Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Oncogene ; 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858591

ABSTRACT

Circulating tumor cells (CTCs) play a critical role as initiators in tumor metastasis, which unlocks an irreversible process of cancer progression. Regarding the fluid environment of intravascular CTCs, a comprehensive understanding of the impact of hemodynamic shear stress on CTCs is of profound significance but remains vague. Here, we report a microfluidic circulatory system that can emulate the CTC microenvironment to research the responses of typical liver cancer cells to varying levels of fluid shear stress (FSS). We observe that HepG2 cells surviving FSS exhibit a marked overexpression of TLR4 and TPPP3, which are shown to be associated with the colony formation, migration, and anti-apoptosis abilities of HepG2. Furthermore, overexpression of these two genes in another liver cancer cell line with normally low TLR4 and TPPP3 expression, SK-Hep-1 cells, by lentivirus-mediated transfection also confirms the critical role of TLR4 and TPPP3 in improving colony formation, migration, and survival capability under a fluid environment. Interestingly, in vivo experiments show SK-Hep-1 cells, overexpressed with these genes, have enhanced metastatic potential to the liver and lungs in mouse models via tail vein injection. Mechanistically, TLR4 and TPPP3 upregulated by FSS may increase FSS-mediated cell survival and metastasis through the p53-Bax signaling pathway. Moreover, elevated levels of these genes correlate with poorer overall survival in liver cancer patients, suggesting that our findings could offer new therapeutic strategies for early cancer diagnosis and targeted treatment development.

2.
Small Methods ; : e2400291, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38779741

ABSTRACT

Triple-negative breast cancer (TNBC) remains a significant challenge in terms of treatment, with limited efficacy of chemotherapy due to side effects and acquired drug resistance. In this study, a threose nucleic acid (TNA)-mediated antisense approach is employed to target therapeutic Akt genes for TNBC therapy. Specifically, two new TNA strands (anti-Akt2 and anti-Akt3) are designed and synthesized that specifically target Akt2 and Akt3 mRNAs. These TNAs exhibit exceptional enzymatic resistance, high specificity, enhance binding affinity with their target RNA molecules, and improve cellular uptake efficiency compared to natural nucleic acids. In both 2D and 3D TNBC cell models, the TNAs effectively inhibit the expression of their target mRNA and protein, surpassing the effects of scrambled TNAs. Moreover, when administered to TNBC-bearing animals in combination with lipid nanoparticles, the targeted anti-Akt TNAs lead to reduced tumor sizes and decreased target protein expression compared to control groups. Silencing the corresponding Akt genes also promotes apoptotic responses in TNBC and suppresses tumor cell proliferation in vivo. This study introduces a novel approach to TNBC therapy utilizing TNA polymers as antisense materials. Compared to conventional miRNA- and siRNA-based treatments, the TNA system holds promise as a cost-effective and scalable platform for TNBC treatment, owing to its remarkable enzymatic resistance, inexpensive synthetic reagents, and simple production procedures. It is anticipated that this TNA-based polymeric system, which targets anti-apoptotic proteins involved in breast tumor development and progression, can represent a significant advancement in the clinical development of effective antisense materials for TNBC, a cancer type that lacks effective targeted therapy.

3.
Bioconjug Chem ; 35(5): 623-632, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38659333

ABSTRACT

Nanodiamonds (NDs) are considered promising delivery platforms, but inaccurate and uncontrolled release of drugs at target sites is the biggest challenge of NDs in precision medicine. This study presents the development of phototriggerable ND-based drug delivery systems, utilizing ortho-nitrobenzyl (o-NB) molecules as photocleavable linkers between drugs and nanocarriers. UV irradiation specifically cleaved o-NB molecules and then was followed by releasing antisense oligonucleotides from ND-based carriers in both buffer and cellular environments. This ND system carried cell nonpermeable therapeutic agents for bypassing lysosomal trapping and degradation. The presence of fluorescent nitrogen-vacancy centers also allowed NDs to serve as biological probes for tracing in cells. We successfully demonstrated phototriggered release of antisense oligonucleotides from ND-based nanocarriers, reactivating their antisense functions. This highlights the potential of NDs, photocleavable linkers, and light stimuli to create advanced drug delivery systems for controlled drug release in disease therapy, opening possibilities for targeted and personalized treatments.


Subject(s)
Drug Delivery Systems , Nanodiamonds , Oligonucleotides, Antisense , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/administration & dosage , Humans , Nanodiamonds/chemistry , Drug Delivery Systems/methods , Drug Liberation , Drug Carriers/chemistry , Ultraviolet Rays , Light
4.
Biosens Bioelectron ; 246: 115831, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38008058

ABSTRACT

Digital DNA amplification is a powerful method for detecting and quantifying rare nucleic acids. In this study, we developed a multi-functional droplet-based platform that integrates the traditional digital DNA amplification workflow into a one-step device. This platform enables efficient droplet generation, transition, and signal detection within a 5-min timeframe, distributing the sample into a uniform array of 4 × 104 droplets (variation <2%) within a chamber. Subsequent in-situ DNA amplification, fluorescence detection, and signal analysis were carried out. To assess the platform's performance, we quantitatively detected the human epidermal growth factor receptor (EGFR) mutation and human papillomavirus (HPV) mutation using digital polymerase chain reaction (dPCR) and digital loop-mediated isothermal amplification (dLAMP), respectively. The fluorescence results exhibited a positive, linear, and statistically significant correlation with target DNA concentrations ranging from 101 to 105 copies/µL, demonstrating the capability and feasibility of the integrated device for dPCR and dLAMP. This platform offers high-throughput droplet generation, eliminates droplet fusion and transition, is user-friendly, reduces costs compared to current methods, and holds potential for thermocycling and isothermal nucleic acid quantification with high sensitivity and accuracy.


Subject(s)
Biosensing Techniques , Microfluidics , Humans , Microfluidics/methods , Nucleic Acid Amplification Techniques/methods , Polymerase Chain Reaction , DNA/genetics
6.
Adv Healthc Mater ; 12(18): e2202609, 2023 07.
Article in English | MEDLINE | ID: mdl-36917657

ABSTRACT

Decades of efforts in engineering in vitro cancer models have advanced drug discovery and the insight into cancer biology. However, the establishment of preclinical models that enable fully recapitulating the tumor microenvironment remains challenging owing to its intrinsic complexity. Recent progress in engineering techniques has allowed the development of a new generation of in vitro preclinical models that can recreate complex in vivo tumor microenvironments and accurately predict drug responses, including spheroids, organoids, and tumor-on-a-chip. These biomimetic 3D tumor models are of particular interest as they pave the way for better understanding of cancer biology and accelerating the development of new anticancer therapeutics with reducing animal use. Here, the recent advances in developing these in vitro platforms for cancer modeling and preclinical drug screening, focusing on incorporating hydrogels are reviewed to reconstitute physiologically relevant microenvironments. The combination of spheroids/organoids with microfluidic technologies is also highlighted to better mimic in vivo tumors and discuss the challenges and future directions in the clinical translation of such models for drug screening and personalized medicine.


Subject(s)
Biomimetics , Neoplasms , Tumor Microenvironment , Animals , Lab-On-A-Chip Devices , Neoplasms/drug therapy , Neoplasms/pathology , Organoids/pathology , Spheroids, Cellular/pathology
7.
Small ; 19(16): e2207194, 2023 04.
Article in English | MEDLINE | ID: mdl-36634971

ABSTRACT

Cancer metastasis is the major cause of cancer-related death. Excessive extracellular matrix deposition and increased stiffness are typical features of solid tumors, creating confined spaces for tumor cell migration and metastasis. Confined migration is involved in all metastasis steps. However, confined and unconfined migration inhibitors are different and drugs available to inhibit confined migration are rare. The main challenges are the modeling of confined migration, the suffering of low throughput, and others. Microfluidic device has the advantage to reduce reagent consumption and enhance throughput. Here, a microfluidic chip that can achieve multi-function drug screening against the collective migration of cancer cells under confined environment is designed. This device is applied to screen out effective drugs on confined migration among a novel mechanoreceptors compound library (166 compounds) in hepatocellular carcinoma, non-small lung cancer, breast cancer, and pancreatic ductal adenocarcinoma cells. Three compounds that can significantly inhibit confined migration in pan-cancer: mitochonic acid 5 (MA-5), SB-705498, and diphenyleneiodonium chloride are found. Finally, it is elucidated that these drugs targeted mitochondria, actin polymerization, and cell viability, respectively. In sum, a high-throughput microfluidic platform for screening drugs targeting confined migration is established and three novel inhibitors of confined migration in multiple cancer types are identified.


Subject(s)
Lung Neoplasms , Microfluidic Analytical Techniques , Humans , Drug Evaluation, Preclinical , Cell Movement , Microfluidics , Lab-On-A-Chip Devices
8.
Cancers (Basel) ; 14(20)2022 Oct 13.
Article in English | MEDLINE | ID: mdl-36291790

ABSTRACT

The PI3K/Akt pathway is frequently deregulated in human cancers, and multiple Akt inhibitors are currently under clinical evaluation. Based on the experience from other molecular targeted therapies, however, it is likely that acquired resistance will be developed in patients treated with Akt inhibitors. We established breast cancer models of acquired resistance by prolonged treatment of cells with allosteric or ATP-competitive Akt inhibitors. Phospho-Receptor tyrosine kinase (Phospho-RTK) arrays revealed hyper-phosphorylation of multiple RTKS, including EGFR, Her2, HFGR, EhpB3 and ROR1, in Akt-inhibitor-resistant cells. Importantly, resistance can be overcome by treatment with an EGFR inhibitor. We further showed that cancer stem cells (CSCs) are enriched in breast tumor cells that have developed resistance to Akt inhibitors. Several candidates of CSC regulators, such as ID4, are identified by RNA sequencing. Cosmic analysis indicated that sensitivity of tumor cells to Akt inhibitors can be predicted by ID4 and stem cell/epithelial-mesenchymal transition pathway targets. These findings indicate the potential of targeting the EGFR pathway and CSC program to circumvent Akt inhibitor resistance in breast cancer.

9.
Mater Today Bio ; 15: 100299, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35637854

ABSTRACT

Compared with siRNAs or other antisense oligonucleotides (ASOs), the chemical simplicity, DNA/RNA binding capability, folding ability of tertiary structure, and excellent physiological stability of threose nucleic acid (TNA) motivate scientists to explore it as a novel molecular tool in biomedical applications. Although ASOs reach the target cells/tumors, insufficient tissue penetration and distribution of ASOs result in poor therapeutic efficacy. Therefore, the study of the time course of drug absorption, biodistribution, metabolism, and excretion is of significantly importance. In this work, the pharmacokinetics and biosafety of TNAs in living organisms are investigated. We found that synthetic TNAs exhibited excellent biological stability, low cytotoxicity, and substantial uptake in living cells without transfection. Using U87 three-dimensional (3D) multicellular spheroids to mimic the in vivo tumor microenvironment, TNAs showed their ability to penetrate efficiently throughout the whole multicellular spheroid as a function of incubation time and concentration when the size of the spheroid is relatively small. Additionally, TNAs could be safely administrated into Balb/c mice and most of them distributed in the kidneys where they supposed to excrete from the body through the renal filtration system. We found that accumulation of TNAs in kidneys induced no pathological changes, and no acute structural and functional damage in renal systems. The favourable biocompatibility of TNA makes it attractive as a safe and effective nucleic acid-based therapeutic agent for practical biological applications.

10.
Ann N Y Acad Sci ; 1506(1): 142-163, 2021 12.
Article in English | MEDLINE | ID: mdl-34850398

ABSTRACT

The test for the cancer stem cell (CSC) hypothesis is to find a target expressed on all, and only CSCs in a patient tumor, then eliminate all cells with that target that eliminates the cancer. That test has not yet been achieved, but CSC diagnostics and targets found on CSCs and some other cells have resulted in a few clinically relevant therapies. However, it has become apparent that eliminating the subset of tumor cells characterized by self-renewal properties is essential for long-term tumor control. CSCs are able to regenerate and initiate tumor growth, recapitulating the heterogeneity present in the tumor before treatment. As great progress has been made in identifying and elucidating the biology of CSCs as well as their interactions with the tumor microenvironment, the time seems ripe for novel therapeutic strategies that target CSCs to find clinical applicability. On May 19-21, 2021, researchers in cancer stem cells met virtually for the Keystone eSymposium "Cancer Stem Cells: Advances in Biology and Clinical Translation" to discuss recent advances in the understanding of CSCs as well as clinical efforts to target these populations.


Subject(s)
Congresses as Topic/trends , Neoplasms/genetics , Neoplastic Stem Cells/physiology , Research Report , Translational Research, Biomedical/trends , Tumor Microenvironment/physiology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Humans , Neoplasms/metabolism , Translational Research, Biomedical/methods
11.
Front Mol Biosci ; 8: 700973, 2021.
Article in English | MEDLINE | ID: mdl-34277708

ABSTRACT

ANLN is frequently upregulated in triple-negative breast cancer (TNBC) and its high expression in tumors are significantly associated with poor survival and recurrence, thereby it has been proposed to function as a prognostic marker for breast cancer. However, the specific function and molecular mechanisms by which ANLN promotes TNBC tumorigenesis remain elusive. Using multiomic profiling, we recently uncovered ANLN as a TNBC-specific gene driven by super-enhancer. Here, by Crispr/Cas9 editing, we showed that knockout of ANLN inhibits spheroid growth of TNBC. Interestingly, its effect on cell proliferation in 2D cultures is minimal. ANLN depletion inhibits mammosphere formation and clonogenicity potently, suggesting its important function in regulating cancer stem cells (CSCs). We screened a panel of stem cell-related genes and uncovered several CSC genes regulated by ANLN. We further identify TWIST1 and BMP2 as essential genes that mediate ANLN's function in stemness but not spheroid growth. These findings may contribute to search for effective targeted therapies to treat TNBC.

12.
Nat Commun ; 12(1): 2242, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33854062

ABSTRACT

Breast cancer is a heterogeneous disease, affecting over 3.5 million women worldwide, yet the functional role of cis-regulatory elements including super-enhancers in different breast cancer subtypes remains poorly characterized. Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with a poor prognosis. Here we apply integrated epigenomic and transcriptomic profiling to uncover super-enhancer heterogeneity between breast cancer subtypes, and provide clinically relevant biological insights towards TNBC. Using CRISPR/Cas9-mediated gene editing, we identify genes that are specifically regulated by TNBC-specific super-enhancers, including FOXC1 and MET, thereby unveiling a mechanism for specific overexpression of the key oncogenes in TNBC. We also identify ANLN as a TNBC-specific gene regulated by super-enhancer. Our studies reveal a TNBC-specific epigenomic landscape, contributing to the dysregulated oncogene expression in breast tumorigenesis.


Subject(s)
Triple Negative Breast Neoplasms/genetics , Animals , Cell Line, Tumor , Enhancer Elements, Genetic , Female , Forkhead Transcription Factors/genetics , Gene Editing , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mice, Nude , Microfilament Proteins/genetics , Proto-Oncogene Proteins c-met/genetics
13.
Mol Cancer Ther ; 15(8): 1964-74, 2016 08.
Article in English | MEDLINE | ID: mdl-27297869

ABSTRACT

Acquired resistance to molecular targeted therapy represents a major challenge for the effective treatment of cancer. Hyperactivation of the PI3K/AKT pathway is frequently observed in virtually all human malignancies, and numerous PI3K and AKT inhibitors are currently under clinical evaluation. However, mechanisms of acquired resistance to AKT inhibitors have yet to be described. Here, we use a breast cancer preclinical model to identify resistance mechanisms to a small molecule allosteric AKT inhibitor, MK2206. Using a step-wise and chronic high-dose exposure, breast cancer cell lines harboring oncogenic PI3K resistant to MK2206 were established. Using this model, we reveal that AKT3 expression is markedly upregulated in AKT inhibitor-resistant cells. Induction of AKT3 is regulated epigenetically by the bromodomain and extra terminal domain proteins. Importantly, knockdown of AKT3, but not AKT1 or AKT2, in resistant cells restores sensitivity to MK2206. AKT inhibitor-resistant cells also display an epithelial to mesenchymal transition phenotype as assessed by alterations in the levels of E-Cadherin, N-Cadherin, and vimentin, as well as enhanced invasiveness of tumor spheroids. Notably, the invasive morphology of resistant spheroids is diminished upon AKT3 depletion. We also show that resistance to MK2206 is reversible because upon drug removal resistant cells regain sensitivity to AKT inhibition, accompanied by reexpression of epithelial markers and reduction of AKT3 expression, implying that epigenetic reprogramming contributes to acquisition of resistance. These findings provide a rationale for developing therapeutics targeting AKT3 to circumvent acquired resistance in breast cancer. Mol Cancer Ther; 15(8); 1964-74. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Heterocyclic Compounds, 3-Ring/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cellular Reprogramming/genetics , Dose-Response Relationship, Drug , Female , Gene Knockdown Techniques , Humans , Phenotype , Proto-Oncogene Proteins c-akt/metabolism
14.
Mol Cell ; 62(6): 929-942, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27237051

ABSTRACT

The retinoblastoma (Rb) protein exerts its tumor suppressor function primarily by inhibiting the E2F family of transcription factors that govern cell-cycle progression. However, it remains largely elusive whether the hyper-phosphorylated, non-E2F1-interacting form of Rb has any physiological role. Here we report that hyper-phosphorylated Rb directly binds to and suppresses the function of mTORC2 but not mTORC1. Mechanistically, Rb, but not p107 or p130, interacts with Sin1 and blocks the access of Akt to mTORC2, leading to attenuated Akt activation and increased sensitivity to chemotherapeutic drugs. As such, inhibition of Rb phosphorylation by depleting cyclin D or using CDK4/6 inhibitors releases Rb-mediated mTORC2 suppression. This, in turn, leads to elevated Akt activation to confer resistance to chemotherapeutic drugs in Rb-proficient cells, which can be attenuated with Akt inhibitors. Therefore, our work provides a molecular basis for the synergistic usage of CDK4/6 and Akt inhibitors in treating Rb-proficient cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Multiprotein Complexes/metabolism , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Retinoblastoma Protein/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Proliferation/drug effects , Cyclin D/genetics , Cyclin D/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/metabolism , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Drug Synergism , Enzyme Activation , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Mechanistic Target of Rapamycin Complex 2 , Mice , Molecular Targeted Therapy , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Signal Transduction , Time Factors , Transfection
15.
Cancer Discov ; 5(11): 1194-209, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26293922

ABSTRACT

UNLABELLED: mTOR serves as a central regulator of cell growth and metabolism by forming two distinct complexes, mTORC1 and mTORC2. Although mechanisms of mTORC1 activation by growth factors and amino acids have been extensively studied, the upstream regulatory mechanisms leading to mTORC2 activation remain largely elusive. Here, we report that the pleckstrin homology (PH) domain of SIN1, an essential and unique component of mTORC2, interacts with the mTOR kinase domain to suppress mTOR activity. More importantly, PtdIns(3,4,5)P3, but not other PtdInsPn species, interacts with SIN1-PH to release its inhibition on the mTOR kinase domain, thereby triggering mTORC2 activation. Mutating critical SIN1 residues that mediate PtdIns(3,4,5)P3 interaction inactivates mTORC2, whereas mTORC2 activity is pathologically increased by patient-derived mutations in the SIN1-PH domain, promoting cell growth and tumor formation. Together, our study unravels a PI3K-dependent mechanism for mTORC2 activation, allowing mTORC2 to activate AKT in a manner that is regulated temporally and spatially by PtdIns(3,4,5)P3. SIGNIFICANCE: The SIN1-PH domain interacts with the mTOR kinase domain to suppress mTOR activity, and PtdIns(3,4,5)P3 binds the SIN1-PH domain to release its inhibition on the mTOR kinase domain, leading to mTORC2 activation. Cancer patient-derived SIN1-PH domain mutations gain oncogenicity by loss of suppressing mTOR activity as a means to facilitate tumorigenesis.


Subject(s)
Multiprotein Complexes/metabolism , Phosphatidylinositol Phosphates/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs , Animals , Binding Sites , Catalysis , Cell Line, Tumor , Disease Models, Animal , Enzyme Activation , Female , Heterografts , Humans , Mechanistic Target of Rapamycin Complex 2 , Mice , Models, Molecular , Molecular Conformation , Multiprotein Complexes/chemistry , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphatidylinositol Phosphates/chemistry , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , TOR Serine-Threonine Kinases/chemistry , TOR Serine-Threonine Kinases/genetics
16.
Mol Cell ; 54(3): 329-30, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24813709

ABSTRACT

In this issue of Molecular Cell, Wang et al. (2014) report that the splicing kinase SRPK1 can function as both an oncogene and a tumor suppressor by modulating the activation state of the protein kinase Akt. This is shown to be mediated by the ability of SRPK1 to bind to the Akt phosphatase PHLPP1.


Subject(s)
Carcinogenesis/metabolism , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Animals , Female , Humans , Male
17.
Cancer Discov ; 4(8): 942-55, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24838891

ABSTRACT

UNLABELLED: Loss of PTEN is a common event in many cancers and leads to hyperactivation of the PI3K-AKT signaling pathway. The mechanisms by which AKT isoforms mediate signaling to phenotypes associated with PTEN inactivation in cancer have not been defined. Here, we show that AKT2 is exclusively required for PTEN-deficient prostate tumor spheroid maintenance, whereas AKT1 is dispensable. shRNA silencing of AKT2 but not AKT1 promotes regression of prostate cancer xenografts. Mechanistically, we show that AKT2 silencing upregulates p21 and the proapoptotic protein BAX and downregulates the insulin-like growth factor receptor-1. We also show that p21 is an effector of AKT2 in mediating prostate tumor maintenance. Moreover, AKT2 is also exclusively required for the maintenance and survival of other PTEN-deficient solid tumors, including breast cancer and glioblastoma. These findings identify a specific function for AKT2 in mediating survival of PTEN-deficient tumors and provide a rationale for developing therapeutics targeting AKT2. SIGNIFICANCE: Depletion of AKT2, but not AKT1, induces potent tumor regression in PTEN-deficient prostate cancer xenografts, concomitant with upregulation of p21, which may serve as a potential biomarker for screening AKT2 activity in clinical samples. The specific role of AKT2 in tumor maintenance provides a rationale for the development of isoform-specific inhibitors for patients with PTEN-deficient cancers.


Subject(s)
Cell Proliferation/genetics , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-akt/genetics , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Male , Prostatic Neoplasms/pathology , Protein Isoforms , Proto-Oncogene Proteins c-akt/biosynthesis , Receptor, IGF Type 1/biosynthesis , Signal Transduction , p21-Activated Kinases/genetics
18.
Cancer Res ; 74(3): 964-73, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24335962

ABSTRACT

Triple-negative breast cancer (TNBC) is currently the only major breast tumor subtype without effective targeted therapy and, as a consequence, in general has a poor outcome. To identify new therapeutic targets in TNBC, we performed a short hairpin RNA (shRNA) screen for protein kinases commonly amplified and overexpressed in breast cancer. Using this approach, we identified AKT3 as a gene preferentially required for the growth of TNBCs. Downregulation of Akt3 significantly inhibits the growth of TNBC lines in three-dimensional (3D) spheroid cultures and in mouse xenograft models, whereas loss of Akt1 or Akt2 have more modest effects. Akt3 silencing markedly upregulates the p27 cell-cycle inhibitor and this is critical for the ability of Akt3 to inhibit spheroid growth. In contrast with Akt1, Akt3 silencing results in only a minor enhancement of migration and does not promote invasion. Depletion of Akt3 in TNBC sensitizes cells to the pan-Akt inhibitor GSK690693. These results imply that Akt3 has a specific function in TNBCs; thus, its therapeutic targeting may provide a new treatment option for this tumor subtype.


Subject(s)
Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Invasiveness , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , RNA Isoforms , Spheroids, Cellular , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Tumor Burden , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
Cancer Res ; 73(1): 50-61, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23135917

ABSTRACT

Tumors are composed of cancer cells but also a larger number of diverse stromal cells in the tumor microenvironment. Stromal cells provide essential supports to tumor pathophysiology but the distinct characteristics of their signaling networks are not usually considered in developing drugs to target tumors. This oversight potentially confounds proof-of-concept studies and increases drug development risks. Here, we show in established murine and human models of breast cancer how differential regulation of Akt by the small GTPase RhoB in cancer cells or stromal endothelial cells determines their dormancy versus outgrowth when angiogenesis becomes critical. In cancer cells in vitro or in vivo, RhoB functions as a tumor suppressor that restricts EGF receptor (EGFR) cell surface occupancy as well as Akt signaling. However, after activation of the angiogenic switch, RhoB functions as a tumor promoter by sustaining endothelial Akt signaling, growth, and survival of stromal endothelial cells that mediate tumor neoangiogenesis. Altogether, the positive impact of RhoB on angiogenesis and progression supercedes its negative impact in cancer cells themselves. Our findings elucidate the dominant positive role of RhoB in cancer. More generally, they illustrate how differential gene function effects on signaling pathways in the tumor stromal component can complicate the challenge of developing therapeutics to target cancer pathophysiology.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/metabolism , Endothelial Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stromal Cells/metabolism , rhoB GTP-Binding Protein/metabolism , Animals , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization , Mice , Mice, Transgenic , Neovascularization, Pathologic/metabolism , Real-Time Polymerase Chain Reaction , Tumor Microenvironment/physiology
20.
Cell ; 150(1): 179-93, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22770219

ABSTRACT

Aberrant Skp2 signaling has been implicated as a driving event in tumorigenesis. Although the underlying molecular mechanisms remain elusive, cytoplasmic Skp2 correlates with more aggressive forms of breast and prostate cancers. Here, we report that Skp2 is acetylated by p300 at K68 and K71, which is a process that can be antagonized by the SIRT3 deacetylase. Inactivation of SIRT3 leads to elevated Skp2 acetylation, which leads to increased Skp2 stability through impairment of the Cdh1-mediated proteolysis pathway. As a result, Skp2 oncogenic function is increased, whereby cells expressing an acetylation-mimetic mutant display enhanced cellular proliferation and tumorigenesis in vivo. Moreover, acetylation of Skp2 in the nuclear localization signal (NLS) promotes its cytoplasmic retention, and cytoplasmic Skp2 enhances cellular migration through ubiquitination and destruction of E-cadherin. Thus, our study identifies an acetylation-dependent regulatory mechanism governing Skp2 oncogenic function and provides insight into how cytoplasmic Skp2 controls cellular migration.


Subject(s)
Breast Neoplasms/pathology , Cell Movement , Prostatic Neoplasms/pathology , S-Phase Kinase-Associated Proteins/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation , Amino Acid Sequence , Animals , Breast Neoplasms/metabolism , Cadherins/metabolism , Casein Kinase I/metabolism , Cell Line, Tumor , Cytoplasm/metabolism , Disease Models, Animal , Humans , Lysine/metabolism , Male , Mice , Molecular Sequence Data , Prostatic Neoplasms/metabolism , Protein Processing, Post-Translational , Protein Sorting Signals , S-Phase Kinase-Associated Proteins/chemistry , S-Phase Kinase-Associated Proteins/genetics , Sequence Alignment , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...