Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cell ; 160(4): 729-744, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25679764

ABSTRACT

Signaling through RAS/MAP kinase pathway is central to biology. ERK has long been perceived as the only substrate for MEK. Here, we report that HSF1, the master regulator of the proteotoxic stress response, is a new MEK substrate. Beyond mediating cell-environment interactions, the MEK-HSF1 regulation impacts malignancy. In tumor cells, MEK blockade inactivates HSF1 and thereby provokes proteomic chaos, presented as protein destabilization, aggregation, and, strikingly, amyloidogenesis. Unlike their non-transformed counterparts, tumor cells are particularly susceptible to proteomic perturbation and amyloid induction. Amyloidogenesis is tumor suppressive, reducing in vivo melanoma growth and contributing to the potent anti-neoplastic effects of proteotoxic stressors. Our findings unveil a key biological function of the oncogenic RAS-MEK signaling in guarding proteostasis and suppressing amyloidogenesis. Thus, proteomic instability is an intrinsic feature of malignant state, and disrupting the fragile tumor proteostasis to promote amyloidogenesis may be a feasible therapeutic strategy.


Subject(s)
Amyloid/metabolism , DNA-Binding Proteins/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasms/metabolism , Protein Stability , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Female , Heat Shock Transcription Factors , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Phosphorylation , Protein Aggregates , Proteome/metabolism , Transplantation, Heterologous
2.
Mol Cell ; 76(4): 546-561.e8, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31561952

ABSTRACT

Through transcriptional control of the evolutionarily conserved heat shock, or proteotoxic stress, response, heat shock factor 1 (HSF1) preserves proteomic stability. Here, we show that HSF1, a physiological substrate for AMP-activated protein kinase (AMPK), constitutively suppresses this central metabolic sensor. By physically evoking conformational switching of AMPK, HSF1 impairs AMP binding to the γ subunits and enhances the PP2A-mediated de-phosphorylation, but it impedes the LKB1-mediated phosphorylation of Thr172, and retards ATP binding to the catalytic α subunits. These immediate and manifold regulations empower HSF1 to both repress AMPK under basal conditions and restrain its activation by diverse stimuli, thereby promoting lipogenesis, cholesterol synthesis, and protein cholesteroylation. In vivo, HSF1 antagonizes AMPK to control body fat mass and drive the lipogenic phenotype and growth of melanomas independently of its intrinsic transcriptional action. Thus, the physical AMPK-HSF1 interaction epitomizes a reciprocal kinase-substrate regulation whereby lipid metabolism and proteomic stability intertwine.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Energy Metabolism , Heat Shock Transcription Factors/metabolism , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/genetics , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Adiposity , Animals , Binding Sites , Cell Proliferation , Cholesterol/biosynthesis , HEK293 Cells , HeLa Cells , Heat Shock Transcription Factors/deficiency , Heat Shock Transcription Factors/genetics , Humans , Lipogenesis , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Phosphorylation , Protein Conformation , Protein Stability , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Structure-Activity Relationship
3.
EMBO J ; 34(3): 275-93, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25425574

ABSTRACT

Numerous extrinsic and intrinsic insults trigger the HSF1-mediated proteotoxic stress response (PSR), an ancient transcriptional program that is essential to proteostasis and survival under such conditions. In contrast to its well-recognized mobilization by proteotoxic stress, little is known about how this powerful adaptive mechanism reacts to other stresses. Surprisingly, we discovered that metabolic stress suppresses the PSR. This suppression is largely mediated through the central metabolic sensor AMPK, which physically interacts with and phosphorylates HSF1 at Ser121. Through AMPK activation, metabolic stress represses HSF1, rendering cells vulnerable to proteotoxic stress. Conversely, proteotoxic stress inactivates AMPK and thereby interferes with the metabolic stress response. Importantly, metformin, a metabolic stressor and popular anti-diabetic drug, inactivates HSF1 and provokes proteotoxic stress within tumor cells, thereby impeding tumor growth. Thus, these findings uncover a novel interplay between the metabolic stress sensor AMPK and the proteotoxic stress sensor HSF1 that profoundly impacts stress resistance, proteostasis, and malignant growth.


Subject(s)
AMP-Activated Protein Kinases/metabolism , DNA-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Stress, Physiological , Transcription Factors/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Cell Line, Tumor , DNA-Binding Proteins/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Heat Shock Transcription Factors , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Phosphorylation/drug effects , Phosphorylation/genetics , Transcription Factors/genetics
4.
Bioessays ; 39(5)2017 05.
Article in English | MEDLINE | ID: mdl-28295473

ABSTRACT

Beyond protein synthesis and autophagy, emerging evidence has implicated mTORC1 in regulating protein folding and proteasomal degradation as well, highlighting its prominent role in cellular proteome homeostasis or proteostasis. In addition to growth signals, mTORC1 senses and responds to a wide array of stresses, including energetic/metabolic stress, genotoxic stress, oxidative stress, osmotic stress, ER stress, proteotoxic stress, and psychological stress. Whereas growth signals unanimously stimulate mTORC1, stresses exert complex impacts on mTORC1, most of which are repressive. mTORC1 suppression, as a generic adaptive strategy, empowers cell survival under various stressful conditions. In this essay, we provide an overview of the emerging role of mTORC1 in proteostasis, the distinct molecular mechanisms through which mTORC1 reacts to diverse stresses, and the schemes exploited by cancer cells to circumvent stress-induced mTORC1 suppression. Hence, acting as a stress sensor, mTORC1 intimately couples stresses to cellular proteostasis.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/metabolism , Proteostasis , Stress, Physiological , Animals , Carcinogenesis , Endoplasmic Reticulum Stress , Humans , Models, Biological , Neoplasms/metabolism , Osmotic Pressure , Oxidative Stress , Stress, Psychological
5.
Cell Mol Life Sci ; 73(22): 4231-4248, 2016 11.
Article in English | MEDLINE | ID: mdl-27289378

ABSTRACT

Proteome homeostasis, or proteostasis, is essential to maintain cellular fitness and its disturbance is associated with a broad range of human health conditions and diseases. Cells are constantly challenged by various extrinsic and intrinsic insults, which perturb cellular proteostasis and provoke proteotoxic stress. To counter proteomic perturbations and preserve proteostasis, cells mobilize the proteotoxic stress response (PSR), an evolutionarily conserved transcriptional program mediated by heat shock factor 1 (HSF1). The HSF1-mediated PSR guards the proteome against misfolding and aggregation. In addition to proteotoxic stress, emerging studies reveal that this proteostatic mechanism also responds to cellular energy state. This regulation is mediated by the key cellular metabolic sensor AMP-activated protein kinase (AMPK). In this review, we present an overview of the maintenance of proteostasis by HSF1, the metabolic regulation of the PSR, particularly focusing on AMPK, and their implications in the two major age-related diseases-diabetes mellitus and neurodegenerative disorders.


Subject(s)
Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Proteins/toxicity , Stress, Physiological/drug effects , Animals , Heat-Shock Proteins/metabolism , Humans , Models, Biological
6.
STAR Protoc ; 4(4): 102692, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37917578

ABSTRACT

Transcription factors (TFs) play a pivotal role in gene expression, and their DNA binding is the prerequisite to instigating gene transcription. Here, we present a protocol that exploits the proximity ligation assay technique to measure the DNA-binding activities of TFs in situ at the single-cell resolution. We describe steps for immunostaining with specific antibodies against double-stranded DNA and the TFs of interest, probe incubation, proximity ligation, and signal amplification. We then detail procedures for imaging and image analysis. For complete details on the use and execution of this protocol, please refer to Dai et al. (2015)1 and Xu et al. (2023).2.


Subject(s)
Antibodies , Transcription Factors , Transcription Factors/genetics , Image Processing, Computer-Assisted , Cells, Cultured
7.
Cell Rep ; 42(6): 112557, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37224019

ABSTRACT

Despite its pivotal roles in biology, how the transcriptional activity of c-MYC is tuned quantitatively remains poorly defined. Here, we show that heat shock factor 1 (HSF1), the master transcriptional regulator of the heat shock response, acts as a prime modifier of the c-MYC-mediated transcription. HSF1 deficiency diminishes c-MYC DNA binding and dampens its transcriptional activity genome wide. Mechanistically, c-MYC, MAX, and HSF1 assemble into a transcription factor complex on genomic DNAs, and surprisingly, the DNA binding of HSF1 is dispensable. Instead, HSF1 physically recruits the histone acetyltransferase general control nonderepressible 5 (GCN5), promoting histone acetylation and augmenting c-MYC transcriptional activity. Thus, we find that HSF1 specifically potentiates the c-MYC-mediated transcription, discrete from its canonical role in countering proteotoxic stress. Importantly, this mechanism of action engenders two distinct c-MYC activation states, primary and advanced, which may be important to accommodate diverse physiological and pathological conditions.


Subject(s)
DNA-Binding Proteins , Heat-Shock Response , Transcription Factors , DNA , DNA-Binding Proteins/metabolism , Heat Shock Transcription Factors/genetics , Heat Shock Transcription Factors/metabolism , Heat-Shock Response/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/metabolism , Humans , Cell Line, Tumor
8.
J Cell Physiol ; 227(8): 2982-7, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22105155

ABSTRACT

Organisms frequently encounter a wide variety of proteotoxic stressors. The heat-shock response, an ancient cytoprotective mechanism, has evolved to augment organismal survival and longevity in the face of proteotoxic stress from without and within. These broadly recognized beneficial effects, ironically, contrast sharply with its emerging role as a culprit in the pathogenesis of cancers. Here, we present an overview of the normal biology of the heat-shock response and highlight its implications in oncogenic processes, including the proteotoxic stress phenotype of cancer; the function of this stress response in helping cancer survive and adapt to proteotoxic stress; and perturbation of proteome homeostasis in cancer as a potential therapeutic avenue.


Subject(s)
Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Heat-Shock Response/genetics , Oxidative Stress , Transcription Factors/genetics , Transcription Factors/metabolism , Cytoprotection , Gene Expression Regulation, Neoplastic , Heat Shock Transcription Factors , Humans , Neoplasms/genetics , Neoplasms/metabolism , Reactive Oxygen Species/metabolism
9.
Bioact Mater ; 18: 138-150, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35387155

ABSTRACT

Despite the recent advances in artificial tissue and organ engineering, how to generate large size viable and functional complex organs still remains as a grand challenge for regenerative medicine. Three-dimensional bioprinting has demonstrated its advantages as one of the major methods in fabricating simple tissues, yet it still faces difficulties to generate vasculatures and preserve cell functions in complex organ production. Here, we overcome the limitations of conventional bioprinting systems by converting a six degree-of-freedom robotic arm into a bioprinter, therefore enables cell printing on 3D complex-shaped vascular scaffolds from all directions. We also developed an oil bath-based cell printing method to better preserve cell natural functions after printing. Together with a self-designed bioreactor and a repeated print-and-culture strategy, our bioprinting system is capable to generate vascularized, contractible, and long-term survived cardiac tissues. Such bioprinting strategy mimics the in vivo organ development process and presents a promising solution for in vitro fabrication of complex organs.

10.
Hepatol Commun ; 6(10): 2781-2797, 2022 10.
Article in English | MEDLINE | ID: mdl-35945902

ABSTRACT

Liver fibrosis is an aberrant wound healing response that results from chronic injury and is mediated by hepatocellular death and activation of hepatic stellate cells (HSCs). While induction of oxidative stress is well established in fibrotic livers, there is limited information on stress-mediated mechanisms of HSC activation. Cellular stress triggers an adaptive defense mechanism via master protein homeostasis regulator, heat shock factor 1 (HSF1), which induces heat shock proteins to respond to proteotoxic stress. Although the importance of HSF1 in restoring cellular homeostasis is well-established, its potential role in liver fibrosis is unknown. Here, we show that HSF1 messenger RNA is induced in human cirrhotic and murine fibrotic livers. Hepatocytes exhibit nuclear HSF1, whereas stellate cells expressing alpha smooth muscle actin do not express nuclear HSF1 in human cirrhosis. Interestingly, despite nuclear HSF1, murine fibrotic livers did not show induction of HSF1 DNA binding activity compared with controls. HSF1-deficient mice exhibit augmented HSC activation and fibrosis despite limited pro-inflammatory cytokine response and display delayed fibrosis resolution. Stellate cell and hepatocyte-specific HSF1 knockout mice exhibit higher induction of profibrogenic response, suggesting an important role for HSF1 in HSC activation and fibrosis. Stable expression of dominant negative HSF1 promotes fibrogenic activation of HSCs. Overactivation of HSF1 decreased phosphorylation of JNK and prevented HSC activation, supporting a protective role for HSF1. Our findings identify an unconventional role for HSF1 in liver fibrosis. Conclusion: Our results show that deficiency of HSF1 is associated with exacerbated HSC activation promoting liver fibrosis, whereas activation of HSF1 prevents profibrogenic HSC activation.


Subject(s)
Actins , Heat Shock Transcription Factors/metabolism , Hepatic Stellate Cells , Actins/genetics , Animals , Cytokines/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Response , Hepatic Stellate Cells/metabolism , Humans , Liver Cirrhosis/genetics , Mice , Mice, Knockout , RNA, Messenger/metabolism
11.
J Biol Chem ; 285(46): 35528-36, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20833711

ABSTRACT

It is conceivable that stimulating proteasome activity for rapid removal of misfolded and oxidized proteins is a promising strategy to prevent and alleviate aging-related diseases. Sulforaphane (SFN), an effective cancer preventive agent derived from cruciferous vegetables, has been shown to enhance proteasome activities in mammalian cells and to reduce the level of oxidized proteins and amyloid ß-induced cytotoxicity. Here, we report that SFN activates heat shock transcription factor 1-mediated heat shock response. Specifically, SFN-induced expression of heat shock protein 27 (Hsp27) underlies SFN-stimulated proteasome activity. SFN-induced proteasome activity was significantly enhanced in Hsp27-overexpressing cells but absent in Hsp27-silenced cells. The role of Hsp27 in regulating proteasome activity was further confirmed in isogenic REG cells, in which SFN-induced proteasome activation was only observed in cells stably overexpressing Hsp27, but not in the Hsp27-free parental cells. Finally, we demonstrated that phosphorylation of Hsp27 is irrelevant to SFN-induced proteasome activation. This study provides a novel mechanism underlying SFN-induced proteasome activity. This is the first report to show that heat shock response by SFN, in addition to the antioxidant response mediated by the Keap1-Nrf2 pathway, may contribute to cytoprotection.


Subject(s)
HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Response/drug effects , Proteasome Endopeptidase Complex/metabolism , Thiocyanates/pharmacology , Animals , Anticarcinogenic Agents/pharmacology , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Cysteine Proteinase Inhibitors/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , HSP27 Heat-Shock Proteins/genetics , HeLa Cells , Heat Shock Transcription Factors , Hot Temperature , Humans , Immunoblotting , Isothiocyanates , Leupeptins/pharmacology , Proteasome Inhibitors , Protein Biosynthesis/drug effects , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Sulfoxides , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation/drug effects
12.
Sci Adv ; 6(46)2020 11.
Article in English | MEDLINE | ID: mdl-33177089

ABSTRACT

The role of proteomic instability in cancer, particularly amyloidogenesis, remains obscure. Heat shock factor 1 (HSF1) transcriptionally governs the proteotoxic stress response to suppress proteomic instability and enhance survival. Paradoxically, HSF1 promotes oncogenesis. Here, we report that AKT activates HSF1 via Ser230 phosphorylation. In vivo, HSF1 enables megalencephaly and hepatomegaly, which are driven by hyperactive phosphatidylinositol 3-kinase/AKT signaling. Hsf1 deficiency exacerbates amyloidogenesis and elicits apoptosis, thereby countering tissue overgrowth. Unexpectedly, HSF1 physically neutralizes soluble amyloid oligomers (AOs). Beyond impeding amyloidogenesis, HSF1 shields HSP60 from direct assault by AOs, averting HSP60 destabilization, collapse of the mitochondrial proteome, and, ultimately, mitophagy and apoptosis. The very same mechanism occurs in Alzheimer's disease. These findings suggest that amyloidogenesis may be a checkpoint mechanism that constrains uncontrolled growth and safeguards tissue homeostasis, congruent with its emerging tumor-suppressive function. HSF1, by acting as an anti-amyloid factor, promotes overgrowth syndromes and cancer but may suppress neurodegenerative disorders.

13.
Philos Trans R Soc Lond B Biol Sci ; 373(1738)2018 Jan 19.
Article in English | MEDLINE | ID: mdl-29203710

ABSTRACT

The heat-shock, or HSF1-mediated proteotoxic stress, response (HSR/HPSR) is characterized by induction of heat-shock proteins (HSPs). As molecular chaperones, HSPs facilitate the folding, assembly, transportation and degradation of other proteins. In mammals, heat shock factor 1 (HSF1) is the master regulator of this ancient transcriptional programme. Upon proteotoxic insults, the HSR/HPSR is essential to proteome homeostasis, or proteostasis, thereby resisting stress and antagonizing protein misfolding diseases and ageing. Contrasting with these benefits, an unexpected pro-oncogenic role of the HSR/HPSR is unfolding. Whereas HSF1 remains latent in primary cells without stress, it becomes constitutively activated within malignant cells, rendering them addicted to HSF1 for their growth and survival. Highlighting the HSR/HPSR as an integral component of the oncogenic network, several key pathways governing HSF1 activation by environmental stressors are causally implicated in malignancy. Importantly, HSF1 impacts the cancer proteome systemically. By suppressing tumour-suppressive amyloidogenesis, HSF1 preserves cancer proteostasis to support the malignant state, both providing insight into how HSF1 enables tumorigenesis and suggesting disruption of cancer proteostasis as a therapeutic strategy. This review provides an overview of the role of HSF1 in oncogenesis, mechanisms underlying its constitutive activation within cancer cells and its pro-oncogenic action, as well as potential HSF1-targeting strategies.This article is part of the theme issue 'Heat shock proteins as modulators and therapeutic targets of chronic disease: an integrated perspective'.


Subject(s)
Carcinogenesis/genetics , Heat Shock Transcription Factors/genetics , Mammals/genetics , Neoplastic Cells, Circulating/metabolism , Animals , Carcinogenesis/metabolism , Heat Shock Transcription Factors/metabolism , Humans , Mammals/metabolism
15.
Methods Mol Biol ; 1487: 195-201, 2017.
Article in English | MEDLINE | ID: mdl-27924568

ABSTRACT

RAS/MAPK signaling responds to diverse extracellular cues and regulates a wide array of cellular processes. Given its biological importance, abnormalities in RAS/MAPK signaling cascade have been intimately implicated in numerous human diseases, including cancer. Herein, we describe a novel methodology to study activation of this pivotal signaling pathway. The Proximity Ligation Assay (PLA) is employed to monitor kinase-substrate interactions between MEK1 and HSF1, or MEK1 and ERK1 in situ.


Subject(s)
Fluorescent Antibody Technique , In Situ Hybridization , Mitogen-Activated Protein Kinases/metabolism , ras Proteins/metabolism , HeLa Cells , Humans
16.
Cancer Res ; 64(14): 4783-9, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15256447

ABSTRACT

Platelet-derived growth factor (PDGF) is expressed in many different tumors, but its precise roles in tumorigenesis remain to be fully defined. Here, we report on a mouse model that demonstrates dose-dependent effects of PDGF-B on glial tumorigenesis. By removing inhibitory regulatory elements in the PDGFB mRNA, we are able to substantially elevate its expression in tumor cells using a retroviral delivery system. This elevation in PDGF-B production results in tumors with shortened latency, increased cellularity, regions of necrosis, and general high-grade character. In addition, elevated PDGF-B in these tumors also mediates vascular smooth muscle cell recruitment that supports tumor angiogenesis. PDGF receptor (PDGFR) signaling appears to be required for the maintenance of these high-grade characteristics, because treatment of high-grade tumors with a small molecule inhibitor of PDGFR results in reversion to a lower grade tumor histology. Our data show that PDGFR signaling quantitatively regulates tumor grade and is required to sustain high-grade oligodendrogliomas.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioma/metabolism , Glioma/pathology , Proto-Oncogene Proteins c-sis/physiology , 5' Untranslated Regions , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/genetics , Cell Division/physiology , Glioma/blood supply , Glioma/genetics , Mice , Mice, Transgenic , NIH 3T3 Cells , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Platelet-Derived Growth Factor/physiology , Proto-Oncogene Proteins c-sis/biosynthesis , Proto-Oncogene Proteins c-sis/genetics , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Signal Transduction/physiology , Transfection
17.
Cancer Res ; 62(19): 5551-8, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12359767

ABSTRACT

Deletion of the INK4a-ARF locus is found in the majority of human malignant gliomas. However, the role of INK4a-ARF loss in gliomagenesis is unclear. Animal modeling has shown that mice with targeted deletions in the Ink4a-Arf gene do not develop spontaneous gliomas. We have previously reported that combined KRas and Akt signaling could induce glioblastoma (GBM) formation from neural progenitor cells but had no effect in differentiated astrocytes. In this investigation, we have studied the effects of Ink4a-Arf loss on the formation of GBM induced by KRas and Akt gene transfer into neural progenitor cells and astrocytes. We show here that Ink4a-Arf deficiency allows for GBM formation from astrocytes and that it enhances tumor incidence in neural progenitor cells. Furthermore, KRas alone can cooperate with deletion of the Ink4a-Arf locus in tumor formation from both neural progenitor cells and astrocytes. The resulting tumors were nestin positive and resembled a spectrum of glioma morphologies ranging in astrocytic character depending on cell-of-origin and presence of activated Akt. Our data strongly supports the view that one role of loss of Ink4a-Arf in gliomagenesis could be to sensitize astrocytes to transformation through dedifferentiation in response to the appropriate oncogenic stimuli.


Subject(s)
Brain Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Glioblastoma/genetics , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p14ARF/genetics , ras Proteins/physiology , Animals , Astrocytes/enzymology , Astrocytes/pathology , Astrocytes/physiology , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Enzyme Activation , Gene Deletion , Gene Expression Regulation , Genes, ras/genetics , Glioblastoma/enzymology , Glioblastoma/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neurons/enzymology , Neurons/pathology , Neurons/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Signal Transduction/genetics , Stem Cells/enzymology , Stem Cells/pathology , Stem Cells/physiology , Transfection
18.
Trends Cell Biol ; 26(1): 17-28, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26597576

ABSTRACT

Proteomic instability is causally related to human diseases. In guarding proteome stability, the heat shock factor 1 (HSF1)-mediated proteotoxic stress response plays a pivotal role. Contrasting with its beneficial role of enhancing cell survival, recent findings have revealed a compelling pro-oncogenic role for HSF1. However, the mechanisms underlying the persistent activation and function of HSF1 within malignancy remain poorly understood. Emerging evidence reveals that oncogenic signaling mobilizes HSF1 and that cancer cells rely on HSF1 to avert proteomic instability and repress tumor-suppressive amyloidogenesis. In aggregate, these new developments suggest that cancer cells endure chronic proteotoxic stress and that proteomic instability is intrinsically associated with the malignant state, a characteristic that could be exploited to combat cancer.


Subject(s)
DNA-Binding Proteins/physiology , Homeostasis , Neoplasms/metabolism , Proteome/metabolism , Transcription Factors/physiology , Animals , Heat Shock Transcription Factors , Humans , Protein Processing, Post-Translational , Signal Transduction
19.
Nat Cell Biol ; 18(5): 527-39, 2016 05.
Article in English | MEDLINE | ID: mdl-27043084

ABSTRACT

To cope with proteotoxic stress, cells attenuate protein synthesis. However, the precise mechanisms underlying this fundamental adaptation remain poorly defined. Here we report that mTORC1 acts as an immediate cellular sensor of proteotoxic stress. Surprisingly, the multifaceted stress-responsive kinase JNK constitutively associates with mTORC1 under normal growth conditions. On activation by proteotoxic stress, JNK phosphorylates both RAPTOR at S863 and mTOR at S567, causing partial disintegration of mTORC1 and subsequent translation inhibition. Importantly, HSF1, the central player in the proteotoxic stress response (PSR), preserves mTORC1 integrity and function by inactivating JNK, independently of its canonical transcriptional action. Thereby, HSF1 translationally augments the PSR. Beyond promoting stress resistance, this intricate HSF1-JNK-mTORC1 interplay, strikingly, regulates cell, organ and body sizes. Thus, these results illuminate a unifying mechanism that controls stress adaptation and growth.


Subject(s)
DNA-Binding Proteins/metabolism , Heat-Shock Response , Multiprotein Complexes/metabolism , Proteins/toxicity , Stress, Physiological/drug effects , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Animals , Body Size/drug effects , Cell Proliferation/drug effects , Cell Size/drug effects , Enzyme Activation/drug effects , HEK293 Cells , HeLa Cells , Heat Shock Transcription Factors , Heat-Shock Response/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/cytology , Liver/drug effects , Liver/growth & development , MAP Kinase Signaling System/drug effects , Mechanistic Target of Rapamycin Complex 1 , Mice , Organ Size/drug effects , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Transcription, Genetic/drug effects
20.
Neoplasia ; 7(4): 397-406, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15967117

ABSTRACT

Chronic platelet-derived growth factor (PDGF) signaling in glial progenitors leads to the formation of oligodendrogliomas in mice, whereas chronic combined Ras and Akt signaling leads to astrocytomas. Different histologies of these tumors imply that the pathways activated by these two oncogenic stimulations are different, and that the apparent lineage of the tumor cells may result from specific signaling activity. Therefore, we have investigated the signaling effects of PDGF in culture and in gliomas in vivo. In culture, PDGF transiently activates ERK1/2 and Akt, and subsequently elevates p21 and PCNA expression similar to chronic PDGF autocrine signaling in cultured astrocytes and PDGF-induced oligodendrogliomas in vivo. Culture experiments show that autocrine PDGF stimulation, and combined active Ras and Akt generate signaling patterns that are in some ways mutually exclusive. Furthermore, forced Akt activity in the context of chronic PDGF stimulation results in cells with an astrocytic differentiation pattern both in culture and in vivo. These data imply that these two interconvertible signaling motifs are distinct in mice and lead to gliomas resembling the two major glioma histologies found in humans. The ability of signaling activity to convert tumor cells from one lineage to another presents a mechanism for the development of tumors apparently comprised of cells from multiple lineages.


Subject(s)
Astrocytoma/metabolism , Oligodendroglioma/metabolism , Animals , Blotting, Western , Brain/metabolism , Cell Differentiation , Cell Lineage , Enzyme Activation , Glioma/metabolism , Immunohistochemistry , MAP Kinase Signaling System , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Plasmids/metabolism , Platelet-Derived Growth Factor/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Time Factors , Transcriptional Activation , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL