Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Nat Commun ; 13(1): 6107, 2022 10 16.
Article in English | MEDLINE | ID: mdl-36245043

ABSTRACT

Acute myeloid leukemia (AML) is maintained by self-renewing leukemic stem cells (LSCs). A fundamental problem in treating AML is that conventional therapy fails to eliminate LSCs, which can reinitiate leukemia. Heat shock transcription factor 1 (HSF1), a central regulator of the stress response, has emerged as an important target in cancer therapy. Using genetic Hsf1 deletion and a direct HSF1 small molecule inhibitor, we show that HSF1 is specifically required for the maintenance of AML, while sparing steady-state and stressed hematopoiesis. Mechanistically, deletion of Hsf1 dysregulates multifaceted genes involved in LSC stemness and suppresses mitochondrial oxidative phosphorylation through downregulation of succinate dehydrogenase C (SDHC), a direct HSF1 target. Forced expression of SDHC largely restores the Hsf1 ablation-induced AML developmental defect. Importantly, the growth and engraftment of human AML cells are suppressed by HSF1 inhibition. Our data provide a rationale for developing efficacious small molecules to specifically target HSF1 in AML.


Subject(s)
Cell Self Renewal , Leukemia, Myeloid, Acute , Humans , Cell Self Renewal/genetics , Heat Shock Transcription Factors/genetics , Heat Shock Transcription Factors/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/metabolism , Succinate Dehydrogenase/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
2.
PLoS Pathog ; 18(6): e1010195, 2022 06.
Article in English | MEDLINE | ID: mdl-35737716

ABSTRACT

Copper homeostasis mechanisms are essential for microbial adaption to changing copper levels within the host during infection. In the opportunistic fungal pathogen Cryptococcus neoformans (Cn), the Cn Cbi1/Bim1 protein is a newly identified copper binding and release protein that is highly induced during copper limitation. Recent studies demonstrated that Cbi1 functions in copper uptake through the Ctr1 copper transporter during copper limitation. However, the mechanism of Cbi1 action is unknown. The fungal cell wall is a dynamic structure primarily composed of carbohydrate polymers, such as chitin and chitosan, polymers known to strongly bind copper ions. We demonstrated that Cbi1 depletion affects cell wall integrity and architecture, connecting copper homeostasis with adaptive changes within the fungal cell wall. The cbi1Δ mutant strain possesses an aberrant cell wall gene transcriptional signature as well as defects in chitin / chitosan deposition and exposure. Furthermore, using Cn strains defective in chitosan biosynthesis, we demonstrated that cell wall chitosan modulates the ability of the fungal cell to withstand copper stress. Given the previously described role for Cbi1 in copper uptake, we propose that this copper-binding protein could be involved in shuttling copper from the cell wall to the copper transporter Ctr1 for regulated microbial copper uptake.


Subject(s)
Chitosan , Cryptococcosis , Cryptococcus neoformans , Cell Wall/metabolism , Chitin/metabolism , Chitosan/metabolism , Copper/metabolism , Copper Transport Proteins , Cryptococcosis/microbiology , Cryptococcus neoformans/genetics , Cryptococcus neoformans/metabolism , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Homeostasis
3.
J Biol Chem ; 296: 100391, 2021.
Article in English | MEDLINE | ID: mdl-33567338

ABSTRACT

Cryptococcus neoformans is an opportunistic fungal pathogen whose pathogenic lifestyle is linked to its ability to cope with fluctuating levels of copper (Cu), an essential metal involved in multiple virulence mechanisms, within distinct host niches. During lethal cryptococcal meningitis in the brain, C. neoformans senses a Cu-deficient environment and is highly dependent on its ability to scavenge trace levels of Cu from its host and adapt to Cu scarcity to successfully colonize this niche. In this study, we demonstrate for this critical adaptation, the Cu-sensing transcription factor Cuf1 differentially regulates the expression of the SOD1 and SOD2 superoxide dismutases in novel ways. Genetic and transcriptional analysis reveals Cuf1 specifies 5'-truncations of the SOD1 and SOD2 mRNAs through specific binding to Cu responsive elements within their respective promoter regions. This results in Cuf1-dependent repression of the highly abundant SOD1 and simultaneously induces expression of two isoforms of SOD2, the canonical mitochondrial targeted isoform and a novel alternative cytosolic isoform, from a single alternative transcript produced specifically under Cu limitation. The generation of cytosolic Sod2 during Cu limitation is required to maintain cellular antioxidant defense against superoxide stress both in vitro and in vivo. Further, decoupling Cuf1 regulation of Sod2 localization compromises the ability of C. neoformans to colonize organs in murine models of cryptococcosis. Our results provide a link between transcription factor-mediated alteration of protein localization and cell proliferation under stress, which could impact tissue colonization by a fungal pathogen.


Subject(s)
Cryptococcus neoformans/enzymology , Fungal Proteins/metabolism , Superoxide Dismutase-1/metabolism , Superoxide Dismutase/metabolism , Transcription Factors/metabolism , Animals , Copper/metabolism , Cryptococcus neoformans/genetics , Cryptococcus neoformans/isolation & purification , Disease Models, Animal , Female , Fungal Proteins/genetics , Male , Mice , Protein Isoforms , Subcellular Fractions/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase-1/genetics
4.
J Biol Chem ; 296: 100097, 2021.
Article in English | MEDLINE | ID: mdl-33208463

ABSTRACT

Heat shock transcription factor 1 (HSF1) orchestrates cellular stress protection by activating or repressing gene transcription in response to protein misfolding, oncogenic cell proliferation, and other environmental stresses. HSF1 is tightly regulated via intramolecular repressive interactions, post-translational modifications, and protein-protein interactions. How these HSF1 regulatory protein interactions are altered in response to acute and chronic stress is largely unknown. To elucidate the profile of HSF1 protein interactions under normal growth and chronic and acutely stressful conditions, quantitative proteomics studies identified interacting proteins in the response to heat shock or in the presence of a poly-glutamine aggregation protein cell-based model of Huntington's disease. These studies identified distinct protein interaction partners of HSF1 as well as changes in the magnitude of shared interactions as a function of each stressful condition. Several novel HSF1-interacting proteins were identified that encompass a wide variety of cellular functions, including roles in DNA repair, mRNA processing, and regulation of RNA polymerase II. One HSF1 partner, CTCF, interacted with HSF1 in a stress-inducible manner and functions in repression of specific HSF1 target genes. Understanding how HSF1 regulates gene repression is a crucial question, given the dysregulation of HSF1 target genes in both cancer and neurodegeneration. These studies expand our understanding of HSF1-mediated gene repression and provide key insights into HSF1 regulation via protein-protein interactions.


Subject(s)
CCCTC-Binding Factor/metabolism , Gene Expression Regulation, Neoplastic , Heat Shock Transcription Factors/metabolism , Heat-Shock Response , Huntington Disease/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Animals , CCCTC-Binding Factor/genetics , HEK293 Cells , Heat Shock Transcription Factors/genetics , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Protein Interaction Maps
5.
Sci Transl Med ; 12(574)2020 12 16.
Article in English | MEDLINE | ID: mdl-33328331

ABSTRACT

Heat shock factor 1 (HSF1) is a cellular stress-protective transcription factor exploited by a wide range of cancers to drive proliferation, survival, invasion, and metastasis. Nuclear HSF1 abundance is a prognostic indicator for cancer severity, therapy resistance, and shortened patient survival. The HSF1 gene was amplified, and nuclear HSF1 abundance was markedly increased in prostate cancers and particularly in neuroendocrine prostate cancer (NEPC), for which there are no available treatment options. Despite genetic validation of HSF1 as a therapeutic target in a range of cancers, a direct and selective small-molecule HSF1 inhibitor has not been validated or developed for use in the clinic. We described the identification of a direct HSF1 inhibitor, Direct Targeted HSF1 InhiBitor (DTHIB), which physically engages HSF1 and selectively stimulates degradation of nuclear HSF1. DTHIB robustly inhibited the HSF1 cancer gene signature and prostate cancer cell proliferation. In addition, it potently attenuated tumor progression in four therapy-resistant prostate cancer animal models, including an NEPC model, where it caused profound tumor regression. This study reports the identification and validation of a direct HSF1 inhibitor and provides a path for the development of a small-molecule HSF1-targeted therapy for prostate cancers and other therapy-resistant cancers.


Subject(s)
Heat Shock Transcription Factors/antagonists & inhibitors , Prostatic Neoplasms , Animals , Cell Nucleus/metabolism , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics
6.
Sci Rep ; 10(1): 16369, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33004835

ABSTRACT

Lytic polysaccharide monooxygenase (LPMO) and copper binding protein CopC share a similar mononuclear copper site. This site is defined by an N-terminal histidine and a second internal histidine side chain in a configuration called the histidine brace. To understand better the determinants of reactivity, the biochemical and structural properties of a well-described cellulose-specific LPMO from Thermoascus aurantiacus (TaAA9A) is compared with that of CopC from Pseudomonas fluorescens (PfCopC) and with the LPMO-like protein Bim1 from Cryptococcus neoformans. PfCopC is not reduced by ascorbate but is a very strong Cu(II) chelator due to residues that interacts with the N-terminus. This first biochemical characterization of Bim1 shows that it is not redox active, but very sensitive to H2O2, which accelerates the release of Cu ions from the protein. TaAA9A oxidizes ascorbate at a rate similar to free copper but through a mechanism that produce fewer reactive oxygen species. These three biologically relevant examples emphasize the diversity in how the proteinaceous environment control reactivity of Cu with O2.


Subject(s)
Copper/metabolism , Histidine/metabolism , Models, Molecular , Oxygenases/metabolism , Escherichia coli , Hydrogen Peroxide/metabolism , Magnetic Resonance Spectroscopy/methods , Oxidation-Reduction
7.
Nat Chem Biol ; 16(3): 337-344, 2020 03.
Article in English | MEDLINE | ID: mdl-31932719

ABSTRACT

Infection by the fungal pathogen Cryptococcus neoformans causes lethal meningitis, primarily in immune-compromised individuals. Colonization of the brain by C. neoformans is dependent on copper (Cu) acquisition from the host, which drives critical virulence mechanisms. While C. neoformans Cu+ import and virulence are dependent on the Ctr1 and Ctr4 proteins, little is known concerning extracellular Cu ligands that participate in this process. We identified a C. neoformans gene, BIM1, that is strongly induced during Cu limitation and which encodes a protein related to lytic polysaccharide monooxygenases (LPMOs). Surprisingly, bim1 mutants are Cu deficient, and Bim1 function in Cu accumulation depends on Cu2+ coordination and cell-surface association via a glycophosphatidyl inositol anchor. Bim1 participates in Cu uptake in concert with Ctr1 and expression of this pathway drives brain colonization in mouse infection models. These studies demonstrate a role for LPMO-like proteins as a critical factor for Cu acquisition in fungal meningitis.


Subject(s)
Copper/metabolism , Cryptococcus neoformans/metabolism , Mixed Function Oxygenases/metabolism , Animals , Cryptococcosis/metabolism , Cryptococcus neoformans/pathogenicity , Disease Models, Animal , Female , Fungal Proteins/genetics , Fungal Proteins/metabolism , Meningitis/metabolism , Meningitis/physiopathology , Mice , Mice, Inbred A , Polysaccharides/metabolism , Virulence
8.
Trends Pharmacol Sci ; 40(12): 986-1005, 2019 12.
Article in English | MEDLINE | ID: mdl-31727393

ABSTRACT

The ability of cancer cells to cope with stressful conditions is critical for their survival, proliferation, and metastasis. The heat shock transcription factor 1 (HSF1) protects cells from stresses such as chemicals, radiation, and temperature. These properties of HSF1 are exploited by a broad spectrum of cancers, which exhibit high levels of nuclear, active HSF1. Functions for HSF1 in malignancy extend well beyond its central role in protein quality control. While HSF1 has been validated as a powerful target in cancers by genetic knockdown studies, HSF1 inhibitors reported to date have lacked sufficient specificity and potency for clinical evaluation. We review the roles of HSF1 in cancer, its potential as a prognostic indicator for cancer treatment, evaluate current HSF1 inhibitors and provide guidelines for the identification of selective HSF1 inhibitors as chemical probes and for clinical development.


Subject(s)
Antineoplastic Agents/pharmacology , Heat Shock Transcription Factors/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Heat Shock Transcription Factors/genetics , Heat Shock Transcription Factors/metabolism , Humans , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction/drug effects
9.
PLoS Biol ; 17(10): e3000104, 2019 10.
Article in English | MEDLINE | ID: mdl-31600193

ABSTRACT

The spliceosome is a large ribonucleoprotein complex that removes introns from pre-mRNAs. At its functional core lies the essential pre-mRNA processing factor 8 (Prp8) protein. Across diverse eukaryotes, this protein cofactor of RNA catalysis harbors a self-splicing element called an intein. Inteins in Prp8 are extremely pervasive and are found at 7 different sites in various species. Here, we focus on the Prp8 intein from Cryptococcus neoformans (Cne), a human fungal pathogen. We solved the crystal structure of this intein, revealing structural homology among protein splicing sequences in eukaryotes, including the Hedgehog C terminus. Working with the Cne Prp8 intein in a reporter assay, we find that the biologically relevant divalent metals copper and zinc inhibit intein splicing, albeit by 2 different mechanisms. Copper likely stimulates reversible modifications on a catalytically important cysteine, whereas zinc binds at the terminal asparagine and the same critical cysteine. Importantly, we also show that copper treatment inhibits Prp8 protein splicing in Cne. Lastly, an intein-containing Prp8 precursor model is presented, suggesting that metal-induced protein splicing inhibition would disturb function of both Prp8 and the spliceosome. These results indicate that Prp8 protein splicing can be modulated, with potential functional implications for the spliceosome.


Subject(s)
Cryptococcus neoformans/genetics , Fungal Proteins/genetics , RNA Splicing , RNA-Binding Proteins/genetics , Spliceosomes/metabolism , Asparagine/chemistry , Asparagine/metabolism , Binding Sites , Cloning, Molecular , Copper/chemistry , Copper/metabolism , Cryptococcus neoformans/metabolism , Crystallography, X-Ray , Cysteine/chemistry , Cysteine/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Gene Expression , Genes, Reporter , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Inteins , Maltose-Binding Proteins/genetics , Maltose-Binding Proteins/metabolism , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Spliceosomes/ultrastructure , Structural Homology, Protein , Zinc/chemistry , Zinc/metabolism
10.
Nat Commun ; 10(1): 1386, 2019 03 27.
Article in English | MEDLINE | ID: mdl-30918258

ABSTRACT

Copper (Cu) is an essential trace element for growth and development and abnormal Cu levels are associated with anemia, metabolic disease and cancer. Evolutionarily conserved from fungi to humans, the high-affinity Cu+ transporter Ctr1 is crucial for both dietary Cu uptake and peripheral distribution, yet the mechanisms for selective permeation of potentially toxic Cu+ ions across cell membranes are unknown. Here we present X-ray crystal structures of Ctr1 from Salmo salar in both Cu+-free and Cu+-bound states, revealing a homo-trimeric Cu+-selective ion channel-like architecture. Two layers of methionine triads form a selectivity filter, coordinating two bound Cu+ ions close to the extracellular entrance. These structures, together with Ctr1 functional characterization, provide a high resolution picture to understand Cu+ import across cellular membranes and suggest therapeutic opportunities for intervention in diseases characterized by inappropriate Cu accumulation.


Subject(s)
Cation Transport Proteins/ultrastructure , Copper/metabolism , Animals , Biological Transport , Cation Transport Proteins/metabolism , Cell Membrane , Copper Transporter 1 , Crystallography, X-Ray , Ion Transport , Salmo salar
11.
mBio ; 9(5)2018 09 18.
Article in English | MEDLINE | ID: mdl-30228242

ABSTRACT

Iron is an indispensable micronutrient for all eukaryotic organisms due to its participation as a redox cofactor in many metabolic pathways. Iron imbalance leads to the most frequent human nutritional deficiency in the world. Adaptation to iron limitation requires a global reorganization of the cellular metabolism directed to prioritize iron utilization for essential processes. In response to iron scarcity, the conserved Saccharomyces cerevisiae mRNA-binding protein Cth2, which belongs to the tristetraprolin family of tandem zinc finger proteins, coordinates a global remodeling of the cellular metabolism by promoting the degradation of multiple mRNAs encoding highly iron-consuming proteins. In this work, we identify a critical mechanism for the degradation of Cth2 protein during the adaptation to iron deficiency. Phosphorylation of a patch of Cth2 serine residues within its amino-terminal region facilitates recognition by the SCFGrr1 ubiquitin ligase complex, accelerating Cth2 turnover by the proteasome. When Cth2 degradation is impaired by either mutagenesis of the Cth2 serine residues or deletion of GRR1, the levels of Cth2 rise and abrogate growth in iron-depleted conditions. Finally, we uncover that the casein kinase Hrr25 phosphorylates and promotes Cth2 destabilization. These results reveal a sophisticated posttranslational regulatory pathway necessary for the adaptation to iron depletion.IMPORTANCE Iron is a vital element for many metabolic pathways, including the synthesis of DNA and proteins, and the generation of energy via oxidative phosphorylation. Therefore, living organisms have developed tightly controlled mechanisms to properly distribute iron, since imbalances lead to nutritional deficiencies, multiple diseases, and vulnerability against pathogens. Saccharomyces cerevisiae Cth2 is a conserved mRNA-binding protein that coordinates a global reprogramming of iron metabolism in response to iron deficiency in order to optimize its utilization. Here we report that the phosphorylation of Cth2 at specific serine residues is essential to regulate the stability of the protein and adaptation to iron depletion. We identify the kinase and ubiquitination machinery implicated in this process to establish a posttranscriptional regulatory model. These results and recent findings for both mammals and plants reinforce the privileged position of E3 ubiquitin ligases and phosphorylation events in the regulation of eukaryotic iron homeostasis.


Subject(s)
Adaptation, Physiological , Proteasome Endopeptidase Complex/metabolism , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/physiology , Tristetraprolin/metabolism , Gene Expression Regulation, Fungal , Iron/metabolism , Mutagenesis , Phosphorylation , Proteasome Endopeptidase Complex/genetics , Protein Processing, Post-Translational , Protein Stability , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Serine/genetics , Tristetraprolin/genetics
12.
J Biol Chem ; 293(40): 15497-15512, 2018 10 05.
Article in English | MEDLINE | ID: mdl-30131336

ABSTRACT

Acquisition of the trace element copper (Cu) is critical to drive essential eukaryotic processes such as oxidative phosphorylation, iron mobilization, peptide hormone biogenesis, and connective tissue maturation. The Ctr1/Ctr3 family of Cu importers, first discovered in fungi and conserved in mammals, are critical for Cu+ movement across the plasma membrane or mobilization from endosomal compartments. Whereas ablation of Ctr1 in mammals is embryonic lethal, and Ctr1 is critical for dietary Cu absorption, cardiac function, and systemic iron distribution, little is known about the intrinsic contribution of Ctr1 for Cu+ permeation through membranes or its mechanism of action. Here, we identify three members of a Cu+ importer family from the thermophilic fungus Chaetomium thermophilum: Ctr3a and Ctr3b, which function on the plasma membrane, and Ctr2, which likely functions in endosomal Cu mobilization. All three proteins drive Cu and isoelectronic silver (Ag) uptake in cells devoid of Cu+ importers. Transport activity depends on signature amino acid motifs that are conserved and essential for all Ctr1/3 transporters. Ctr3a is stable and amenable to purification and was incorporated into liposomes to reconstitute an in vitro Ag+ transport assay characterized by stopped-flow spectroscopy. Ctr3a has intrinsic high-affinity metal ion transport activity that closely reflects values determined in vivo, with slow turnover kinetics. Given structural models for mammalian Ctr1, Ctr3a likely functions as a low-efficiency Cu+ ion channel. The Ctr1/Ctr3 family may be tuned to import essential yet potentially toxic Cu+ ions at a slow rate to meet cellular needs, while minimizing labile intracellular Cu+ pools.


Subject(s)
Antiporters/metabolism , Cation Transport Proteins/metabolism , Cell Membrane/metabolism , Chaetomium/metabolism , Copper/metabolism , Fungal Proteins/metabolism , Amino Acid Sequence , Antiporters/genetics , Cation Transport Proteins/genetics , Cations, Divalent , Cations, Monovalent , Chaetomium/genetics , Fungal Proteins/genetics , Gene Expression , Genetic Complementation Test , Ion Transport , Kinetics , Plasmids/chemistry , Plasmids/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proteolipids/chemistry , Proteolipids/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Silver/metabolism , Transformation, Genetic
13.
Mol Microbiol ; 108(5): 473-494, 2018 06.
Article in English | MEDLINE | ID: mdl-29608794

ABSTRACT

The ability of the human fungal pathogen Cryptococcus neoformans to adapt to variable copper (Cu) environments within the host is key for successful dissemination and colonization. During pulmonary infection, host alveolar macrophages compartmentalize Cu into the phagosome and C. neoformans Cu-detoxifying metallothioneins, MT1 and MT2, are required for survival of the pathogen. In contrast, during brain colonization the C. neoformans Cu+ importers Ctr1 and Ctr4 are required for virulence. Central for the regulation and expression of both the Cu detoxifying MT1/2 and the Cu acquisition Ctr1/4 proteins is the Cu-metalloregulatory transcription factor Cuf1, an established C. neoformans virulence factor. Due to the importance of the distinct C. neoformans Cu homeostasis mechanisms during host colonization and virulence, and to the central role of Cuf1 in regulating Cu homeostasis, we performed a combination of RNA-Seq and ChIP-Seq experiments to identify differentially transcribed genes between conditions of high and low Cu. We demonstrate that the transcriptional regulation exerted by Cuf1 is intrinsically complex and that Cuf1 also functions as a transcriptional repressor. The Cu- and Cuf1-dependent regulon in C. neoformans reveals new adaptive mechanisms for Cu homeostasis in this pathogenic fungus and identifies potential new pathogen-specific targets for therapeutic intervention in fungal infections.


Subject(s)
Copper/metabolism , Cryptococcosis/microbiology , Cryptococcus neoformans/genetics , Fungal Proteins/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Cryptococcus neoformans/pathogenicity , Cryptococcus neoformans/physiology , Fungal Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation, Fungal/genetics , Genome-Wide Association Study , Humans , RNA, Fungal , Transcription Factors/genetics , Virulence/genetics
14.
Bioorg Med Chem ; 26(19): 5299-5306, 2018 10 15.
Article in English | MEDLINE | ID: mdl-29661622

ABSTRACT

Heat shock factor 1 (HSF1) is a stress-responsive transcription factor that regulates expression of protein chaperones and cell survival factors. In cancer, HSF1 plays a unique role, hijacking the normal stress response to drive a cancer-specific transcriptional program. These observations suggest that HSF1 inhibitors could be promising therapeutics. However, HSF1 is activated through a complex mechanism, which involves release of a negative regulatory domain, leucine zipper 4 (LZ4), from a masked oligomerization domain (LZ1-3), and subsequent binding of the oligomer to heat shock elements (HSEs) in HSF1-responsive genes. Recent crystal structures have suggested that HSF1 oligomers are held together by extensive, buried contact surfaces, making it unclear whether there are any possible binding sites for inhibitors. Here, we have rationally designed a series of peptide-based molecules based on the LZ4 and LZ1-3 motifs. Using a plate-based, fluorescence polarization (FP) assay, we identified a minimal region of LZ4 that suppresses binding of HSF1 to the HSE. Using this information, we converted this peptide into a tracer and used it to understand how binding of LZ4 to LZ1-3 suppresses HSF1 activation. Together, these results suggest a previously unexplored avenue in the development of HSF1 inhibitors. Furthermore, the findings highlight how native interactions can inspire the design of inhibitors for even the most challenging protein-protein interactions (PPIs).


Subject(s)
Drug Design , Heat Shock Transcription Factors/antagonists & inhibitors , Peptides/chemistry , Amino Acid Sequence , Binding Sites , Fluorescence Polarization , Heat Shock Transcription Factors/metabolism , Humans , Leucine Zippers , Peptides/chemical synthesis , Peptides/metabolism
15.
Nat Rev Mol Cell Biol ; 19(1): 4-19, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28852220

ABSTRACT

The heat shock transcription factors (HSFs) were discovered over 30 years ago as direct transcriptional activators of genes regulated by thermal stress, encoding heat shock proteins. The accepted paradigm posited that HSFs exclusively activate the expression of protein chaperones in response to conditions that cause protein misfolding by recognizing a simple promoter binding site referred to as a heat shock element. However, we now realize that the mammalian family of HSFs comprises proteins that independently or in concert drive combinatorial gene regulation events that activate or repress transcription in different contexts. Advances in our understanding of HSF structure, post-translational modifications and the breadth of HSF-regulated target genes have revealed exciting new mechanisms that modulate HSFs and shed new light on their roles in physiology and pathology. For example, the ability of HSF1 to protect cells from proteotoxicity and cell death is impaired in neurodegenerative diseases but can be exploited by cancer cells to support their growth, survival and metastasis. These new insights into HSF structure, function and regulation should facilitate the development tof new disease therapeutics to manipulate this transcription factor family.


Subject(s)
Gene Expression Regulation/genetics , Heat Shock Transcription Factors/genetics , Heat-Shock Proteins/genetics , Transcription, Genetic/genetics , Animals , Heat-Shock Response/genetics , Humans , Protein Processing, Post-Translational/genetics
16.
mBio ; 8(5)2017 10 31.
Article in English | MEDLINE | ID: mdl-29089435

ABSTRACT

Copper (Cu) ions serve as catalytic cofactors to drive key biochemical processes, and yet Cu levels that exceed cellular homeostatic control capacity are toxic. The underlying mechanisms for Cu toxicity are poorly understood. During pulmonary infection by the fungal pathogen Cryptococcus neoformans, host alveolar macrophages compartmentalize Cu to the phagosome, and the ability to detoxify Cu is critical for its survival and virulence. Here, we report that iron-sulfur (Fe-S) clusters are critical targets of Cu toxicity in both Saccharomyces cerevisiae and C. neoformans in a manner that depends on the accessibility of Cu to the Fe-S cofactor. To respond to this Cu-dependent Fe-S stress, C. neoformans induces the transcription of mitochondrial ABC transporter Atm1, which functions in cytosolic-nuclear Fe-S protein biogenesis in response to Cu and in a manner dependent on the Cu metalloregulatory transcription factor Cuf1. As Atm1 functions in exporting an Fe-S precursor from the mitochondrial matrix to the cytosol, C. neoformans cells depleted for Atm1 are sensitive to Cu even while the Cu-detoxifying metallothionein proteins are highly expressed. We provide evidence for a previously unrecognized microbial defense mechanism to deal with Cu toxicity, and we highlight the importance for C. neoformans of having several distinct mechanisms for coping with Cu toxicity which together could contribute to the success of this microbe as an opportunistic human fungal pathogen.IMPORTANCEC. neoformans is an opportunistic pathogen that causes lethal meningitis in over 650,000 people annually. The severity of C. neoformans infections is further compounded by the use of toxic or poorly effective systemic antifungal agents as well as by the difficulty of diagnosis. Cu is a natural potent antimicrobial agent that is compartmentalized within the macrophage phagosome and used by innate immune cells to neutralize microbial pathogens. While the Cu detoxification machinery of C. neoformans is essential for virulence, little is known about the mechanisms by which Cu kills fungi. Here we report that Fe-S cluster-containing proteins, including members of the Fe-S protein biogenesis machinery itself, are critical targets of Cu toxicity and therefore that this biosynthetic process provides an important layer of defense against high Cu levels. Given the role of Cu ionophores as antimicrobials, understanding how Cu is toxic to microorganisms could lead to the development of effective, broad-spectrum antimicrobials. Moreover, understanding Cu toxicity could provide additional insights into the pathophysiology of human diseases of Cu overload such as Wilson's disease.


Subject(s)
Copper/metabolism , Cryptococcus neoformans/drug effects , Cryptococcus neoformans/metabolism , Iron-Sulfur Proteins/metabolism , Stress, Physiological , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Cryptococcus neoformans/pathogenicity , Cytosol/metabolism , Gene Expression Regulation, Fungal , Host-Pathogen Interactions , Humans , Iron-Sulfur Proteins/genetics , Macrophages/immunology , Macrophages/microbiology , Metallothionein/genetics , Metallothionein/metabolism , Mice , Mitochondria/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Virulence
17.
Annu Rev Microbiol ; 71: 597-623, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28886682

ABSTRACT

Fungal cells colonize and proliferate in distinct niches, from soil and plants to diverse tissues in human hosts. Consequently, fungi are challenged with the goal of obtaining nutrients while simultaneously elaborating robust regulatory mechanisms to cope with a range of availability of nutrients, from scarcity to excess. Copper is essential for life but also potentially toxic. In this review we describe the sophisticated homeostatic mechanisms by which fungi acquire, utilize, and control this biochemically versatile trace element. Fungal pathogens, which can occupy distinct host tissues that have their own intrinsic requirements for copper homeostasis, have evolved mechanisms to acquire copper to successfully colonize the host, disseminate to other tissues, and combat host copper bombardment mechanisms that would otherwise mitigate virulence.


Subject(s)
Copper/metabolism , Fungi/metabolism , Trace Elements/metabolism , Homeostasis
18.
Neuroimage ; 155: 245-256, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28473286

ABSTRACT

The clinical use of Magnetic Resonance Imaging (MRI) and Positron Emission Tomography (PET) has proven to be a strong diagnostic tool in the field of neurology. The reliability of these methods to confirm clinical diagnoses has guided preclinical research to utilize these techniques for the characterization of animal disease models. Previously, we demonstrated that an endothelial cell-specific ablation of the murine Serum Response Factor (SrfiECKO) results in blood brain barrier (BBB) breakdown and hemorrhagic stroke. Taking advantage of this mouse model we here perform a comprehensive longitudinal, multiparametric and in vivo imaging approach to reveal pathophysiological processes occurring before and during the appearance of cerebral microbleeds using combined PET and MRI. We complement our imaging results with data regarding animal behavior and immunohistochemistry. Our results demonstrate diffusion abnormalities in the cortical brain tissue prior to the onset of cerebral microbleeds. Diffusion reductions were accompanied by significant increments of [18F]FAZA uptake before the onset of the lesions in T2WI. The Open Field behavioral tests revealed reduced activity of SrfiECKO animals, whereas histology confirmed the presence of hemorrhages in cortical regions of the mouse brain and iron deposition at lesion sites with increased hypoxia inducible factor 1α, CD31 and glial fibrillary acidic protein expression. For the first time, we performed a thorough evaluation of the prodromal period before the occurrence of spontaneous cerebral microbleeds. Using in vivo PET and MRI, we show the pathological tissue changes that occur previous to gross blood brain barrier (BBB) disruption and breakage. In addition, our results show that apparent diffusion coefficient (ADC) reduction may be an early biomarker of BBB disruption proposing an alternate clinical interpretation. Furthermore, our findings remark the usefulness of this novel SrfiECKO mouse model to study underlying mechanisms of hemorrhagic stroke.


Subject(s)
Blood-Brain Barrier/diagnostic imaging , Cerebral Cortex/diagnostic imaging , Intracranial Hemorrhages/diagnostic imaging , Magnetic Resonance Imaging/methods , Positron-Emission Tomography/methods , Prodromal Symptoms , Stroke/diagnosis , Animals , Disease Models, Animal , Male , Mice , Mice, Transgenic
19.
J Biol Chem ; 292(27): 11531-11546, 2017 07 07.
Article in English | MEDLINE | ID: mdl-28507097

ABSTRACT

Copper is an essential element for proper organismal development and is involved in a range of processes, including oxidative phosphorylation, neuropeptide biogenesis, and connective tissue maturation. The copper transporter (Ctr) family of integral membrane proteins is ubiquitously found in eukaryotes and mediates the high-affinity transport of Cu+ across both the plasma membrane and endomembranes. Although mammalian Ctr1 functions as a Cu+ transporter for Cu acquisition and is essential for embryonic development, a homologous protein, Ctr2, has been proposed to function as a low-affinity Cu transporter, a lysosomal Cu exporter, or a regulator of Ctr1 activity, but its functional and evolutionary relationship to Ctr1 is unclear. Here we report a biochemical, genetic, and phylogenetic comparison of metazoan Ctr1 and Ctr2, suggesting that Ctr2 arose over 550 million years ago as a result of a gene duplication event followed by loss of Cu+ transport activity. Using a random mutagenesis and growth selection approach, we identified amino acid substitutions in human and mouse Ctr2 proteins that support copper-dependent growth in yeast and enhance copper accumulation in Ctr1-/- mouse embryonic fibroblasts. These mutations revert Ctr2 to a more ancestral Ctr1-like state while maintaining endogenous functions, such as stimulating Ctr1 cleavage. We suggest key structural aspects of metazoan Ctr1 and Ctr2 that discriminate between their biological roles, providing mechanistic insights into the evolutionary, biochemical, and functional relationships between these two related proteins.


Subject(s)
Cation Transport Proteins , Copper/metabolism , Evolution, Molecular , Gene Duplication , Phylogeny , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Copper Transporter 1 , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Humans , Ion Transport/physiology , Mice , Mice, Knockout , SLC31 Proteins
20.
Nat Commun ; 8: 14405, 2017 02 13.
Article in English | MEDLINE | ID: mdl-28194040

ABSTRACT

Huntington's Disease (HD) is a neurodegenerative disease caused by poly-glutamine expansion in the Htt protein, resulting in Htt misfolding and cell death. Expression of the cellular protein folding and pro-survival machinery by heat shock transcription factor 1 (HSF1) ameliorates biochemical and neurobiological defects caused by protein misfolding. We report that HSF1 is degraded in cells and mice expressing mutant Htt, in medium spiny neurons derived from human HD iPSCs and in brain samples from patients with HD. Mutant Htt increases CK2α' kinase and Fbxw7 E3 ligase levels, phosphorylating HSF1 and promoting its proteasomal degradation. An HD mouse model heterozygous for CK2α' shows increased HSF1 and chaperone levels, maintenance of striatal excitatory synapses, clearance of Htt aggregates and preserves body mass compared with HD mice homozygous for CK2α'. These results reveal a pathway that could be modulated to prevent neuronal dysfunction and muscle wasting caused by protein misfolding in HD.


Subject(s)
Brain/metabolism , Heat Shock Transcription Factors/metabolism , Huntington Disease/metabolism , Neurons/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Female , HEK293 Cells , Heat Shock Transcription Factors/genetics , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , PC12 Cells , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...