Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
bioRxiv ; 2023 Nov 03.
Article in English | MEDLINE | ID: mdl-37961427

ABSTRACT

The role of autophagy in tumorigenesis and tumor metastasis remains poorly understood. Here we show that inhibition of autophagy stabilizes the transcription factor Twist1 through Sequestosome-1 (SQSTM1, also known as p62) and thus increases cell proliferation, migration, and epithelial-mesenchymal transition (EMT) in tumor development and metastasis. Inhibition of autophagy or p62 overexpression blocks Twist1 protein degradation in the proteasomes, while p62 inhibition enhances it. SQSTM1/p62 interacts with Twist1 via the UBA domain of p62, in a Twist1-ubiquitination-dependent manner. Lysine 175 in Twist1 is critical for Twist1 ubiquitination, degradation, and SQSTM1/p62 interaction. For squamous skin cancer and melanoma cells that express Twist1, SQSTM1/p62 increases tumor growth and metastasis in mice. Together, our results identified Twist1 as a key downstream protein for autophagy and suggest a critical role of the autophagy/p62/Twist1 axis in cancer development and metastasis.

2.
Dev Cell ; 56(14): 2029-2042.e5, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34171288

ABSTRACT

Mitochondria are critical metabolic and signaling hubs, and dysregulated mitochondrial homeostasis is implicated in many diseases. Degradation of damaged mitochondria by selective GABARAP/LC3-dependent macro-autophagy (mitophagy) is critical for maintaining mitochondrial homeostasis. To identify alternate forms of mitochondrial quality control that functionally compensate if mitophagy is inactive, we selected for autophagy-dependent cancer cells that survived loss of LC3-dependent autophagosome formation caused by inactivation of ATG7 or RB1CC1/FIP200. We discovered rare surviving autophagy-deficient clones that adapted to maintain mitochondrial homeostasis after gene inactivation and identified two enhanced mechanisms affecting mitochondria including mitochondrial dynamics and mitochondrial-derived vesicles (MDVs). To further understand these mechanisms, we quantified MDVs via flow cytometry and confirmed an SNX9-mediated mechanism necessary for flux of MDVs to lysosomes. We show that the autophagy-dependent cells acquire unique dependencies on these processes, indicating that these alternate forms of mitochondrial homeostasis compensate for loss of autophagy to maintain mitochondrial health.


Subject(s)
Autophagy , Microtubule-Associated Proteins/metabolism , Mitochondria/pathology , Mitochondrial Dynamics , Mitophagy , Sorting Nexins/metabolism , Transport Vesicles/physiology , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Endosomes/metabolism , Humans , Lysosomes , Microtubule-Associated Proteins/genetics , Mitochondria/metabolism , Sorting Nexins/genetics
3.
Dev Cell ; 56(7): 906-918, 2021 04 05.
Article in English | MEDLINE | ID: mdl-33689692

ABSTRACT

Beginning with the earliest studies of autophagy in cancer, there have been indications that autophagy can both promote and inhibit cancer growth and progression; autophagy regulation of organelle homeostasis is similarly complicated. In this review we discuss pro- and antitumor effects of organelle-targeted autophagy and how this contributes to several hallmarks of cancer, such as evading cell death, genomic instability, and altered metabolism. Typically, the removal of damaged or dysfunctional organelles prevents tumor development but can also aid in proliferation or drug resistance in established tumors. By better understanding how organelle-specific autophagy takes place and can be manipulated, it may be possible to go beyond the brute-force approach of trying to manipulate all autophagy in order to improve therapeutic targeting of this process in cancer.


Subject(s)
Autophagy , Neoplasms/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Homeostasis , Humans , Macroautophagy , Mitophagy
4.
Cell Death Dis ; 12(3): 248, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33674554

ABSTRACT

Many surgical models are used to study kidney and other diseases in mice, yet the effects of the surgical procedure itself on the kidney and other tissues have not been elucidated. In the present study, we found that both sham surgery and unilateral nephrectomy (UNX), which is used as a model of renal compensatory hypertrophy, in mice resulted in increased mammalian target of rapamycin complex 1/2 (mTORC1/2) in the remaining kidney. mTORC1 is known to regulate lysosomal biogenesis and autophagy. Genes associated with lysosomal biogenesis and function were decreased in sham surgery and UNX kidneys. In both sham surgery and UNX, there was suppressed autophagic flux in the kidney as indicated by the lack of an increase in LC3-II or autophagosomes seen on immunoblot, IF and EM after bafilomycin A1 administration and a concomitant increase in p62, a marker of autophagic cargo. There was a massive increase in pro-inflammatory cytokines, which are known to activate ERK1/2, in the serum after sham surgery and UNX. There was a large increase in ERK1/2 in sham surgery and UNX kidneys, which was blocked by the MEK1/2 inhibitor, trametinib. Trametinib also resulted in a significant decrease in p62. In summary, there was an intense systemic inflammatory response, an ERK-mediated increase in p62 and suppressed autophagic flux in the kidney after sham surgery and UNX. It is important that researchers are aware that changes in systemic pro-inflammatory cytokines, ERK1/2 and autophagy can be caused by sham surgery as well as the kidney injury/disease itself.


Subject(s)
Autophagy-Related Proteins/metabolism , Autophagy , Kidney Diseases/metabolism , Kidney/surgery , Nephrectomy/adverse effects , Animals , Autophagy-Related Proteins/genetics , Cell Line , Cytokines/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Inflammation Mediators/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Diseases/etiology , Kidney Diseases/genetics , Kidney Diseases/pathology , Lysosomes/genetics , Lysosomes/metabolism , Lysosomes/pathology , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/genetics , Mechanistic Target of Rapamycin Complex 2/metabolism , Metabolomics , Mice, Inbred C57BL , Signal Transduction
6.
Cell Signal ; 75: 109760, 2020 11.
Article in English | MEDLINE | ID: mdl-32866627

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is a common inherited disorder characterized by kidney cyst growth often resulting in end-stage renal disease. There is growing attention on understanding the role of impaired autophagy in ADPKD. Trehalose (TRE) has been shown to increase both protein stability and aggregate clearance and induce autophagy in neurodegenerative diseases. TRE treatment in wild type mice compared to vehicle resulted in increased expression in the kidney of Atg12-5 complex and increased Rab9a, autophagy-related proteins that play a role in the formation of autophagosomes. Thus, the aim of the study was to determine the effect of TRE on cyst growth and autophagy-related proteins, in the hypomorphic Pkd1RC/RC mouse model of ADPKD. Pkd1RC/RC mice were treated 2% TRE in water from days 50 to 120 of age. TRE did not slow cyst growth or improve kidney function or affect proliferation and apoptosis in Pkd1RC/RC kidneys. In Pkd1RC/RC vs. wild type kidneys, expression of the Atg12-5 complex was inhibited by TRE resulting in increased free Atg12 and TRE was unable to rescue the deficiency of the Atg12-5 complex. Rab9a was decreased in Pkd1RC/RC vs. wild type kidneys and unaffected by TRE. The TRE-induced increase in p62, a marker of autophagic cargo, that was seen in normal kidneys was blocked in Pkd1RC/RC kidneys. In summary, the autophagy phenotype in Pkd1RC/RC kidneys was characterized by decreases in crucial autophagy-related proteins (Atg12-5 complex, Atg5, Atg16L1), decreased Rab9a and increased mTORC1 (pS6S240/244, pmTORS2448) proteins. TRE increased Atg12-5 complex, Rab9a and p62 in normal kidneys, but was unable to rescue the deficiency in autophagy proteins or suppress mTORC1 in Pkd1RC/RC kidneys and did not protect against cyst growth.


Subject(s)
Polycystic Kidney, Autosomal Dominant/drug therapy , Protein Kinase C/metabolism , Trehalose/pharmacology , Animals , Autophagy/drug effects , Autophagy-Related Protein 12/metabolism , Autophagy-Related Protein 5/metabolism , Autophagy-Related Proteins , Mice , Mice, Inbred C57BL , rab GTP-Binding Proteins/metabolism
7.
Autophagy ; 16(12): 2282-2284, 2020 12.
Article in English | MEDLINE | ID: mdl-32866428

ABSTRACT

In this commentary I discuss a recent paper that describes a new mechanism for how macroautophagy/autophagy regulates the immune response to cancer, and relate it to other recent studies in this area. These recent developments may allow more effective strategies to manipulate autophagy to improve cancer therapy.


Subject(s)
Autophagy , Immunity , Neoplasms/immunology , Neoplasms/pathology , Humans , Immune System/metabolism , Immune System/pathology
8.
Prog Mol Biol Transl Sci ; 172: 55-65, 2020.
Article in English | MEDLINE | ID: mdl-32620250

ABSTRACT

The cellular recycling process of macroautophagy, which is the mechanism by which cellular material is delivered to lysosomes via double membraned vesicles called autophagosomes, is intimately connected to programmed cell death pathways, especially apoptosis. In this article, I discuss some underlying mechanisms and their implications for improving cancer therapy and propose that the approaches that have been taken to understand the autophagy-apoptosis connection to enhance cancer drug action can serve as a model for the kinds of information that should be developed to understand how autophagy controls other biological processes as well.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/physiology , Autophagy/drug effects , Autophagy-Related Proteins/antagonists & inhibitors , Autophagy-Related Proteins/physiology , Chloroquine/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Humans , Hydroxychloroquine/pharmacology , Lysosomes/drug effects , Lysosomes/physiology , Microtubules/drug effects , Microtubules/physiology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/physiology , Neoplasms/drug therapy , Neoplasms/pathology , Phagocytosis/physiology , Therapeutic Index, Drug
9.
Cell Signal ; 74: 109730, 2020 10.
Article in English | MEDLINE | ID: mdl-32730856

ABSTRACT

Cardiac hypertrophy is common in autosomal dominant polycystic kidney disease (ADPKD) patients. We found increased heart weight in Pkd1RC/RC and Pkd2WS25/+ mouse models of ADPKD. As there is a link between increased heart weight and mammalian target of rapamycin (mTOR), the aim of the study was to determine mTOR complex 1 and 2 signaling proteins in the heart in the Pkd1RC/RC mouse model of PKD. In 70 day old Pkd1RC/RC hearts, on immunoblot analysis, there was a large increase in p-AMPKThr172, a known autophagy inducer, and an increase in p-AktSer473 and p-AktThr308, but no increase in other mTORC1/2 proteins (p-S6Ser240/244, p-mTORSer2448). In 150 day old Pkd1RC/RC hearts, there was an increase in mTORC1 (p-S6Ser240/244) and mTOR-related proteins (p-AktThr308, p-GSK3ßSer9, p-AMPKThr172). As the mTOR pathway is the master regulator of autophagy, autophagy proteins were measured. There was an increase in p-Beclin-1 (BECN1), an autophagy regulator and activating molecule in Beclin-1-regulated autophagy (AMBRA1), a regulator of Beclin that play a role in autophagosome formation, an early stage of autophagy. There was a defect in the later stage of autophagy, the fusion of the autophagosome with the lysosome, known as autophagic flux, as evidenced by the lack of an increase in LC3-II, a marker of autophagosomes, with the lysosomal inhibitor bafilomycin, in both 70 day old and 150 day old hearts. To determine the role of autophagy in causing increased heart weight, Pkd1RC/RC were treated with 2-deoxyglucose (2-DG) or Tat-Beclin1 peptide, agents known to induce autophagy. 2-DG treatment from 150 to 350 days of age, a time period when increased heart weight developed, did not reduce the increased heart weight. Unexpectedly, Tat-Beclin 1 peptide treatment from 70 to 120 days of age resulted in increased heart weight. In summary, there is suppressed autophagic flux in the heart at an early age in Pkd1RC/RC mice. Increased mTOR signaling in older mice is associated suppressed autophagic flux. There was a large increase in p-AMPKThr172, a known autophagy inducer, in both young and old mice. 2-DG treatment did not impact increased heart weight and Tat-Beclin1 peptide increased heart weight.


Subject(s)
Cardiomegaly/metabolism , Polycystic Kidney, Autosomal Dominant , TOR Serine-Threonine Kinases/physiology , Animals , Autophagy , Disease Models, Animal , Mice , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology
10.
Int Rev Cell Mol Biol ; 352: 159-187, 2020.
Article in English | MEDLINE | ID: mdl-32334815

ABSTRACT

Autophagy, the process of macromolecular degradation through the lysosome, has been extensively studied for the past decade or two. Autophagy can regulate cell death, especially apoptosis, through selective degradation of both positive and negative apoptosis regulators. However, multiple other programmed cell death pathways exist. As knowledge of these other types of cell death expand, it has been suggested that they also interact with autophagy. In this review, we discuss the molecular mechanisms that comprise three non-apoptotic forms of cell death (necroptosis, pyroptosis and ferroptosis) focusing on how the autophagy machinery regulates these different cell death mechanisms through (i) its degradative functions, i.e., true autophagy, and (ii) other non-degradative functions of the autophagy machinery such as serving as a signaling scaffold or by participating in other autophagy-independent cellular processes.


Subject(s)
Autophagy , Ferroptosis , Necrosis/metabolism , Pyroptosis , Animals , Humans
11.
Autophagy ; 16(7): 1332-1340, 2020 07.
Article in English | MEDLINE | ID: mdl-32160093

ABSTRACT

Macroautophagy/autophagy degrades proteins and organelles to generate macromolecular building blocks. As such, some cancer cells are particularly dependent on autophagy. In a previous paper, we found that even highly autophagy-dependent cancer cells can adapt to circumvent autophagy inhibition. However, it remains unclear if autophagy-dependent cancer cells could survive the complete elimination of autophagosome formation. We extended our previous findings to show that knockout (KO) of both the upstream autophagy regulator RB1CC1/FIP200 and the downstream regulator and mediator of LC3 conjugation, ATG7, strongly inhibits growth in highly autophagy-dependent cells within one week of editing. However, rare clones survived the loss of ATG7 or RB1CC1 and maintained growth even under autophagy-inducing conditions. Autophagy-dependent cells circumvent the complete loss of autophagy that is mediated by RB1CC1 KO, similar to the loss of ATG7, by upregulating NFE2L2/NRF2 signaling. These results indicate that cancer cell lines could adapt to the complete loss of autophagy by changing their biology to adopt alternative ways of dealing with autophagy-mediated cellular functions. ABBREVIATIONS: CGS: CRISPR growth score; CQ: chloroquine; CRISPR: clustered regularly interspaced short palindromic repeats; EBSS: Earl's balanced salt solution; EEF2: eukaryotic translation elongation factor 2; FOXO3/FOXO3a: forkhead box O3; GFP: green fluorescent protein; KEAP1: kelch Like ECH associated protein 1; KO: knockout; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MEFs: mouse embryonic fibroblasts; NFE2L2/NRF2: nuclear factor, erythroid 2 like 2; NLS: nuclear localization signal; PCNA: proliferating cell nuclear antigen; PE: phosphatidylethanolamine; POLR2A: RNA polymerase II subunit A; PTEN: phosphatase and tensin homolog; ROS: reactive oxygen species; SNARE: soluble NSF attachment protein receptor; SQSTM1: sequestosome 1; STX17: syntaxin 17; TBHP: tert-butyl hydroperoxide; ULK1: unc-51 like autophagy activating kinase 1; ULK2: unc-51 like autophagy activating kinase 2; WT: wild type.


Subject(s)
Autophagy-Related Proteins/metabolism , Autophagy , Microtubule-Associated Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Cell Line, Tumor , Female , Humans , NF-E2-Related Factor 2/metabolism
12.
Cell Death Differ ; 27(3): 843-857, 2020 03.
Article in English | MEDLINE | ID: mdl-31836831

ABSTRACT

Autophagy allows for cellular material to be delivered to lysosomes for degradation resulting in basal or stress-induced turnover of cell components that provide energy and macromolecular precursors. These activities are thought to be particularly important in cancer where both tumor-promoting and tumor-inhibiting functions of autophagy have been described. Autophagy has also been intricately linked to apoptosis and programmed cell death, and understanding these interactions is becoming increasingly important in improving cancer therapy and patient outcomes. In this review, we consider how recent discoveries about how autophagy manipulation elicits its effects on cancer cell behavior can be leveraged to improve therapeutic responses.


Subject(s)
Autophagy , Neoplasms/pathology , Neoplasms/therapy , Animals , Apoptosis , Clinical Trials as Topic , Humans , Molecular Targeted Therapy , Neoplasms/immunology
13.
J Cell Biol ; 219(1)2020 01 06.
Article in English | MEDLINE | ID: mdl-31753861

ABSTRACT

Autophagy is intricately linked with many intracellular signaling pathways, particularly nutrient-sensing mechanisms and cell death signaling cascades. In cancer, the roles of autophagy are context dependent. Tumor cell-intrinsic effects of autophagy can be both tumor suppressive and tumor promotional. Autophagy can therefore not only activate and inhibit cell death, but also facilitate the switch between cell death mechanisms. Moreover, autophagy can play opposing roles in the tumor microenvironment via non-cell-autonomous mechanisms. Preclinical data support a tumor-promotional role of autophagy in established tumors and during cancer therapy; this has led to the launch of dozens of clinical trials targeting autophagy in multiple cancer types. However, many questions remain: which tumors and genetic backgrounds are the most sensitive to autophagy inhibition, and which therapies should be combined with autophagy inhibitors? Additionally, since cancer cells are under selective pressure and are prone to adaptation, particularly after treatment, it is unclear if and how cells adapt to autophagy inhibition. Here we review recent literature addressing these issues.


Subject(s)
Adaptation, Physiological , Autophagy , Cell Transformation, Neoplastic/pathology , Neoplasms/pathology , Signal Transduction , Tumor Microenvironment , Animals , Cell Death , Humans
14.
Yale J Biol Med ; 92(4): 707-718, 2019 12.
Article in English | MEDLINE | ID: mdl-31866785

ABSTRACT

In cancer therapy, a principle goal is to kill cancer cells while minimizing death of normal cells. Traditional cytotoxic therapies and the newer agents that target specific signaling proteins that are critical for cancer cell growth do this by activating a specific type of programmed cell death - apoptosis. However, it has been well established that cancer cells have varying levels of responses to apoptotic stimuli, with some being close to an "apoptotic threshold" and others being further away and that this ultimately determines whether cancer therapy is successful or not. In this review, we will highlight how the underlying mechanisms that control apoptosis thresholds relate to another important homeostatic process in cell survival and cell death, autophagy, and discuss recent evidence suggesting how inhibition of autophagy can enhance the action of anti-cancer drugs by modulating the apoptotic response.


Subject(s)
Apoptosis , Autophagy , Neoplasms/pathology , Neoplasms/therapy , Animals , Clinical Trials as Topic , Humans , Models, Biological , Molecular Targeted Therapy
15.
Cell Cycle ; 18(24): 3421-3431, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31736401

ABSTRACT

Autophagy is cellular recycling process that plays a complex role in cancer. Pre-clinical studies indicating a pro-tumorigenic role of autophagy have led to the launch of dozens of clinical trials combining autophagy inhibition with other standard of care therapies in different tumor types. A recent publication utilized a novel, acute, CRISPR/Cas9 assay to identify cancer cell lines that are exquisitely sensitive to loss of core autophagy genes within the first 7 days. However, weeks later, rare populations of originally autophagy dependent cells were found that could circumvent autophagy inhibition. Analysis of these rare clones revealed that in the process of circumventing loss of autophagy, the cells upregulated NRF2 signaling to maintain protein homeostasis and consequently become more sensitive to proteasome inhibition as well as knock down of NRF2. This review highlights recent publications regarding the role of autophagy in cancer and potential mechanisms cancer cells may be able to commandeer to circumvent autophagy inhibition. We hope to make significant clinical advances by understanding if and when cancer cells will become resistant to autophagy inhibition, and pre-clinical studies may be able to provide insight into the best combinatorial therapies to prevent tumor relapse while on autophagy inhibitors.


Subject(s)
Autophagy/genetics , Carcinogenesis/genetics , NF-E2-Related Factor 2/genetics , Neoplasms/genetics , CRISPR-Cas Systems/genetics , Cell Lineage/genetics , Clonal Evolution/genetics , Humans , Neoplasms/pathology , Neoplasms/therapy , Signal Transduction/genetics
16.
Dev Cell ; 50(6): 690-703.e6, 2019 09 23.
Article in English | MEDLINE | ID: mdl-31378590

ABSTRACT

While autophagy is thought to be an essential process in some cancer cells, it is unknown if or how such cancer cells can circumvent autophagy inhibition. To address this, we developed a CRISPR/Cas9 assay with dynamic live-cell imaging to measure acute effects of knockout (KO) of autophagy genes compared to known essential and non-essential genes. In some cancer cells, autophagy is as essential for cancer cell growth as mRNA transcription or translation or DNA replication. However, even these highly autophagy-dependent cancer cells evolve to circumvent loss of autophagy by upregulating NRF2, which is necessary and sufficient for autophagy-dependent cells to circumvent ATG7 KO and maintain protein homeostasis. Importantly, however, this adaptation increases susceptibly to proteasome inhibitors. These studies identify a common mechanism of acquired resistance to autophagy inhibition and show that selection to avoid tumor cell dependency on autophagy creates new, potentially actionable cancer cell susceptibilities.


Subject(s)
Adaptation, Physiological , Autophagy , NF-E2-Related Factor 2/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Up-Regulation , Adaptation, Physiological/drug effects , Autophagy/drug effects , Autophagy-Related Protein 7/metabolism , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Cell Survival/drug effects , Clone Cells , Gene Knockout Techniques , Genes, Essential , Humans , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Ribonucleoproteins/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects
17.
Autophagy ; 15(4): 735-737, 2019 04.
Article in English | MEDLINE | ID: mdl-30653391

ABSTRACT

SQSTM1/p62 facilitates responses to various cellular stresses and has been implicated in human diseases. This protein functions as a major cytoplasmic signaling hub and has multiple binding partners, including arginylated (Nt-R) proteins that are recognized by the ZZ domain of SQSTM1/p62 (SQSTM1/p62ZZ). We have determined the molecular mechanism of Nt-R recognition using a combination of biochemical and NMR approaches and obtained the crystal structure of SQSTM1/p62ZZ in complex with Nt-R. We found that binding of SQSTM1/p62ZZ to Nt-R induces SQSTM1/p62 puncta formation and macroautophagy/autophagy and identified a regulatory linker (RL) region of SQSTM1/p62 that associates with SQSTM1/p62ZZ in vitro. Our findings suggest a mechanism for SQSTM1/p62 autoregulation that can be essential in mediating autophagy.


Subject(s)
Autophagy , Humans , Sequestosome-1 Protein , Signal Transduction
18.
Nat Commun ; 9(1): 4373, 2018 10 22.
Article in English | MEDLINE | ID: mdl-30349045

ABSTRACT

Autophagic receptor p62 is a critical mediator of cell detoxification, stress response, and metabolic programs and is commonly deregulated in human diseases. The diverse functions of p62 arise from its ability to interact with a large set of ligands, such as arginylated (Nt-R) substrates. Here, we describe the structural mechanism for selective recognition of Nt-R by the ZZ domain of p62 (p62ZZ). We show that binding of p62ZZ to Nt-R substrates stimulates p62 aggregation and macroautophagy and is required for autophagic targeting of p62. p62 is essential for mTORC1 activation in response to arginine, but it is not a direct sensor of free arginine in the mTORC1 pathway. We identified a regulatory linker (RL) region in p62 that binds p62ZZ in vitro and may modulate p62 function. Our findings shed new light on the mechanistic and functional significance of the major cytosolic adaptor protein p62 in two fundamental signaling pathways.


Subject(s)
Autophagy/physiology , Sequestosome-1 Protein/metabolism , Autophagy/genetics , Cell Line , Crystallography, X-Ray , Flow Cytometry , HEK293 Cells , Humans , Immunohistochemistry , Magnetic Resonance Spectroscopy , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Protein Binding , Sequestosome-1 Protein/genetics , Signal Transduction , Spectrometry, Fluorescence
19.
Proc Natl Acad Sci U S A ; 115(36): E8479-E8488, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30127018

ABSTRACT

Molecular alterations that confer phenotypic advantages to tumors can also expose specific therapeutic vulnerabilities. To search for potential treatments that would selectively affect metastatic cells, we examined the sensitivity of lineage-related human bladder cancer cell lines with different lung colonization abilities to chloroquine (CQ) or bafilomycin A1, which are inhibitors of lysosome function and autophagy. Both CQ and bafilomycin A1 were more cytotoxic in vitro to highly metastatic cells compared with their less metastatic counterparts. Genetic inactivation of macroautophagy regulators and lysosomal proteins indicated that this was due to greater reliance on the lysosome but not upon macroautophagy. To identify the mechanism underlying these effects, we generated cells resistant to CQ in vitro. Surprisingly, selection for in vitro CQ resistance was sufficient to alter gene expression patterns such that unsupervised cluster analysis of whole-transcriptome data indicated that selection for CQ resistance alone created tumor cells that were more similar to the poorly metastatic parental cells from which the metastatic cells were derived; importantly, these tumor cells also had diminished metastatic ability in vivo. These effects were mediated in part by differential expression of the transcriptional regulator ID4 (inhibitor of DNA binding 4); depletion of ID4 both promoted in vitro CQ sensitivity and restored lung colonization and metastasis of CQ-resistant cells. These data demonstrate that selection for metastasis ability confers selective vulnerability to lysosomal inhibitors and identify ID4 as a potential biomarker for the use of lysosomal inhibitors to reduce metastasis in patients.


Subject(s)
Chloroquine/pharmacology , Drug Resistance, Neoplasm/drug effects , Lung Neoplasms , Lysosomes/metabolism , Macrolides/pharmacology , Urinary Bladder Neoplasms , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitor of Differentiation Proteins/biosynthesis , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lysosomes/pathology , Mice , Neoplasm Metastasis , Neoplasm Proteins/biosynthesis , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
20.
Mol Cancer Ther ; 17(10): 2112-2122, 2018 10.
Article in English | MEDLINE | ID: mdl-30026382

ABSTRACT

Antiangiogenic therapy used in treatment of metastatic colorectal cancer (mCRC) inevitably succumbs to treatment resistance. Upregulation of MET may play an essential role to acquired anti-VEGF resistance. We previously reported that cabozantinib (XL184), an inhibitor of receptor tyrosine kinases (RTK) including MET, AXL, and VEGFR2, had potent antitumor effects in mCRC patient-derived tumor explant models. In this study, we examined the mechanisms of cabozantinib sensitivity, using regorafenib as a control. The tumor growth inhibition index (TGII) was used to compare treatment effects of cabozantinib 30 mg/kg daily versus regorafenib 10 mg/kg daily for a maximum of 28 days in 10 PDX mouse models. In vivo angiogenesis and glucose uptake were assessed using dynamic contrast-enhanced (DCE)-MRI and [18F]-FDG-PET imaging, respectively. RNA-Seq, RTK assay, and immunoblotting analysis were used to evaluate gene pathway regulation in vivo and in vitro Analysis of TGII demonstrated significant antitumor effects with cabozantinib compared with regorafenib (average TGII 3.202 vs. 48.48, respectively; P = 0.007). Cabozantinib significantly reduced vascularity and glucose uptake compared with baseline. Gene pathway analysis showed that cabozantinib significantly decreased protein activity involved in glycolysis and upregulated proteins involved in autophagy compared with control, whereas regorafenib did not. The combination of two separate antiautophagy agents, SBI-0206965 and chloroquine, plus cabozantinib increased apoptosis in vitro Cabozantinib demonstrated significant antitumor activity, reduction in tumor vascularity, increased autophagy, and altered cell metabolism compared with regorafenib. Our findings support further evaluation of cabozantinib and combinational approaches targeting autophagy in colorectal cancer. Mol Cancer Ther; 17(10); 2112-22. ©2018 AACR.


Subject(s)
Anilides/pharmacology , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis , Biomarkers, Tumor , Cell Line, Tumor , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/drug therapy , Disease Models, Animal , Female , Humans , Magnetic Resonance Imaging , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Positron-Emission Tomography , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL