Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters










Publication year range
1.
Cancer Res Commun ; 3(11): 2400-2411, 2023 11 24.
Article in English | MEDLINE | ID: mdl-37882674

ABSTRACT

We have previously identified alveolar type II cell as the cell-of-origin of KrasG12D-induced lung adenocarcinoma using cell lineage-specific inducible Cre mouse models. Using gain-of-function and loss-of-function genetic models, we discovered that active Notch signaling and low Sox2 levels dictate the ability of type II cells to proliferate and progress into lung adenocarcinoma upon KrasG12D activation. Here, we examine the phenotype of type II cells after Kras activation and find evidence for proliferation of cells that coexpress type I and type II markers. Three-dimensional organoid culture and transplantation studies determine that these dual-positive cells are highly plastic and tumor initiating in vivo. RNA sequencing analysis reveals that these dual-positive cells are enriched in Ras/MAPK, EGFR, and Notch pathways. Furthermore, the proliferation of these cells requires active Notch signaling and is inhibited by genetic/chemical Sox2 upregulation. Our findings could provide new therapeutic strategies to target KRAS-activated lung adenocarcinomas. SIGNIFICANCE: Identification of progenitor like tumor-initiating cells in KRAS-mutant lung adenocarcinoma may allow development of novel targeted therapeutics.


Subject(s)
Adenocarcinoma of Lung , Adenocarcinoma , Lung Neoplasms , Mice , Animals , Proto-Oncogene Proteins p21(ras)/genetics , Adenocarcinoma/genetics , Lung Neoplasms/genetics , Cell Plasticity , Cell Proliferation/genetics , Adenocarcinoma of Lung/genetics
2.
Neurooncol Adv ; 5(1): vdad095, 2023.
Article in English | MEDLINE | ID: mdl-37781087

ABSTRACT

Background: Medulloblastoma is the most common malignant pediatric brain tumor, and leptomeningeal dissemination (LMD) of medulloblastoma both portends a poorer prognosis at diagnosis and is incurable at recurrence. The biological mechanisms underlying LMD are unclear. The Abelson (ABL) tyrosine kinase family members, ABL1 and ABL2, have been implicated in cancer cell migration, invasion, adhesion, metastasis, and chemotherapy resistance, and are upstream mediators of the oncogene c-MYC in fibroblasts and lung cancer cells. However, their role in medulloblastoma has not yet been explored. The purpose of this work was to elucidate the role of ABL1/2 in medulloblastoma LMD. Methods: ABL1 and ABL2 mRNA expression of patient specimens was analyzed. shRNA knockdowns of ABL1/2 and pharmacologic inhibition of ABL1/2 were used for in vitro and in vivo analyses of medulloblastoma LMD. RNA sequencing of ABL1/2 genetic knockdown versus scrambled control medulloblastoma was completed. Results: ABL1/2 mRNA is highly expressed in human medulloblastoma and pharmacologic inhibition of ABL kinases resulted in cytotoxicity. Knockdown of ABL1/2 resulted in decreased adhesion of medulloblastoma cells to the extracellular matrix protein, vitronectin (P = .0013), and significantly decreased tumor burden in a mouse model of medulloblastoma LMD with improved overall survival (P = .0044). Furthermore, both pharmacologic inhibition of ABL1/2 and ABL1/2 knockdown resulted in decreased expression of c-MYC, identifying a putative signaling pathway, and genes/pathways related to oncogenesis and neurodevelopment were differentially expressed between ABL1/2 knockdown and control medulloblastoma cells. Conclusions: ABL1 and ABL2 have potential roles in medulloblastoma LMD upstream of c-MYC expression.

3.
Cancers (Basel) ; 15(19)2023 Sep 24.
Article in English | MEDLINE | ID: mdl-37835395

ABSTRACT

The Hippo pathway transcriptional co-activators, YES-associated protein (YAP) and Transcriptional Co-Activator with PDZ Binding Motif (TAZ), have both been linked to tumor progression and metastasis. These two proteins possess overlapping and distinct functions, and their activities lead to the expression of genes involved in multiple cellular processes, including cell proliferation, survival, and migration. The dysregulation of YAP/TAZ-dependent cellular processes can result in altered tumor growth and metastasis. In addition to their well-documented roles in the regulation of cancer cell growth, survival, migration, and invasion, the YAP/TAZ-dependent signaling pathways have been more recently implicated in cellular processes that promote metastasis and therapy resistance in several solid tumor types. This review highlights the role of YAP/TAZ signaling networks in the regulation of tumor cell plasticity mediated by hybrid and reversible epithelial-mesenchymal transition (EMT) states, and the promotion of cancer stem cell/progenitor phenotypes. Mechanistically, YAP and TAZ regulate these cellular processes by targeting transcriptional networks. In this review, we detail recently uncovered mechanisms whereby YAP and TAZ mediate tumor growth, metastasis, and therapy resistance, and discuss new therapeutic strategies to target YAP/TAZ function in various solid tumor types. Understanding the distinct and overlapping roles of YAP and TAZ in multiple cellular processes that promote tumor progression to metastasis is expected to enable the identification of effective therapies to treat solid tumors through the hyper-activation of YAP and TAZ.

4.
Proc Natl Acad Sci U S A ; 120(16): e2210418120, 2023 04 18.
Article in English | MEDLINE | ID: mdl-37040401

ABSTRACT

The hypoxia-inducible factor 1-α (HIF-1α) enables cells to adapt and respond to hypoxia (Hx), and the activity of this transcription factor is regulated by several oncogenic signals and cellular stressors. While the pathways controlling normoxic degradation of HIF-1α are well understood, the mechanisms supporting the sustained stabilization and activity of HIF-1α under Hx are less clear. We report that ABL kinase activity protects HIF-1α from proteasomal degradation during Hx. Using a fluorescence-activated cell sorting (FACS)-based CRISPR/Cas9 screen, we identified HIF-1α as a substrate of the cleavage and polyadenylation specificity factor-1 (CPSF1), an E3-ligase which targets HIF-1α for degradation in the presence of an ABL kinase inhibitor in Hx. We show that ABL kinases phosphorylate and interact with CUL4A, a cullin ring ligase adaptor, and compete with CPSF1 for CUL4A binding, leading to increased HIF-1α protein levels. Further, we identified the MYC proto-oncogene protein as a second CPSF1 substrate and show that active ABL kinase protects MYC from CPSF1-mediated degradation. These studies uncover a role for CPSF1 in cancer pathobiology as an E3-ligase antagonizing the expression of the oncogenic transcription factors, HIF-1α and MYC.


Subject(s)
Gene Expression Regulation , Transcription Factors , Humans , Cullin Proteins/metabolism , Hypoxia , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Genes, abl , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Cleavage And Polyadenylation Specificity Factor/metabolism
5.
bioRxiv ; 2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36711936

ABSTRACT

Targeted therapies have revolutionized cancer chemotherapy. Unfortunately, most patients develop multifocal resistance to these drugs within a matter of months. Here, we used a high-throughput phenotypic small molecule screen to identify MCB-613 as a compound that selectively targets EGFR-mutant, EGFR inhibitor-resistant non-small cell lung cancer (NSCLC) cells harboring diverse resistance mechanisms. Subsequent proteomic and functional genomic screens involving MCB-613 identified its target in this context to be KEAP1, revealing that this gene is selectively essential in the setting of EGFR inhibitor resistance. In-depth molecular characterization demonstrated that (1) MCB-613 binds KEAP1 covalently; (2) a single molecule of MCB-613 is capable of bridging two KEAP1 monomers together; and, (3) this modification interferes with the degradation of canonical KEAP1 substrates such as NRF2. Surprisingly, NRF2 knockout sensitizes cells to MCB-613, suggesting that the drug functions through modulation of an alternative KEAP1 substrate. Together, these findings advance MCB-613 as a new tool for exploiting the selective essentiality of KEAP1 in drug-resistant, EGFR-mutant NSCLC cells.

6.
iScience ; 25(10): 105114, 2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36185377

ABSTRACT

Epithelial cells of diverse tissues are characterized by the presence of a single apical domain. In the lung, electron microscopy studies have suggested that alveolar type-2 epithelial cells (AT2s) en face multiple alveolar sacs. However, apical and basolateral organization of the AT2s and their establishment during development and remodeling after injury repair remain unknown. Thick tissue imaging and electron microscopy revealed that a single AT2 can have multiple apical domains that enface multiple alveoli. AT2s gradually establish multi-apical domains post-natally, and they are maintained throughout life. Lineage tracing, live imaging, and selective cell ablation revealed that AT2s dynamically reorganize multi-apical domains during injury repair. Single-cell transcriptome signatures of residual AT2s revealed changes in cytoskeleton and cell migration. Significantly, cigarette smoke and oncogene activation lead to dysregulation of multi-apical domains. We propose that the multi-apical domains of AT2s enable them to be poised to support the regeneration of a large array of alveolar sacs.

7.
Cell Rep ; 40(9): 111268, 2022 08 30.
Article in English | MEDLINE | ID: mdl-36044842

ABSTRACT

Patients with human epidermal growth factor receptor 2-positive (HER2+/ERBB2) breast cancer often present with brain metastasis. HER2-targeted therapies have not been successful to treat brain metastases in part due to poor blood-brain barrier (BBB) penetrance and emergence of resistance. Here, we report that Abelson (ABL) kinase allosteric inhibitors improve overall survival and impair HER2+ brain metastatic outgrowth in vivo. Mechanistically, ABL kinases phosphorylate the RNA-binding protein Y-box-binding protein 1 (YB-1). ABL kinase inhibition disrupts binding of YB-1 to the ERBB2 mRNA and impairs translation, leading to a profound decrease in HER2 protein levels. ABL-dependent tyrosine phosphorylation of YB-1 promotes HER2 translation. Notably, loss of YB-1 inhibits brain metastatic outgrowth and impairs expression of a subset of ABL-dependent brain metastatic targets. These data support a role for ABL kinases in the translational regulation of brain metastatic targets through YB-1 and offer a therapeutic target for HER2+ brain metastasis patients.


Subject(s)
Brain Neoplasms , Breast Neoplasms , Proto-Oncogene Proteins c-abl , Y-Box-Binding Protein 1 , Brain/metabolism , Brain Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/secondary , Cell Line, Tumor , Female , Humans , Proto-Oncogene Proteins c-abl/metabolism , Receptor, ErbB-2/metabolism , Y-Box-Binding Protein 1/genetics
8.
Cell Rep ; 37(4): 109880, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34706244

ABSTRACT

Targeting mitochondrial metabolism has emerged as a treatment option for cancer patients. The ABL tyrosine kinases promote metastasis, and enhanced ABL signaling is associated with a poor prognosis in lung adenocarcinoma patients. Here we show that ABL kinase allosteric inhibitors impair mitochondrial integrity and decrease oxidative phosphorylation. To identify metabolic vulnerabilities that enhance this phenotype, we utilized a CRISPR/Cas9 loss-of-function screen and identified HMG-CoA reductase, the rate-limiting enzyme of the mevalonate pathway and target of statin therapies, as a top-scoring sensitizer to ABL inhibition. Combination treatment with ABL allosteric inhibitors and statins decreases metastatic lung cancer cell survival in vitro in a synergistic manner. Notably, combination therapy in mouse models of lung cancer brain metastasis and therapy resistance impairs metastatic colonization with a concomitant increase in animal survival. Thus, metabolic combination therapy might be effective to decrease metastatic outgrowth, leading to increased survival for lung cancer patients with advanced disease.


Subject(s)
Apoptosis/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Oncogene Proteins v-abl/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Allosteric Regulation/drug effects , Allosteric Regulation/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Drug Synergism , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Nude , Neoplasm Metastasis , Oncogene Proteins v-abl/genetics , Oncogene Proteins v-abl/metabolism , Signal Transduction/genetics , Xenograft Model Antitumor Assays
9.
Cell Commun Signal ; 19(1): 59, 2021 05 22.
Article in English | MEDLINE | ID: mdl-34022881

ABSTRACT

The ABL kinases, ABL1 and ABL2, promote tumor progression and metastasis in various solid tumors. Recent reports have shown that ABL kinases have increased expression and/or activity in solid tumors and that ABL inactivation impairs metastasis. The therapeutic effects of ABL inactivation are due in part to ABL-dependent regulation of diverse cellular processes related to the epithelial to mesenchymal transition and subsequent steps in the metastatic cascade. ABL kinases target multiple signaling pathways required for promoting one or more steps in the metastatic cascade. These findings highlight the potential utility of specific ABL kinase inhibitors as a novel treatment paradigm for patients with advanced metastatic disease. Video abstract.


Subject(s)
Epithelial-Mesenchymal Transition , Proto-Oncogene Proteins c-abl/metabolism , Cytoskeleton/metabolism , Disease Progression , Humans , Molecular Targeted Therapy , Neoplasm Metastasis , Proto-Oncogene Proteins c-abl/chemistry
10.
Mol Cancer Ther ; 20(3): 455-466, 2021 03.
Article in English | MEDLINE | ID: mdl-33402399

ABSTRACT

Paget's "seed and soil" hypothesis of metastatic spread has acted as a foundation of the field for over a century, with continued evolution as mechanisms of the process have been elucidated. The central nervous system (CNS) presents a unique soil through this lens, relatively isolated from peripheral circulation and immune surveillance with distinct cellular and structural composition. Research in primary and metastatic brain tumors has demonstrated that this tumor microenvironment (TME) plays an essential role in the growth of CNS tumors. In each case, the cancerous cells develop complex and bidirectional relationships that reorganize the local TME and reprogram the CNS cells, including endothelial cells, pericytes, astrocytes, microglia, infiltrating monocytes, and lymphocytes. These interactions create a structurally and immunologically permissive TME with malignant processes promoting positive feedback loops and systemic consequences. Strategies to interrupt interactions with the native CNS components, on "salting the soil," to create an inhospitable environment are promising in the preclinical setting. This review aims to examine the general and specific pathways thus far investigated in brain metastases and related work in glioma to identify targetable mechanisms that may have general application across the spectrum of intracranial tumors.


Subject(s)
Brain Neoplasms/pathology , Humans , Neoplasm Metastasis , Tumor Microenvironment
11.
Proc Natl Acad Sci U S A ; 117(52): 33486-33495, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33318173

ABSTRACT

Brain metastases are the most common intracranial tumors in adults and are associated with increased patient morbidity and mortality. Limited therapeutic options are currently available for the treatment of brain metastasis. Here, we report on the discovery of an actionable signaling pathway utilized by metastatic tumor cells whereby the transcriptional regulator Heat Shock Factor 1 (HSF1) drives a transcriptional program, divergent from its canonical role as the master regulator of the heat shock response, leading to enhanced expression of a subset of E2F transcription factor family gene targets. We find that HSF1 is required for survival and outgrowth by metastatic lung cancer cells in the brain parenchyma. Further, we identify the ABL2 tyrosine kinase as an upstream regulator of HSF1 protein expression and show that the Src-homology 3 (SH3) domain of ABL2 directly interacts with HSF1 protein at a noncanonical, proline-independent SH3 interaction motif. Pharmacologic inhibition of the ABL2 kinase using small molecule allosteric inhibitors, but not ATP-competitive inhibitors, disrupts this interaction. Importantly, knockdown as well as pharmacologic inhibition of ABL2 using allosteric inhibitors impairs expression of HSF1 protein and HSF1-E2F transcriptional gene targets. Collectively, these findings reveal a targetable ABL2-HSF1-E2F signaling pathway required for survival by brain-metastatic tumor cells.


Subject(s)
Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Brain Neoplasms/secondary , Heat Shock Transcription Factors/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Protein-Tyrosine Kinases/metabolism , Transcription, Genetic , Allosteric Regulation , Animals , Cell Line, Tumor , Cell Survival , E2F Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice, Nude , Up-Regulation/genetics
12.
PLoS One ; 15(10): e0241423, 2020.
Article in English | MEDLINE | ID: mdl-33119681

ABSTRACT

Mesenchymal stem cells (MSCs) are recruited and activated by solid tumors and play a role in tumor progression and metastasis. Here we show that MSCs promote metastasis in a panel of non-small cell lung cancer (NSCLC) cells. MSCs elicit transcriptional alterations in lung cancer cells leading to increased expression of factors implicated in the epithelial-to-mesenchymal transition (EMT) and secreted proteins including matrix metalloproteinase-9 (MMP9). MSCs enhance secretion of enzymatically active MMP9 in a panel of lung adenocarcinoma cells. High expression of MMP9 is linked to low survival rates in lung adenocarcinoma patients. Notably, we found that ABL tyrosine kinases are activated in MSC-primed lung cancer cells and functional ABL kinases are required for MSC-induced MMP9 expression, secretion and proteolytic activity. Importantly, ABL kinases are required for MSC-induced NSCLC metastasis. These data reveal an actionable target for inhibiting MSC-induced metastatic activity of lung adenocarcinoma cells through disruption of an ABL kinase-MMP9 signaling axis activated in MSC-primed lung cancer cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Matrix Metalloproteinase 9/metabolism , Mesenchymal Stem Cells/pathology , Proto-Oncogene Proteins c-abl/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Metastasis
13.
Cell Rep ; 29(11): 3421-3434.e8, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31825826

ABSTRACT

Brain metastases are a common consequence of advanced lung cancer, resulting in cranial neuropathies and increased mortality. Currently, there are no effective therapies to treat brain metastases due to a lack of actionable targets and a failure of systemic therapies to penetrate the blood-brain barrier (BBB). Here we identify an autocrine signaling axis required for lung adenocarcinoma brain metastasis, whereby nuclear accumulation of the TAZ transcriptional co-activator drives expression of a panel of transcripts enriched in brain metastases, including ABL2 and AXL, encoding for protein tyrosine kinases that engage in bidirectional signaling. Activation of ABL2 in turn promotes TAZ tyrosine phosphorylation and nuclear localization, establishing an autocrine AXL-ABL2-TAZ feed-forward signaling loop required for brain metastasis colonization. Notably, treatment with a BBB-penetrant ABL allosteric inhibitor or knockdown of ABL2, AXL, or TAZ markedly decreases brain metastases. These findings suggest that ABL and AXL inhibitors might be effective against brain metastases.


Subject(s)
Adenocarcinoma of Lung/metabolism , Brain Neoplasms/metabolism , Lung Neoplasms/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Transcription Factors/metabolism , Acyltransferases , Adenocarcinoma of Lung/pathology , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/secondary , Cell Line, Tumor , Female , HEK293 Cells , Humans , Lung Neoplasms/pathology , Mice , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pyrazoles/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction , Axl Receptor Tyrosine Kinase
14.
Oncotarget ; 10(57): 6045-6046, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31666936

ABSTRACT

[This corrects the article DOI: 10.18632/oncotarget.26740.].

15.
Clin Cancer Res ; 25(22): 6570-6580, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31213459

ABSTRACT

The incidence of brain metastases is increasing as cancer therapies improve and patients live longer, providing new challenges to the multidisciplinary teams that care for these patients. Brain metastatic cancer cells possess unique characteristics that allow them to penetrate the blood-brain barrier, colonize the brain parenchyma, and persist in the intracranial environment. In addition, brain metastases subvert the innate and adaptive immune system, permitting evasion of the antitumor immune response. Better understanding of the above mechanisms will allow for development and delivery of more effective therapies for brain metastases. In this review, we outline the molecular mechanisms underlying development, survival, and immunosuppression of brain metastases. We also discuss current and emerging treatment strategies, including surgery, radiation, disease-specific and mutation-targeted systemic therapy, and immunotherapy.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/secondary , Brain Neoplasms/therapy , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain Neoplasms/metabolism , Cellular Microenvironment , Combined Modality Therapy , Disease Management , Disease Susceptibility , Humans , Prognosis , Treatment Outcome , Tumor Escape
16.
Oncotarget ; 10(20): 1874-1886, 2019 Mar 08.
Article in English | MEDLINE | ID: mdl-30956771

ABSTRACT

Lung cancer is the leading cause of cancer mortality in the United States, with an overall five-year survival rate of ~16%. Non-small cell lung cancer (NSCLC) accounts for ~80% of all lung cancer cases, and the majority (40%) of these are adenocarcinomas. Loss of function point mutations in TP53 (46%) and activating mutations in KRAS (33%) are the most common mutations in human lung adenocarcinomas. Because neither of these genetic alterations are clinically actionable, chemotherapy remains the mainstay of treatment in patients with oncogenic KRAS driver mutations. However, chemoresistance to genotoxic agents such as docetaxel remains a major clinical challenge facing lung cancer patients. Here we show that ABL kinase allosteric inhibitors can be effectively used for the treatment of KrasG12D/+; p53-/- lung adenocarcinomas in an autochthonous mouse model. Unexpectedly, we found that treatment of tumor-bearing mice with an ABL allosteric inhibitor promoted differentiation of lung adenocarcinomas from poorly differentiated tumors expressing basal cell markers to tumors expressing terminal differentiation markers in vivo, which rendered lung adenocarcinomas susceptible to chemotherapy. These findings uncover a novel therapeutic approach for the treatment of lung adenocarcinomas with poor response to chemotherapy.

17.
Proc Natl Acad Sci U S A ; 116(5): 1603-1612, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30655340

ABSTRACT

Current therapeutic interventions for the treatment of respiratory infections are hampered by the evolution of multidrug resistance in pathogens as well as the lack of effective cellular targets. Despite the identification of multiple region-specific lung progenitor cells, the identity of molecules that might be therapeutically targeted in response to infections to promote activation of progenitor cell types remains elusive. Here, we report that loss of Abl1 specifically in SCGB1A1-expressing cells leads to a significant increase in the proliferation and differentiation of bronchiolar epithelial cells, resulting in dramatic expansion of an SCGB1A1+ airway cell population that coexpresses SPC, a marker for type II alveolar cells that promotes alveolar regeneration following bacterial pneumonia. Furthermore, treatment with an Abl-specific allosteric inhibitor enhanced regeneration of the alveolar epithelium and promoted accelerated recovery of mice following pneumonia. These data reveal a potential actionable target that may be exploited for efficient recovery after pathogen-induced infections.


Subject(s)
Lung/metabolism , Lung/physiopathology , Pneumonia, Bacterial/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Regeneration/physiology , Stem Cells/metabolism , Uteroglobin/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/physiology , Animals , Bronchioles/metabolism , Bronchioles/physiopathology , Cell Differentiation/physiology , Cell Line , Female , Humans , Male , Mice , Mice, Inbred C57BL , Pneumonia, Bacterial/physiopathology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/physiopathology , Respiratory Mucosa/metabolism , Respiratory Mucosa/physiopathology , Stem Cells/physiology
18.
JCI Insight ; 1(21): e89647, 2016 Dec 22.
Article in English | MEDLINE | ID: mdl-28018973

ABSTRACT

Current therapies to treat non-small cell lung carcinoma (NSCLC) have proven ineffective owing to transient, variable, and incomplete responses. Here we show that ABL kinases, ABL1 and ABL2, promote metastasis of lung cancer cells harboring EGFR or KRAS mutations. Inactivation of ABL kinases suppresses NSCLC metastasis to brain and bone, and other organs. ABL kinases are required for expression of prometastasis genes. Notably, ABL1 and ABL2 depletion impairs extravasation of lung adenocarcinoma cells into the lung parenchyma. We found that ABL-mediated activation of the TAZ and ß-catenin transcriptional coactivators is required for NSCLC metastasis. ABL kinases activate TAZ and ß-catenin by decreasing their interaction with the ß-TrCP ubiquitin ligase, leading to increased protein stability. High-level expression of ABL1, ABL2, and a subset of ABL-dependent TAZ- and ß-catenin-target genes correlates with shortened survival of lung adenocarcinoma patients. Thus, ABL-specific allosteric inhibitors might be effective to treat metastatic lung cancer with an activated ABL pathway signature.

19.
J Clin Invest ; 126(12): 4482-4496, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27797343

ABSTRACT

Cellular identity in metazoan organisms is frequently established through lineage-specifying transcription factors, which control their own expression through transcriptional positive feedback, while antagonizing the developmental networks of competing lineages. Here, we have uncovered a distinct positive feedback loop that arises from the reciprocal stabilization of the tyrosine kinase ABL and the transcriptional coactivator TAZ. Moreover, we determined that this loop is required for osteoblast differentiation and embryonic skeletal formation. ABL potentiated the assembly and activation of the RUNX2-TAZ master transcription factor complex that is required for osteoblastogenesis, while antagonizing PPARγ-mediated adipogenesis. ABL also enhanced TAZ nuclear localization and the formation of the TAZ-TEAD complex that is required for osteoblast expansion. Last, we have provided genetic data showing that regulation of the ABL-TAZ amplification loop lies downstream of the adaptor protein 3BP2, which is mutated in the craniofacial dysmorphia syndrome cherubism. Our study demonstrates an interplay between ABL and TAZ that controls the mesenchymal maturation program toward the osteoblast lineage and is mechanistically distinct from the established model of lineage-specific maturation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Nucleus/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Osteoblasts/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Nucleus/genetics , Cherubism/genetics , Cherubism/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , HEK293 Cells , Humans , Mice , Mice, Knockout , PPAR gamma/genetics , PPAR gamma/metabolism , Proto-Oncogene Proteins c-abl/genetics , Trans-Activators
20.
Sci Signal ; 9(413): ra12, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26838548

ABSTRACT

Bone metastases occur in up to 70% of advanced breast cancer. For most patients with breast cancer, bone metastases are predominantly osteolytic. Interactions between tumor cells and stromal cells in the bone microenvironment drive osteolytic bone metastasis, a process that requires the activation of osteoclasts, cells that break down bone. We report that ABL kinases promoted metastasis of breast cancer cells to bone by regulating the crosstalk between tumor cells and the bone microenvironment. ABL kinases protected tumor cells from apoptosis induced by TRAIL (TNF-related apoptosis-inducing ligand), activated the transcription factor STAT5, and promoted osteolysis through the STAT5-dependent expression of genes encoding the osteoclast-activating factors interleukin-6 (IL-6) and matrix metalloproteinase 1 (MMP1). Furthermore, in breast cancer cells, ABL kinases increased the abundance of the Hippo pathway mediator TAZ and the expression of TAZ-dependent target genes that promote bone metastasis. Knockdown of ABL kinases or treatment with ABL-specific allosteric inhibitor impaired osteolytic metastasis of breast cancer cells in mice. These findings revealed a role for ABL kinases in regulating tumor-bone interactions and provide a rationale for using ABL-specific inhibitors to limit breast cancer metastasis to bone.


Subject(s)
Bone Neoplasms/metabolism , Breast Neoplasms/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Osteolysis/metabolism , Proto-Oncogene Proteins c-abl/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Animals , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Heterografts , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Osteolysis/genetics , Osteolysis/pathology , Proto-Oncogene Proteins c-abl/genetics , STAT5 Transcription Factor/genetics , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...