binding, suggesting that the PYK2-NDRG1-NEDD4 circuit has a critical role in receptor degradation, drug response, and resistance mechanism. Our studies offer a preclinical proof of concept for a strategy of cotargeting the EGFR and PYK2/FAK kinases to improve TNBC therapy. Cancer Res; 77(1); 86-99. ©2016 AACR.


Subject(s)
Drug Resistance, Neoplasm/physiology , ErbB Receptors/antagonists & inhibitors , Focal Adhesion Kinase 2/antagonists & inhibitors , Signal Transduction/physiology , Triple Negative Breast Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Endosomal Sorting Complexes Required for Transport/metabolism , Female , Fluorescent Antibody Technique , Gefitinib , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Nedd4 Ubiquitin Protein Ligases , Oligonucleotide Array Sequence Analysis , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Receptor, ErbB-3/genetics , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/metabolism , Ubiquitin-Protein Ligases/metabolism , Xenograft Model Antitumor Assays
8.
Nat Commun ; 6: 6064, 2015 Feb 04.
Article in English | MEDLINE | ID: mdl-25648557

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is a central developmental process implicated in cancer metastasis. Here we show that the tyrosine kinase PYK2 enhances cell migration and invasion and potentiates EMT in human breast carcinoma. EMT inducer, such as EGF, induces rapid phosphorylation of PYK2 and its translocation to early endosomes where it co-localizes with EGFR and sustains its downstream signals. Furthermore, PYK2 enhances EGF-induced STAT3-phosphorylation, while phospho-STAT3 directly binds to PYK2 promoter and regulates PYK2 transcription. STAT3 and PYK2 also enhance c-Met expression, while c-Met augments their phosphorylation, suggesting a positive feedback loop between PYK2-STAT3-c-Met. We propose that PYK2 sustains endosomal-derived receptor signalling and participates in a positive feedback that links cell surface receptor(s) to transcription factor(s) activation, thereby prolonging signalling duration and potentiating EMT. Given the role of EMT in breast cancer metastasis, we also found a significant correlation between PYK2 expression, tumour grade and lymph node metastasis, thus, demonstrating the clinicopathological implication of our findings.


Subject(s)
Endosomes/metabolism , Epithelial-Mesenchymal Transition/physiology , Focal Adhesion Kinase 2/metabolism , Cell Line , Cell Line, Tumor , Chromatin Immunoprecipitation , Epithelial-Mesenchymal Transition/genetics , Focal Adhesion Kinase 2/genetics , Humans , Protein Binding/genetics , Protein Binding/physiology , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , Signal Transduction/physiology
9.
Fish Shellfish Immunol ; 41(1): 64-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24657259

ABSTRACT

Evaluation of functional mechanism of immunostimulant in fish is essential for realizing their therapeutic potential. The impact of dietary supplementation of seeds of Achyranthes aspera on the immune system of Catla catla was studied. Catla (37.73 ± 6.7 g) were fed with test diets containing 0.5 (D1) and 1.0% (D2) seeds of A. aspera and control diet (D3) for 50 days. Then fish were immunized with heat-killed Aeromonas hydrophila. Blood and tissue samples were collected after 7 days of immunization. Three fish were sacrificed for each parameter. Respiratory burst activity, bactericidal activity and antibody response were significantly (P < 0.05) higher in test diets fed catla compared to the control one. Nitric oxide synthase and serum lysozyme were significantly (P < 0.05) higher in fish fed with diet D2 compared to the other treatments. There were up-regulations of TNFα, lysozyme c and g gene expressions in kidney and IL-10, lysozyme c and g expressions in hepatopancreas of test diets fed fish compared to the control one. The expression of IL-10 was down-regulated in the kidney of D2 diet fed fish compared to others. In the gill, significantly (P < 0.05) higher expressions of TNFα was found in D1 diet feed fish and lysozyme c and in D2 diet fed fish compared to the other groups. This study showed tissue-specific gene expression pattern in catla. An inverse relationship was found between the expression of TNFα and IL-10 in kidney of catla.


Subject(s)
Achyranthes/immunology , Aeromonas hydrophila/immunology , Carps , Fish Diseases/microbiology , Gene Expression Regulation/immunology , Gram-Negative Bacterial Infections/veterinary , Animals , Aquaculture , Fish Diseases/blood , Fish Diseases/genetics , Fish Diseases/immunology , Gills/immunology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/microbiology , Hepatopancreas/immunology , Immunomodulation/immunology , Interleukin-10/blood , Kidney/immunology , Muramidase/blood , Nitric Oxide Synthase/blood , Random Allocation , Respiratory Burst/immunology , Tumor Necrosis Factor-alpha/blood
10.
J Complement Integr Med ; 9: Article 4, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-22499721

ABSTRACT

Shea butter is traditionally used in Africa for its anti-inflammatory and analgesic effects. In this study we explored the anti-inflammatory activities of the methanolic extract of shea butter (SBE) using lipopolysaccharide (LPS)-induced murine macrophage cell line J774. It was observed that SBE significantly reduced the levels of LPS-induced nitric oxide, Tumor necrosis factor-α (TNF-α), interleukins, 1ß (IL-1ß), and -12 (IL-12) in the culture supernatants in a dose dependent manner. Expression of pro-inflammatory enzymes, inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) were also inhibited by SBE. These anti-inflammatory effects were due to an inhibitory action of SBE on LPS-induced iNOS, COX-2, TNF-α, IL-1ß, and IL-12 mRNA expressions. Moreover, SBE efficiently suppressed IκB phosphorylation and NF-κB nuclear translocation induced by LPS. These findings explain the molecular bases of shea butter's bioactivity against various inflammatory conditions and substantiate it as a latent source of novel therapeutic agents.


Subject(s)
Macrophages/drug effects , Plant Extracts/pharmacology , Sapotaceae , Animals , Biomarkers/metabolism , Blotting, Western , Cell Line , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Interleukin-12/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Plants, Medicinal , Reverse Transcriptase Polymerase Chain Reaction , Seeds , Tumor Necrosis Factor-alpha/metabolism
11.
Shock ; 33(5): 493-9, 2010 May.
Article in English | MEDLINE | ID: mdl-19823115

ABSTRACT

Sepsis and/or systemic inflammatory response syndrome are leading causes of death in intensive care unit patients. NO is a critical player in the pathogenesis of bacterial sepsis. Several studies demonstrate elevation of iNOS in LPS-induced acute inflammatory responses and mortality; however, the effectiveness of its therapeutic suppression in systemic inflammation is largely controversial. Earlier, we have reported that DNAzymes specific to iNOS mRNA efficiently suppress iNOS expression in LPS-stimulated J774 murine macrophages. In the present study, we explored the effects of two of these DNAzymes in BALB/c mice model of LPS-induced lethal systemic inflammation. Experimental animal groups receiving previous injections of iNOS-specific DNAzyme (100 microg, i.p.) showed significantly reduced mortality. Total cell counts of peritoneal lavage and histopathological studies of tissues demonstrated substantial reduction in the leukocytic infiltration and edema in DNAzyme-treated mice. In addition, DNAzyme-injected animals displayed significantly decreased IL-12 serum level, whereas the levels of IL-1[beta], IFN-[gamma], and TNF-[alpha] also declined to a great extent. DNAzyme treatment resulted in significantly reduced NO levels in serum and peritoneal lavage, confirming functional suppression of iNOS gene in LPS-injected mice. These DNAzymes were also able to limit excessive NO production by cytokine and LPS co-challenges in cultured peritoneal macrophages from DNAzyme-treated mice. Estimation of iNOS mRNA and protein expression in the peritoneal macrophages of DNAzyme-administered animals further confirmed the iNOS gene knockdown. All these results indicated that iNOS-specific DNAzymes reduce inflammatory responses and enhance survival in murine model of LPS-induced lethal systemic inflammation.


Subject(s)
DNA, Catalytic/therapeutic use , DNA, Single-Stranded/therapeutic use , Inflammation/drug therapy , Lipopolysaccharides/toxicity , Nitric Oxide Synthase Type II/genetics , Animals , Cells, Cultured , Female , Gene Knockdown Techniques , Inflammation/chemically induced , Inflammation/mortality , Inflammation/pathology , Inflammation Mediators/antagonists & inhibitors , Interleukin-12/metabolism , Macrophages, Peritoneal , Mice , Mice, Inbred BALB C , Nitric Oxide/biosynthesis , Nitric Oxide/blood
12.
Mol Cell Biochem ; 336(1-2): 127-35, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19813079

ABSTRACT

Whole plant methanolic extracts of 14 traditionally used medicinal herbs were evaluated for their anti-inflammatory activity. Extracts of Grindelia robusta, Salix nigra, Arnica montana, and Quassia amara showed up to 4.5-fold inhibition of nitric oxide (NO) production in the J774 murine macrophage cells challenged with LPS without cytotoxicity. These four selected extracts significantly reduced the protein levels of inducible NO synthase (iNOS) and the cyclooxygenase-2 (COX-2) as observed by Western blot analysis. Culture supernatants from cells treated with these extracts indicated 3-5-fold reduction of tumor necrosis factor-alpha (TNF-alpha). However, only G. robusta and Q. amara extracts significantly inhibited (by 50%) IL-1beta and IL-12 secretions. Furthermore, all these plant extracts were shown to prevent the LPS-mediated nuclear translocation of nuclear factor-kappaB (NF-kappaB). All the above observations indicate the anti-inflammatory potential of these plant extracts.


Subject(s)
Anti-Infective Agents/pharmacology , Inflammation Mediators/metabolism , Inflammation/prevention & control , Lipopolysaccharides/toxicity , Macrophages/drug effects , Plant Extracts/pharmacology , Animals , Arnica/chemistry , Cell Line , Cyclooxygenase 2/metabolism , Grindelia/chemistry , Inflammation/chemically induced , Interleukins/metabolism , Macrophages/metabolism , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitrites/metabolism , Plants, Medicinal/chemistry , Protein Transport/drug effects , Quassia/chemistry , Salix/chemistry , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
13.
FEBS Lett ; 583(17): 2968-74, 2009 Sep 03.
Article in English | MEDLINE | ID: mdl-19683526

ABSTRACT

Persistently elevated level of TNF-alpha has been implicated in several inflammatory disorders, however, its autocrine production through TNF-alpha receptors signaling is poorly understood. Here we report that simultaneous silencing of TNF-receptors, R1 and R2 by DNAzyme or siRNA suppressed TNF-alpha expression more efficiently than silencing them individually in lipopolysaccharides (LPS) stimulated THP-1 macrophages. Co-silencing of TNF-receptors also inhibited TNF-alpha induced NF-kappaB activation to a higher extent. It was further observed that NF-kappaB inhibitor but not c-Jun N-terminal kinase inhibitor (SP600125) suppressed TNF-alpha expression. All these results suggest that TNF-alpha expression is regulated by synergistic signaling of TNF receptors through downstream NF-kappaB activation.


Subject(s)
Macrophages/metabolism , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor-alpha/metabolism , Anthracenes/metabolism , Cells, Cultured , Enzyme Inhibitors/metabolism , Gene Silencing , Humans , Lipopolysaccharides/metabolism , Macrophages/cytology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/genetics
14.
Free Radic Res ; 43(2): 100-5, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19148843

ABSTRACT

All-trans-canthaxanthin (4, 4'-diketo beta-carotene) but not 9-cis-canthaxanthin has been shown to induce apoptosis in some cell lines. In this study apoptotic activity of 9-cis-canthaxanthin on THP-1 macrophage is reported. Comparison of apoptotic activities of the two canthaxanthin isomers on this cell line by annexin V-cy3 and TUNEL assays indicated the higher pro-apoptotic activity of 9-cis-isomer than the all-trans-isomer. Canthaxanthin-induced apoptosis in this cell line was found to be accompanied by increased caspase-3 and caspase-8 activities, indicating its progression via caspase cascade. Induction of both caspase activities was higher by 9-cis-canthaxanthin than that by trans-canthaxanthin. All these results suggest that canthaxanthin stereoisomers differentially induce apoptosis of THP-1 monocyte/macrophage.


Subject(s)
Apoptosis/drug effects , Canthaxanthin/pharmacology , Macrophages/drug effects , Antioxidants/pharmacology , Caspase 3/metabolism , Caspase 8/metabolism , Cell Line , Cells, Cultured , Humans , Isomerism , Macrophages/cytology , Macrophages/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...