Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Proteome Res ; 23(5): 1547-1558, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38619923

ABSTRACT

Circadian misalignment due to night work has been associated with an elevated risk for chronic diseases. We investigated the effects of circadian misalignment using shotgun protein profiling of peripheral blood mononuclear cells taken from healthy humans during a constant routine protocol, which was conducted immediately after participants had been subjected to a 3-day simulated night shift schedule or a 3-day simulated day shift schedule. By comparing proteomic profiles between the simulated shift conditions, we identified proteins and pathways that are associated with the effects of circadian misalignment and observed that insulin regulation pathways and inflammation-related proteins displayed markedly different temporal patterns after simulated night shift. Further, by integrating the proteomic profiles with previously assessed metabolomic profiles in a network-based approach, we found key associations between circadian dysregulation of protein-level pathways and metabolites of interest in the context of chronic metabolic diseases. Endogenous circadian rhythms in circulating glucose and insulin differed between the simulated shift conditions. Overall, our results suggest that circadian misalignment is associated with a tug of war between central clock mechanisms controlling insulin secretion and peripheral clock mechanisms regulating insulin sensitivity, which may lead to adverse long-term outcomes such as diabetes and obesity. Our study provides a molecular-level mechanism linking circadian misalignment and adverse long-term health consequences of night work.


Subject(s)
Circadian Rhythm , Inflammation , Insulin , Leukocytes, Mononuclear , Humans , Leukocytes, Mononuclear/metabolism , Insulin/metabolism , Insulin/blood , Inflammation/metabolism , Inflammation/blood , Male , Adult , Shift Work Schedule , Female , Proteomics/methods , Blood Glucose/metabolism , Signal Transduction , Insulin Resistance , Young Adult
2.
Pigment Cell Melanoma Res ; 34(5): 955-965, 2021 09.
Article in English | MEDLINE | ID: mdl-34160901

ABSTRACT

Solar ultraviolet B radiation (UVB) is one of the leading causes of various skin conditions, including photoaging, sunburn erythema, and melanoma. As a protective response, the skin has inbuilt defense mechanisms, including DNA repair, cell cycle, apoptosis, and melanin synthesis. Though DNA repair, cell cycle, and apoptosis are clock controlled, the circadian mechanisms associated with melanin synthesis are not well understood. Using human melanocytes and melanoma cells under synchronized clock conditions, we observed that the microphthalmia-associated transcription factor (MITF), a rate-limiting protein in melanin synthesis, is expressed rhythmically with 24-hr periodicity in the presence of circadian clock protein, BMAL1. Furthermore, we demonstrated that BMAL1 binds to the promoter region of MITF and transcriptionally regulates its expression, which positively influences melanin synthesis. Finally, we report that an increase in melanin levels due to BMAL1 overexpression protects human melanoma cells from UVB. In conclusion, our studies provide novel insights into the mechanistic role of the circadian clock in melanin synthesis and protection against UVB-mediated DNA damage and genomic instability.


Subject(s)
ARNTL Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Neoplasm Proteins/metabolism , ARNTL Transcription Factors/genetics , Animals , Humans , Melanoma/genetics , Melanoma/pathology , Mice , Microphthalmia-Associated Transcription Factor/genetics , Neoplasm Proteins/genetics
3.
J Pineal Res ; 70(3): e12726, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33638890

ABSTRACT

Circadian disruption has been identified as a risk factor for health disorders such as obesity, cardiovascular disease, and cancer. Although epidemiological studies suggest an increased risk of various cancers associated with circadian misalignment due to night shift work, the underlying mechanisms have yet to be elucidated. We sought to investigate the potential mechanistic role that circadian disruption of cancer hallmark pathway genes may play in the increased cancer risk in shift workers. In a controlled laboratory study, we investigated the circadian transcriptome of cancer hallmark pathway genes and associated biological pathways in circulating leukocytes obtained from healthy young adults during a 24-hour constant routine protocol following 3 days of simulated day shift or night shift. The simulated night shift schedule significantly altered the normal circadian rhythmicity of genes involved in cancer hallmark pathways. A DNA repair pathway showed significant enrichment of rhythmic genes following the simulated day shift schedule, but not following the simulated night shift schedule. In functional assessments, we demonstrated that there was an increased sensitivity to both endogenous and exogenous sources of DNA damage after exposure to simulated night shift. Our results suggest that circadian dysregulation of DNA repair may increase DNA damage and potentiate elevated cancer risk in night shift workers.


Subject(s)
Biomarkers, Tumor/genetics , Chronobiology Disorders/etiology , Circadian Rhythm , DNA Damage , DNA Repair , Neoplasms/etiology , Shift Work Schedule/adverse effects , Transcriptome , Activity Cycles , Adult , Chronobiology Disorders/genetics , Chronobiology Disorders/physiopathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasms/genetics , Neoplasms/pathology , Risk Assessment , Risk Factors , Sleep , Time Factors , Young Adult
4.
FASEB J ; 34(2): 3347-3358, 2020 02.
Article in English | MEDLINE | ID: mdl-31919902

ABSTRACT

Radiation therapy (RT) is commonly used to treat solid tumors of the breast, lung, and esophagus; however, the heart is an unintentional target of ionizing radiation (IR). IR exposure to the heart results in chronic toxicities including heart failure. We hypothesize that the circadian system plays regulatory roles in minimizing the IR-induced cardiotoxicity. We treated mice in control (Day Shift), environmentally disrupted (Rotating Shift), and genetically disrupted (Per 1/2 mutant) circadian conditions with 18 Gy of IR to the heart. Compared to control mice, circadian clock disruption significantly exacerbated post-IR systolic dysfunction (by ultrasound echocardiography) and increased fibrosis in mice. At the cellular level, Bmal1 protein bound to Atm, Brca1, and Brca2 promoter regions and its expression level was inversely correlated with the DNA damage levels based on the state of the clock. Further studies with circadian synchronized cardiomyocytes revealed that Bmal1 depletion increased the IR-induced DNA damage and apoptosis. Collectively, these findings suggest that the circadian clock protects from IR-induced toxicity and potentially impacts RT treatment outcome in cancer patients through IR-induced DNA damage responses.


Subject(s)
Myocytes, Cardiac/metabolism , Period Circadian Proteins/genetics , Radiation Injuries, Experimental/genetics , Animals , Apoptosis , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Cell Line , DNA Damage , Mice , Mice, Inbred C57BL , Mutation , Myocytes, Cardiac/physiology , Myocytes, Cardiac/radiation effects , Promoter Regions, Genetic , Radiation Injuries, Experimental/metabolism , Radiation, Ionizing , Rats , Systole
5.
PLoS Biol ; 16(10): e2004204, 2018 10.
Article in English | MEDLINE | ID: mdl-30296263

ABSTRACT

Long noncoding RNAs constitute a major fraction of the eukaryotic transcriptome, and together with proteins, they intricately fine-tune various growth regulatory signals to control cellular homeostasis. Here, we describe the functional characterisation of a novel pair of long intergenic noncoding RNAs (lincRNAs) comprised of complementary, fully overlapping sense and antisense transcripts Genomic Instability Inducing RNA (Ginir) and antisense RNA of Ginir (Giniras), respectively, from mouse cells. This transcript pair is expressed in a spatiotemporal manner during embryonic development. The individual levels of the sense and antisense transcripts are finely balanced during embryonic growth and in adult tissues. Functional studies of the individual transcripts performed using overexpression and knock-down strategies in mouse cells has led to the discovery that Ginir RNA is a regulator of cellular proliferation and can act as an oncogene having a preeminent role in malignant transformation. Mechanistically, we demonstrate that the oncogenic function of Ginir is mediated by its interaction with centrosomal protein 112 (Cep112). Additionally, we establish here a specific interaction between Cep112 with breast cancer type 1 susceptibility protein (Brca1), another centrosome-associated protein. Next, we prove that the mutual interaction between Cep112 with Brca1 is significant for mitotic regulation and maintenance of genomic stability. Furthermore, we demonstrate that the Cep112 protein interaction with Brca1 protein is impaired when an elevated level of Ginir RNA is present in the cells, resulting in severe deregulation and abnormality in mitosis, leading to malignant transformation. Inhibiting the Ginir RNA function in transformed cells attenuates transformation and restores genomic stability. Together, these findings unravel, to our knowledge, a hitherto-unknown mechanism of oncogenesis mediated by a long noncoding RNA and establishes a unique role of Cep112-Brca1 interaction being modulated by Ginir RNA in maintaining mitotic fidelity.


Subject(s)
RNA, Long Noncoding/genetics , Animals , BRCA1 Protein , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Centrosome , Genome , Genomic Instability , Genomics/methods , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , RNA, Antisense/genetics , RNA, Untranslated/genetics , Transcriptome , Tumor Suppressor Proteins/physiology
6.
Proc Natl Acad Sci U S A ; 115(30): 7825-7830, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29991600

ABSTRACT

Misalignment between internal circadian rhythmicity and externally imposed behavioral schedules, such as occurs in shift workers, has been implicated in elevated risk of metabolic disorders. To determine underlying mechanisms, it is essential to assess whether and how peripheral clocks are disturbed during shift work and to what extent this is linked to the central suprachiasmatic nuclei (SCN) pacemaker and/or misaligned behavioral time cues. Investigating rhythms in circulating metabolites as biomarkers of peripheral clock disturbances may offer new insights. We evaluated the impact of misaligned sleep/wake and feeding/fasting cycles on circulating metabolites using a targeted metabolomics approach. Sequential plasma samples obtained during a 24-h constant routine that followed a 3-d simulated night-shift schedule, compared with a simulated day-shift schedule, were analyzed for 132 circulating metabolites. Nearly half of these metabolites showed a 24-h rhythmicity under constant routine following either or both simulated shift schedules. However, while traditional markers of the circadian clock in the SCN-melatonin, cortisol, and PER3 expression-maintained a stable phase alignment after both schedules, only a few metabolites did the same. Many showed reversed rhythms, lost their rhythms, or showed rhythmicity only under constant routine following the night-shift schedule. Here, 95% of the metabolites with a 24-h rhythmicity showed rhythms that were driven by behavioral time cues externally imposed during the preceding simulated shift schedule rather than being driven by the central SCN circadian clock. Characterization of these metabolite rhythms will provide insight into the underlying mechanisms linking shift work and metabolic disorders.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Fasting/blood , Gene Expression Regulation/physiology , Hydrocortisone/blood , Melatonin/blood , Period Circadian Proteins/biosynthesis , Adult , Female , Humans , Male
7.
Oncotarget ; 9(18): 14524-14538, 2018 Mar 06.
Article in English | MEDLINE | ID: mdl-29581861

ABSTRACT

Cisplatin is one of the most commonly used chemotherapeutic drugs; however, toxicity and tumor resistance limit its use. Studies using murine models and human subjects have shown that the time of day of cisplatin treatment influences renal and blood toxicities. We hypothesized that the mechanisms responsible for these outcomes are driven by the circadian clock. We conducted experiments using wild-type and circadian disrupted Per1/2-/- mice treated with cisplatin at selected morning (AM) and evening (PM) times. Wild-type mice treated in the evening showed an enhanced rate of removal of cisplatin-DNA adducts and less toxicity than the morning-treated mice. This temporal variation in toxicity was lost in the Per1/2-/- clock-disrupted mice, suggesting that the time-of-day effect is linked to the circadian clock. Observations in blood cells from humans subjected to simulated day and night shift schedules corroborated this view. Per1/2-/- mice also exhibited a more robust immune response and slower tumor growth rate, indicating that the circadian clock also influences the immune response to melanoma tumors. Our findings indicate that cisplatin chronopharmacology involves the circadian clock control of DNA repair as well as immune responses, and thus affects both cisplatin toxicity and tumor growth. This has important implications for chronochemotherapy in cancer patients, and also suggests that influencing the circadian clock (e.g., through bright light treatment) may be explored as a tool to improve patient outcomes.

8.
Cancer Res ; 77(12): 3181-3193, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28484075

ABSTRACT

TWIST1 is a transcription factor critical for development that can promote prostate cancer metastasis. During embryonic development, TWIST1 and HOXA9 are coexpressed in mouse prostate and then silenced postnatally. Here we report that TWIST1 and HOXA9 coexpression are reactivated in mouse and human primary prostate tumors and are further enriched in human metastases, correlating with survival. TWIST1 formed a complex with WDR5 and the lncRNA Hottip/HOTTIP, members of the MLL/COMPASS-like H3K4 methylases, which regulate chromatin in the Hox/HOX cluster during development. TWIST1 overexpression led to coenrichment of TWIST1 and WDR5 as well as increased H3K4me3 chromatin at the Hoxa9/HOXA9 promoter, which was dependent on WDR5. Expression of WDR5 and Hottip/HOTTIP was also required for TWIST1-induced upregulation of HOXA9 and aggressive cellular phenotypes such as invasion and migration. Pharmacologic inhibition of HOXA9 prevented TWIST1-induced aggressive prostate cancer cellular phenotypes in vitro and metastasis in vivo This study demonstrates a novel mechanism by which TWIST1 regulates chromatin and gene expression by cooperating with the COMPASS-like complex to increase H3K4 trimethylation at target gene promoters. Our findings highlight a TWIST1-HOXA9 embryonic prostate developmental program that is reactivated during prostate cancer metastasis and is therapeutically targetable. Cancer Res; 77(12); 3181-93. ©2017 AACR.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Homeodomain Proteins/metabolism , Neoplasm Invasiveness/genetics , Nuclear Proteins/metabolism , Prostatic Neoplasms/pathology , RNA, Long Noncoding/metabolism , Twist-Related Protein 1/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Chromatin , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/genetics , Gene Knockout Techniques , Heterografts , Histone-Lysine N-Methyltransferase/genetics , Homeodomain Proteins/genetics , Humans , Immunohistochemistry , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , Male , Mice , Neoplasm Invasiveness/pathology , Nuclear Proteins/genetics , Polymerase Chain Reaction , Prostatic Neoplasms/genetics , RNA, Long Noncoding/genetics , Twist-Related Protein 1/genetics
10.
Neoplasia ; 17(1): 16-31, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25622896

ABSTRACT

The TWIST1 gene has diverse roles in development and pathologic diseases such as cancer. TWIST1 is a dimeric basic helix-loop-helix (bHLH) transcription factor existing as TWIST1-TWIST1 or TWIST1-E12/47. TWIST1 partner choice and DNA binding can be influenced during development by phosphorylation of Thr125 and Ser127 of the Thr-Gln-Ser (TQS) motif within the bHLH of TWIST1. The significance of these TWIST1 phosphorylation sites for metastasis is unknown. We created stable isogenic prostate cancer cell lines overexpressing TWIST1 wild-type, phospho-mutants, and tethered versions. We assessed these isogenic lines using assays that mimic stages of cancer metastasis. In vitro assays suggested the phospho-mimetic Twist1-DQD mutation could confer cellular properties associated with pro-metastatic behavior. The hypo-phosphorylation mimic Twist1-AQA mutation displayed reduced pro-metastatic activity compared to wild-type TWIST1 in vitro, suggesting that phosphorylation of the TWIST1 TQS motif was necessary for pro-metastatic functions. In vivo analysis demonstrates that the Twist1-AQA mutation exhibits reduced capacity to contribute to metastasis, whereas the expression of the Twist1-DQD mutation exhibits proficient metastatic potential. Tethered TWIST1-E12 heterodimers phenocopied the Twist1-DQD mutation for many in vitro assays, suggesting that TWIST1 phosphorylation may result in heterodimerization in prostate cancer cells. Lastly, the dual phosphatidylinositide 3-kinase (PI3K)-mammalian target of rapamycin (mTOR) inhibitor BEZ235 strongly attenuated TWIST1-induced migration that was dependent on the TQS motif. TWIST1 TQS phosphorylation state determines the intensity of TWIST1-induced pro-metastatic ability in prostate cancer cells, which may be partly explained mechanistically by TWIST1 dimeric partner choice.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Nuclear Proteins/metabolism , Prostatic Neoplasms/metabolism , Protein Interaction Domains and Motifs , Twist-Related Protein 1/metabolism , Amino Acid Motifs , Animals , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cluster Analysis , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Heterografts , Humans , Male , Mutation , Neoplasm Metastasis , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein Multimerization , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , TOR Serine-Threonine Kinases/metabolism , Transcriptome , Twist-Related Protein 1/chemistry , Twist-Related Protein 1/genetics
11.
Mol Cancer Res ; 11(11): 1387-400, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23982216

ABSTRACT

UNLABELLED: Twist1, a basic helix-loop-helix transcription factor, plays a key role during development and is a master regulator of the epithelial-mesenchymal transition (EMT) that promotes cancer metastasis. Structure-function relationships of Twist1 to cancer-related phenotypes are underappreciated, so we studied the requirement of the conserved Twist box domain for metastatic phenotypes in prostate cancer. Evidence suggests that Twist1 is overexpressed in clinical specimens and correlated with aggressive/metastatic disease. Therefore, we examined a transactivation mutant, Twist1-F191G, in prostate cancer cells using in vitro assays, which mimic various stages of metastasis. Twist1 overexpression led to elevated cytoskeletal stiffness and cell traction forces at the migratory edge of cells based on biophysical single-cell measurements. Twist1 conferred additional cellular properties associated with cancer cell metastasis including increased migration, invasion, anoikis resistance, and anchorage-independent growth. The Twist box mutant was defective for these Twist1 phenotypes in vitro. Importantly, we observed a high frequency of Twist1-induced metastatic lung tumors and extrathoracic metastases in vivo using the experimental lung metastasis assay. The Twist box was required for prostate cancer cells to colonize metastatic lung lesions and extrathoracic metastases. Comparative genomic profiling revealed transcriptional programs directed by the Twist box that were associated with cancer progression, such as Hoxa9. Mechanistically, Twist1 bound to the Hoxa9 promoter and positively regulated Hoxa9 expression in prostate cancer cells. Finally, Hoxa9 was important for Twist1-induced cellular phenotypes associated with metastasis. These data suggest that the Twist box domain is required for Twist1 transcriptional programs and prostate cancer metastasis. IMPLICATIONS: Targeting the Twist box domain of Twist1 may effectively limit prostate cancer metastatic potential.


Subject(s)
Neoplasm Metastasis/genetics , Nuclear Proteins/metabolism , Prostatic Neoplasms/genetics , Protein Structure, Tertiary/genetics , Twist-Related Protein 1/metabolism , Amino Acid Substitution , Animals , Biomarkers, Tumor , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis/pathology , Nuclear Proteins/genetics , Promoter Regions, Genetic , Prostatic Neoplasms/pathology , Transcriptional Activation , Twist-Related Protein 1/genetics
12.
PLoS One ; 8(6): e65726, 2013.
Article in English | MEDLINE | ID: mdl-23762417

ABSTRACT

Sorafenib (SOR) is the only systemic agent known to improve survival for hepatocellular carcinoma (HCC). However, SOR prolongs survival by less than 3 months and does not alter symptomatic progression. To improve outcomes, several phase I-II trials are currently examining SOR with radiation (RT) for HCC utilizing heterogeneous concurrent and sequential treatment regimens. Our study provides preclinical data characterizing the effects of concurrent versus sequential RT-SOR on HCC cells both in vitro and in vivo. Concurrent and sequential RT-SOR regimens were tested for efficacy among 4 HCC cell lines in vitro by assessment of clonogenic survival, apoptosis, cell cycle distribution, and γ-H2AX foci formation. Results were confirmed in vivo by evaluating tumor growth delay and performing immunofluorescence staining in a hind-flank xenograft model. In vitro, concurrent RT-SOR produced radioprotection in 3 of 4 cell lines, whereas sequential RT-SOR produced decreased colony formation among all 4. Sequential RT-SOR increased apoptosis compared to RT alone, while concurrent RT-SOR did not. Sorafenib induced reassortment into less radiosensitive phases of the cell cycle through G1-S delay and cell cycle slowing. More double-strand breaks (DSBs) persisted 24 h post-irradiation for RT alone versus concurrent RT-SOR. In vivo, sequential RT-SOR produced the greatest tumor growth delay, while concurrent RT-SOR was similar to RT alone. More persistent DSBs were observed in xenografts treated with sequential RT-SOR or RT alone versus concurrent RT-SOR. Sequential RT-SOR additionally produced a greater reduction in xenograft tumor vascularity and mitotic index than either concurrent RT-SOR or RT alone. In conclusion, sequential RT-SOR demonstrates greater efficacy against HCC than concurrent RT-SOR both in vitro and in vivo. These results may have implications for clinical decision-making and prospective trial design.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/therapy , Combined Modality Therapy/methods , Gamma Rays/therapeutic use , Histones/genetics , Liver Neoplasms/therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Clinical Trials as Topic , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , Gene Expression/drug effects , Gene Expression/radiation effects , Hindlimb/blood supply , Hindlimb/metabolism , Hindlimb/pathology , Histones/metabolism , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Nude , Neovascularization, Pathologic/prevention & control , Niacinamide/therapeutic use , Radiation Tolerance , Sorafenib , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Mol Cancer Res ; 11(4): 329-38, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23364532

ABSTRACT

A large fraction of non-small cell lung cancers (NSCLC) are dependent on defined oncogenic driver mutations. Although targeted agents exist for EGFR- and EML4-ALK-driven NSCLCs, no therapies target the most frequently found driver mutation, KRAS. Furthermore, acquired resistance to the currently targetable driver mutations is nearly universally observed. Clearly a novel therapeutic approach is needed to target oncogene-driven NSCLCs. We recently showed that the basic helix-loop-helix transcription factor Twist1 cooperates with mutant Kras to induce lung adenocarcinoma in transgenic mouse models and that inhibition of Twist1 in these models led to Kras-induced senescence. In the current study, we examine the role of TWIST1 in oncogene-driven human NSCLCs. Silencing of TWIST1 in KRAS-mutant human NSCLC cell lines resulted in dramatic growth inhibition and either activation of a latent oncogene-induced senescence program or, in some cases, apoptosis. Similar effects were observed in EGFR mutation-driven and c-Met-amplified NSCLC cell lines. Growth inhibition by silencing of TWIST1 was independent of p53 or p16 mutational status and did not require previously defined mediators of senescence, p21 and p27, nor could this phenotype be rescued by overexpression of SKP2. In xenograft models, silencing of TWIST1 resulted in significant growth inhibition of KRAS-mutant, EGFR-mutant, and c-Met-amplified NSCLCs. Remarkably, inducible silencing of TWIST1 resulted in significant growth inhibition of established KRAS-mutant tumors. Together these findings suggest that silencing of TWIST1 in oncogene driver-dependent NSCLCs represents a novel and promising therapeutic strategy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Oncogenes , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/genetics , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cellular Senescence/genetics , Gene Silencing , HEK293 Cells , Heterografts , Humans , Lung Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Nuclear Proteins/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Twist-Related Protein 1/metabolism
14.
Cancer Biol Ther ; 14(4): 347-56, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23358469

ABSTRACT

Outcomes for poor-risk localized prostate cancers treated with radiation are still insufficient. Targeting the "non-oncogene" addiction or stress response machinery is an appealing strategy for cancer therapeutics. Heat-shock-protein-90 (Hsp90), an integral member of this machinery, is a molecular chaperone required for energy-driven stabilization and selective degradation of misfolded "client" proteins, that is commonly overexpressed in tumor cells. Hsp90 client proteins include critical components of pathways implicated in prostate cancer cell survival and radioresistance, such as androgen receptor signaling and the PI3K-Akt-mTOR pathway. We examined the effects of a novel non-geldanamycin Hsp90 inhibitor, AUY922, combined with radiation (RT) on two prostate cancer cell lines, Myc-CaP and PC3, using in vitro assays for clonogenic survival, apoptosis, cell cycle distribution, γ-H2AX foci kinetics and client protein expression in pathways important for prostate cancer survival and radioresistance. We then evaluated tumor growth delay and effects of the combined treatment (RT-AUY922) on the PI3K-Akt-mTOR and AR pathways in a hind-flank tumor graft model. We observed that AUY922 caused supra-additive radiosensitization in both cell lines at low nanomolar doses with enhancement ratios between 1.4-1.7 (p < 0.01). RT-AUY922 increased apoptotic cell death compared with either therapy alone, induced G 2-M arrest and produced marked changes in client protein expression. These results were confirmed in vivo, where RT-AUY922 combination therapy produced supra-additive tumor growth delay compared with either therapy by itself in Myc-CaP and PC3 tumor grafts (both p < 0.0001). Our data suggest that combined RT-AUY922 therapy exhibits promising activity against prostate cancer cells, which should be investigated in clinical studies.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoxazoles/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Resorcinols/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Cycle Checkpoints/drug effects , Cell Division/drug effects , Cell Growth Processes/drug effects , Cell Growth Processes/radiation effects , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Down-Regulation , G2 Phase/drug effects , Humans , Male , Mice , Mice, Transgenic , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Random Allocation , Signal Transduction/drug effects , Signal Transduction/radiation effects , Xenograft Model Antitumor Assays
15.
Int J Radiat Oncol Biol Phys ; 86(1): 143-9, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23182391

ABSTRACT

PURPOSE: Despite improvements in chemoradiation, local control remains a major clinical problem in locally advanced non-small cell lung cancer. The Hedgehog pathway has been implicated in tumor recurrence by promoting survival of tumorigenic precursors and through effects on tumor-associated stroma. Whether Hedgehog inhibition can affect radiation efficacy in vivo has not been reported. METHODS AND MATERIALS: We evaluated the effects of a targeted Hedgehog inhibitor (HhAntag) and radiation on clonogenic survival of human non-small cell lung cancer lines in vitro. Using an A549 cell line xenograft model, we examined tumor growth, proliferation, apoptosis, and gene expression changes after concomitant HhAntag and radiation. In a transgenic mouse model of Kras(G12D)-induced and Twist1-induced lung adenocarcinoma, we assessed tumor response to radiation and HhAntag by serial micro-computed tomography (CT) scanning. RESULTS: In 4 human lung cancer lines in vitro, HhAntag showed little or no effect on radiosensitivity. By contrast, in both the human tumor xenograft and murine inducible transgenic models, HhAntag enhanced radiation efficacy and delayed tumor growth. By use of the human xenograft model to differentiate tumor and stromal effects, mouse stromal cells, but not human tumor cells, showed significant and consistent downregulation of Hedgehog pathway gene expression. This was associated with increased tumor cell apoptosis. CONCLUSIONS: Targeted Hedgehog pathway inhibition can increase in vivo radiation efficacy in lung cancer preclinical models. This effect is associated with pathway suppression in tumor-associated stroma. These data support clinical testing of Hedgehog inhibitors as a component of multimodality therapy for locally advanced non-small cell lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Hedgehog Proteins/antagonists & inhibitors , Lung Neoplasms/radiotherapy , Neoplasm Proteins/antagonists & inhibitors , Radiation Tolerance/physiology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/radiotherapy , Anilides/pharmacology , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/physiology , Female , Hedgehog Proteins/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Mice, Transgenic , Neoplasm Proteins/metabolism , Pyridines/pharmacology , Transplantation, Heterologous
16.
Cell Rep ; 2(6): 1657-69, 2012 Dec 27.
Article in English | MEDLINE | ID: mdl-23260667

ABSTRACT

Chromatin dynamics play a central role in maintaining genome integrity, but how this is achieved remains largely unknown. Here, we report that microrchidia CW-type zinc finger 2 (MORC2), an uncharacterized protein with a derived PHD finger domain and a conserved GHKL-type ATPase module, is a physiological substrate of p21-activated kinase 1 (PAK1), an important integrator of extracellular signals and nuclear processes. Following DNA damage, MORC2 is phosphorylated on serine 739 in a PAK1-dependent manner, and phosphorylated MORC2 regulates its DNA-dependent ATPase activity to facilitate chromatin remodeling. Moreover, MORC2 associates with chromatin and promotes gamma-H2AX induction in a PAK1 phosphorylation-dependent manner. Consequently, cells expressing MORC2-S739A mutation displayed a reduction in DNA repair efficiency and were hypersensitive to DNA-damaging agent. These findings suggest that the PAK1-MORC2 axis is critical for orchestrating the interplay between chromatin dynamics and the maintenance of genomic integrity through sequentially integrating multiple essential enzymatic processes.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromatin Assembly and Disassembly , DNA Damage , Transcription Factors/metabolism , Adenosine Triphosphatases/genetics , Amino Acid Substitution , DNA Repair/genetics , HeLa Cells , Humans , Mutation, Missense , Phosphorylation/genetics , Protein Structure, Tertiary , Transcription Factors/genetics , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
17.
Cancer Biol Ther ; 12(7): 657-63, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21811091

ABSTRACT

Pituitary adenomas with local invasion and high secretory activity remain a therapeutic challenge. The HIV protease inhibitor nelfinavir is a radiosensitizer in multiple tumor models. We tested nelfinavir as a radiosensitizer in pituitary adenoma cells in vitro and in vivo. We examined the effect of nelfinavir with radiation on in vitro cell viability, clonogenic survival, apoptosis, prolactin secretion, cell cycle distribution, and the PI3K-AKT-mTOR pathway. We evaluated tumor growth delay and confirmed nelfinavir's effect on the PI3K-AKT-mTOR pathway in a hind-flank model. Nelfinavir sensitized pituitary adenoma cells to ionizing radiation as shown by viability assays and clonogenic assay with an enhancement ratio of 1.2 (p < 0.05). There is increased apoptotic cell death, as determined by annexin-V expression and cleaved caspase-3 levels. Nelfinavir does not affect prolactin secretion or cell cycle distribution. In vivo, untreated tumors reached 4-fold volume in 12 days, 17 days with nelfinavir treatment, 27 days with radiation 6 Gy, and 41 days with nelfinavir plus radiation (one-way ANOVA p < 0.001). Decreased phospho-S6 on Western blotting in vitro and immunohistochemistry in vivo demonstrated nelfinavir inhibition of the PI3K-AKT-mTOR pathway. Our data suggests a promising combination therapy with nelfinavir plus radiation in pituitary adenomas, which should be investigated in clinical studies.


Subject(s)
Adenoma/drug therapy , Adenoma/radiotherapy , Nelfinavir/pharmacology , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Adenoma/metabolism , Adenoma/pathology , Animals , Annexin A5/metabolism , Apoptosis/drug effects , Apoptosis/radiation effects , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Female , Humans , Mice , Mice, Nude , Neoplasms, Experimental , Phosphatidylinositol 3-Kinases/metabolism , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Prolactin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
18.
Cancer Biol Ther ; 9(7): 479-82, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20190563

ABSTRACT

MicroRNAs are small, non-coding RNAs that regulate gene expression by degrading and/or suppressing the translation of target mRNA by Watson-Crick base pairing in the 3-'UTR of mRNA. The recent explosion of information about the biochemistry and action of microRNAs has implicated these regulatory molecules in many unexpected biologic processes, ranging from development and homeostasis to diseases such as cancer. In general, microRNAs are down regulated or deleted in cancer while a few are upregulated. However, some microRNAs suppress oncogenesis or metastasis, while others are involved in promoting tumorigenesis. All these developments make microRNAs attractive diagnostic markers as well as therapeutic targets. Here we will briefly review the opportunities and potential limitations of using microRNAs in cancer therapeutics.


Subject(s)
Genetic Therapy , MicroRNAs/genetics , Neoplasms/therapy , Animals , Gene Targeting , Humans , MicroRNAs/antagonists & inhibitors , Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...