Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2693: 81-94, 2023.
Article in English | MEDLINE | ID: mdl-37540428

ABSTRACT

Mammalian heat shock factor HSF1 transcriptional activity is controlled by a multitude of phosphorylations that occur under physiological conditions or following exposure of cells to a variety of stresses. One set of HSF1 phosphorylation is on serine 303 and serine 307 (S303/S307). These HSF1 phosphorylation sites are known to repress its transcriptional activity. Here, we describe a knock-in mouse model where these two serine residues were replaced by alanine residues and have determined the impact of these mutations on cellular proliferation and drug resistance. Our previous study using this mouse model indicated the susceptibility of the mutant mice to become obese with age due to an increase in basal levels of heat shock proteins (HSPs) and chronic inflammation. Since HSF1 transcriptional activity is increased in many tumor types, this mouse model may be a useful tool for studies related to cellular transformation and cancer.


Subject(s)
DNA-Binding Proteins , Transcription Factors , Mice , Animals , Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Heat Shock Transcription Factors/genetics , Heat Shock Transcription Factors/metabolism , Phosphorylation , Drug Resistance , Cell Proliferation , Serine/metabolism , Mammals/metabolism
2.
Cell Mol Life Sci ; 79(4): 198, 2022 Mar 21.
Article in English | MEDLINE | ID: mdl-35313355

ABSTRACT

The dual specificity protein phosphatases (Dusps) control dephosphorylation of mitogen-activated protein kinases (MAPKs) as well as other substrates. Here, we report that Dusp26, which is highly expressed in neuroblastoma cells and primary neurons is targeted to the mitochondrial outer membrane via its NH2-terminal mitochondrial targeting sequence. Loss of Dusp26 has a significant impact on mitochondrial function that is associated with increased levels of reactive oxygen species (ROS), reduction in ATP generation, reduction in mitochondria motility and release of mitochondrial HtrA2 protease into the cytoplasm. The mitochondrial dysregulation in dusp26-deficient neuroblastoma cells leads to the inhibition of cell proliferation and cell death. In vivo, Dusp26 is highly expressed in neurons in different brain regions, including cortex and midbrain (MB). Ablation of Dusp26 in mouse model leads to dopaminergic (DA) neuronal cell loss in the substantia nigra par compacta (SNpc), inflammatory response in MB and striatum, and phenotypes that are normally associated with Neurodegenerative diseases. Consistent with the data from our mouse model, Dusp26 expressing cells are significantly reduced in the SNpc of Parkinson's Disease patients. The underlying mechanism of DA neuronal death is that loss of Dusp26 in neurons increases mitochondrial ROS and concurrent activation of MAPK/p38 signaling pathway and inflammatory response. Our results suggest that regulation of mitochondrial-associated protein phosphorylation is essential for the maintenance of mitochondrial homeostasis and dysregulation of this process may contribute to the initiation and development of neurodegenerative diseases.


Subject(s)
Dopaminergic Neurons/physiology , Dual-Specificity Phosphatases/physiology , Mitochondria/metabolism , Mitogen-Activated Protein Kinase Phosphatases/physiology , Animals , Cell Death/genetics , Cell Respiration/genetics , Cells, Cultured , Cytoprotection/genetics , HEK293 Cells , Humans , Male , Mice , Mice, 129 Strain , Mice, Knockout , Mitochondria/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Oxidative Stress/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology
3.
Mol Cancer Res ; 18(3): 463-476, 2020 03.
Article in English | MEDLINE | ID: mdl-31744878

ABSTRACT

Deregulated oncogenic signaling linked to PI3K/AKT and mTORC1 pathway activation is a hallmark of human T-cell acute leukemia (T-ALL) pathogenesis and contributes to leukemic cell resistance and adverse prognosis. Notably, although the multiagent chemotherapy of leukemia leads to a high rate of complete remission, options for salvage therapy for relapsed/refractory disease are limited due to the serious side effects of augmenting cytotoxic chemotherapy. We report that ablation of HSF1, a key transcriptional regulator of the chaperone response and cellular bioenergetics, from mouse T-ALL tumors driven by PTEN loss or human T-ALL cell lines, has significant therapeutic effects in reducing tumor burden and sensitizing malignant cell death. From a mechanistic perspective, the enhanced sensitivity of T-ALLs to HSF1 depletion resides in the reduced MAPK-ERK signaling and metabolic and ATP-producing capacity of malignant cells lacking HSF1 activity. Impaired mitochondrial ATP production and decreased intracellular amino acid content in HSF1-deficient T-ALL cells trigger an energy-saving adaptive response featured by attenuation of the mTORC1 activity, which is coregulated by ATP, and its downstream target proteins (p70S6K and 4E-BP). This leads to protein translation attenuation that diminishes oncogenic signals and malignant cell growth. Collectively, these metabolic alterations in the absence of HSF1 activity reveal cancer cell liabilities and have a profound negative impact on T-ALL progression. IMPLICATIONS: Targeting HSF1 and HSF1-dependent cancer-specific anabolic and protein homeostasis programs has a significant therapeutic potential for T-ALL and may prevent progression of relapsed/refractory disease.


Subject(s)
Heat Shock Transcription Factors/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Animals , Cell Line, Tumor , Disease Progression , Energy Metabolism , Female , Humans , Male , Mice , Signal Transduction
4.
Mol Cell Biol ; 39(9)2019 05 01.
Article in English | MEDLINE | ID: mdl-30745413

ABSTRACT

Delineating the mechanisms that drive hepatic injury and hepatocellular carcinoma (HCC) progression is critical for development of novel treatments for recurrent and advanced HCC but also for the development of diagnostic and preventive strategies. Heat shock protein 70 (HSP70) acts in concert with several cochaperones and nucleotide exchange factors and plays an essential role in protein quality control that increases survival by protecting cells against environmental stressors. Specifically, the HSP70-mediated response has been implicated in the pathogenesis of cancer, but the specific mechanisms by which HSP70 may support malignant cell transformation remains to be fully elucidated. Here, we show that genetic ablation of HSP70 markedly impairs HCC initiation and progression by distinct but overlapping pathways. This includes the potentiation of the carcinogen-induced DNA damage response, at the tumor initiation stage, to increase the p53-dependent surveillance response leading to the cell cycle exit or death of genomically damaged differentiated pericentral hepatocytes, and this may also prevent their conversion into more proliferating HCC progenitor cells. Subsequently, activation of a mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) negative feedback pathway diminishes oncogenic signals, thereby attenuating premalignant cell transformation and tumor progression. Modulation of HSP70 function may be a strategy for interfering with oncogenic signals driving liver cell transformation and tumor progression, thus providing an opportunity for human cancer control.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/genetics , Diethylnitrosamine/adverse effects , HSP70 Heat-Shock Proteins/genetics , Liver Neoplasms/pathology , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , DNA Damage , Disease Progression , Gene Knockdown Techniques , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , MAP Kinase Signaling System , Male , Mice , Tumor Suppressor Protein p53/metabolism
5.
Mol Cell Biol ; 38(18)2018 09 15.
Article in English | MEDLINE | ID: mdl-29941492

ABSTRACT

Activation of the adaptive response to cellular stress orchestrated by heat shock factor 1 (HSF1), which is an evolutionarily conserved transcriptional regulator of chaperone response and cellular bioenergetics in diverse model systems, is a central feature of organismal defense from environmental and cellular stress. HSF1 activity, induced by proteostatic, metabolic, and growth factor signals, is regulated by posttranscriptional modifications, yet the mechanisms that regulate HSF1 and particularly the functional significance of these modifications in modulating its biological activity in vivo remain unknown. HSF1 phosphorylation at both Ser303 (S303) and Ser307 (S307) has been shown to repress HSF1 transcriptional activity under normal physiological growth conditions. To determine the biological relevance of these HSF1 phosphorylation events, we generated a knock-in mouse model in which S303 and S307 were replaced with alanine (HSF1303A/307A). Our results confirmed that loss of phosphorylation in HSF1303A/307A cells and tissues increases protein stability but also markedly sensitizes HSF1 activation under normal and heat- or nutrient-induced stress conditions. Interestingly, the enhanced HSF1 activation in HSF1303A/307A mice activates a supportive metabolic program that aggravates the development of age-dependent obesity, fatty liver diseases, and insulin resistance. Thus, these findings highlight the importance of a posttranslational mechanism (through phosphorylation at S303 and S307 sites) of regulation of the HSF1-mediated transcriptional program that moderates the severity of nutrient-induced metabolic diseases.


Subject(s)
Heat Shock Transcription Factors/genetics , Heat Shock Transcription Factors/metabolism , Aging/genetics , Aging/metabolism , Amino Acid Substitution , Animals , Cells, Cultured , Disease Models, Animal , Female , Gene Knock-In Techniques , Heat Shock Transcription Factors/chemistry , Heat-Shock Response , Humans , Insulin Resistance/genetics , Insulin Resistance/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/genetics , Obesity/metabolism , Phosphorylation , Protein Stability , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/chemistry
6.
Methods Mol Biol ; 1709: 1-22, 2018.
Article in English | MEDLINE | ID: mdl-29177647

ABSTRACT

Heat shock transcription factors (Hsfs) regulate transcription of heat shock proteins as well as other genes whose promoters contain heat shock elements (HSEs). There are at least five Hsfs in mammalian cells, Hsf1, Hsf2, Hsf3, Hsf4, and Hsfy (Wu, Annu Rev Cell Dev Biol 11:441-469, 1995; Morimoto, Genes Dev 12:3788-3796, 1998; Tessari et al., Mol Hum Repord 4:253-258, 2004; Fujimoto et al., Mol Biol Cell 21:106-116, 2010; Nakai et al., Mol Cell Biol 17:469-481, 1997; Sarge et al., Genes Dev 5:1902-1911, 1991). To understand the physiological roles of Hsf1, Hsf2, and Hsf4 in vivo, we generated knockout mouse lines for these factors (Zhang et al., J Cell Biochem 86:376-393, 2002; Wang et al., Genesis 36:48-61, 2003; Min et al., Genesis 40:205-217, 2004). Numbers of other laboratories have also generated Hsf1 (Xiao et al., EMBO J 18:5943-5952, 1999; Sugahara et al., Hear Res 182:88-96, 2003), Hsf2 (McMillan et al., Mol Cell Biol 22:8005-8014, 2002; Kallio et al., EMBO J 21:2591-2601, 2002), and Hsf4 (Fujimoto et al., EMBO J 23:4297-4306, 2004) knockout mouse models. In this chapter, we describe the design of the targeting vectors, the plasmids used, and the successful generation of mice lacking the individual genes. We also briefly describe what we have learned about the physiological functions of these genes in vivo.


Subject(s)
Gene Deletion , Gene Knockout Techniques/methods , Heat Shock Transcription Factors/genetics , Heat-Shock Proteins/genetics , Transcription Factors/genetics , Animals , Genetic Vectors , Male , Mice , Mice, Knockout
7.
J Biol Chem ; 2017 Jul 19.
Article in English | MEDLINE | ID: mdl-28724629

ABSTRACT

This article has been withdrawn by the authors. During preparation of this manuscript, a number of errors occurred in the preparation/assembly of Figs 2D, 2E, S1C, S1E, and S4. The authors apologize for not acknowledging that Fig. 6E and 6J represented the same samples and therefore the ß-actin immunoblot was reused. These presentation errors do not impact the underlying scientific findings of the article and the article is being withdrawn so that a corrected manuscript can be submitted for publication. We are sorry for any problems or issues that this may have caused the scientific community.

8.
J Cell Biol ; 216(3): 723-741, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28183717

ABSTRACT

Metabolic energy reprogramming facilitates adaptations to a variety of stress conditions and cellular dysfunction, but how the energetic demands are monitored and met in response to physiological stimuli remains elusive. Our data support a model demonstrating that heat shock factor 1 (HSF1), a master transcriptional regulator of the chaperone response, has been coopted from its role as a critical protein quality-control regulator to having a central role in systemic energy sensing and for metabolic adaptation to nutrient availability. We found that in the absence of HSF1, levels of NAD+ and ATP are not efficiently sustained in hepatic cells, largely because of transcriptional repression of nicotinamide phosphoribosyltransferase in the NAD+ salvage pathway. Mechanistically, the defect in NAD+ and ATP synthesis linked to a loss of NAD+-dependent deacetylase activity, increased protein acetylation, and impaired mitochondrial integrity. Remarkably, the drop in ATP level caused by HSF1 loss invoked an adaptive response featuring the inhibition of energetically demanding processes, including gluconeogenesis, translation, and lipid synthesis. Our work identifies HSF1 as a central regulator of cellular bioenergetics and protein homeostasis that benefits malignant cell progression and exacerbates development of metabolic diseases.


Subject(s)
DNA-Binding Proteins/genetics , Energy Metabolism/genetics , Homeostasis/genetics , Liver/physiology , Molecular Chaperones/genetics , Transcription Factors/genetics , Transcription, Genetic/genetics , Adenosine Triphosphate/metabolism , Animals , Gene Expression Regulation/genetics , Heat Shock Transcription Factors , Heat-Shock Proteins/genetics , Heat-Shock Response/genetics , Liver/metabolism , Mice , Mice, Inbred C57BL , NAD/metabolism , Protein Processing, Post-Translational/genetics
9.
J Neurochem ; 130(5): 626-41, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24903326

ABSTRACT

Traumatic brain injury (TBI) induces severe harm and disability in many accident victims and combat-related activities. The heat-shock proteins Hsp70/Hsp110 protect cells against death and ischemic damage. In this study, we used mice deficient in Hsp110 or Hsp70 to examine their potential requirement following TBI. Data indicate that loss of Hsp110 or Hsp70 increases brain injury and death of neurons. One of the mechanisms underlying the increased cell death observed in the absence of Hsp110 and Hsp70 following TBI is the increased expression of reactive oxygen species-induced p53 target genes Pig1, Pig8, and Pig12. To examine whether drugs that increase the levels of Hsp70/Hsp110 can protect cells against TBI, we subjected mice to TBI and administered Celastrol or BGP-15. In contrast to Hsp110- or Hsp70i-deficient mice that were not protected following TBI and Celastrol treatment, there was a significant improvement of wild-type mice following administration of these drugs during the first week following TBI. In addition, assessment of neurological injury shows significant improvement in contextual and cued fear conditioning tests and beam balance in wild-type mice that were treated with Celastrol or BGP-15 following TBI compared to TBI-treated mice. These studies indicate a significant role of Hsp70/Hsp110 in neuronal survival following TBI and the beneficial effects of Hsp70/Hsp110 inducers toward reducing the pathological consequences of TBI. Our data indicate that loss of Hsp110 or Hsp70 in mice increases brain injury following TBI. (a) One of the mechanisms underlying the increased cell death observed in the absence of these Hsps following TBI is the increased expression of ROS-induced p53 target genes known as Pigs. In addition, (b) using drugs (Celastrol or BGP-15) to increase Hsp70/Hsp110 levels protect cells against TBI, suggesting the beneficial effects of Hsp70/Hsp110 inducers to reduce the pathological consequences of TBI.


Subject(s)
Brain Injuries/metabolism , HSP110 Heat-Shock Proteins/metabolism , HSP72 Heat-Shock Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiplex Polymerase Chain Reaction , Oligonucleotide Array Sequence Analysis , Oximes/pharmacology , Pentacyclic Triterpenes , Piperidines/pharmacology , Reactive Oxygen Species/metabolism , Triterpenes/pharmacology
10.
Dev Biol ; 386(2): 448-60, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24380799

ABSTRACT

Heat shock factor binding protein 1 (HSBP1) is a 76 amino acid polypeptide that contains two arrays of hydrophobic heptad repeats and was originally identified through its interaction with the oligomerization domain of heat shock factor 1 (Hsf1), suppressing Hsf1's transcriptional activity following stress. To examine the function of HSBP1 in vivo, we generated mice with targeted disruption of the hsbp1 gene and examined zebrafish embryos treated with HSBP1-specific morpholino oligonucleotides. Our results show that hsbp1 is critical for preimplantation embryonic development. Embryonic stem (ES) cells deficient in hsbp1 survive and proliferate normally into the neural lineage in vitro; however, lack of hsbp1 in embryoid bodies (EBs) leads to disorganization of the germ layers and a reduction in the endoderm-specific markers (such as α-fetoprotein). We further show that hsbp1-deficient mouse EBs and knockdown of HSBP1 in zebrafish leads to an increase in the expression of the neural crest inducers Snail2, Tfap2α and Foxd3, suggesting a potential role for HSBP1 in the Wnt pathway. The hsbp1-deficient ES cells, EBs and zebrafish embryos with reduced HSBP1 levels exhibit elevated levels of Hsf1 activity and expression of heat shock proteins (Hsps). We conclude that HSBP1 plays an essential role during early mouse and zebrafish embryonic development.


Subject(s)
Embryonic Development/physiology , Endoderm/embryology , Gene Expression Regulation, Developmental/physiology , Heat-Shock Proteins/metabolism , Neoplasm Proteins/metabolism , Neural Crest/embryology , Animals , Blotting, Western , DNA-Binding Proteins/metabolism , Embryoid Bodies/metabolism , Gene Expression Regulation, Developmental/genetics , Genotype , Heat Shock Transcription Factors , Heat-Shock Proteins/genetics , Immunohistochemistry , In Situ Hybridization , Mice , Molecular Chaperones , Morpholinos/genetics , Neoplasm Proteins/genetics , Real-Time Polymerase Chain Reaction , Transcription Factors/metabolism , Wnt Signaling Pathway/genetics , Zebrafish , alpha-Fetoproteins/metabolism
11.
J Biol Chem ; 287(42): 35646-35657, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-22847003

ABSTRACT

ErbB2/Neu oncogene is overexpressed in 25% of invasive/metastatic breast cancers. We have found that deletion of heat shock factor Hsf1 in mice overexpressing ErbB2/Neu significantly reduces mammary tumorigenesis and metastasis. Hsf1(+/-)ErbB2/Neu(+) tumors exhibit reduced cellular proliferative and invasive properties associated with reduced activated ERK1/2 and reduced epithelial-mesenchymal transition (EMT). Hsf1(+/+)Neu(+) mammary epithelial cells exposed to TGFß show high levels of ERK1/2 activity and EMT; this is associated with reduced expression of E-cadherin and increased expression of Slug and vimentin, a mesenchymal marker. In contrast, Hsf1(-/-)Neu(+) or Hsf1(+/+)Neu(+) cells do not exhibit activated ERK1/2 and show reduced EMT in the presence of TGFß. The ineffective activation of the RAS/RAF/MEK/ERK1/2 signaling pathway in cells with reduced levels of HSF1 is due to the low levels of HSP90 in complex with RAF1 that are required for RAF1 stability and maturation. These results indicate a powerful inhibitory effect conferred by HSF1 downstream target genes in the inhibition of ErbB2-induced breast cancers in the absence of the Hsf1 gene.


Subject(s)
Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , Mammary Neoplasms, Animal/metabolism , Receptor, ErbB-2/metabolism , Transcription Factors/biosynthesis , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , DNA-Binding Proteins/genetics , Female , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Metastasis , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Receptor, ErbB-2/genetics , Transcription Factors/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
12.
Mol Cancer Res ; 10(4): 523-34, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22355043

ABSTRACT

Studies suggest that Hsf4 expression correlates with its role in cell growth and differentiation. However, the role of Hsf4 in tumorigenesis in vivo remains unexplored. In this article, we provide evidence that absence of the Hsf4 gene suppresses evolution of spontaneous tumors arising in p53- or Arf-deficient mice. Furthermore, deletion of hsf4 alters the tumor spectrum by significantly inhibiting development of lymphomas that are normally observed in the majority of mice lacking p53 or Arf tumor suppressor genes. Using mouse embryo fibroblasts deficient in the hsf4 gene, we have found that these cells exhibit reduced proliferation that is associated with induction of senescence and senescence-associated ß-galactosidase (SA-ß-gal). Cellular senescence in hsf4-deficient cells is associated with the increased expression of the cyclin-dependent kinase inhibitors, p21 and p27 proteins. Consistent with the cellular senescence observed in vitro, specific normal tissues of hsf4(-/-) mice and tumors that arose in mice deficient in both hsf4 and p53 genes exhibit increased SA-ß-gal activity and elevated levels of p27 compared with wild-type mice. These results suggest that hsf4 deletion-induced senescence is also present in vivo. Our results therefore indicate that Hsf4 is involved in modulation of cellular senescence, which can be exploited during cancer therapy.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cells, Cultured , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA-Binding Proteins/deficiency , Heat Shock Transcription Factors , Mice , Mice, Inbred C57BL , Mice, Transgenic , Transcription Factors/deficiency , Tumor Suppressor Protein p53/metabolism
13.
Methods Mol Biol ; 787: 1-20, 2011.
Article in English | MEDLINE | ID: mdl-21898223

ABSTRACT

Heat-shock transcription factors (Hsfs) regulate transcription of heat-shock proteins as well as other genes whose promoters contain heat-shock elements. There are at least five Hsfs in mammalian cells, Hsf1, Hsf2, Hsf3, Hsf4, and Hsfy. To understand the physiological roles of Hsf1, Hsf2, and Hsf4 in vivo, we generated knockout mouse lines for these factors. In this chapter, we describe the design of the targeting vectors, the plasmids used, and the successful generation of mice lacking the individual genes. We also briefly describe what we have learned about the physiological functions of these genes in vivo.


Subject(s)
DNA-Binding Proteins/genetics , Heat-Shock Proteins/genetics , Sequence Deletion , Transcription Factors/genetics , Animals , Base Sequence , Cell Line , Genetic Vectors , Heat Shock Transcription Factors , Heat-Shock Response/genetics , Mice , Mice, Knockout
14.
Cell Metab ; 14(1): 91-103, 2011 Jul 06.
Article in English | MEDLINE | ID: mdl-21723507

ABSTRACT

Hepatocellular carcinoma (HCC) occurrence and progression are linked tightly to progressive hepatic metabolic syndrome associated with insulin resistance, hepatic steatosis, and chronic inflammation. Heat shock transcription factor 1 (HSF1), a major transactivator of stress proteins, increases survival by protecting cells against environmental stressors. It has been implicated in the pathogenesis of cancer, but specific mechanisms by which HSF1 supports cancer development remain elusive. We propose a pathogenic mechanism whereby HSF1 activation promotes growth of premalignant cells and HCC development by stimulating lipid biosynthesis and perpetuating chronic hepatic metabolic disease induced by carcinogens. Our work shows that inactivation of HSF1 impairs cancer progression, mitigating adverse effects of carcinogens on hepatic metabolism by enhancing insulin sensitivity and sensitizing activation of AMP-activated protein kinase (AMPK), an important regulator of energy homeostasis and inhibitor of lipid synthesis. HSF1 is a potential target for the control of hepatic steatosis, hepatic insulin resistance, and HCC development.


Subject(s)
Carcinoma, Hepatocellular/etiology , DNA-Binding Proteins/metabolism , Fatty Liver/metabolism , Liver Neoplasms/etiology , Metabolic Syndrome/metabolism , Transcription Factors/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/pathology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Fatty Liver/chemically induced , Fatty Liver/prevention & control , Heat Shock Transcription Factors , Lipid Metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Mice , Mice, Knockout , Receptor, Insulin/metabolism , Signal Transduction , Transcription Factors/deficiency , Transcription Factors/genetics
15.
Mol Cell Biol ; 30(19): 4626-43, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20679486

ABSTRACT

Accumulation of tau into neurofibrillary tangles is a pathological consequence of Alzheimer's disease and other tauopathies. Failures of the quality control mechanisms by the heat shock proteins (Hsps) positively correlate with the appearance of such neurodegenerative diseases. However, in vivo genetic evidence for the roles of Hsps in neurodegeneration remains elusive. Hsp110 is a nucleotide exchange factor for Hsp70, and direct substrate binding to Hsp110 may facilitate substrate folding. Hsp70 complexes have been implicated in tau phosphorylation state and amyloid precursor protein (APP) processing. To provide evidence for a role for Hsp110 in central nervous system homeostasis, we have generated hsp110(-)(/)(-) mice. Our results show that hsp110(-)(/)(-) mice exhibit accumulation of hyperphosphorylated-tau (p-tau) and neurodegeneration. We also demonstrate that Hsp110 is in complexes with tau, other molecular chaperones, and protein phosphatase 2A (PP2A). Surprisingly, high levels of PP2A remain bound to tau but with significantly reduced activity in brain extracts from aged hsp110(-)(/)(-) mice compared to brain extracts from wild-type mice. Mice deficient in the Hsp110 partner (Hsp70) also exhibit a phenotype comparable to that of hsp110(-)(/)(-) mice, confirming a critical role for Hsp110-Hsp70 in maintaining tau in its unphosphorylated form during aging. In addition, crossing hsp110(-)(/)(-) mice with mice overexpressing mutant APP (APPßsw) leads to selective appearance of insoluble amyloid ß42 (Aß42), suggesting an essential role for Hsp110 in APP processing and Aß generation. Thus, our findings provide in vivo evidence that Hsp110 plays a critical function in tau phosphorylation state through maintenance of efficient PP2A activity, confirming its role in pathogenesis of Alzheimer's disease and other tauopathies.


Subject(s)
Aging , Amyloid beta-Peptides/metabolism , HSP110 Heat-Shock Proteins/metabolism , tau Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Brain/cytology , Brain/metabolism , Cells, Cultured , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HSP110 Heat-Shock Proteins/genetics , Humans , Immunoblotting , Immunohistochemistry , Male , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Motor Activity/genetics , Motor Activity/physiology , Neurons/cytology , Neurons/metabolism , Phosphorylation , Protein Binding , Protein Phosphatase 2/metabolism , Solubility
16.
J Cell Biochem ; 107(3): 504-15, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19343786

ABSTRACT

Heat shock factor Hsf1 regulates the stress-inducibility of heat shock proteins (Hsps) or molecular chaperones. One of the functions attributed to Hsps is their participation in folding and degradation of proteins. We recently showed that hsf1(-/-) cells accumulate ubiquitinated proteins. However, a direct role for Hsf1 in stability of specific proteins such as p53 has not been elucidated. We present evidence that cells deficient in hsf1 accumulate wild-type p53 protein. We further show that hsf1(-/-) cells express lower levels of alphaB-crystallin and cells deficient in alphaB-crystallin also accumulate p53 protein. Reports indicate that alphaB-crystallin binds to Fbx4 ubiquitin ligase, and they target cyclin D1 for degradation through a pathway involving the SCF (Skp1-Cul1-F-box) complex. Towards determining a mechanism for p53 degradation involving alphaB-crystallin and Hsf1, we have found that ectopic expression of Fbx4 in wild-type mouse embryo fibroblasts (MEFs) expressing mutant p53 (p53R175H) leads to increase in its degradation, while MEFs deficient in hsf1 or alphaBcry are defective in degradation of this p53 protein. In addition, immunoprecipitated p53R175H from wild-type MEFs is able to pull-down both alphaB-crystallin and Fbx4. Finally, immunoprecipitated wild-type p53 from doxorubicin treated U2OS cells can pull-down endogenous alphaB-crystallin and Fbx4. These results indicate that hsf1- and alphaBcry-deficient cells accumulate p53 due to reduced levels of alphaB-crystallin in these cells. Elevated levels of p53 in hsf1- and alphaBcry-deficient cells lead to their increased sensitivity to DNA damaging agents. These data reveal a novel mechanism for protein degradation through Hsf1 and alphaB-crystallin.


Subject(s)
DNA-Binding Proteins/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/metabolism , alpha-Crystallin B Chain/genetics , Animals , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , F-Box Proteins/metabolism , Heat Shock Transcription Factors , Mice , Mice, Knockout , Transcription Factors/deficiency , Transfection , alpha-Crystallin B Chain/metabolism
17.
J Gen Virol ; 89(Pt 6): 1421-1433, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18474558

ABSTRACT

Arenaviruses include several causative agents of haemorrhagic fever disease in humans. In addition, the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) is a superb model for the study of virus-host interactions, including the basis of viral persistence and associated diseases. There is little understanding about the molecular mechanisms concerning the regulation and specific role of viral proteins in modulating arenavirus-host cell interactions either associated with an acute or persistent infection, and associated disease. Here, we report the genomic and biological characterization of LCMV strains 'Docile' (persistent) and 'Aggressive' (not persistent) recovered from cloned cDNA via reverse genetics. Our results confirmed that the cloned viruses accurately recreated the in vivo phenotypes associated with the corresponding natural Docile and Aggressive viral isolates. In addition, we provide evidence that the ability of the Docile strain to persist is determined by the nature of both S and L RNA segments. Thus, our findings provide the foundation for studies aimed at gaining a detailed understanding of viral determinants of LCMV persistence in its natural host, which may aid in the development of vaccines to prevent or treat the diseases caused by arenaviruses in humans.


Subject(s)
Lymphocytic Choriomeningitis/veterinary , Lymphocytic choriomeningitis virus/physiology , Plasmids , Reassortant Viruses/physiology , Rodent Diseases/virology , Animals , Cell Line , Cricetinae , Host-Pathogen Interactions , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/pathogenicity , Mice , Mice, Inbred C57BL , RNA, Viral/chemistry , Reassortant Viruses/pathogenicity , Sequence Analysis, RNA , Virulence
18.
J Virol ; 82(6): 2952-65, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18216105

ABSTRACT

The migration of activated antigen-specific immune cells to the target tissues of virus replication is controlled by the expression of adhesion molecules on the vascular endothelium that bind to ligands on circulating lymphocytes. Here, we demonstrate that the adhesion pathway mediated by vascular cell adhesion molecule 1 (VCAM-1) plays a role in regulating T-cell-mediated inflammation and pathology in nonlymphoid tissues, including the central nervous system (CNS) during viral infection. The ablation of VCAM-1 expression from endothelial and hematopoietic cells using a loxP-Cre recombination strategy had no major effect on the induction or overall tissue distribution of antigen-specific T cells during a systemic infection with lymphocytic choriomeningitis virus (LCMV), except in the case of lung tissue. However, enhanced resistance to lethal LCM and the significantly reduced magnitude and duration of footpad swelling observed in VCAM-1 mutant mice compared to B6 controls suggest a significant role for VCAM-1 in promoting successful local inflammatory reactions associated with efficient viral clearance and even life-threatening immunopathology under particular infection conditions. Interestingly, analysis of the infiltrating populations in the brains of intracerebrally infected mice revealed that VCAM-1 deletion significantly delayed migration into the CNS of antigen-presenting cells (macrophages and dendritic cells), which are critical for optimal stimulation of migrating virus-specific CD8(+) T cells initiating a pathological cascade. We propose that the impaired migration of these accessory cells in the brain may explain the improved clinical outcome of infection in VCAM-1 mutant mice. Thus, these results underscore the potential role of VCAM-1 in regulating the immune response and inflammatory reactions against viral infections.


Subject(s)
Inflammation/immunology , Lymphocytic Choriomeningitis/prevention & control , Lymphocytic choriomeningitis virus/immunology , Vascular Cell Adhesion Molecule-1/physiology , Animals , Antibodies, Viral/blood , CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Virus Replication
19.
J Virol ; 82(7): 3353-68, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18199651

ABSTRACT

A characteristic feature in the immune response to many persistent viral infections is the dysfunction or deletion of antigen-specific T cells (exhaustion). This down-regulation of virus-specific T-cell response represents a critical control mechanism that exists within T-cell activation pathways to prevent lethal disease by inappropriate responses against disseminating virus infections. However, the molecular mechanisms by which the immune system determines whether to mount a full response to such infections remain largely unexplored. Here, we have established that in the murine lymphocytic choriomeningitis virus (LCMV) model, induction of the T-cell receptor signaling inhibitor molecule E3 ligase Cbl-b is critically involved in this decision. In particular, our data revealed that Cbl-b controls the program responsible for T-cell tolerance (exhaustion) induction during a chronic viral infection. Thus, Cbl-b(-/-) mice infected with a low dose of LCMV Docile mount a strong CD8(+) T-cell response that rapidly clears the infection, and the animals remain healthy; in contrast, down-regulation of the epitope-specific CD8(+) T-cell population in persistently infected Cbl-b(-/-) mice, compared to that in chronically infected B6 mice, was significantly delayed, and this was associated with increased morbidity and eventual death in nearly 20% of the animals. Interestingly, infection of Cbl-b(-/-) mice with a moderate virus dose resulted in rapid death with 100% mortality by 7 to 8 days after infection, caused by a dysregulated antiviral T-cell response, whereas the infected B6 mice survived and remained healthy. In conclusion, our results suggest that Cbl-b is critically involved in T-cell exhaustion and prevention of lethal disease.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Arenaviridae Infections/immunology , Arenaviridae Infections/pathology , CD8-Positive T-Lymphocytes/immunology , Lymphocytic choriomeningitis virus/immunology , Proto-Oncogene Proteins c-cbl/immunology , Adaptor Proteins, Signal Transducing/deficiency , Adoptive Transfer , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Cell Proliferation , Liver/pathology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-cbl/deficiency , Survival Analysis
20.
Genesis ; 45(8): 487-501, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17661394

ABSTRACT

The mammalian small heat shock protein (sHSPs) family is comprised of 10 members and includes HSPB1, which is proposed to play an essential role in cellular physiology, acting as a molecular chaperone to regulate diverse cellular processes. Whilst differential roles for sHSPs are suggested for specific tissues, the relative contribution of individual sHSP family members in cellular and organ physiology remains unclear. To address the function of HSPB1 in vivo and determine its tissue-specific expression during development and in the adult, we generated knock-in mice where the coding sequence of hspb1 is replaced by a lacZ reporter gene. Hspb1 expression marks myogenic differentiation with specific expression first confined to developing cardiac muscles and the vascular system, and later in skeletal muscles with specific expression at advanced stages of myoblast differentiation. In the adult, hspb1 expression was observed in other tissues, such as stratified squamous epithelium of skin, oronasal cavity, tongue, esophagus, and uterine cervix but its expression was most prominent in the musculature. Interestingly, in cardiac muscle hsbp1 expression was down-regulated during the neonatal period and maintained to a relatively low steady-level throughout adulthood. Despite this widespread expression, hspb1-/- mice were viable and fertile with no apparent morphological abnormalities in tissues under physiological conditions. However, at the cellular level and under stress conditions (heat challenge), HSPB1 act synergistically with the stress-induced HSPA1 (HSP70) in thermotolerance development, protecting cells from apoptosis. Our data thus indicate a nonessential role for HSPB1 in embryonic development and for maintenance of tissues under physiological conditions, but also shows that it plays an important role by acting synergistically with other HSPs during stress conditions to exert cytoprotection and anti-apoptotic effects.


Subject(s)
Gene Expression Regulation, Developmental , Gene Targeting , Heat-Shock Proteins/physiology , Muscle, Skeletal/metabolism , Neoplasm Proteins/physiology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Blastocyst , Blotting, Southern , Blotting, Western , Bone Marrow/metabolism , Crosses, Genetic , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/radiation effects , Etoposide/pharmacology , Female , Fever , Heat-Shock Proteins/genetics , Integrases/metabolism , Lac Operon/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Chaperones , Muscle, Skeletal/cytology , Neoplasm Proteins/genetics , Radiation, Ionizing , beta-Galactosidase
SELECTION OF CITATIONS
SEARCH DETAIL
...