Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Nat Genet ; 33(3): 357-65, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12567188

ABSTRACT

The gene TP53, encoding p53, has a common sequence polymorphism that results in either proline or arginine at amino-acid position 72. This polymorphism occurs in the proline-rich domain of p53, which is necessary for the protein to fully induce apoptosis. We found that in cell lines containing inducible versions of alleles encoding the Pro72 and Arg72 variants, and in cells with endogenous p53, the Arg72 variant induces apoptosis markedly better than does the Pro72 variant. Our data indicate that at least one source of this enhanced apoptotic potential is the greater ability of the Arg72 variant to localize to the mitochondria; this localization is accompanied by release of cytochrome c into the cytosol. These data indicate that the two polymorphic variants of p53 are functionally distinct, and these differences may influence cancer risk or treatment.


Subject(s)
Apoptosis/genetics , Genes, p53 , Polymorphism, Genetic , Apoptosis/drug effects , Arginine/genetics , Cell Line , Chaperonin 60/metabolism , Codon/genetics , Fatty Acids, Unsaturated/pharmacology , Genetic Variation , HSP70 Heat-Shock Proteins/metabolism , Humans , Membrane Proteins/metabolism , Mitochondria/metabolism , Neoplasms/etiology , Neoplasms/genetics , Proline/genetics , Suppression, Genetic , Transcriptional Activation , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin/chemistry
7.
ACS Chem Biol ; 17(5): 1038-1044, 2022 05 20.
Article in English | MEDLINE | ID: mdl-35420772

ABSTRACT

Melanoma is a serious health challenge. Ferroptosis is a regulated form of oxidative cell death that shows varied efficacy in melanoma. We aimed to better understand the molecular basis for this differential ferroptosis sensitivity. We find that elevated expression of ErbB3 (V-Erb-B2 Avian Erythroblastic Leukemia Viral Oncogene Homologue 3) associates with ferroptosis resistance and that ErbB3 knockdown sensitizes to ferroptosis inducers. ErbB3 depletion also promotes a marked reduction in the cellular ratio of GSH/GSSG (reduced/oxidized glutathione) and that of NADPH/NADP+ (reduced/oxidized nicotinamide adenine dinucleotide phosphate), together with an increase in the abundance of the lipid peroxidation product malondialdehyde (MDA). We identify several small molecule inhibitors targeting ErbB3 signaling pathways that also reduce the NADPH/NADP+ and GSH/GSSG ratios, concomitantly sensitizing the melanomas to ferroptosis activators. These findings point to a previously unrecognized role of ErbB3 in ferroptosis sensitivity and provide new insight into pathways that regulate this cell death process.


Subject(s)
Ferroptosis , Melanoma , Skin Neoplasms , Glutathione Disulfide/metabolism , Humans , Melanoma/drug therapy , NADP/metabolism , Receptor, ErbB-3 , Melanoma, Cutaneous Malignant
8.
Nat Cell Biol ; 6(5): 443-50, 2004 May.
Article in English | MEDLINE | ID: mdl-15077116

ABSTRACT

The tumour suppressor activity of the p53 protein has been explained by its ability to induce apoptosis in response to a variety of cellular stresses. Thus, understanding the mechanism by which p53 functions in the execution of cell death pathways is of considerable importance in cancer biology. Recent studies have indicated that p53 has a direct signalling role at mitochondria in the induction of apoptosis, although the mechanisms involved are not completely understood. Here we show that, after cell stress, p53 interacts with the pro-apoptotic mitochondrial membrane protein Bak. Interaction of p53 with Bak causes oligomerization of Bak and release of cytochrome c from mitochondria. Notably, we show that formation of the p53-Bak complex coincides with loss of an interaction between Bak and the anti-apoptotic Bcl2-family member Mcl1. These results are consistent with a model in which p53 and Mcl1 have opposing effects on mitochondrial apoptosis by interacting with, and modulating the activity of, the death effector Bak.


Subject(s)
Apoptosis/physiology , Membrane Proteins/metabolism , Mitochondria/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2 , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Humans , Macromolecular Substances , Membrane Proteins/genetics , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/genetics , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/genetics , bcl-2 Homologous Antagonist-Killer Protein
9.
Mol Cell Oncol ; 8(2): 1877076, 2021.
Article in English | MEDLINE | ID: mdl-33860080

ABSTRACT

The tumor protein P53 (TP53, or p53) has complex and at times seemingly contradictory roles in the regulation of metabolism and ferroptosis sensitivity. We find that the actions of p53 influence the redox state, which can trigger changes in redox-sensitive proteins, thereby modifying metabolic processes and response to ferroptosis.

10.
Cancer Res Commun ; 1(1): 17-29, 2021 10.
Article in English | MEDLINE | ID: mdl-35187538

ABSTRACT

NRAS-mutant melanoma is currently a challenge to treat. This is due to an absence of inhibitors directed against mutant NRAS, along with adaptive and acquired resistance of this tumor type to inhibitors in the MAPK pathway. Inhibitors to MEK (mitogen-activated protein kinase kinase) have shown some promise for NRAS-mutant melanoma. In this work we explored the use of MEK inhibitors for NRAS-mutant melanoma. At the same time we investigated the impact of the brain microenvironment, specifically astrocytes, on the response of a melanoma brain metastatic cell line to MEK inhibition. These parallel avenues led to the surprising finding that astrocytes enhance the sensitivity of melanoma tumors to MEK inhibitors (MEKi). We show that MEKi cause an upregulation of the transcription factor ID3, which confers resistance. This upregulation of ID3 is blocked by conditioned media from astrocytes. We show that silencing ID3 enhances the sensitivity of melanoma to MEK inhibitors, thus mimicking the effect of the brain microenvironment. Moreover, we report that ID3 is a client protein of the chaperone HSP70, and that HSP70 inhibition causes ID3 to misfold and accumulate in a detergent-insoluble fraction in cells. We show that HSP70 inhibitors synergize with MEK inhibitors against NRAS-mutant melanoma, and that this combination significantly enhances the survival of mice in two different models of NRAS-mutant melanoma. These studies highlight ID3 as a mediator of adaptive resistance, and support the combined use of MEK and HSP70 inhibitors for the therapy of NRAS-mutant melanoma. SIGNIFICANCE: MEK inhibitors are currently used for NRAS-mutant melanoma, but have shown modest efficacy as single agents. This research shows a synergistic effect of combining HSP70 inhibitors with MEK inhibitors for the treatment of NRAS mutant melanoma.


Subject(s)
Melanoma , Mitogen-Activated Protein Kinase Kinases , Mice , Animals , GTP Phosphohydrolases/genetics , Membrane Proteins/genetics , Mutation , Melanoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Tumor Microenvironment
12.
Nat Commun ; 11(1): 473, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980600

ABSTRACT

A variant at amino acid 47 in human TP53 exists predominantly in individuals of African descent. P47S human and mouse cells show increased cancer risk due to defective ferroptosis. Here, we show that this ferroptotic defect causes iron accumulation in P47S macrophages. This high iron content alters macrophage cytokine profiles, leads to higher arginase level and activity, and decreased nitric oxide synthase activity. This leads to more productive intracellular bacterial infections but is protective against malarial toxin hemozoin. Proteomics of macrophages reveal decreased liver X receptor (LXR) activation, inflammation and antibacterial defense in P47S macrophages. Both iron chelators and LXR agonists improve the response of P47S mice to bacterial infection. African Americans with elevated saturated transferrin and serum ferritin show higher prevalence of the P47S variant (OR = 1.68 (95%CI 1.07-2.65) p = 0.023), suggestive of its role in iron accumulation in humans. This altered macrophage phenotype may confer an advantage in malaria-endemic sub-Saharan Africa.


Subject(s)
Iron/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Africa South of the Sahara , Black or African American/genetics , Animals , Bacterial Infections/etiology , Bacterial Infections/genetics , Bacterial Infections/metabolism , Ferritins/blood , Ferroptosis/drug effects , Ferroptosis/genetics , Ferroptosis/physiology , Genetic Variation , Hemeproteins/toxicity , Humans , Listeriosis/etiology , Liver X Receptors/agonists , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Malaria/genetics , Malaria/metabolism , Mice , Mice, Transgenic , Transferrin/metabolism
13.
Elife ; 92020 11 10.
Article in English | MEDLINE | ID: mdl-33170774

ABSTRACT

The Pro47Ser variant of p53 (S47) exists in African-descent populations and is associated with increased cancer risk in humans and mice. Due to impaired repression of the cystine importer Slc7a11, S47 cells show increased glutathione (GSH) accumulation compared to cells with wild -type p53. We show that mice containing the S47 variant display increased mTOR activity and oxidative metabolism, as well as larger size, improved metabolic efficiency, and signs of superior fitness. Mechanistically, we show that mTOR and its positive regulator Rheb display increased association in S47 cells; this is due to an altered redox state of GAPDH in S47 cells that inhibits its ability to bind and sequester Rheb. Compounds that decrease glutathione normalize GAPDH-Rheb complexes and mTOR activity in S47 cells. This study reveals a novel layer of regulation of mTOR by p53, and raises the possibility that this variant may have been selected for in early Africa.


Subject(s)
TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/genetics , Amino Acid Substitution/genetics , Animals , Black People/genetics , Cell Line , Glutathione/metabolism , Glycolysis , Humans , Mitochondria/metabolism , Oxidation-Reduction , TOR Serine-Threonine Kinases/genetics , Tumor Suppressor Protein p53/metabolism
14.
Cancer Res ; 80(23): 5270-5281, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33023943

ABSTRACT

The protein chaperone HSP70 is overexpressed in many cancers including colorectal cancer, where overexpression is associated with poor survival. We report here the creation of a uniquely acting HSP70 inhibitor (HSP70i) that targets multiple compartments in the cancer cell, including mitochondria. This inhibitor was mitochondria toxic and cytotoxic to colorectal cancer cells, but not to normal colon epithelial cells. Inhibition of HSP70 was efficacious as a single agent in primary and metastatic models of colorectal cancer and enabled identification of novel mitochondrial client proteins for HSP70. In a syngeneic colorectal cancer model, the inhibitor increased immune cell recruitment into tumors. Cells treated with the inhibitor secreted danger-associated molecular patterns (DAMP), including ATP and HMGB1, and functioned effectively as a tumor vaccine. Interestingly, the unique properties of this HSP70i in the disruption of mitochondrial function and the inhibition of proteostasis both contributed to DAMP release. This HSP70i constitutes a promising therapeutic opportunity in colorectal cancer and may exhibit antitumor activity against other tumor types. SIGNIFICANCE: These findings describe a novel HSP70i that disrupts mitochondrial proteostasis, demonstrating single-agent efficacy that induces immunogenic cell death in treated tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Mitochondria/drug effects , Adenosine Triphosphate/metabolism , Alarmins/metabolism , Animals , Cell-Free System , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , HMGB1 Protein/metabolism , HT29 Cells , High-Throughput Screening Assays , Humans , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Mitochondria/metabolism , Xenograft Model Antitumor Assays
15.
Cancer Res ; 78(19): 5694-5705, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30115697

ABSTRACT

The tumor suppressor TP53 is the most frequently mutated gene in human cancer and serves to restrict tumor initiation and progression. Single-nucleotide polymorphisms (SNP) in TP53 and p53 pathway genes can have a marked impact on p53 tumor suppressor function, and some have been associated with increased cancer risk and impaired response to therapy. Approximately 6% of Africans and 1% of African Americans express a p53 allele with a serine instead of proline at position 47 (Pro47Ser). This SNP impairs p53-mediated apoptosis in response to radiation and genotoxic agents and is associated with increased cancer risk in humans and in a mouse model. In this study, we compared the ability of wild-type (WT) and S47 p53 to suppress tumor development and respond to therapy. Our goal was to find therapeutic compounds that are more, not less, efficacious in S47 tumors. We identified the superior efficacy of two agents, cisplatin and BET inhibitors, on S47 tumors compared with WT. Cisplatin caused dramatic decreases in the progression of S47 tumors by activating the p53/PIN1 axis to drive the mitochondrial cell death program. These findings serve as important proof of principle that chemotherapy can be tailored to p53 genotype.Significance: A rare African-derived radioresistant p53 SNP provides proof of principle that chemotherapy can be tailored to TP53 genotype. Cancer Res; 78(19); 5694-705. ©2018 AACR.


Subject(s)
Polymorphism, Single Nucleotide , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Africa , Black or African American/genetics , Alleles , Animals , Apoptosis , Black People/genetics , Cell Death/drug effects , Cell Line, Tumor , Cell Transformation, Neoplastic , Cisplatin/pharmacology , Disease Progression , Fibroblasts/metabolism , Genotype , Humans , Mice , Mitochondria/metabolism , Mutation/drug effects , Neoplasm Transplantation , Pharmacogenetics , Precision Medicine , Risk
16.
Oncotarget ; 8(28): 45656-45669, 2017 Jul 11.
Article in English | MEDLINE | ID: mdl-28484090

ABSTRACT

Protein quality control is an important component of survival for all cells. The use of proteasome inhibitors for cancer therapy derives from the fact that tumor cells generally exhibit greater levels of proteotoxic stress than do normal cells, and thus cancer cells tend to be more sensitive to proteasome inhibition. However, this approach has been limited in some cases by toxicity to normal cells. Recently, the concept of inhibiting proteostasis in organelles for cancer therapy has been advanced, in part because it is predicted to have reduced toxicity for normal cells. Here we demonstrate that a fraction of the major stress-induced chaperone HSP70 (also called HSPA1A or HSP72, but hereafter HSP70) is abundantly present in mitochondria of tumor cells, but is expressed at quite low or undetectable levels in mitochondria of most normal tissues and non-tumor cell lines. We show that treatment of tumor cells with HSP70 inhibitors causes a marked change in mitochondrial protein quality control, loss of mitochondrial membrane potential, reduced oxygen consumption rate, and loss of ATP production. We identify several nuclear-encoded mitochondrial proteins, including polyadenylate binding protein-1 (PABPC1), which exhibit decreased abundance in mitochondria following treatment with HSP70 inhibitors. We also show that targeting HSP70 function leads to reduced levels of several mitochondrial-encoded RNA species that encode components of the electron transport chain. Our data indicate that small molecule inhibitors of HSP70 represent a new class of organelle proteostasis inhibitors that impair mitochondrial function in cancer cells, and therefore constitute novel therapeutics.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Proteasome Inhibitors/pharmacology , Proteostasis/drug effects , Stress, Physiological , Adenosine Triphosphate/metabolism , Cell Line, Tumor , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/mortality , Poly(A)-Binding Protein I/metabolism , Prognosis , Protein Binding
17.
Cell Rep ; 14(10): 2413-25, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26947067

ABSTRACT

p53 is well known for its tumor suppressor role, but this protein also has a poorly understood role in the regulation of metabolism. Human studies have implicated a common polymorphism at codon 72 of p53 in diabetic and pre-diabetic phenotypes. To understand this role, we utilized a humanized mouse model of the p53 codon 72 variants and monitored these mice following challenge with a high-fat diet (HFD). Mice with the arginine 72 (R72) variant of p53 developed more-severe obesity and glucose intolerance on a HFD, compared to mice with the proline 72 variant (P72). R72 mice developed insulin resistance, islet hypertrophy, increased infiltration of immune cells, and fatty liver disease. Gene expression analyses and studies with small-molecule inhibitors indicate that the p53 target genes Tnf and Npc1l1 underlie this phenotype. These results shed light on the role of p53 in obesity, metabolism, and inflammation.


Subject(s)
Genetic Predisposition to Disease , Obesity/genetics , Polymorphism, Single Nucleotide , Tumor Suppressor Protein p53/genetics , Animals , Body Weight/genetics , Diet, High-Fat , Glucose Tolerance Test , Humans , Insulin Resistance , Liver/metabolism , Liver/pathology , Male , Membrane Proteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Models, Animal , Obesity/physiopathology , Pancreas/metabolism , Pancreas/pathology , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism
18.
Cancer Res ; 76(9): 2720-30, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26984758

ABSTRACT

The stress-inducible chaperone protein HSP70 (HSPA1) is implicated in melanoma development, and HSP70 inhibitors exert tumor-specific cytotoxic activity in cancer. In this study, we documented that a significant proportion of melanoma tumors express high levels of HSP70, particularly at advanced stages, and that phospho-FAK (PTK2) and BRAF are HSP70 client proteins. Treatment of melanoma cells with HSP70 inhibitors decreased levels of phospho-FAK along with impaired migration, invasion, and metastasis in vitro and in vivo Moreover, the HSP70 inhibitor PET-16 reduced levels of mutant BRAF, synergized with the BRAF inhibitor PLX4032 in vitro, and enhanced the durability of response to BRAF inhibition in vivo Collectively, these findings provide strong support for HSP70 inhibition as a therapeutic strategy in melanoma, especially as an adjuvant approach for overcoming the resistance to BRAF inhibitors frequently observed in melanoma patients. Cancer Res; 76(9); 2720-30. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Focal Adhesion Kinase 1/metabolism , HSP70 Heat-Shock Proteins/metabolism , Indoles/pharmacology , Melanoma/pathology , Sulfonamides/pharmacology , Animals , Blotting, Western , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness/pathology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Tissue Array Analysis , Vemurafenib
19.
ACS Chem Biol ; 9(11): 2508-16, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25148104

ABSTRACT

The stress-inducible mammalian heat shock protein 70 (HSP70) and its bacterial orthologue DnaK are highly conserved nucleotide binding molecular chaperones. They represent critical regulators of cellular proteostasis, especially during conditions of enhanced stress. Cancer cells rely on HSP70 for survival, and this chaperone represents an attractive new therapeutic target. We have used a structure-activity approach and biophysical methods to characterize a class of inhibitors that bind to a unique allosteric site within the C-terminus of HSP70 and DnaK. Data from X-ray crystallography together with isothermal titration calorimetry, mutagenesis, and cell-based assays indicate that these inhibitors bind to a previously unappreciated allosteric pocket formed within the non-ATP-bound protein state. Moreover, binding of inhibitor alters the local protein conformation, resulting in reduced chaperone-client interactions and impairment of proteostasis. Our findings thereby provide a new chemical scaffold and target platform for both HSP70 and DnaK; these will be important tools with which to interrogate chaperone function and to aid ongoing efforts to optimize potency and efficacy in developing modulators of these chaperones for therapeutic use.


Subject(s)
Escherichia coli Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Allosteric Site , Crystallography, X-Ray , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/metabolism , Protein Conformation
20.
PLoS One ; 9(7): e103518, 2014.
Article in English | MEDLINE | ID: mdl-25058147

ABSTRACT

The HSP70 family of molecular chaperones function to maintain protein quality control and homeostasis. The major stress-induced form, HSP70 (also called HSP72 or HSPA1A) is considered an important anti-cancer drug target because it is constitutively overexpressed in a number of human cancers and promotes cancer cell survival. All HSP70 family members contain two functional domains: an N-terminal nucleotide binding domain (NBD) and a C-terminal protein substrate-binding domain (SBD); the latter is subdivided into SBDα and SBDß subdomains. The NBD and SBD structures of the bacterial ortholog, DnaK, have been characterized, but only the isolated NBD and SBDα segments of eukaryotic HSP70 proteins have been determined. Here we report the crystal structure of the substrate-bound human HSP70-SBD to 2 angstrom resolution. The overall fold of this SBD is similar to the corresponding domain in the substrate-bound DnaK structures, confirming a similar overall architecture of the orthologous bacterial and human HSP70 proteins. However, conformational differences are observed in the peptide-HSP70-SBD complex, particularly in the loop L(α, ß) that bridges SBDα to SBDß, and the loop L(L,1) that connects the SBD and NBD. The interaction between the SBDα and SBDß subdomains and the mode of substrate recognition is also different between DnaK and HSP70. This suggests that differences may exist in how different HSP70 proteins recognize their respective substrates. The high-resolution structure of the substrate-bound-HSP70-SBD complex provides a molecular platform for the rational design of small molecule compounds that preferentially target this C-terminal domain, in order to modulate human HSP70 function.


Subject(s)
Crystallography, X-Ray/methods , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/metabolism , Peptides/metabolism , Adenosine Triphosphatases/chemistry , Bacterial Proteins/chemistry , Binding Sites , Humans , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary
SELECTION OF CITATIONS
SEARCH DETAIL