Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 836
Filter
2.
Nat Commun ; 13(1): 5469, 2022 09 17.
Article in English | MEDLINE | ID: mdl-36115844

ABSTRACT

Oncogenic RAS mutations are common in multiple myeloma (MM), an incurable malignancy of plasma cells. However, the mechanisms of pathogenic RAS signaling in this disease remain enigmatic and difficult to inhibit therapeutically. We employ an unbiased proteogenomic approach to dissect RAS signaling in MM. We discover that mutant isoforms of RAS organize a signaling complex with the amino acid transporter, SLC3A2, and MTOR on endolysosomes, which directly activates mTORC1 by co-opting amino acid sensing pathways. MM tumors with high expression of mTORC1-dependent genes are more aggressive and enriched in RAS mutations, and we detect interactions between RAS and MTOR in MM patient tumors harboring mutant RAS isoforms. Inhibition of RAS-dependent mTORC1 activity synergizes with MEK and ERK inhibitors to quench pathogenic RAS signaling in MM cells. This study redefines the RAS pathway in MM and provides a mechanistic and rational basis to target this mode of RAS signaling.


Subject(s)
Genes, ras , Multiple Myeloma , Transcription Factors , Amino Acids/metabolism , Genes, ras/genetics , Genes, ras/physiology , Humans , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mitogen-Activated Protein Kinase Kinases , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Mutation , Protein Isoforms , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Bioengineered ; 13(2): 3122-3136, 2022 02.
Article in English | MEDLINE | ID: mdl-35037835

ABSTRACT

Nasopharyngeal carcinoma (NPC) refers to a malignancy initiating from the superior mucosal epithelium of the nasopharynx. Optimal therapies for NPC are still needed. In this investigation, we attempted to explore whether BarH-like homeobox 2 (BARX2), a well-known tumor suppressor, had anti-cancer properties on NPC, and the possible mechanisms. After searching for NPC-related databases, we determined BARX2 as one of the core genes in NPC. The results of RT-qPCR and immunohistochemistry or Western blot demonstrated that BARX2 was reduced in NPC patients and cells. Ectopic expression of BARX2 reverted the malignant phenotype of NPC cells. Mechanistically, BARX2 bound to the keratin 16 (KRT16) promoter to downregulate its expression. In addition, BARX2 was found to reduce the phosphorylation levels of MEK and ERK. Further KRT16 upregulation in cells overexpressing BARX2 promoted malignant aggressiveness of C666-1 and HNE3 cells and activated the Ras signaling pathway. BARX2 inhibited the growth and metastasis of tumors and suppressed the Ras signaling pathway in vivo. In conclusion, our findings indicate that BARX2 reverts malignant phenotypes of NPC cells by downregulating KRT16 in a Ras-dependent fashion. BARX2 might act as a possible therapeutic regulator for NPC.


Subject(s)
Genes, ras/physiology , Homeodomain Proteins/physiology , Keratin-16/genetics , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Progression , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Neoplasms/genetics , Signal Transduction/genetics
4.
PLoS Genet ; 17(8): e1009738, 2021 08.
Article in English | MEDLINE | ID: mdl-34411095

ABSTRACT

Activation of Ras signaling occurs in ~30% of human cancers. However, activated Ras alone is insufficient to produce malignancy. Thus, it is imperative to identify those genes cooperating with activated Ras in driving tumoral growth. In this work, we have identified a novel EGFR inhibitor, which we have named EGFRAP, for EGFR adaptor protein. Elimination of EGFRAP potentiates activated Ras-induced overgrowth in the Drosophila wing imaginal disc. We show that EGFRAP interacts physically with the phosphorylated form of EGFR via its SH2 domain. EGFRAP is expressed at high levels in regions of maximal EGFR/Ras pathway activity, such as at the presumptive wing margin. In addition, EGFRAP expression is up-regulated in conditions of oncogenic EGFR/Ras activation. Normal and oncogenic EGFR/Ras-mediated upregulation of EGRAP levels depend on the Notch pathway. We also find that elimination of EGFRAP does not affect overall organogenesis or viability. However, simultaneous downregulation of EGFRAP and its ortholog PVRAP results in defects associated with increased EGFR function. Based on these results, we propose that EGFRAP is a new negative regulator of the EGFR/Ras pathway, which, while being required redundantly for normal morphogenesis, behaves as an important modulator of EGFR/Ras-driven tissue hyperplasia. We suggest that the ability of EGFRAP to functionally inhibit the EGFR pathway in oncogenic cells results from the activation of a feedback loop leading to increase EGFRAP expression. This could act as a surveillance mechanism to prevent excessive EGFR activity and uncontrolled cell growth.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Genes, ras/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle , Cell Proliferation/genetics , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, ras/physiology , Imaginal Discs/metabolism , Morphogenesis , Phosphorylation , Receptors, Invertebrate Peptide/antagonists & inhibitors , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism , Signal Transduction/genetics , ras Proteins/genetics
5.
Dev Cell ; 56(15): 2223-2236.e5, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34324859

ABSTRACT

Cancer tissue often comprises multiple tumor clones with distinct oncogenic alterations such as Ras or Src activation, yet the mechanism by which tumor heterogeneity drives cancer progression remains elusive. Here, we show in Drosophila imaginal epithelium that clones of Ras- or Src-activated benign tumors interact with each other to mutually promote tumor malignancy. Mechanistically, Ras-activated cells upregulate the cell-surface ligand Delta while Src-activated cells upregulate its receptor Notch, leading to Notch activation in Src cells. Elevated Notch signaling induces the transcriptional repressor Zfh1/ZEB1, which downregulates E-cadherin and cell death gene hid, leading to Src-activated invasive tumors. Simultaneously, Notch activation in Src cells upregulates the cytokine Unpaired/IL-6, which activates JAK-STAT signaling in neighboring Ras cells. Elevated JAK-STAT signaling upregulates the BTB-zinc-finger protein Chinmo, which downregulates E-cadherin and thus generates Ras-activated invasive tumors. Our findings provide a mechanistic explanation for how tumor heterogeneity triggers tumor progression via cell-cell interactions.


Subject(s)
Neoplasms/metabolism , Oncogene Protein pp60(v-src)/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Cadherins/metabolism , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Epithelium/metabolism , Gene Expression Regulation, Neoplastic/genetics , Genes, ras/genetics , Genes, ras/physiology , Imaginal Discs/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Oncogene Protein pp60(v-src)/physiology , Proto-Oncogene Proteins p21(ras)/physiology , Receptors, Notch/genetics , Receptors, Notch/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Zinc Fingers
6.
Gene ; 765: 145116, 2021 Jan 10.
Article in English | MEDLINE | ID: mdl-32896589

ABSTRACT

In contrast to most mammals including human, fish cell lines have long been known to be immortal, with little sign of cellular senescence, despite the absence of transformation. Recently, our laboratory reported that DNA demethylation with 5-aza-2'-deoxycytidine (5-Aza-dC) induces telomere-independent cellular senescence and senescence-associated secretory phenotype (SASP) in an immortal fish cell line, EPC (Epithelioma papulosum cyprini). However, it is not known how fish derived cultured cells are usually resistant to aging in vitro. In this study, we focused on Ras, which carries out the main role of Ras-induced senescence (RIS), and investigated the role of Ras in the regulation of senescence in EPC cells. Our results show that 5-Aza-dC induced the expression of the ras (hras, kras, nras) gene in EPC cells. EPC cells overexpressing HRas or its constitutively active form (HRasV12) showed p53-dependent senescence-like growth arrest and senescence-associated ß-galactosidase (SA-ß-gal) activity with a large and/or flat morphology characteristic of cell senescence. On the other hand, the SASP was not induced. These results imply that the increased expression of HRas contributes to early senescence in EPC cells, but it alone may not be sufficient for the full senescence, even if HRas is aberrantly activated. Thus, the limited mechanism of RIS may play a role in the senescence-resistance of fish cell lines.


Subject(s)
Cellular Senescence/genetics , Genes, ras/genetics , Genes, ras/physiology , Aging/genetics , Aging/metabolism , Animals , Cell Line , Cells, Cultured , Cellular Senescence/physiology , Fishes/genetics , Humans , Proto-Oncogene Proteins p21(ras)/genetics
7.
Cancer Res ; 80(18): 3799-3802, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32732221

ABSTRACT

Myc and Ras are two of the most commonly activated oncogenes in tumorigenesis. Together and independently they regulate many cancer hallmarks including proliferation, apoptosis, and self-renewal. Recently, they were shown to cooperate to regulate host tumor microenvironment programs including host immune responses. But, is their partnership always cooperative or do they have distinguishable functions? Here, we provide one perspective that Myc and Ras cooperation depends on the genetic evolution of a particular cancer. This in turn, dictates when they cooperate via overlapping and identifiably distinct cellular- and host immune-dependent mechanisms that are cancer type specific.


Subject(s)
Breast Neoplasms/genetics , Genes, myc/physiology , Genes, ras/physiology , Hematologic Neoplasms/genetics , Lung Neoplasms/genetics , Tumor Microenvironment/genetics , Adenocarcinoma/genetics , Carcinogenesis/genetics , Carcinogenesis/immunology , Cell Physiological Phenomena/genetics , Female , Humans , Transcriptional Activation
8.
Cancer Res ; 80(18): 3820-3829, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32661137

ABSTRACT

The enzyme glucose-6-phosphate dehydrogenase (G6PD) is a major contributor to NADPH production and redox homeostasis and its expression is upregulated and correlated with negative patient outcomes in multiple human cancer types. Despite these associations, whether G6PD is essential for tumor initiation, growth, or metastasis remains unclear. Here, we employ modern genetic tools to evaluate the role of G6PD in lung, breast, and colon cancer driven by oncogenic K-Ras. Human HCT116 colorectal cancer cells lacking G6PD exhibited metabolic indicators of oxidative stress, but developed into subcutaneous xenografts with growth comparable with that of wild-type controls. In a genetically engineered mouse model of non-small cell lung cancer driven by K-Ras G12D and p53 deficiency, G6PD knockout did not block formation or proliferation of primary lung tumors. In MDA-MB-231-derived human triple-negative breast cancer cells implanted as orthotopic xenografts, loss of G6PD modestly decreased primary site growth without ablating spontaneous metastasis to the lung and moderately impaired the ability of breast cancer cells to colonize the lung when delivered via tail vein injection. Thus, in the studied K-Ras tumor models, G6PD was not strictly essential for tumorigenesis and at most modestly promoted disease progression. SIGNIFICANCE: K-Ras-driven tumors can grow and metastasize even in the absence of the oxidative pentose pathway, a main NADPH production route.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Colonic Neoplasms/enzymology , Genes, ras/physiology , Glucosephosphate Dehydrogenase/physiology , Lung Neoplasms/enzymology , Triple Negative Breast Neoplasms/enzymology , Animals , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Disease Models, Animal , Female , Gene Knockdown Techniques , Glucosephosphate Dehydrogenase/genetics , HCT116 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Mice , Mice, Nude , Neoplasm Transplantation , Neoplastic Cells, Circulating , Oxidative Stress , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
9.
Toxicol Appl Pharmacol ; 402: 115121, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32621833

ABSTRACT

Aberrant activation of H-Ras is often associated with tumor aggressiveness in breast cancer. Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1  (Pin1) is a unique enzyme that interacts with phosphorylated serine or threonine of a target protein and isomerizes the adjacent proline residue. Pin1 is prevalently overexpressed in human cancers, and its overexpression correlates with poor prognosis. Nuclear factor E2-related factor 2 (Nrf2) is a master regulator of cellular redox homeostasis. The sustained activation/accumulation of Nrf2 has been observed in many different types of human malignancies, conferring an advantage for growth and survival of cancer cells. The activated form of H-Ras (GTP-H-Ras) is highly overexpressed in human breast cancer tissues. In our present study, silencing of H-Ras decreased the invasiveness of MDA-MB-231 human breast cancer cells and abrogated the interaction between Pin1 and Nrf2 in these cells. Pin1 knockdown blocked the accumulation of Nrf2, thereby suppressing proliferation and clonogenicity of MCF10A-Ras human mammary epithelial cells. We found that Pin1 binds to Nrf2 which stabilizes this transcription factor by hampering proteasomal degradation. In conclusion, H-Ras activation in cooperation with the Pin1-Nrf2 complex represents a novel mechanism underlying breast cancer progression and constitutive activation of Nrf2 and can be exploited as a therapeutic target.


Subject(s)
Breast Neoplasms/metabolism , Genes, ras/physiology , NF-E2-Related Factor 2/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Silencing , Genes, ras/genetics , HEK293 Cells , Humans , NF-E2-Related Factor 2/genetics , NIMA-Interacting Peptidylprolyl Isomerase/genetics
10.
J Neuropathol Exp Neurol ; 79(7): 734-745, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32417918

ABSTRACT

Medulloblastomas (MBs) are currently divided into 4 molecular subgroups: WNT, SHH, Group 3, and Group 4. Among them, Group 3 MB has the worst prognosis, and 40%-50% of Group 3 cases are already metastatic at the time of diagnosis. Emerging evidence indicates that exosomes drive tumor invasion, but very little is known about exosomes in MBs. In this study, we initially discovered that exosomes isolated from Group 3 MB cell lines altered in vitro behaviors of a less invasive SHH MB cell line and yielded a much more aggressive phenotype. RNA-sequencing analysis revealed 7 exosomal miRNAs with markedly different expression levels between the SHH and Group 3 MB cell lines. They were all predicted to be related to the Ras/MAPK pathway according to the Kyoto Encyclopedia of Genes and Genomes data analysis. Increased expression of miR-181a-5p, miR-125b-5p, and let-7b-5p was further confirmed in Group 3 MB cells with real-time PCR and was shown to increase in vitro invasion and migratory abilities of tumor cells through the activation of ERK in Ras/MAPK pathway. Collectively, our findings suggest that exosomal miRNAs have a critical role in MB progression in vitro and might serve as diagnostic biomarkers and therapeutic targets.


Subject(s)
Cell Movement/physiology , Cerebellar Neoplasms/metabolism , Exosomes/metabolism , Genes, ras/physiology , MAP Kinase Signaling System/physiology , Medulloblastoma/metabolism , MicroRNAs/metabolism , Cell Line, Tumor , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Exosomes/genetics , Humans , Medulloblastoma/genetics , Medulloblastoma/pathology , MicroRNAs/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology
11.
Curr Opin Pediatr ; 32(1): 48-56, 2020 02.
Article in English | MEDLINE | ID: mdl-31815779

ABSTRACT

PURPOSE OF REVIEW: The current review aims to highlight the frequency of RAS mutations in pediatric leukemias and solid tumors and to propose strategies for targeting oncogenic RAS in pediatric cancers. RECENT FINDINGS: The three RAS genes (HRAS, NRAS, and KRAS) comprise the most frequently mutated oncogene family in human cancer. RAS mutations are commonly observed in three of the leading causes of cancer death in the United States, namely lung cancer, pancreatic cancer, and colorectal cancer. The association of RAS mutations with these aggressive malignancies inspired the creation of the National Cancer Institute RAS initiative and spurred intense efforts to develop strategies to inhibit oncogenic RAS, with much recent success. RAS mutations are frequently observed in pediatric cancers; however, recent advances in anti-RAS drug development have yet to translate into pediatric clinical trials. SUMMARY: We find that RAS is mutated in common and rare pediatric malignancies and that oncogenic RAS confers a functional dependency in these cancers. Many strategies for targeting RAS are being pursued for malignancies that primarily affect adults and there is a clear need for inclusion of pediatric patients in clinical trials of these agents.


Subject(s)
Genes, ras/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Child , GTP Phosphohydrolases/genetics , Genes, ras/physiology , Humans , Membrane Proteins/genetics , Molecular Targeted Therapy , Mutation , Proto-Oncogene Proteins p21(ras)/genetics
12.
Exp Gerontol ; 128: 110752, 2019 12.
Article in English | MEDLINE | ID: mdl-31648009

ABSTRACT

Oncogene-induced senescence (OIS) serves as an important barrier to tumor progression in cells that have acquired activating mutations in RAS and other oncogenes. Senescent cells also produce a secretome known as the senescence-associated secretory phenotype (SASP) that includes pro-inflammatory cytokines and chemokines. SASP factors reinforce and propagate the senescence program and identify senescent cells to the immune system for clearance. The OIS program is executed by several transcriptional effectors that include p53, RB, NF-κB and C/EBPß. In this review, we summarize the critical role of C/EBPß in regulating OIS and the SASP. Post-translational modifications induced by oncogenic RAS signaling control C/EBPß activity and dimerization, and these alterations switch C/EBPß to a pro-senescence form during OIS. In addition, C/EBPß is regulated by a unique 3'UTR-mediated mechanism that restrains its activity in tumor cells to facilitate senescence bypass and suppression of the SASP.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/physiology , Cellular Senescence/physiology , Oncogenes/physiology , Animals , CCAAT-Enhancer-Binding Protein-beta/chemistry , Genes, ras/physiology , Humans , MAP Kinase Signaling System/physiology , NF-kappa B/physiology , Phosphorylation , Protein Multimerization , Signal Transduction/physiology
13.
BMC Cancer ; 18(1): 1201, 2018 Dec 03.
Article in English | MEDLINE | ID: mdl-30509235

ABSTRACT

BACKGROUND: Peritoneal dissemination is a critical prognostic factor in ovarian cancer. Although stabilized spheroid formation promotes cancer cell peritoneal dissemination in ovarian cancer, the associated oncogenes are unknown. In this study, we assessed the role of the KRAS oncogene in ovarian cancer cell dissemination, focusing on the stability of cells in spheroid condition, as well as the modulation of intracellular signaling following spheroid transformation. METHODS: We used ID8, a murine ovarian cancer cell line, and ID8-KRAS, an oncogenic KRAS (G12 V)-transduced ID8 cell line in this study. Spheroid-forming (3D) culture and cell proliferation assays were performed to evaluate the growth characteristics of these cells. cDNA microarray analysis was performed to identify genes involved in KRAS-associated signal transduction in floating condition. A MEK inhibitor was used to evaluate the effect on cancer peritoneal dissemination. RESULTS: Cell viability and proliferation in monolayer (2D) cultures did not differ between ID8 and ID8-KRAS cells. However, the proportions of viable and proliferating ID8-KRAS cells in 3D culture were approximately 2-fold and 5-fold higher than that of ID8, respectively. Spheroid-formation was increased in ID8-KRAS cells. Analysis of peritoneal floating cells obtained from mice intra-peritoneally injected with cancer cells revealed that the proportion of proliferating cancer cells was approximately 2-fold higher with ID8-KRAS than with ID8 cells. Comprehensive cDNA microarray analysis revealed that pathways related to cell proliferation, and cell cycle checkpoint and regulation were upregulated specifically in ID8-KRAS cells in 3D culture, and that some genes partially regulated by the MEK-ERK pathway were upregulated only in ID8-KRAS cells in 3D culture. Furthermore, a MEK inhibitor, trametinib, suppressed spheroid formation in 3D culture of ID8-KRAS cells, although trametinib did not affect 2D-culture cell proliferation. Finally, we demonstrated that trametinib dramatically improved the prognosis for mice with ID8-KRAS tumors in an in vivo mouse model. CONCLUSIONS: Our data indicated that KRAS promoted ovarian cancer dissemination by stabilizing spheroid formation and that the MEK pathway is important for stabilized spheroid formation. Disruption of spheroid formation by a MEK inhibitor could be a therapeutic target for cancer peritoneal dissemination.


Subject(s)
Cell Proliferation/physiology , Genes, ras/physiology , MAP Kinase Signaling System/physiology , Ovarian Neoplasms/metabolism , Spheroids, Cellular/metabolism , Animals , Cell Culture Techniques/methods , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred C57BL , Ovarian Neoplasms/pathology
14.
Biochem Pharmacol ; 158: 217-228, 2018 12.
Article in English | MEDLINE | ID: mdl-30352234

ABSTRACT

RAS is the most commonly mutated driver of tumorigenesis, seen in about 30% of all cancer cases. There is a subset of tumors termed RAS-driven cancers in which RAS mutation or overactivation is evident, including as much as 95% in pancreatic and 50% in colon cancer. RAS is a family of small membrane bound GTPases that act as a signaling node to control both normal and cancer biology. Since the discovery of RAS' overall prominence in many tumor types and specifically in RAS-dependent cancers, it has been an obvious therapeutic target for drug development. However, RAS has proved a very elusive target, and after a few prominent RAS targeted drugs failed in clinical trials after decades of research, RAS was termed "undruggable" and research in this field was greatly hampered. An increase in knowledge about basic RAS biology has led to a resurgence in the generation of novel therapeutics targeting RAS signaling utilizing various and distinct approaches. These new drugs target RAS activation directly, block downstream signaling effectors and inhibit proper post-translational processing and trafficking/recycling of RAS. This review will cover how these new drugs were developed and how they have fared in preclinical and early phase clinical trials.


Subject(s)
Antineoplastic Agents/administration & dosage , Genes, ras/physiology , Molecular Targeted Therapy/trends , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Genes, ras/drug effects , Humans , Molecular Targeted Therapy/methods , Neoplasms/genetics
15.
Cell Syst ; 7(1): 49-62.e8, 2018 07 25.
Article in English | MEDLINE | ID: mdl-29960885

ABSTRACT

Altered glycolysis is a hallmark of diseases including diabetes and cancer. Despite intensive study of the contributions of individual glycolytic enzymes, systems-level analyses of flux control through glycolysis remain limited. Here, we overexpress in two mammalian cell lines the individual enzymes catalyzing each of the 12 steps linking extracellular glucose to excreted lactate, and find substantial flux control at four steps: glucose import, hexokinase, phosphofructokinase, and lactate export (and not at any steps of lower glycolysis). The four flux-controlling steps are specifically upregulated by the Ras oncogene: optogenetic Ras activation rapidly induces the transcription of isozymes catalyzing these four steps and enhances glycolysis. At least one isozyme catalyzing each of these four steps is consistently elevated in human tumors. Thus, in the studied contexts, flux control in glycolysis is concentrated in four key enzymatic steps. Upregulation of these steps in tumors likely underlies the Warburg effect.


Subject(s)
Glycolysis/physiology , Hexokinase/metabolism , Phosphofructokinase-1/metabolism , Animals , Biological Transport , Cell Line , Genes, ras/genetics , Genes, ras/physiology , Glucose/metabolism , Glycolysis/genetics , HEK293 Cells , Hexokinase/genetics , Humans , Isoenzymes/metabolism , Lactic Acid/biosynthesis , Mammals , Mice , Models, Biological , NIH 3T3 Cells , Neoplasms/enzymology
16.
Crit Rev Oncol Hematol ; 125: 69-77, 2018 May.
Article in English | MEDLINE | ID: mdl-29650279

ABSTRACT

BACKGROUND: At present, there is uncertainty on the best systemic treatment in first-line setting for RAS wild-type (WT) metastatic colorectal cancer (mCRC) patients. Indeed, several chemotherapy and biologics combinations showed an improvement on survival. We performed a systematic review with a pair-wise and bayesan meta-analysis to rank the best strategy for these patients. METHODS: A systematic literature search through March 2017 was performed to evaluate the association between several treatment combinations and overall survival (OS), progression-free survival (PFS), overall response rate (ORR) and toxicity rate (TR) in RAS WT mCRC patients. Data were extracted from studies and pooled using the random-effect model for pair-wise meta-analyses and bayesan model for network meta-analysis (NMA). RESULTS: Eight studies with a total of 2518 individuals were included in the meta-analyses. Pooled analyses for subgroups stratified by type of schedule and tumor location demonstrated that anti-EGFR + doublet had the best OS when compared to doublet ±â€¯bevacizumab (0.767; 95%CI, 0.695-0.846; P < 0.0001). This benefit is limited to LSCC when compared to a doublet-based schedule and doublet + bevacizumab (HRs, 0.692; 95%CI, 0.596-0.804; P < 0.001; 0.706; 95%CI, 0.584-0.854; P < 0.001; respectively). No significant differences are detected in PFS, whereas the cetuximab-based regimens showed the highest ORR and TR. In NMA our ranking showed the best performance for FOLFOX + panitumumab. CONCLUSIONS: Our study indicates that FOLFOX + panitumumab has the major probability to provide an improvement of survival with a good safety profile in patients with RAS WT mCRC with an added value from selection based on sidedness.


Subject(s)
Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Genes, ras/physiology , Neoadjuvant Therapy/methods , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bayes Theorem , Colorectal Neoplasms/genetics , Disease-Free Survival , Humans , Neoplasm Metastasis
17.
Mol Cancer ; 17(1): 33, 2018 02 19.
Article in English | MEDLINE | ID: mdl-29455666

ABSTRACT

Lung neoplasms are the leading cause of death by cancer worldwide. Non-small cell lung cancer (NSCLC) constitutes more than 80% of all lung malignancies and the majority of patients present advanced disease at onset. However, in the last decade, multiple oncogenic driver alterations have been discovered and each of them represents a potential therapeutic target. Although KRAS mutations are the most frequently oncogene aberrations in lung adenocarcinoma patients, effective therapies targeting KRAS have yet to be developed. Moreover, the role of KRAS oncogene in NSCLC remains unclear and its predictive and prognostic impact remains controversial. The study of the underlying biology of KRAS in NSCLC patients could help to determine potential candidates to evaluate novel targeted agents and combinations that may allow a tailored treatment for these patients. The aim of this review is to update the current knowledge about KRAS-mutated lung adenocarcinoma, including a historical overview, the biology of the molecular pathways involved, the clinical relevance of KRAS mutations as a prognostic and predictive marker and the potential therapeutic approaches for a personalized treatment of KRAS-mutated NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Genes, ras/genetics , Lung Neoplasms/genetics , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Genes, ras/physiology , Humans , Lung Neoplasms/drug therapy , Mutation/genetics
18.
Cancer Res ; 78(1): 7-14, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29263151

ABSTRACT

KRAS is one of the most frequently mutated oncogenes in cancer, being a potent initiator of tumorigenesis, a strong inductor of malignancy, and a predictive biomarker of response to therapy. Despite the large investment to understand the effects of KRAS activation in cancer cells, pharmacologic targeting of KRAS or its downstream effectors has not yet been successful at the clinical level. Recent studies are now describing new mechanisms of KRAS-induced tumorigenesis by analyzing its effects on the components of the tumor microenvironment. These studies revealed that the activation of KRAS on cancer cells extends to the surrounding microenvironment, affecting the properties and functions of its constituents. Herein, we discuss the most emergent perspectives on the relationship between KRAS-mutant cancer cells and their microenvironment components. Cancer Res; 78(1); 7-14. ©2017 AACR.


Subject(s)
Genes, ras/physiology , Neoplasms/pathology , Tumor Microenvironment/genetics , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Fibroblasts/pathology , Humans , Mutation , Myeloid Cells/pathology , Neoplasms/genetics , Neoplasms/immunology , Signal Transduction , Tumor Escape/genetics
19.
Article in English | MEDLINE | ID: mdl-29229665

ABSTRACT

The primary site of Ras signal transduction is the plasma membrane (PM). On the PM, the ubiquitously expressed Ras isoforms, H-, N-, and K-Ras, spatially segregate to nonoverlapping nanometer-sized domains, called nanoclusters, with further lateral segregation into nonoverlapping guanosine triphosphate (GTP)-bound and guanosine diphosphate (GDP)-bound nanoclusters. Effector binding and activation is restricted to GTP nanoclusters, rendering the underlying assembly mechanism essential to Ras signaling. Ras nanoclusters have distinct lipid compositions as a result of lipid-sorting specificity encoded in each Ras carboxy-terminal membrane anchor. The role of the G-domain in regulating anchor-membrane interactions is becoming clearer. Ras G-domains undergo significant conformational orientation changes on guanine nucleotide switch, leading to differential direct contacts between the G-domain and reorganization of the membrane anchor. Ras G-domains also contain weak dimer interfaces, resulting in homodimerization, which is an obligate step of nanoclustering. Modulating the formation of Ras dimers, the lipid composition of the PM or lateral dynamics of key PM phospholipids represent novel mechanisms whereby the extent of Ras nanoclustering can be regulated to tune the gain in Ras signaling circuits.


Subject(s)
Cell Membrane/physiology , Genes, ras/physiology , Signal Transduction
20.
Biomed Pharmacother ; 96: 489-496, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29032332

ABSTRACT

The high prevalence of end-stage renal disease emphasizes the failure to provide therapies to effectively prevent and/or reverse renal fibrosis. Therefore, the aim of this study was to evaluate the effect of long-term treatment with chaethomellic acid A (CAA), which selectively blocks Ha-Ras farnesylation, on renal mass reduction-induced renal fibrosis. Male Wistar rats were sham-operated (SO) or subjected to 5/6 renal mass reduction (RMR). One week after surgery, rats were placed in four experimental groups: SO:SO rats without treatment (n=13); SO+CAA: SO rats treated with CAA (n=13); RMR:RMR rats without treatment (n=14); and RMR+CAA:RMR rats treated with CAA (n=13). CAA was intraperitoneally administered in a dose of 0.23µg/kg three times a week for six months. Renal fibrosis was evaluated by two-dimensional ultrasonography and histopathological analysis. The kidneys of the RMR animals treated with CAA showed a significantly decrease in the medullary echogenicity (p<0.05) compared with the RMR rats that received no treatment. Glomerulosclerosis and arteriolosclerosis scores were significantly lower (p<0.001) in the RMR+CAA group when compared with the RMR group. There were no significant differences in interstitial fibrosis, interstitial inflammation and tubular dilatation scores between the RMR+CAA and RMR groups. These data suggest that CAA can be a potential future drug to attenuate the progression of chronic kidney disease.


Subject(s)
Arteriolosclerosis/diagnostic imaging , Disease Models, Animal , Glomerulosclerosis, Focal Segmental/diagnostic imaging , Renal Agents/therapeutic use , Renal Insufficiency, Chronic/diagnostic imaging , Animals , Arteriolosclerosis/drug therapy , Arteriolosclerosis/metabolism , Drug Administration Schedule , Genes, ras/drug effects , Genes, ras/physiology , Glomerulosclerosis, Focal Segmental/drug therapy , Glomerulosclerosis, Focal Segmental/metabolism , Male , Protein Prenylation/drug effects , Protein Prenylation/physiology , Rats , Rats, Wistar , Renal Agents/pharmacology , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Time Factors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...