Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
Add more filters

Publication year range
1.
Cell ; 171(2): 273-285, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28985560

ABSTRACT

Ferroptosis is a form of regulated cell death characterized by the iron-dependent accumulation of lipid hydroperoxides to lethal levels. Emerging evidence suggests that ferroptosis represents an ancient vulnerability caused by the incorporation of polyunsaturated fatty acids into cellular membranes, and cells have developed complex systems that exploit and defend against this vulnerability in different contexts. The sensitivity to ferroptosis is tightly linked to numerous biological processes, including amino acid, iron, and polyunsaturated fatty acid metabolism, and the biosynthesis of glutathione, phospholipids, NADPH, and coenzyme Q10. Ferroptosis has been implicated in the pathological cell death associated with degenerative diseases (i.e., Alzheimer's, Huntington's, and Parkinson's diseases), carcinogenesis, stroke, intracerebral hemorrhage, traumatic brain injury, ischemia-reperfusion injury, and kidney degeneration in mammals and is also implicated in heat stress in plants. Ferroptosis may also have a tumor-suppressor function that could be harnessed for cancer therapy. This Primer reviews the mechanisms underlying ferroptosis, highlights connections to other areas of biology and medicine, and recommends tools and guidelines for studying this emerging form of regulated cell death.


Subject(s)
Cell Death , Animals , Apoptosis , Humans , Iron/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism
2.
Genes Dev ; 35(7-8): 528-541, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33737385

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is one of the most lethal cancers worldwide and evolves often to lung metastasis. P53R175H (homologous to Trp53R172H in mice) is a common hot spot mutation. How metastasis is regulated by p53R175H in ESCC remains to be investigated. To investigate p53R175H-mediated molecular mechanisms, we used a carcinogen-induced approach in Trp53R172H/- mice to model ESCC. In the primary Trp53R172H/- tumor cell lines, we depleted Trp53R172H (shTrp53) and observed a marked reduction in cell invasion in vitro and lung metastasis burden in a tail-vein injection model in comparing isogenic cells (shCtrl). Furthermore, we performed bulk RNA-seq to compare gene expression profiles of metastatic and primary shCtrl and shTrp53 cells. We identified the YAP-BIRC5 axis as a potential mediator of Trp53R172H -mediated metastasis. We demonstrate that expression of Survivin, an antiapoptotic protein encoded by BIRC5, increases in the presence of Trp53R172H Furthermore, depletion of Survivin specifically decreases Trp53R172H-driven lung metastasis. Mechanistically, Trp53R172H but not wild-type Trp53, binds with YAP in ESCC cells, suggesting their cooperation to induce Survivin expression. Furthermore, Survivin high expression level is associated with increased metastasis in several GI cancers. Taken together, this study unravels new insights into how mutant p53 mediates metastasis.


Subject(s)
Lung Neoplasms/physiopathology , Survivin/genetics , Survivin/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Neoplastic/genetics , Lung Neoplasms/genetics , Mice , Mutation , Neoplasm Metastasis , Transcriptome , Tumor Suppressor Protein p53/metabolism
3.
Mol Cell ; 77(3): 633-644.e5, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31836388

ABSTRACT

Metastatic melanoma is an aggressive disease, despite recent improvements in therapy. Eradicating all melanoma cells even in drug-sensitive tumors is unsuccessful in patients because a subset of cells can transition to a slow-cycling state, rendering them resistant to most targeted therapy. It is still unclear what pathways define these subpopulations and promote this resistant phenotype. In the current study, we show that Wnt5A, a non-canonical Wnt ligand that drives a metastatic, therapy-resistant phenotype, stabilizes the half-life of p53 and uses p53 to initiate a slow-cycling state following stress (DNA damage, targeted therapy, and aging). Inhibiting p53 blocks the slow-cycling phenotype and sensitizes melanoma cells to BRAF/MEK inhibition. In vivo, this can be accomplished with a single dose of p53 inhibitor at the commencement of BRAF/MEK inhibitor therapy. These data suggest that taking the paradoxical approach of inhibiting rather than activating wild-type p53 may sensitize previously resistant metastatic melanoma cells to therapy.


Subject(s)
Melanoma/metabolism , Tumor Suppressor Protein p53/genetics , Wnt-5a Protein/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Humans , MAP Kinase Kinase Kinases/metabolism , Melanoma/genetics , Melanoma/pathology , Molecular Targeted Therapy , Mutation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction/drug effects , Sulfonamides/pharmacology , Tumor Microenvironment/drug effects , Tumor Suppressor Protein p53/physiology
4.
Nature ; 589(7843): 597-602, 2021 01.
Article in English | MEDLINE | ID: mdl-33361818

ABSTRACT

Isoprenoids are vital for all organisms, in which they maintain membrane stability and support core functions such as respiration1. IspH, an enzyme in the methyl erythritol phosphate pathway of isoprenoid synthesis, is essential for Gram-negative bacteria, mycobacteria and apicomplexans2,3. Its substrate, (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMBPP), is not produced in metazoans, and in humans and other primates it activates cytotoxic Vγ9Vδ2 T cells at extremely low concentrations4-6. Here we describe a class of IspH inhibitors and refine their potency to nanomolar levels through structure-guided analogue design. After modification of these compounds into prodrugs for delivery into bacteria, we show that they kill clinical isolates of several multidrug-resistant bacteria-including those from the genera Acinetobacter, Pseudomonas, Klebsiella, Enterobacter, Vibrio, Shigella, Salmonella, Yersinia, Mycobacterium and Bacillus-yet are relatively non-toxic to mammalian cells. Proteomic analysis reveals that bacteria treated with these prodrugs resemble those after conditional IspH knockdown. Notably, these prodrugs also induce the expansion and activation of human Vγ9Vδ2 T cells in a humanized mouse model of bacterial infection. The prodrugs we describe here synergize the direct killing of bacteria with a simultaneous rapid immune response by cytotoxic γδ T cells, which may limit the increase of antibiotic-resistant bacterial populations.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/immunology , Lymphocyte Activation/drug effects , Microbial Viability/drug effects , Oxidoreductases/antagonists & inhibitors , T-Lymphocytes, Cytotoxic/drug effects , Animals , Drug Resistance, Microbial , Drug Resistance, Multiple , Enzyme Inhibitors/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Female , Half-Life , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/microbiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microbial Sensitivity Tests , Molecular Docking Simulation , Oxidoreductases/deficiency , Oxidoreductases/genetics , Oxidoreductases/metabolism , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Substrate Specificity , Swine/blood , T-Lymphocytes, Cytotoxic/immunology
5.
Crit Rev Biochem Mol Biol ; 59(1-2): 128-138, 2024.
Article in English | MEDLINE | ID: mdl-38661126

ABSTRACT

TP53 encodes a transcription factor that is centrally-involved in several pathways, including the control of metabolism, the stress response, DNA repair, cell cycle arrest, senescence, programmed cell death, and others. Since the discovery of TP53 as the most frequently-mutated tumor suppressor gene in cancer over four decades ago, the field has focused on uncovering target genes of this transcription factor that are essential for tumor suppression. This search has been fraught with red herrings, however. Dozens of p53 target genes were discovered that had logical roles in tumor suppression, but subsequent data showed that most were not tumor suppressive, and were dispensable for p53-mediated tumor suppression. In this review, we focus on p53 transcriptional targets in two categories: (1) canonical targets like CDKN1A (p21) and BBC3 (PUMA), which clearly play critical roles in p53-mediated cell cycle arrest/senescence and cell death, but which are not mutated in cancer, and for which knockout mice fail to develop spontaneous tumors; and (2) a smaller category of recently-described p53 target genes that are mutated in human cancer, and which appear to be critical for tumor suppression by p53. Interestingly, many of these genes encode proteins that control broad cellular pathways, like splicing and protein degradation, and several of them encode proteins that feed back to regulate p53. These include ZMAT3, GLS2, PADI4, ZBXW7, RFX7, and BTG2. The findings from these studies provide a more complex, but exciting, potential framework for understanding the role of p53 in tumor suppression.


Subject(s)
Neoplasms , Tumor Suppressor Protein p53 , Humans , Animals , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Genes, Tumor Suppressor , Gene Expression Regulation, Neoplastic
6.
Proc Natl Acad Sci U S A ; 120(7): e2212940120, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36749725

ABSTRACT

Missense mutations that inactivate p53 occur commonly in cancer, and germline mutations in TP53 cause Li Fraumeni syndrome, which is associated with early-onset cancer. In addition, there are over two hundred germline missense variants of p53 that remain uncharacterized. In some cases, these germline variants have been shown to encode lesser-functioning, or hypomorphic, p53 protein, and these alleles are associated with increased cancer risk in humans and mouse models. However, most hypomorphic p53 variants remain un- or mis-classified in clinical genetics databases. There thus exists a significant need to better understand the behavior of p53 hypomorphs and to develop a functional assay that can distinguish hypomorphs from wild-type p53 or benign variants. We report the surprising finding that two different African-centric genetic hypomorphs of p53 that occur in distinct functional domains of the protein share common activities. Specifically, the Pro47Ser variant, located in the transactivation domain, and the Tyr107His variant, located in the DNA binding domain, both share increased propensity to misfold into a conformation specific for mutant, misfolded p53. Additionally, cells and tissues containing these hypomorphic variants show increased NF-κB activity. We identify a common gene expression signature from unstressed lymphocyte cell lines that is shared between multiple germline hypomorphic variants of TP53, and which successfully distinguishes wild-type p53 and a benign variant from lesser-functioning hypomorphic p53 variants. Our findings will allow us to better understand the contribution of p53 hypomorphs to disease risk and should help better inform cancer risk in the carriers of p53 variants.


Subject(s)
Li-Fraumeni Syndrome , Tumor Suppressor Protein p53 , Animals , Mice , Humans , Tumor Suppressor Protein p53/metabolism , Genetic Predisposition to Disease , Li-Fraumeni Syndrome/genetics , Genes, p53 , Heterozygote , Germ-Line Mutation
7.
Genes Dev ; 32(3-4): 230-243, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29463573

ABSTRACT

Mutant forms of p53 protein often possess protumorigenic functions, conferring increased survival and migration to tumor cells via their "gain-of-function" activity. Whether and how a common polymorphism in TP53 at amino acid 72 (Pro72Arg; referred to here as P72 and R72) impacts this gain of function has not been determined. We show that mutant p53 enhances migration and metastasis of tumors through the ability to bind and regulate PGC-1α and that this regulation is markedly impacted by the codon 72 polymorphism. Tumor cells with the R72 variant of mutant p53 show increased PGC-1α function along with greatly increased mitochondrial function and metastatic capability. Breast cancers containing mutant p53 and the R72 variant show poorer prognosis compared with P72. The combined results reveal PGC-1α as a novel "gain-of-function" partner of mutant p53 and indicate that the codon 72 polymorphism influences the impact of mutant p53 on metabolism and metastasis.


Subject(s)
Genes, p53 , Mutation , Neoplasms/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Movement , Female , Hepatocyte Nuclear Factor 4/metabolism , Humans , Male , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/pathology , Oxidative Phosphorylation , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/antagonists & inhibitors
8.
Nature ; 569(7754): 73-78, 2019 05.
Article in English | MEDLINE | ID: mdl-30996346

ABSTRACT

Polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) are pathologically activated neutrophils that are crucial for the regulation of immune responses in cancer. These cells contribute to the failure of cancer therapies and are associated with poor clinical outcomes. Despite recent advances in the understanding of PMN-MDSC biology, the mechanisms responsible for the pathological activation of neutrophils are not well defined, and this limits the selective targeting of these cells. Here we report that mouse and human PMN-MDSCs exclusively upregulate fatty acid transport protein 2 (FATP2). Overexpression of FATP2 in PMN-MDSCs was controlled by granulocyte-macrophage colony-stimulating factor, through the activation of the STAT5 transcription factor. Deletion of FATP2 abrogated the suppressive activity of PMN-MDSCs. The main mechanism of FATP2-mediated suppressive activity involved the uptake of arachidonic acid and the synthesis of prostaglandin E2. The selective pharmacological inhibition of FATP2 abrogated the activity of PMN-MDSCs and substantially delayed tumour progression. In combination with checkpoint inhibitors, FATP2 inhibition blocked tumour progression in mice. Thus, FATP2 mediates the acquisition of immunosuppressive activity by PMN-MDSCs and represents a target to inhibit the functions of PMN-MDSCs selectively and to improve the efficiency of cancer therapy.


Subject(s)
Fatty Acid Transport Proteins/metabolism , Fatty Acids/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neutrophils/metabolism , Aged , Animals , Arachidonic Acid/metabolism , Dinoprostone/metabolism , Fatty Acid Transport Proteins/antagonists & inhibitors , Female , Humans , Lipid Metabolism , Lipids , Male , Mice , Middle Aged , Neutrophils/pathology , STAT5 Transcription Factor/metabolism
9.
Genes Dev ; 30(8): 918-30, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27034505

ABSTRACT

A nonsynonymous single-nucleotide polymorphism at codon 47 in TP53 exists in African-descent populations (P47S, rs1800371; referred to here as S47). Here we report that, in human cell lines and a mouse model, the S47 variant exhibits a modest decrease in apoptosis in response to most genotoxic stresses compared with wild-type p53 but exhibits a significant defect in cell death induced by cisplatin. We show that, compared with wild-type p53, S47 has nearly indistinguishable transcriptional function but shows impaired ability to transactivate a subset of p53 target genes, including two involved in metabolism:Gls2(glutaminase 2) and Sco2 We also show that human and mouse cells expressing the S47 variant are markedly resistant to cell death by agents that induce ferroptosis (iron-mediated nonapoptotic cell death). We show that mice expressing S47 in homozygous or heterozygous form are susceptible to spontaneous cancers of diverse histological types. Our data suggest that the S47 variant may contribute to increased cancer risk in individuals of African descent, and our findings highlight the need to assess the contribution of this variant to cancer risk in these populations. These data also confirm the potential relevance of metabolism and ferroptosis to tumor suppression by p53.


Subject(s)
Genes, p53/genetics , Polymorphism, Single Nucleotide , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Black People/genetics , Carcinoma, Hepatocellular/genetics , Cell Death/drug effects , Cell Death/genetics , Cell Line , Cisplatin/pharmacology , Codon/chemistry , Codon/genetics , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Neoplasms/genetics , Protein Binding/genetics , Risk Factors , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
10.
J Biol Chem ; 298(12): 102637, 2022 12.
Article in English | MEDLINE | ID: mdl-36309086

ABSTRACT

The tumor suppressor protein p53 suppresses cancer by regulating processes such as apoptosis, cell cycle arrest, senescence, and ferroptosis, which is an iron-mediated and lipid peroxide-induced cell death pathway. Whereas numerous p53 target genes have been identified, only a few appear to be critical for the suppression of tumor growth. Additionally, while ferroptosis is clearly implicated in tumor suppression by p53, few p53 target genes with roles in ferroptosis have been identified. We have previously studied germline missense p53 variants that are hypomorphic or display reduced activity. These hypomorphic variants are associated with increased risk for cancer, but they retain the majority of p53 transcriptional function; as such, study of the transcriptional targets of these hypomorphs has the potential to reveal the identity of other genes important for p53-mediated tumor suppression. Here, using RNA-seq in lymphoblastoid cell lines, we identify PLTP (phospholipid transfer protein) as a p53 target gene that shows impaired transactivation by three different cancer-associated p53 hypomorphs: P47S (Pro47Ser, rs1800371), Y107H (Tyr107His, rs368771578), and G334R (Gly334Arg, rs78378222). We show that enforced expression of PLTP potently suppresses colony formation in human tumor cell lines. We also demonstrate that PLTP regulates the sensitivity of cells to ferroptosis. Taken together, our findings reveal PLTP to be a p53 target gene that is extremely sensitive to p53 transcriptional function and which has roles in growth suppression and ferroptosis.


Subject(s)
Ferroptosis , Neoplasms , Phospholipid Transfer Proteins , Humans , Apoptosis , Cell Death/genetics , Cell Line, Tumor , Neoplasms/genetics , Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Phospholipid Transfer Proteins/metabolism
12.
Proc Natl Acad Sci U S A ; 117(43): 26804-26811, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33055209

ABSTRACT

The p53 tumor suppressor protein is a transcription factor and master stress response mediator, and it is subject to reduction-oxidation (redox)-dependent regulation. The P47S variant of TP53, which exists primarily in African-descent populations, associates with an elevated abundance of low molecular weight (LMW) thiols, including glutathione (GSH) and coenzyme A (CoA). Here we show that S47 and P47 cells exhibit distinct metabolic profiles, controlled by their different redox states and expression of Activating Transcription Factor-4 (ATF4). We find that S47 cells exhibit decreased catabolic glycolysis but increased use of the pentose phosphate pathway (PPP), and an enhanced abundance of the antioxidant, NADPH. We identify ATF4 as differentially expressed in P47 and S47 cells and show that ATF4 can reverse the redox status and rescue metabolism of S47 cells, as well as increase sensitivity to ferroptosis. This adaptive metabolic switch is rapid, reversible, and accompanied by thiol-mediated changes in the structures and activities of key glycolytic signaling pathway proteins, including GAPDH and G6PD. The results presented here unveil the important functional interplay among pathways regulating thiol-redox status, metabolic adaptation, and cellular responses to oxidative stress.


Subject(s)
Activating Transcription Factor 4/metabolism , Ferroptosis , Genes, p53 , Oxidation-Reduction , Sulfhydryl Compounds/metabolism , Animals , Cell Line , Coenzyme A/metabolism , Glutathione/metabolism , Glycolysis , Homeostasis , Humans , Male , Mice , Protein Processing, Post-Translational
14.
Proc Natl Acad Sci U S A ; 116(17): 8390-8396, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30962386

ABSTRACT

A population-restricted single-nucleotide coding region polymorphism (SNP) at codon 47 exists in the human TP53 gene (P47S, hereafter P47 and S47). In studies aimed at identifying functional differences between these variants, we found that the African-specific S47 variant associates with an impaired response to agents that induce the oxidative stress-dependent, nonapoptotic cell death process of ferroptosis. This phenotype is manifested as a greater resistance to glutamate-induced cytotoxicity in cultured cells as well as increased carbon tetrachloride-mediated liver damage in a mouse model. The differential ferroptotic responses associate with intracellular antioxidant differences between P47 and S47 cells, including elevated abundance of the low molecular weight thiols coenzyme A (CoA) and glutathione in S47 cells. Importantly, the disparate ferroptosis phenotypes related to the P47S polymorphism are reversible. Exogenous administration of CoA provides protection against ferroptosis in cultured mouse and human cells, as well as in a mouse model. The combined data support a positive role for p53 in ferroptosis and identify CoA as a regulator of this cell death process. Together, these findings provide mechanistic insight linking redox regulation of p53 to small molecule antioxidants and stress signaling pathways. They also identify potential therapeutic approaches to redox-related pathologies.


Subject(s)
Ferroptosis/physiology , Tumor Suppressor Protein p53 , Animals , Carbon Tetrachloride/toxicity , Cells, Cultured , Coenzyme A/metabolism , Disease Models, Animal , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice , Oxidation-Reduction , Polymorphism, Single Nucleotide/genetics , Polymorphism, Single Nucleotide/physiology , Sulfhydryl Compounds/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
15.
Nurs Adm Q ; 45(3): 219-225, 2021.
Article in English | MEDLINE | ID: mdl-34060505

ABSTRACT

The COVID-19 pandemic has overwhelmed communities. Physical, emotional, and financial struggles have heightened, especially with our vulnerable populations. People have been afraid to return to their provider's office. For children, there has been an interruption of well-visits and immunizations. As the nation saw a decline in immunization uptake, a pilot nurse-led program was designed to increase vaccinations and address the social determinant needs during a global pandemic. The purpose of this article is to describe the planning and implementation of a curbside immunization event. The Logic model was used as a framework to ensure an efficient and replicable process. Initial observations showed an overall increase in immunization uptake and 97% of participants current with recommended vaccinations. Most parents (93%) would attend again and recommend it to others. They also felt that infection control precautions helped make the care delivered safe and efficient. Social determinants of health were assessed and addressed. This method of vaccine delivery is a viable model going into the future. Others may replicate this model, and it may also serve as a platform regarding flu or COVID-19 vaccine distribution.


Subject(s)
Immunization/nursing , Models, Nursing , Social Determinants of Health/statistics & numerical data , Humans , Immunization/statistics & numerical data , Immunization Programs/methods , Immunization Programs/standards , Immunization Programs/statistics & numerical data , Michigan , Pilot Projects
16.
EMBO J ; 35(2): 193-207, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26658110

ABSTRACT

Telomeres and tumor suppressor protein TP53 (p53) function in genome protection, but a direct role of p53 at telomeres has not yet been described. Here, we have identified non-canonical p53-binding sites within the human subtelomeres that suppress the accumulation of DNA damage at telomeric repeat DNA. These non-canonical subtelomeric p53-binding sites conferred transcription enhancer-like functions that include an increase in local histone H3K9 and H3K27 acetylation and stimulation of subtelomeric transcripts, including telomere repeat-containing RNA (TERRA). p53 suppressed formation of telomere-associated γH2AX and prevented telomere DNA degradation in response to DNA damage stress. Our findings indicate that p53 provides a direct chromatin-associated protection to human telomeres, as well as other fragile genomic sites. We propose that p53-associated chromatin modifications enhance local DNA repair or protection to provide a previously unrecognized tumor suppressor function of p53.


Subject(s)
Carrier Proteins/metabolism , DNA Damage/genetics , Telomere/metabolism , Tumor Suppressor Protein p53/metabolism , Carrier Proteins/genetics , HCT116 Cells , Humans , Protein Binding , Telomere/genetics , Tumor Suppressor Protein p53/genetics
17.
Mol Cell ; 36(1): 15-27, 2009 Oct 09.
Article in English | MEDLINE | ID: mdl-19818706

ABSTRACT

The multifunctional, stress-inducible molecular chaperone HSP70 has important roles in aiding protein folding and maintaining protein homeostasis. HSP70 expression is elevated in many cancers, contributing to tumor cell survival and resistance to therapy. We have determined that a small molecule called 2-phenylethynesulfonamide (PES) interacts selectively with HSP70 and leads to a disruption of the association between HSP70 and several of its cochaperones and substrate proteins. Treatment of cultured tumor cells with PES promotes cell death that is associated with protein aggregation, impaired autophagy, and inhibition of lysosomal function. Moreover, this small molecule is able to suppress tumor development and enhance survival in a mouse model of Myc-induced lymphomagenesis. The data demonstrate that PES disrupts actions of HSP70 in multiple cell signaling pathways, offering an opportunity to better understand the diverse functions of this molecular chaperone and also to aid in the development of new cancer therapies.


Subject(s)
HSP70 Heat-Shock Proteins/antagonists & inhibitors , Sulfonamides/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptotic Protease-Activating Factor 1/metabolism , Autophagy/drug effects , Caspases/metabolism , Cathepsin L/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , DNA-Binding Proteins/metabolism , HSP40 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Lymphoma/pathology , Lymphoma/prevention & control , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Mice, Inbred Strains , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , NF-kappa B/metabolism , Protein Binding/drug effects , Protein Binding/physiology , Protein Interaction Domains and Motifs , Protein Multimerization/drug effects , Sequestosome-1 Protein , Sulfonamides/metabolism , Sulfonamides/therapeutic use , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism
18.
Public Health Nutr ; 20(18): 3304-3315, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28879832

ABSTRACT

OBJECTIVE: To investigate dietary intake, BMI and supermarket access at varying geographic scales and transport modes across areas of socio-economic disadvantage, and to evaluate the implementation of an urban planning policy that provides guidance on spatial access to supermarkets. DESIGN: Cross-sectional study used generalised estimating equations to investigate associations between supermarket density and proximity, vegetable and fruit intake and BMI at five geographic scales representing distances people travel to purchase food by varying transport modes. A stratified analysis by area-level disadvantage was conducted to detect optimal distances to supermarkets across socio-economic areas. Spatial distribution of supermarket and transport access was analysed using a geographic information system. SETTING: Melbourne, Australia. SUBJECTS: Adults (n 3128) from twelve local government areas (LGA) across Melbourne. RESULTS: Supermarket access was protective of BMI for participants in high disadvantaged areas within 800 m (P=0·040) and 1000 m (P=0·032) road network buffers around the household but not for participants in less disadvantaged areas. In urban growth area LGA, only 26 % of dwellings were within 1 km of a supermarket, far less than 80-90 % of dwellings suggested in the local urban planning policy. Low public transport access compounded disadvantage. CONCLUSIONS: Rapid urbanisation is a global health challenge linked to increases in dietary risk factors and BMI. Our findings highlight the importance of identifying the most appropriate geographic scale to inform urban planning policy for optimal health outcomes across socio-economic strata. Urban planning policy implementation in disadvantaged areas within cities has potential for reducing health inequities.


Subject(s)
Body Mass Index , City Planning , Environment Design , Socioeconomic Factors , Urban Population , Australia , Cross-Sectional Studies , Diet , Female , Fruit , Geographic Information Systems , Humans , Male , Middle Aged , Residence Characteristics , Risk Factors , Transportation , Urbanization , Vegetables
19.
Bioorg Med Chem Lett ; 26(20): 4884-4887, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27650927

ABSTRACT

Dimeric ß-carbolines are cytotoxic against multiple NSCLC cell lines, and we report herein our preliminary studies on the mechanism of action of these dimeric structures. Dimeric ß-carboline 1, which is more potent than the corresponding monomer in NSCLC cell lines, is a lysosomotropic agent that inhibits autophagy and mediates cell death by apoptosis, upregulating the pro-apoptotic BH3-only protein PUMA (p53 upregulated modulator of apoptosis) in a dose dependent manner.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/physiology , Carbolines/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Proto-Oncogene Proteins/physiology , Apoptosis/drug effects , Carbolines/chemistry , Cell Line, Tumor , Dimerization , Humans
20.
Tetrahedron Lett ; 56(23): 3515-3517, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-26257442

ABSTRACT

The design, synthesis and biological evaluation (anticancer and antimalarial activity) of bis-ß-carbolines, based on the structure of the naturally occurring alkaloid neokauluamine, is described.

SELECTION OF CITATIONS
SEARCH DETAIL