833 results on '"Genes, ras physiology"'
Search Results
2. Oncogenic RAS commandeers amino acid sensing machinery to aberrantly activate mTORC1 in multiple myeloma.
- Author
-
Yang Y, Bolomsky A, Oellerich T, Chen P, Ceribelli M, Häupl B, Wright GW, Phelan JD, Huang DW, Lord JW, Van Winkle CK, Yu X, Wisniewski J, Wang JQ, Tosto FA, Beck E, Wilson K, McKnight C, Travers J, Klumpp-Thomas C, Smith GA, Pittaluga S, Maric I, Kazandjian D, Thomas CJ, and Young RM
- Subjects
- Amino Acids metabolism, Humans, Mechanistic Target of Rapamycin Complex 1 genetics, Mechanistic Target of Rapamycin Complex 1 metabolism, Mitogen-Activated Protein Kinase Kinases, Mutation, Protein Isoforms, Genes, ras genetics, Genes, ras physiology, Multiple Myeloma genetics, Multiple Myeloma metabolism, Transcription Factors genetics, Transcription Factors metabolism
- 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., (© 2022. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2022
- Full Text
- View/download PDF
3. BarH-like homeobox 2 represses the transcription of keratin 16 and affects Ras signaling pathway to suppress nasopharyngeal carcinoma progression.
- Author
-
Lu Z, Peng H, Li R, Xu X, and Peng J
- Subjects
- 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, Genes, ras physiology, Homeodomain Proteins physiology, Keratin-16 genetics, Nasopharyngeal Carcinoma pathology, Nasopharyngeal Neoplasms pathology
- 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.
- Published
- 2022
- Full Text
- View/download PDF
4. EGFRAP encodes a new negative regulator of the EGFR acting in both normal and oncogenic EGFR/Ras-driven tissue morphogenesis.
- Author
-
Soler Beatty J, Molnar C, Luque CM, de Celis JF, and Martín-Bermudo MD
- Subjects
- 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, ErbB Receptors antagonists & inhibitors, Genes, ras genetics
- 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., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
5. Interaction between Ras and Src clones causes interdependent tumor malignancy via Notch signaling in Drosophila.
- Author
-
Enomoto M, Takemoto D, and Igaki T
- Subjects
- 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, Neoplasms metabolism, Oncogene Protein pp60(v-src) metabolism, Proto-Oncogene Proteins p21(ras) metabolism
- 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., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
6. Increased expression of hras induces early, but not full, senescence in the immortal fish cell line, EPC.
- Author
-
Futami K, Aoyama K, Fukuda K, Maita M, and Katagiri T
- Subjects
- Aging genetics, Aging metabolism, Animals, Cell Line, Cells, Cultured, Cellular Senescence physiology, Fishes genetics, Humans, Proto-Oncogene Proteins p21(ras) genetics, Cellular Senescence genetics, Genes, ras genetics, Genes, ras physiology
- 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 (HRas
V12 ) showed p53-dependent senescence-like growth arrest and senescence-associated β-galactosidase (SA-β-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., (Copyright © 2020 Elsevier B.V. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
7. Glucose-6-Phosphate Dehydrogenase Is Not Essential for K-Ras-Driven Tumor Growth or Metastasis.
- Author
-
Ghergurovich JM, Esposito M, Chen Z, Wang JZ, Bhatt V, Lan T, White E, Kang Y, Guo JY, and Rabinowitz JD
- Subjects
- 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, Carcinoma, Non-Small-Cell Lung genetics, Colonic Neoplasms enzymology, Genes, ras physiology, Glucosephosphate Dehydrogenase physiology, Lung Neoplasms enzymology, Triple Negative Breast Neoplasms enzymology
- 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., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
8. The Myc and Ras Partnership in Cancer: Indistinguishable Alliance or Contextual Relationship?
- Author
-
Mahauad-Fernandez WD and Felsher DW
- Subjects
- Adenocarcinoma genetics, Carcinogenesis genetics, Carcinogenesis immunology, Cell Physiological Phenomena genetics, Female, Humans, Transcriptional Activation, Breast Neoplasms genetics, Genes, myc physiology, Genes, ras physiology, Hematologic Neoplasms genetics, Lung Neoplasms genetics, Tumor Microenvironment genetics
- 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., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
9. H-Ras induces Nrf2-Pin1 interaction: Implications for breast cancer progression.
- Author
-
Saeidi S, Kim SJ, Han HJ, Kim SH, Zheng J, Lee HB, Han W, Noh DY, Na HK, and Surh YJ
- Subjects
- 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, Breast Neoplasms metabolism, Genes, ras physiology, NF-E2-Related Factor 2 metabolism, NIMA-Interacting Peptidylprolyl Isomerase metabolism
- 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., Competing Interests: Declaration of Competing Interest The authors declare no potential conflicts of interest., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
10. Aggressive Medulloblastoma-Derived Exosomal miRNAs Promote In Vitro Invasion and Migration of Tumor Cells Via Ras/MAPK Pathway.
- Author
-
Zhu LY, Wu XY, Liu XD, Zheng DF, Li HS, Yang B, Zhang J, and Chang Q
- Subjects
- Cell Line, Tumor, Cerebellar Neoplasms genetics, Cerebellar Neoplasms pathology, Exosomes genetics, Humans, Medulloblastoma genetics, Medulloblastoma pathology, MicroRNAs genetics, Neoplasm Invasiveness genetics, Neoplasm Invasiveness pathology, Cell Movement physiology, Cerebellar Neoplasms metabolism, Exosomes metabolism, Genes, ras physiology, MAP Kinase Signaling System physiology, Medulloblastoma metabolism, MicroRNAs metabolism
- 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., (© 2020 American Association of Neuropathologists, Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
11. Targeting RAS in pediatric cancer: is it becoming a reality?
- Author
-
Vaseva AV and Yohe ME
- Subjects
- 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, Genes, ras genetics, Neoplasms drug therapy, Neoplasms genetics
- 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.
- Published
- 2020
- Full Text
- View/download PDF
12. Regulation of senescence and the SASP by the transcription factor C/EBPβ.
- Author
-
Salotti J and Johnson PF
- Subjects
- 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, CCAAT-Enhancer-Binding Protein-beta physiology, Cellular Senescence physiology, Oncogenes physiology
- 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/EBPβ. In this review, we summarize the critical role of C/EBPβ in regulating OIS and the SASP. Post-translational modifications induced by oncogenic RAS signaling control C/EBPβ activity and dimerization, and these alterations switch C/EBPβ to a pro-senescence form during OIS. In addition, C/EBPβ 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., (Published by Elsevier Inc.)
- Published
- 2019
- Full Text
- View/download PDF
13. The oncogene KRAS promotes cancer cell dissemination by stabilizing spheroid formation via the MEK pathway.
- Author
-
Ogishima J, Taguchi A, Kawata A, Kawana K, Yoshida M, Yoshimatsu Y, Sato M, Nakamura H, Kawata Y, Nishijima A, Fujimoto A, Tomio K, Adachi K, Nagamatsu T, Oda K, Kiyono T, Osuga Y, and Fujii T
- Subjects
- Animals, Cell Culture Techniques methods, Cell Line, Tumor, Female, Humans, Mice, Mice, Inbred C57BL, Ovarian Neoplasms pathology, Cell Proliferation physiology, Genes, ras physiology, MAP Kinase Signaling System physiology, Ovarian Neoplasms metabolism, Spheroids, Cellular metabolism
- 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.
- Published
- 2018
- Full Text
- View/download PDF
14. RASpecting the oncogene: New pathways to therapeutic advances.
- Author
-
Stout MC and Campbell PM
- Subjects
- Animals, Genes, ras drug effects, Humans, Molecular Targeted Therapy methods, Neoplasms genetics, Antineoplastic Agents administration & dosage, Genes, ras physiology, Molecular Targeted Therapy trends, Neoplasms drug therapy, Neoplasms metabolism
- 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., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
15. Ras and the Plasma Membrane: A Complicated Relationship.
- Author
-
Zhou Y, Prakash P, Gorfe AA, and Hancock JF
- Subjects
- Signal Transduction, Cell Membrane physiology, Genes, ras physiology
- 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., (Copyright © 2018 Cold Spring Harbor Laboratory Press; all rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
16. Four Key Steps Control Glycolytic Flux in Mammalian Cells.
- Author
-
Tanner LB, Goglia AG, Wei MH, Sehgal T, Parsons LR, Park JO, White E, Toettcher JE, and Rabinowitz JD
- Subjects
- 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, Glycolysis physiology, Hexokinase metabolism, Phosphofructokinase-1 metabolism
- 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., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
17. The best strategy for RAS wild-type metastatic colorectal cancer patients in first-line treatment: A classic and Bayesian meta-analysis.
- Author
-
Ciliberto D, Staropoli N, Caglioti F, Chiellino S, Ierardi A, Ingargiola R, Botta C, Arbitrio M, Correale P, Tassone P, and Tagliaferri P
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bayes Theorem, Colorectal Neoplasms genetics, Disease-Free Survival, Humans, Neoplasm Metastasis, Colorectal Neoplasms pathology, Colorectal Neoplasms therapy, Genes, ras physiology, Neoadjuvant Therapy methods
- 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 ± 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., (Copyright © 2018 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
18. KRAS oncogene in non-small cell lung cancer: clinical perspectives on the treatment of an old target.
- Author
-
Román M, Baraibar I, López I, Nadal E, Rolfo C, Vicent S, and Gil-Bazo I
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Carcinoma, Non-Small-Cell Lung drug therapy, Genes, ras physiology, Humans, Lung Neoplasms drug therapy, Mutation genetics, Carcinoma, Non-Small-Cell Lung genetics, Genes, ras genetics, Lung Neoplasms genetics
- 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.
- Published
- 2018
- Full Text
- View/download PDF
19. KRAS Oncogenic Signaling Extends beyond Cancer Cells to Orchestrate the Microenvironment.
- Author
-
Dias Carvalho P, Guimarães CF, Cardoso AP, Mendonça S, Costa ÂM, Oliveira MJ, and Velho S
- Subjects
- Extracellular Matrix genetics, Extracellular Matrix pathology, Fibroblasts pathology, Humans, Mutation, Myeloid Cells pathology, Neoplasms genetics, Neoplasms immunology, Signal Transduction, Tumor Escape genetics, Genes, ras physiology, Neoplasms pathology, Tumor Microenvironment genetics
- 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 ., (©2017 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
20. Long-term treatment with chaethomellic acid A reduces glomerulosclerosis and arteriolosclerosis in a rat model of chronic kidney disease.
- Author
-
Nogueira A, Vala H, Vasconcelos-Nóbrega C, Faustino-Rocha AI, Pires CA, Colaço A, Oliveira PA, and Pires MJ
- Subjects
- 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, Arteriolosclerosis diagnostic imaging, Disease Models, Animal, Glomerulosclerosis, Focal Segmental diagnostic imaging, Renal Agents therapeutic use, Renal Insufficiency, Chronic diagnostic imaging
- 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., (Copyright © 2017 Elsevier Masson SAS. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
21. Comprehensive profiling of H-Ras signalling in angiosarcoma endothelium.
- Author
-
da Costa A, Bonner M, and Arbiser JL
- Subjects
- Animals, Blotting, Western, Cell Line, Tumor, Hemangiosarcoma genetics, Mice, Neovascularization, Pathologic, Oligonucleotide Array Sequence Analysis, Rabbits, Genes, ras physiology, Hemangiosarcoma metabolism, Signal Transduction
- Abstract
The MS1/SVR system, in which MS1 represents immortalized endothelial cells and SVR represents MS1 cells transformed with oncogenic human-rat sarcoma protein (H-Ras), has been used for around 20 years as a valuable tool to study angiogenesis and carcinogenesis. Despite the use of these cells in numerous studies, a comprehensive profile of the signalling differences due to oncogenic H-Ras transformation has not been performed previously. In this study, we profiled the well-known MS1 and SVR cell lines using a combination of both Western blot and gene chip assays., (© 2017 British Association of Dermatologists.)
- Published
- 2017
- Full Text
- View/download PDF
22. Blockade of the IL-6 trans-signalling/STAT3 axis suppresses cachexia in Kras-induced lung adenocarcinoma.
- Author
-
Miller A, McLeod L, Alhayyani S, Szczepny A, Watkins DN, Chen W, Enriori P, Ferlin W, Ruwanpura S, and Jenkins BJ
- Subjects
- Adenocarcinoma drug therapy, Adenocarcinoma genetics, Adenocarcinoma pathology, Adenocarcinoma of Lung, Animals, Cachexia etiology, Cachexia pathology, Cell Transformation, Neoplastic genetics, Disease Models, Animal, Female, Humans, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms pathology, Male, Mice, Mice, Transgenic, STAT3 Transcription Factor antagonists & inhibitors, STAT3 Transcription Factor metabolism, Signal Transduction drug effects, Adenocarcinoma complications, Cachexia prevention & control, Genes, ras physiology, Interleukin-6 antagonists & inhibitors, Lung Neoplasms complications
- Abstract
Lung cancer is the leading cause of cancer death worldwide, and is frequently associated with the devastating paraneoplastic syndrome of cachexia. The potent immunomodulatory cytokine interleukin (IL)-6 has been linked with the development of lung cancer as well as cachexia; however, the mechanisms by which IL-6 promotes muscle wasting in lung cancer cachexia are ill-defined. In this study, we report that the gp130
F/F knock-in mouse model displaying hyperactivation of the latent transcription factor STAT3 via the common IL-6 cytokine family signalling receptor, gp130, develops cachexia during Kras-driven lung carcinogenesis. Specifically, exacerbated weight loss, early mortality and reduced muscle and adipose tissue mass were features of the gp130F/F :KrasG12D model, but not parental KrasG12D mice in which STAT3 was not hyperactivated. Gene expression profiling of muscle tissue in cachectic gp130F/F :KrasG12D mice revealed the upregulation of IL-6 and STAT3-target genes compared with KrasG12D muscle tissue. These cachectic features of gp130F/F :KrasG12D mice were abrogated upon the genetic normalization of STAT3 activation or ablation of IL-6 in gp130F/F :KrasG12D :Stat3-/+ or gp130F/F :KrasG12D :Il6-/- mice, respectively. Furthermore, protein levels of the soluble IL-6 receptor (sIL-6R), which is the central facilitator of IL-6 trans-signalling, were elevated in cachectic muscle from gp130F/F :KrasG12D mice, and the specific blockade of IL-6 trans-signalling, but not classical signalling, with an anti-IL-6R antibody ameliorated cachexia-related characteristics in gp130F/F :KrasG12D mice. Collectively, these preclinical findings identify trans-signalling via STAT3 as the signalling modality by which IL-6 promotes muscle wasting in lung cancer cachexia, and therefore support the clinical evaluation of the IL-6 trans-signalling/STAT3 axis as a therapeutic target in advanced lung cancer patients presenting with cachexia.- Published
- 2017
- Full Text
- View/download PDF
23. RAS and ROS-A Story of Pseudomonas Survival.
- Author
-
Kolar SL and Liu GY
- Subjects
- ADP-Ribosylation, NADPH Oxidases metabolism, Neutrophils immunology, Neutrophils metabolism, Neutrophils microbiology, Pneumonia, Pneumococcal immunology, Pneumonia, Pneumococcal metabolism, Pseudomonas genetics, Pseudomonas metabolism, Reactive Oxygen Species metabolism, Signal Transduction, Genes, ras physiology, Pseudomonas immunology, Reactive Oxygen Species immunology
- Abstract
Some pathogens block generation of reactive oxygen species to evade neutrophil killing, but how that is accomplished is poorly understood. In this issue of Cell Host & Microbe, Vareechon et al. (2017) describe ADP-ribosylation of Ras as a strategy to inhibit assembly of neutrophil NADPH oxidase., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
24. Nontypeable Haemophilus influenzae-Promoted Proliferation of Kras-Induced Early Adenomatous Lesions Is Completely Dependent on Toll-Like Receptor Signaling.
- Author
-
Jungnickel C, Schnabel PA, Bohle R, Wiewrodt R, Herr C, Bals R, and Beisswenger C
- Subjects
- Adenocarcinoma physiopathology, Animals, Cell Proliferation physiology, Haemophilus Infections physiopathology, Haemophilus influenzae physiology, Lung Neoplasms physiopathology, Mice, Neutrophils physiology, Pulmonary Disease, Chronic Obstructive virology, Signal Transduction physiology, Toll-Like Receptor 2 deficiency, Toll-Like Receptor 4 deficiency, ras Proteins metabolism, Genes, ras physiology, Pulmonary Disease, Chronic Obstructive physiopathology, Toll-Like Receptor 2 physiology, Toll-Like Receptor 4 physiology
- Abstract
Chronic obstructive pulmonary disease (COPD) is a risk factor for lung cancer. COPD is characterized by chronic airway inflammation and lung infections. The airways of patients with COPD are frequently colonized with bacteria [eg, nontypeable Haemophilus influenzae (NTHi)] that cause pulmonary inflammation and exacerbations. Pulmonary adenocarcinomas are frequently associated with an activating mutation in the KRAS gene. We determined the function of Toll-like receptor (TLR) signaling on the progression of Kras-induced early adenomatous lesions in the lung. Wild-type (WT) mice and mice doubly deficient in Tlr-2 and -4 (Tlr2/4
-/- ), both with an oncogenic Kras allele in lung epithelium, were exposed to NTHi for 4 weeks. Exposure to NTHi resulted in increased tumor proliferation and growth in WT mice, but not in Tlr2/4-/- mice. Alveolar adenomatous hyperplasia and adenocarcinoma were significantly increased in WT mice compared with Tlr2/4-/- mice. The average size of tumors was significantly larger in WT mice, whereas there was no difference in the number of alveolar lesions between WT and Tlr2/4-/- mice. NTHi-induced pulmonary neutrophilic inflammation and tumor-associated neutrophils were reduced in Tlr2/4-/- mice. Thus, subsequent to a driver mutation, NTHi-induced inflammation promotes proliferation of early adenomatous lesions in a TLR-dependent manner., (Copyright © 2017 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
25. Individual S-acylated cysteines differentially contribute to H-Ras endomembrane trafficking and acylation/deacylation cycles.
- Author
-
Pedro MP, Vilcaes AA, Gomez GA, and Daniotti JL
- Subjects
- Acylation, Animals, CHO Cells, Cell Line, Cell Membrane physiology, Cricetulus, Cysteine metabolism, Golgi Apparatus metabolism, Mutation, Proteins metabolism, Signal Transduction, Thiolester Hydrolases metabolism, Cell Membrane metabolism, Genes, ras physiology, Protein Transport physiology
- Abstract
S-acylation/deacylation cycles and vesicular transport are critical for an adequate subcellular distribution of S-acylated Ras proteins. H-Ras is dually acylated on cysteines 181 and 184, but it is unknown how these residues individually contribute to H-Ras trafficking. In this study, we characterized the acylation and deacylation rates and membrane trafficking of monoacylated H-Ras mutants to analyze their contributions to H-Ras plasma membrane and endomembrane distribution. We demonstrated that dually acylated H-Ras interacts with acyl-protein thioesterases (APTs) 1 and 2 at the plasma membrane. Moreover, single-acylation mutants of H-Ras differed not only in their subcellular distribution, where both proteins localized to different extents at both the Golgi complex and plasma membrane, but also in their deacylation rates, which we showed to be due to different sensitivities to APT1 and APT2. Fluorescence photobleaching and photoactivation experiments also revealed that 1) although S-acylated, single-acylation mutants are incorporated with different efficiencies into Golgi complex to plasma membrane vesicular carriers, and 2) the different deacylation rates of single-acylated H-Ras influence differentially its overall exchange between different compartments by nonvesicular transport. Taken together, our results show that individual S-acylation sites provide singular information about H-Ras subcellular distribution that is required for GTPase signaling., (© 2017 Pedro et al. This article is distributed by The American Society for Cell Biology under license from the author(s). Two months after publication it is available to the public under an Attribution–Noncommercial–Share Alike 3.0 Unported Creative Commons License (http://creativecommons.org/licenses/by-nc-sa/3.0).)
- Published
- 2017
- Full Text
- View/download PDF
26. The RAS-Effector Interaction as a Drug Target.
- Author
-
Keeton AB, Salter EA, and Piazza GA
- Subjects
- Animals, Cell Transformation, Neoplastic metabolism, Genes, ras drug effects, Genes, ras physiology, Humans, Mutation, Antineoplastic Agents pharmacology, Cell Transformation, Neoplastic genetics, Signal Transduction drug effects, ras Proteins genetics, ras Proteins metabolism
- Abstract
About a third of all human cancers harbor mutations in one of the K-, N-, or HRAS genes that encode an abnormal RAS protein locked in a constitutively activated state to drive malignant transformation and tumor growth. Despite more than three decades of intensive research aimed at the discovery of RAS-directed therapeutics, there are no FDA-approved drugs that are broadly effective against RAS-driven cancers. Although RAS proteins are often said to be "undruggable," there is mounting evidence suggesting it may be feasible to develop direct inhibitors of RAS proteins. Here, we review this evidence with a focus on compounds capable of inhibiting the interaction of RAS proteins with their effectors that transduce the signals of RAS and that drive and sustain malignant transformation and tumor growth. These reports of direct-acting RAS inhibitors provide valuable insight for further discovery and development of clinical candidates for RAS-driven cancers involving mutations in RAS genes or otherwise activated RAS proteins. Cancer Res; 77(2); 221-6. ©2017 AACR., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
27. Oncogenic RAS: From Its Activation to Its Direct Targeting.
- Author
-
Zaravinos A
- Subjects
- Animals, Antineoplastic Agents administration & dosage, Carcinogenesis drug effects, Drug Delivery Systems methods, Gene Expression Regulation, Neoplastic, Genes, ras drug effects, Humans, Mutation genetics, ras Proteins antagonists & inhibitors, Carcinogenesis genetics, Carcinogenesis metabolism, Drug Delivery Systems trends, Genes, ras physiology, ras Proteins genetics, ras Proteins metabolism
- Abstract
The RAS oncogenes are the most commonly mutated genes across human cancers. This review is a look back at the discovery of the RAS genes, how we came to understand their function, and how these oncogenes are aberrantly activated, driving cancer development and growth. It also highlights the comprehensive efforts made during the last 30 years to develop therapeutic strategies against the oncogenic mutant RAS. Although there have been many lost battles in the war against mutant RAS-bearing cancer cells, and we have yet to determine which is the "best" path for inhibition of the mutated RAS, a better understanding of the protein's structure combined with technological advances in the field has allowed the development of new strategies to make anti-RAS therapies a reality. The state-of-the-art for each strategy is discussed, focusing on the promises and potential pitfalls of each of them.
- Published
- 2017
- Full Text
- View/download PDF
28. CD99 triggering induces methuosis of Ewing sarcoma cells through IGF-1R/RAS/Rac1 signaling.
- Author
-
Manara MC, Terracciano M, Mancarella C, Sciandra M, Guerzoni C, Pasello M, Grilli A, Zini N, Picci P, Colombo MP, Morrione A, and Scotlandi K
- Subjects
- 12E7 Antigen antagonists & inhibitors, 12E7 Antigen physiology, Antineoplastic Agents, Immunological pharmacology, Apoptosis drug effects, Bone Neoplasms metabolism, Cells, Cultured, Genes, ras physiology, HEK293 Cells, Humans, Pinocytosis drug effects, Proteolysis drug effects, Receptor, IGF Type 1, Receptors, Somatomedin metabolism, Sarcoma, Ewing metabolism, Signal Transduction drug effects, rac1 GTP-Binding Protein metabolism, 12E7 Antigen immunology, Antibodies, Monoclonal pharmacology, Bone Neoplasms pathology, Cell Death drug effects, Sarcoma, Ewing pathology
- Abstract
CD99 is a cell surface molecule that has emerged as a novel target for Ewing sarcoma (EWS), an aggressive pediatric bone cancer. This report provides the first evidence of methuosis in EWS, a non-apoptotic form of cell death induced by an antibody directed against the CD99 molecule. Upon mAb triggering, CD99 induces an IGF-1R/RAS/Rac1 complex, which is internalized into RAB5-positive endocytic vacuoles. This complex is then dissociated, with the IGF-1R recycling to the cell membrane while CD99 and RAS/Rac1 are sorted into immature LAMP-1-positive vacuoles, whose excessive accumulation provokes methuosis. This process, which is not detected in CD99-expressing normal mesenchymal cells, is inhibited by disruption of the IGF-1R signaling, whereas enhanced by IGF-1 stimulation. Induction of IGF-1R/RAS/Rac1 was also observed in the EWS xenografts that respond to anti-CD99 mAb, further supporting the role of the IGF/RAS/Rac1 axis in the hyperstimulation of macropinocytosis and selective death of EWS cells. Thus, we describe a vulnerability of EWS cells, including those resistant to standard chemotherapy, to a treatment with anti-CD99 mAb, which requires IGF-1R/RAS signaling but bypasses the need for their direct targeting. Overall, we propose CD99 targeting as new opportunity to treat EWS patients resistant to canonical apoptosis-inducing agents.
- Published
- 2016
- Full Text
- View/download PDF
29. Roads to melanoma: Key pathways and emerging players in melanoma progression and oncogenic signaling.
- Author
-
Paluncic J, Kovacevic Z, Jansson PJ, Kalinowski D, Merlot AM, Huang ML, Lok HC, Sahni S, Lane DJ, and Richardson DR
- Subjects
- Animals, Disease Progression, Genes, ras physiology, Humans, MAP Kinase Signaling System physiology, Phosphatidylinositol 3-Kinases metabolism, Signal Transduction genetics, Wnt Signaling Pathway physiology, raf Kinases physiology, Melanoma genetics, Melanoma pathology, Oncogenes physiology, Skin Neoplasms genetics, Skin Neoplasms pathology
- Abstract
Melanoma has markedly increased worldwide during the past several decades in the Caucasian population and is responsible for 80% of skin cancer deaths. Considering that metastatic melanoma is almost completely resistant to most current therapies and is linked with a poor patient prognosis, it is crucial to further investigate potential molecular targets. Major cell-autonomous drivers in the pathogenesis of this disease include the classical MAPK (i.e., RAS-RAF-MEK-ERK), WNT, and PI3K signaling pathways. These pathways play a major role in defining the progression of melanoma, and some have been the subject of recent pharmacological strategies to treat this belligerent disease. This review describes the latest advances in the understanding of melanoma progression and the major molecular pathways involved. In addition, we discuss the roles of emerging molecular players that are involved in melanoma pathogenesis, including the functional role of the melanoma tumor antigen, p97/MFI2 (melanotransferrin)., (Copyright © 2016 Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
30. [KRAS and bronchial adenocarcinoma. Between disappointments and hopes].
- Author
-
Guibert N, Ilie M, Léna H, Didier A, Hofman P, and Mazieres J
- Subjects
- Adenocarcinoma diagnosis, Adenocarcinoma epidemiology, Biomarkers, Tumor genetics, Bronchial Neoplasms diagnosis, Bronchial Neoplasms epidemiology, Carcinogenesis genetics, Carcinoma, Non-Small-Cell Lung diagnosis, Carcinoma, Non-Small-Cell Lung epidemiology, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung therapy, Humans, Lung Neoplasms diagnosis, Lung Neoplasms epidemiology, Lung Neoplasms genetics, Lung Neoplasms therapy, Mutation, Prognosis, Adenocarcinoma genetics, Adenocarcinoma therapy, Bronchial Neoplasms genetics, Bronchial Neoplasms therapy, Genes, ras physiology, Molecular Targeted Therapy methods, Molecular Targeted Therapy trends
- Abstract
A better understanding of oncogenesis and the development of targeted therapies have led to improved outcomes in the treatment of lung cancer. KRAS mutation has the potential to drive the oncogenesis of almost one third of lung adenocarcinomas but it leads to a highly complex proliferation signal involving multiple signaling pathways, explaining the disappointing results of various inhibition strategies of K-ras or its effectors. Nevertheless, recent data suggest different roles of distinct KRAS mutation subtypes and KRAS interactions with new genes in the field of synthetic lethality mechanisms open the way to new therapeutic possibilities. This review aims to provide an overview of: 1) epidemiological data and particularly the prognostic impact of KRAS mutations in non-small cell lung cancer, 2) the results of different drugs either being tested in humans or sources of hope., (Copyright © 2015 Elsevier Masson SAS. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
31. Age-associated inflammation connects RAS-induced senescence to stem cell dysfunction and epidermal malignancy.
- Author
-
Golomb L, Sagiv A, Pateras IS, Maly A, Krizhanovsky V, Gorgoulis VG, Oren M, and Ben-Yehuda A
- Subjects
- Animals, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic pathology, Cellular Senescence genetics, Cellular Senescence physiology, Genes, ras genetics, Inflammation genetics, Mice, Reverse Transcriptase Polymerase Chain Reaction, Skin Neoplasms metabolism, Aging physiology, Genes, ras physiology, Inflammation metabolism, Skin Neoplasms pathology
- Abstract
Aging is the single biggest risk factor for malignant transformation. Among the most common age-associated malignancies are non-melanoma skin cancers, comprising the most common types of human cancer. Here we show that mutant H-Ras activation in mouse epidermis, a frequent event in cutaneous squamous cell carcinoma (SCC), elicits a differential outcome in aged versus young mice. Whereas H-Ras activation in the young skin results in hyperplasia that is mainly accompanied by rapid hair growth, H-Ras activation in the aged skin results in more dysplasia and gradual progression to in situ SCC. Progression is associated with increased inflammation, pronounced accumulation of immune cells including T cells, macrophages and mast cells as well as excessive cell senescence. We found not only an age-dependent increase in expression of several pro-inflammatory mediators, but also activation of a strong anti-inflammatory response involving enhanced IL4/IL10 expression and immune skewing toward a Th2 response. In addition, we observed an age-dependent increase in the expression of Pdl1, encoding an immune suppressive ligand that promotes cancer immune evasion. Moreover, upon switching off oncogenic H-Ras activity, young but not aged skin regenerates successfully, suggesting a failure of the aged epidermal stem cells to repair damaged tissue. Our findings support an age-dependent link between accumulation of senescent cells, immune infiltration and cancer progression, which may contribute to the increased cancer risk associated with old age.
- Published
- 2015
- Full Text
- View/download PDF
32. BTB-Zinc Finger Oncogenes Are Required for Ras and Notch-Driven Tumorigenesis in Drosophila.
- Author
-
Doggett K, Turkel N, Willoughby LF, Ellul J, Murray MJ, Richardson HE, and Brumby AM
- Subjects
- Animals, Animals, Genetically Modified, Drosophila Proteins genetics, Drosophila melanogaster, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Microarray Analysis, Nuclear Proteins chemistry, Nuclear Proteins genetics, Protein Interaction Domains and Motifs genetics, Carcinogenesis genetics, Drosophila Proteins physiology, Genes, ras physiology, Oncogenes physiology, Receptors, Notch physiology, Zinc Fingers genetics
- Abstract
During tumorigenesis, pathways that promote the epithelial-to-mesenchymal transition (EMT) can both facilitate metastasis and endow tumor cells with cancer stem cell properties. To gain a greater understanding of how these properties are interlinked in cancers we used Drosophila epithelial tumor models, which are driven by orthologues of human oncogenes (activated alleles of Ras and Notch) in cooperation with the loss of the cell polarity regulator, scribbled (scrib). Within these tumors, both invasive, mesenchymal-like cell morphology and continual tumor overgrowth, are dependent upon Jun N-terminal kinase (JNK) activity. To identify JNK-dependent changes within the tumors we used a comparative microarray analysis to define a JNK gene signature common to both Ras and Notch-driven tumors. Amongst the JNK-dependent changes was a significant enrichment for BTB-Zinc Finger (ZF) domain genes, including chronologically inappropriate morphogenesis (chinmo). chinmo was upregulated by JNK within the tumors, and overexpression of chinmo with either RasV12 or Nintra was sufficient to promote JNK-independent epithelial tumor formation in the eye/antennal disc, and, in cooperation with RasV12, promote tumor formation in the adult midgut epithelium. Chinmo primes cells for oncogene-mediated transformation through blocking differentiation in the eye disc, and promoting an escargot-expressing stem or enteroblast cell state in the adult midgut. BTB-ZF genes are also required for Ras and Notch-driven overgrowth of scrib mutant tissue, since, although loss of chinmo alone did not significantly impede tumor development, when loss of chinmo was combined with loss of a functionally related BTB-ZF gene, abrupt, tumor overgrowth was significantly reduced. abrupt is not a JNK-induced gene, however, Abrupt is present in JNK-positive tumor cells, consistent with a JNK-associated oncogenic role. As some mammalian BTB-ZF proteins are also highly oncogenic, our work suggests that EMT-promoting signals in human cancers could similarly utilize networks of these proteins to promote cancer stem cell states.
- Published
- 2015
- Full Text
- View/download PDF
33. Enhanced MET Translation and Signaling Sustains K-Ras-Driven Proliferation under Anchorage-Independent Growth Conditions.
- Author
-
Fujita-Sato S, Galeas J, Truitt M, Pitt C, Urisman A, Bandyopadhyay S, Ruggero D, and McCormick F
- Subjects
- Animals, Cell Adhesion genetics, Cells, Cultured, HCT116 Cells, HEK293 Cells, Humans, Mice, Protein Biosynthesis, Signal Transduction physiology, Tissue Culture Techniques, Cell Proliferation genetics, Genes, ras physiology, Proto-Oncogene Proteins c-met genetics, Proto-Oncogene Proteins c-met metabolism
- Abstract
Oncogenic K-Ras mutation occurs frequently in several types of cancers, including pancreatic and lung cancers. Tumors with K-Ras mutation are resistant to chemotherapeutic drugs as well as molecular targeting agents. Although numerous approaches are ongoing to find effective ways to treat these tumors, there are still no effective therapies for K-Ras mutant cancer patients. Here we report that K-Ras mutant cancers are more dependent on K-Ras in anchorage-independent culture conditions than in monolayer culture conditions. In seeking to determine mechanisms that contribute to the K-Ras dependency in anchorage-independent culture conditions, we discovered the involvement of Met in K-Ras-dependent, anchorage-independent cell growth. The Met signaling pathway is enhanced and plays an indispensable role in anchorage-independent growth even in cells in which Met is not amplified. Indeed, Met expression is elevated under anchorage-independent growth conditions and is regulated by K-Ras in a MAPK/ERK kinase (MEK)-dependent manner. Remarkably, in spite of a global downregulation of mRNA translation during anchorage-independent growth, we find that Met mRNA translation is specifically enhanced under these conditions. Importantly, ectopic expression of an active Met mutant rescues K-Ras ablation-derived growth suppression, indicating that K-Ras-mediated Met expression drives "K-Ras addiction" in anchorage-independent conditions. Our results indicate that enhanced Met expression and signaling is essential for anchorage-independent growth of K-Ras mutant cancer cells and suggests that pharmacological inhibitors of Met could be effective for K-Ras mutant tumor patients., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
34. Loss of RhoA Exacerbates, Rather Than Dampens, Oncogenic K-Ras Induced Lung Adenoma Formation in Mice.
- Author
-
Zandvakili I, Davis AK, Hu G, and Zheng Y
- Subjects
- Animals, Blotting, Western, Genes, ras genetics, Immunohistochemistry, Mice, ras Proteins genetics, ras Proteins metabolism, rhoA GTP-Binding Protein deficiency, rhoC GTP-Binding Protein, Adenoma genetics, Adenoma pathology, Genes, ras physiology, Lung Neoplasms genetics, Lung Neoplasms pathology, rhoA GTP-Binding Protein genetics
- Abstract
Numerous cellular studies have indicated that RhoA signaling is required for oncogenic Ras-induced transformation, suggesting that RhoA is a useful target in Ras induced neoplasia. However, to date very limited data exist to genetically attribute RhoA function to Ras-mediated tumorigenesis in mammalian models. In order to assess whether RhoA is required for K-Ras-induced lung cancer initiation, we utilized the K-RasG12D Lox-Stop-Lox murine lung cancer model in combination with a conditional RhoAflox/flox and RhoC-/- knockout mouse models. Deletion of the floxed Rhoa gene and expression of K-RasG12D was achieved by either CCSP-Cre or adenoviral Cre, resulting in simultaneous expression of K-RasG12D and deletion of RhoA from the murine lung. We found that deletion of RhoA, RhoC or both did not adversely affect normal lung development. Moreover, we found that deletion of either RhoA or RhoC alone did not suppress K-RasG12D induced lung adenoma initiation. Rather, deletion of RhoA alone exacerbated lung adenoma formation, whereas dual deletion of RhoA and RhoC together significantly reduced K-RasG12D induced adenoma formation. Deletion of RhoA appears to induce a compensatory mechanism that exacerbates adenoma formation. The compensatory mechanism is at least partly mediated by RhoC. This study suggests that targeting of RhoA alone may allow for compensation and a paradoxical exacerbation of neoplasia, while simultaneous targeting of both RhoA and RhoC is likely to lead to more favorable outcomes.
- Published
- 2015
- Full Text
- View/download PDF
35. Ras induces experimental lung metastasis through up-regulation of RbAp46 to suppress RECK promoter activity.
- Author
-
Yeh HH, Tseng YF, Hsu YC, Lan SH, Wu SY, Raghavaraju G, Cheng DE, Lee YR, Chang TY, Chow NH, Hung WC, and Liu HS
- Subjects
- Animals, Female, GPI-Linked Proteins antagonists & inhibitors, Humans, Lung Neoplasms pathology, MCF-7 Cells, Mice, Mice, Nude, NIH 3T3 Cells, Urinary Bladder Neoplasms metabolism, Urinary Bladder Neoplasms pathology, Urinary Bladder Neoplasms secondary, GPI-Linked Proteins metabolism, Genes, ras physiology, Lung Neoplasms metabolism, Promoter Regions, Genetic physiology, Retinoblastoma-Binding Protein 7 biosynthesis, Up-Regulation physiology
- Abstract
Background: Mutant Ras plays multiple functions in tumorigenesis including tumor formation and metastasis. Reversion-inducing cysteine-rich protein with Kazal motifs (RECK), a metastasis inhibitor gene, suppresses matrix metalloproteinase (MMP) activity in the metastatic cascade. Clarifying the relationship between Ras and RECK and understanding the underlying molecular mechanism may lead to the development of better treatment for Ras-related tumors., Methods: Suppression subtractive hybridization PCR (SSH PCR) was conducted to identify Ha-ras (val12) up-regulated genes in bladder cancer cells. Stable cell lines of human breast cancer (MCF-7-ras) and mouse NIH3T3 fibroblasts (7-4) harboring the inducible Ha-ras (val12) oncogene, which could be induced by isopropylthio-β-D-galactoside (IPTG), were used to clarify the relationship between Ras and the up-regulated genes. Chromatin immunoprecipitation (ChIP) assay, DNA affinity precipitation assay (DAPA) and RECK reporter gene assay were utilized to confirm the complex formation and binding with promoters., Results: Retinoblastoma binding protein-7 (RbAp46) was identified and confirmed as a Ha-ras (val12) up-regulated gene. RbAp46 could bind with histone deacetylase (HDAC1) and Sp1, followed by binding to RECK promoter at the Sp1 site resulting in repression of RECK expression. High expression of Ras protein accompanied with high RbAp46 and low RECK expression were detected in 75% (3/4) of the clinical bladder cancer tumor tissues compared to the adjacent normal parts. Ras induced RbAp46 expression increases invasion of the bladder cancer T24 cells and MMP-9 activity was increased, which was confirmed by specific lentiviral shRNAs inhibitors against Ras and RbAp46. Similarly, knockdown of RbAp46 expression in the stable NIH3T3 cells "7-4" by shRNA decreased Ras-related lung metastasis using a xenograft nude mice model., Conclusions: We confirmed that RbAp46 is a Ha-ras (val12) up-regulated gene and binds with HDAC1 and Sp1. Furthermore, RbAp46 binds to the RECK promoter at the Sp1 site via recruitment by Sp1. RECK is subsequently activated, leading to increased MMP9 activity, which may lead to increased metastasis in vivo. Our findings of Ras upregulation of RbAp46 may lead to revealing a novel mechanism of Ras-related tumor cell metastasis.
- Published
- 2015
- Full Text
- View/download PDF
36. Discoidin domain receptor 1 is a novel transcriptional target of ZEB1 in breast epithelial cells undergoing H-Ras-induced epithelial to mesenchymal transition.
- Author
-
Koh M, Woo Y, Valiathan RR, Jung HY, Park SY, Kim YN, Kim HR, Fridman R, and Moon A
- Subjects
- Cell Line, Tumor, Cytoskeleton physiology, Discoidin Domain Receptors, Epithelial Cells pathology, Female, Humans, MicroRNAs physiology, Morphogenesis, Zinc Finger E-box-Binding Homeobox 1, Breast pathology, Epithelial-Mesenchymal Transition, Genes, ras physiology, Homeodomain Proteins physiology, Receptor Protein-Tyrosine Kinases physiology, Receptors, Mitogen physiology, Transcription Factors physiology
- Abstract
The epithelial-to-mesenchymal transition (EMT) process allows carcinoma cells to dissociate from the primary tumor thereby facilitating tumor cell invasion and metastasis. Ras-dependent hyperactive signaling is commonly associated with tumorigenesis, invasion, EMT, and metastasis. However, the downstream effectors by which Ras regulates EMT remain ill defined. In this study, we show that the H-Ras pathway leads to mesenchymal-like phenotypic changes in human breast epithelial cells by controlling the ZEB1/microRNA-200c axis. Moreover, H-Ras suppresses the expression of the discoidin domain receptor 1 (DDR1), a collagen receptor tyrosine kinase, via ZEB1, thus identifying ZEB1 as a novel transcriptional repressor of DDR1. Mutation studies on the putative promoter of the DDR1 gene revealed that bipartite Z- and E-box elements play a key role in transcriptional repression of DDR1 in Hs578T and MDA-MB-231 breast carcinoma cell lines by ZEB1. Furthermore, we found an inverse correlation between ZEB1 and DDR1 expression in various cancer cell lines and in human breast carcinoma tissues. Consistently, overexpression of DDR1 reduced the invasive phenotype of mesenchymal-like triple-negative breast cancer cells in 3D cultures and in vivo. Thus, ZEB1's role in maintenance of EMT in breast carcinoma cells is mediated in part by its ability to suppress DDR1 expression and consequently contribute to the activation of the invasive phenotype. Taken together, our results unveil a novel H-Ras/ZEB1/DDR1 network that contributes to breast cancer progression in triple-negative breast cancers., Competing Interests: The authors disclose no potential conflicts of interest., (© 2014 UICC.)
- Published
- 2015
- Full Text
- View/download PDF
37. Carabin protects against cardiac hypertrophy by blocking calcineurin, Ras, and Ca2+/calmodulin-dependent protein kinase II signaling.
- Author
-
Bisserier M, Berthouze-Duquesnes M, Breckler M, Tortosa F, Fazal L, de Régibus A, Laurent AC, Varin A, Lucas A, Branchereau M, Marck P, Schickel JN, Deloménie C, Cazorla O, Soulas-Sprauel P, Crozatier B, Morel E, Heymes C, and Lezoualc'h F
- Subjects
- Animals, Calcium-Calmodulin-Dependent Protein Kinase Type 2 antagonists & inhibitors, Cells, Cultured, Female, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Myocytes, Cardiac metabolism, Rats, Signal Transduction physiology, Calcineurin metabolism, Calcium-Calmodulin-Dependent Protein Kinase Type 2 metabolism, Cardiomegaly metabolism, Cardiomegaly prevention & control, GTPase-Activating Proteins biosynthesis, Genes, ras physiology
- Abstract
Background: Cardiac hypertrophy is an early hallmark during the clinical course of heart failure and is regulated by various signaling pathways. However, the molecular mechanisms that negatively regulate these signal transduction pathways remain poorly understood., Methods and Results: Here, we characterized Carabin, a protein expressed in cardiomyocytes that was downregulated in cardiac hypertrophy and human heart failure. Four weeks after transverse aortic constriction, Carabin-deficient (Carabin(-/-)) mice developed exaggerated cardiac hypertrophy and displayed a strong decrease in fractional shortening (14.6±1.6% versus 27.6±1.4% in wild type plus transverse aortic constriction mice; P<0.0001). Conversely, compensation of Carabin loss through a cardiotropic adeno-associated viral vector encoding Carabin prevented transverse aortic constriction-induced cardiac hypertrophy with preserved fractional shortening (39.9±1.2% versus 25.9±2.6% in control plus transverse aortic constriction mice; P<0.0001). Carabin also conferred protection against adrenergic receptor-induced hypertrophy in isolated cardiomyocytes. Mechanistically, Carabin carries out a tripartite suppressive function. Indeed, Carabin, through its calcineurin-interacting site and Ras/Rab GTPase-activating protein domain, functions as an endogenous inhibitor of calcineurin and Ras/extracellular signal-regulated kinase prohypertrophic signaling. Moreover, Carabin reduced Ca(2+)/calmodulin-dependent protein kinase II activation and prevented nuclear export of histone deacetylase 4 after adrenergic stimulation or myocardial pressure overload. Finally, we showed that Carabin Ras-GTPase-activating protein domain and calcineurin-interacting domain were both involved in the antihypertrophic action of Carabin., Conclusions: Our study identifies Carabin as a negative regulator of key prohypertrophic signaling molecules, calcineurin, Ras, and Ca(2+)/calmodulin-dependent protein kinase II and implicates Carabin in the development of cardiac hypertrophy and failure., (© 2014 American Heart Association, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
38. Mitofusin 2 ameliorates aortic remodeling by suppressing ras/raf/ERK pathway and regulating mitochondrial function in vascular smooth muscle cells.
- Author
-
Wang Z, Niu Q, Peng X, Li M, Liu Y, Liu J, Wen S, and Wei Y
- Subjects
- Animals, Aorta metabolism, GTP Phosphohydrolases, Myocytes, Smooth Muscle metabolism, Random Allocation, Rats, Rats, Inbred SHR, Rats, Inbred WKY, Genes, ras physiology, MAP Kinase Signaling System physiology, Membrane Proteins biosynthesis, Mitochondria physiology, Mitochondrial Proteins biosynthesis, Muscle, Smooth, Vascular metabolism, Vascular Remodeling physiology, raf Kinases physiology
- Published
- 2015
- Full Text
- View/download PDF
39. [Gastrointestinal Cancers Symposium 2015: New analysis from PEAK study: Early, deep and sustained tumor shrinkage by panitumumab].
- Subjects
- Colorectal Neoplasms metabolism, Colorectal Neoplasms secondary, ErbB Receptors metabolism, Genes, ras physiology, Humans, Neoplasm Metastasis drug therapy, Panitumumab, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Colorectal Neoplasms drug therapy
- Published
- 2015
- Full Text
- View/download PDF
40. [Tumor dynamics: parameters for the overall benefit of therapy].
- Author
-
Karthaus M
- Subjects
- Colorectal Neoplasms secondary, ErbB Receptors metabolism, Genes, ras physiology, Humans, Panitumumab, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Colorectal Neoplasms drug therapy, Neoplasm Metastasis drug therapy
- Published
- 2015
41. Flavors of EGFR-Ras signals impacting intestinal homeostasis.
- Author
-
Depeille P and Roose JP
- Subjects
- Animals, Humans, Intestinal Mucosa cytology, Intestines cytology, ErbB Receptors metabolism, Genes, ras physiology, Homeostasis physiology, Intestinal Mucosa metabolism, Signal Transduction physiology
- Published
- 2015
- Full Text
- View/download PDF
42. Antroquinonol blocks Ras and Rho signaling via the inhibition of protein isoprenyltransferase activity in cancer cells.
- Author
-
Ho CL, Wang JL, Lee CC, Cheng HY, Wen WC, Cheng HH, and Chen MC
- Subjects
- Dimethylallyltranstransferase chemistry, Dimethylallyltranstransferase metabolism, Dose-Response Relationship, Drug, Enzyme Inhibitors pharmacology, Genes, ras physiology, Hep G2 Cells, Humans, K562 Cells, Protein Structure, Secondary, Protein Structure, Tertiary, Signal Transduction physiology, Ubiquinone pharmacology, rho-Associated Kinases metabolism, Dimethylallyltranstransferase antagonists & inhibitors, Genes, ras drug effects, Signal Transduction drug effects, Ubiquinone analogs & derivatives, rho-Associated Kinases antagonists & inhibitors
- Abstract
Antroquinonol is the smallest anticancer molecule isolated from Antrodia camphorata thus far. The ubiquinone-like structure of Antroquinonol exhibits a broad spectrum of activity against malignancies in vivo and in vitro. However, the mechanism of action of Antroquinonol remains unclear. Here, we provide evidence that Antroquinonol plays a role in the inhibition of Ras and Ras-related small GTP-binding protein functions through the inhibition of protein isoprenyl transferase activity in cancer cells. Using cell line-based assays, we found that the inactive forms of Ras and Rho proteins were significantly elevated after treatment with Antroquinonol. We also demonstrated that Antroquinonol binds directly to farnesyltransferase and geranylgeranyltransferase-I, which are key enzymes involved in activation of Ras-related proteins, and inhibits enzymes activities in vitro. Furthermore, a molecular docking analysis illustrated that the isoprenoid moiety of Antroquinonol binds along the hydrophobic cavity of farnesyltransferase similar to its natural substrate, farnesyl pyrophosphate. In contrast, the ring structure of Antroquinonol lies adjacent to the Ras-CAAX motif-binding site on farnesyltransferase. The molecular docking study also showed a reasonable correlation with the IC50 values of Antroquinonol analogues. We also found that the levels of LC3B-II and the autophagosome-associated LC3 form were also significantly increased in H838 after Antroquinonol administration. In conclusion, Antroquinonol inhibited Ras and Ras-related GTP-binding protein activation through inhibition of protein isoprenyl transferase activity, leading to activation of autophagy and associated mode of cell death in cancer cells., (Copyright © 2014 Elsevier Masson SAS. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
43. Parp-1 genetic ablation in Ela-myc mice unveils novel roles for Parp-1 in pancreatic cancer.
- Author
-
Martínez-Bosch N, Iglesias M, Munné-Collado J, Martínez-Cáceres C, Moreno M, Guerra C, Yélamos J, and Navarro P
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Apoptosis genetics, Disease Models, Animal, Female, Gene Expression Regulation, Neoplastic, Genes, ras physiology, Humans, Male, Mice, Mice, Knockout, Middle Aged, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Poly (ADP-Ribose) Polymerase-1, Pancreatic Neoplasms genetics, Poly(ADP-ribose) Polymerases genetics, Proto-Oncogene Proteins c-myc metabolism
- Abstract
Pancreatic cancer has a dismal prognosis and is currently the fourth leading cause of cancer-related death in developed countries. The inhibition of poly(ADP-ribose) polymerase-1 (Parp-1), the major protein responsible for poly(ADP-ribosy)lation in response to DNA damage, has emerged as a promising treatment for several tumour types. Here we aimed to elucidate the involvement of Parp-1 in pancreatic tumour progression. We assessed Parp-1 protein expression in normal, preneoplastic and pancreatic tumour samples from humans and from K-Ras- and c-myc-driven mouse models of pancreatic cancer. Parp-1 was highly expressed in acinar cells in normal and cancer tissues. In contrast, ductal cells expressed very low or undetectable levels of this protein, both in a normal and in a tumour context. The Parp-1 expression pattern was similar in human and mouse samples, thereby validating the use of animal models for further studies. To determine the in vivo effects of Parp-1 depletion on pancreatic cancer progression, Ela-myc-driven pancreatic tumour development was analysed in a Parp-1 knock-out background. Loss of Parp-1 resulted in increased tumour necrosis and decreased proliferation, apoptosis and angiogenesis. Interestingly, Ela-myc:Parp-1(-/-) mice displayed fewer ductal tumours than their Ela-myc:Parp-1(+/+) counterparts, suggesting that Parp-1 participates in promoting acinar-to-ductal metaplasia, a key event in pancreatic cancer initiation. Moreover, impaired macrophage recruitment can be responsible for the ADM blockade found in the Ela-myc:Parp-1(-/-) mice. Finally, molecular analysis revealed that Parp-1 modulates ADM downstream of the Stat3-MMP7 axis and is also involved in transcriptional up-regulation of the MDM2, VEGFR1 and MMP28 cancer-related genes. In conclusion, the expression pattern of Parp-1 in normal and cancer tissue and the in vivo functional effects of Parp-1 depletion point to a novel role for this protein in pancreatic carcinogenesis and shed light into the clinical use of Parp-1 inhibitors., (Copyright © 2014 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.)
- Published
- 2014
- Full Text
- View/download PDF
44. Two cases of neuroblastoma comprising two distinct clones.
- Author
-
Yamazaki F, Nakazawa A, Osumi T, Shimojima N, Tanaka T, Nakagawara A, and Shimada H
- Subjects
- Adrenal Gland Neoplasms classification, Adrenal Gland Neoplasms genetics, Anaplastic Lymphoma Kinase, Biomarkers, Tumor genetics, Gene Amplification, Genes, ras physiology, Humans, Infant, Male, N-Myc Proto-Oncogene Protein, Neuroblastoma classification, Neuroblastoma genetics, Nuclear Proteins genetics, Oncogene Proteins genetics, Prognosis, Receptor Protein-Tyrosine Kinases metabolism, Receptor, trkA metabolism, Adrenal Gland Neoplasms pathology, Biomarkers, Tumor metabolism, Neuroblastoma secondary
- Abstract
We report two cases of high-risk metastatic neuroblastoma, comprising two biologically distinct components in the adrenal primary tumor, which showed clear differences not only histologically but also in MYCN amplification and HA-RAS/TRKA immunoreactivity (Case 1), anaplastic lymphoma kinase (ALK) immunoreactivity (Case 2). These two cases with multiple separated components were similar to cases classified as ganglioneuroblastoma, nodular subtype (GNBn), in terms of composite tumor. Comparable to the GNBn category, the prognosis of the patients described here may depend on the components with unfavorable histology according to International Neuroblastoma Pathology Classification. Further analyses of such composite neuroblastoma cases are important for assessing disease prognosis., (© 2013 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
45. Paris saponin VII inhibits growth of colorectal cancer cells through Ras signaling pathway.
- Author
-
Li Y, Sun Y, Fan L, Zhang F, Meng J, Han J, Guo X, Zhang D, Zhang R, Yue Z, and Mei Q
- Subjects
- Animals, Antineoplastic Agents, Phytogenic isolation & purification, Antineoplastic Agents, Phytogenic pharmacology, Colorectal Neoplasms pathology, Female, Genes, ras drug effects, Growth Inhibitors isolation & purification, Growth Inhibitors pharmacology, HT29 Cells, Humans, Mice, Mice, Inbred ICR, Mice, Nude, Plant Extracts isolation & purification, Plant Extracts pharmacology, Plant Extracts therapeutic use, Saponins isolation & purification, Saponins pharmacology, Signal Transduction drug effects, Signal Transduction physiology, Xenograft Model Antitumor Assays methods, Antineoplastic Agents, Phytogenic therapeutic use, Colorectal Neoplasms drug therapy, Genes, ras physiology, Growth Inhibitors therapeutic use, Saponins therapeutic use, Trillium
- Abstract
Dysregulation of the Ras signaling pathway plays a key role in the progression of colorectal cancer. When bound to GTP, Ras is activated and stimulates several downstream effectors' pathways, including the Raf/MEK/ERK kinase cascade, the PI3-kinase/AKT/mTor pathway, and the Ral GTPase pathway. Saponins extracted from Liliaceae family herbs have strong antitumor activities with low toxicity. In this study, Paris saponin VII (PSVII), isolated from Trillium tschonoskii Maxim., was evaluated on human colorectal cancer cells (HT-29 and SW-620), a mouse model of colitis associated colorectal cancer (CACC) and a murine model of xenograft tumor. It was found that PSVII inhibited colorectal cancer cell growth in a concentration-dependent manner. The IC50 values of PSVII for growth inhibition of HT-29 and SW-620 cells were 1.02 ± 0.05 μM and 4.90 ± 0.23 μM. It could induce cell apoptosis, together with cell cycle arrest in G1 phase, and trigger apoptosis in a caspase-3-dependent manner. PSVII-induced growth inhibitory effect was associated with disturbance of MAPK pathway by down-regulating MEK1/2, ERK1/2 phosphorylation, and suppression of AKT pathway by reducing AKT and GSK-3β phosphorylation. In the CACC mouse model, PSVII protected mice from intestinal toxicities and carcinogenesis induced by 1,2-dimethylhydrazine (DMH) and dextran sodium sulfate (DSS). In the model of xenograft tumor, PSVII remarkably decreased the xenograft tumor size and triggered the apoptosis of tumor cells. Both in vitro and in vivo study showed that PSVII inhibited Ras activity. Taken together, PSVII might be a potential therapeutic reagent for colorectal cancer through targeting Ras signaling pathway., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
46. RAS transformation requires CUX1-dependent repair of oxidative DNA damage.
- Author
-
Ramdzan ZM, Vadnais C, Pal R, Vandal G, Cadieux C, Leduy L, Davoudi S, Hulea L, Yao L, Karnezis AN, Paquet M, Dankort D, and Nepveu A
- Subjects
- Animals, Cell Line, Tumor, Cell Transformation, Neoplastic genetics, Cells, Cultured, Cellular Senescence genetics, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Humans, Mammary Neoplasms, Experimental genetics, Mice, Transgenic, Nuclear Proteins genetics, Nuclear Proteins metabolism, Oxidative Stress, Repressor Proteins genetics, Repressor Proteins metabolism, Transcription Factors, DNA Damage, DNA Repair physiology, Genes, ras physiology, Homeodomain Proteins physiology, Nuclear Proteins physiology, Repressor Proteins physiology
- Abstract
The Cut homeobox 1 (CUX1) gene is a target of loss-of-heterozygosity in many cancers, yet elevated CUX1 expression is frequently observed and is associated with shorter disease-free survival. The dual role of CUX1 in cancer is illustrated by the fact that most cell lines with CUX1 LOH display amplification of the remaining allele, suggesting that decreased CUX1 expression facilitates tumor development while increased CUX1 expression is needed in tumorigenic cells. Indeed, CUX1 was found in a genome-wide RNAi screen to identify synthetic lethal interactions with oncogenic RAS. Here we show that CUX1 functions in base excision repair as an ancillary factor for the 8-oxoG-DNA glycosylase, OGG1. Single cell gel electrophoresis (comet assay) reveals that Cux1⁺/⁻ MEFs are haploinsufficient for the repair of oxidative DNA damage, whereas elevated CUX1 levels accelerate DNA repair. In vitro base excision repair assays with purified components demonstrate that CUX1 directly stimulates OGG1's enzymatic activity. Elevated reactive oxygen species (ROS) levels in cells with sustained RAS pathway activation can cause cellular senescence. We show that elevated expression of either CUX1 or OGG1 prevents RAS-induced senescence in primary cells, and that CUX1 knockdown is synthetic lethal with oncogenic RAS in human cancer cells. Elevated CUX1 expression in a transgenic mouse model enables the emergence of mammary tumors with spontaneous activating Kras mutations. We confirmed cooperation between Kras(G12V) and CUX1 in a lung tumor model. Cancer cells can overcome the antiproliferative effects of excessive DNA damage by inactivating a DNA damage response pathway such as ATM or p53 signaling. Our findings reveal an alternate mechanism to allow sustained proliferation in RAS-transformed cells through increased DNA base excision repair capability. The heightened dependency of RAS-transformed cells on base excision repair may provide a therapeutic window that could be exploited with drugs that specifically target this pathway., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2014
- Full Text
- View/download PDF
47. Realgar bioleaching solution suppress ras excessive activation by increasing ROS in Caenorhabditis elegans.
- Author
-
Zhi de J, Feng N, Liu DL, Hou RL, Wang MZ, Ding XX, and Li HY
- Subjects
- Animals, Animals, Genetically Modified, Dose-Response Relationship, Drug, Genes, ras physiology, Oxidative Stress drug effects, Oxidative Stress physiology, Pharmaceutical Solutions pharmacology, Arsenicals pharmacology, Caenorhabditis elegans drug effects, Caenorhabditis elegans metabolism, Genes, ras drug effects, Reactive Oxygen Species metabolism, Sulfides pharmacology
- Abstract
Although realgar bioleaching solution (RBS) has been proved to be a potential candidate for cancer therapy, the mechanisms of RBS anticancer are still far from being completely understood. Dosed with RBS in C. elegans, the multivulva phenotype resulting from oncogenic ras gain-of-function was inhibited in a dose dependent manner. It could be abrogated by concurrent treatment C. elegans with RBS and the radical scavenger DMSO. However, RBS could not induce DAF-16 nuclear translocation in TJ356 or the increase of HSP 16.2 expression in CL2070, which both could be aroused visible GFP fluorescent variation to represent for oxidative stress generation. Treatment C. elegans with superoxide anion generator paraquat, similar results were also obtained. Our results indicated that RBS suppress excessive activated ras by increasing reactive oxygen species (ROS) in C. elegans. Secondly, ROS induced by RBS significantly accumulated on a higher level in C. elegans with a mutational ras than that with wild ras, thus leading to oxidative stress on ras gain-of-function background rather than on normal ras context. Our results firstly demonstrated that using C. elegans as a model organism for evaluating prooxidant drug candidates for cancer therapy.
- Published
- 2014
- Full Text
- View/download PDF
48. CRR9/CLPTM1L regulates cell survival signaling and is required for Ras transformation and lung tumorigenesis.
- Author
-
James MA, Vikis HG, Tate E, Rymaszewski AL, and You M
- Subjects
- Animals, Carcinoma, Non-Small-Cell Lung pathology, Cell Survival genetics, HEK293 Cells, Humans, Lung Neoplasms pathology, Mice, Mice, Nude, NIH 3T3 Cells, Signal Transduction genetics, Tumor Cells, Cultured, Carcinoma, Non-Small-Cell Lung genetics, Cell Transformation, Neoplastic genetics, Genes, ras physiology, Lung Neoplasms genetics, Membrane Proteins physiology, Neoplasm Proteins physiology
- Abstract
The transmembrane protein CLPTM1L is overexpressed in non-small cell lung cancer, where it protects tumor cells from genotoxic apoptosis. Here, we show that RNA interference-mediated blockade of CLPTM1L inhibits K-Ras-induced lung tumorigenesis. CLPTM1L expression was required in vitro for morphologic transformation by H-RasV12 or K-RasV12, anchorage-independent growth, and survival of anoikis of lung tumor cells. Mechanistic investigations indicated that CLPTM1L interacts with phosphoinositide 3-kinase and is essential for Ras-induced AKT phosphorylation. Furthermore that the anti-apoptotic protein Bcl-xL is regulated by CLPTM1L independently of AKT activation. Constitutive activation of AKT or Bcl-xL rescued the transformed phenotype in CLPTM1L-depleted cells. The CLPTM1L gene lies within a cancer susceptibility locus at chromosome 5p15.33 defined by genome-wide association studies. The risk genotype at the CLPTM1L locus was associated with high expression of CLPTM1L in normal lung tissue, suggesting that cis-regulation of CLPTM1L may contribute to lung cancer risk. Taken together, our results establish a protumorigenic role for CLPTM1L that is critical for Ras-driven lung cancers, with potential implications for therapy and chemosensitization., (©2013 AACR.)
- Published
- 2014
- Full Text
- View/download PDF
49. RAS/ERK signaling controls proneural genetic programs in cortical development and gliomagenesis.
- Author
-
Li S, Mattar P, Dixit R, Lawn SO, Wilkinson G, Kinch C, Eisenstat D, Kurrasch DM, Chan JA, and Schuurmans C
- Subjects
- Animals, Brain Neoplasms pathology, Cerebral Cortex embryology, Cerebral Cortex pathology, Female, Glioma pathology, HEK293 Cells, Humans, Male, Mice, Mice, Transgenic, Pregnancy, Brain Neoplasms metabolism, Cerebral Cortex metabolism, Genes, ras physiology, Glioma metabolism, MAP Kinase Signaling System physiology, Neurons metabolism
- Abstract
Neural cell fate specification is well understood in the embryonic cerebral cortex, where the proneural genes Neurog2 and Ascl1 are key cell fate determinants. What is less well understood is how cellular diversity is generated in brain tumors. Gliomas and glioneuronal tumors, which are often localized in the cerebrum, are both characterized by a neoplastic glial component, but glioneuronal tumors also have an intermixed neuronal component. A core abnormality in both tumor groups is overactive RAS/ERK signaling, a pro-proliferative signal whose contributions to cell differentiation in oncogenesis are largely unexplored. We found that RAS/ERK activation levels differ in two distinct human tumors associated with constitutively active BRAF. Pilocytic astrocytomas, which contain abnormal glial cells, have higher ERK activation levels than gangliogliomas, which contain abnormal neuronal and glial cells. Using in vivo gain of function and loss of function in the mouse embryonic neocortex, we found that RAS/ERK signals control a proneural genetic switch, inhibiting Neurog2 expression while inducing Ascl1, a competing lineage determinant. Furthermore, we found that RAS/ERK levels control Ascl1's fate specification properties in murine cortical progenitors--at higher RAS/ERK levels, Ascl1(+) progenitors are biased toward proliferative glial programs, initiating astrocytomas, while at moderate RAS/ERK levels, Ascl1 promotes GABAergic neuronal and less glial differentiation, generating glioneuronal tumors. Mechanistically, Ascl1 is phosphorylated by ERK, and ERK phosphoacceptor sites are necessary for Ascl1's GABAergic neuronal and gliogenic potential. RAS/ERK signaling thus acts as a rheostat to influence neural cell fate selection in both normal cortical development and gliomagenesis, controlling Neurog2-Ascl1 expression and Ascl1 function.
- Published
- 2014
- Full Text
- View/download PDF
50. Regulation of polarized morphogenesis by protein kinase C iota in oncogenic epithelial spheroids.
- Author
-
Linch M, Sanz-Garcia M, Rosse C, Riou P, Peel N, Madsen CD, Sahai E, Downward J, Khwaja A, Dillon C, Roffey J, Cameron AJ, and Parker PJ
- Subjects
- Animals, Cell Transformation, Neoplastic metabolism, Cells, Cultured, Cysts metabolism, Dogs, Epithelial Cells metabolism, Genes, ras physiology, Humans, Isoenzymes antagonists & inhibitors, Isoenzymes genetics, Kidney metabolism, Kidney pathology, Phosphatidylinositol 3-Kinase metabolism, Protein Kinase C antagonists & inhibitors, Protein Kinase C genetics, RNA, Small Interfering genetics, Receptor, ErbB-2 metabolism, Spheroids, Cellular metabolism, Cell Polarity, Cell Transformation, Neoplastic pathology, Cysts pathology, Epithelial Cells pathology, Isoenzymes metabolism, Morphogenesis physiology, Protein Kinase C metabolism, Spheroids, Cellular pathology
- Abstract
Protein kinase C iota (PKCι), a serine/threonine kinase required for cell polarity, proliferation and migration, is commonly up- or downregulated in cancer. PKCι is a human oncogene but whether this is related to its role in cell polarity and what repertoire of oncogenes acts in concert with PKCι is not known. We developed a panel of candidate oncogene expressing Madin-Darby canine kidney (MDCK) cells and demonstrated that H-Ras, ErbB2 and phosphatidylinositol 3-kinase transformation led to non-polar spheroid morphogenesis (dysplasia), whereas MDCK spheroids expressing c-Raf or v-Src were largely polarized. We show that small interfering RNA (siRNA)-targeting PKCι decreased the size of all spheroids tested and partially reversed the aberrant polarity phenotype in H-Ras and ErbB2 spheroids only. This indicates distinct requirements for PKCι and moreover that different thresholds of PKCι activity are required for these phenotypes. By manipulating PKCι function using mutant constructs, siRNA depletion or chemical inhibition, we have demonstrated that PKCι is required for polarization of parental MDCK epithelial cysts in a 3D matrix and that there is a threshold of PKCι activity above and below which, disorganized epithelial morphogenesis results. Furthermore, treatment with a novel PKCι inhibitor, CRT0066854, was able to restore polarized morphogenesis in the dysplastic H-Ras spheroids. These results show that tightly regulated PKCι is required for normal-polarized morphogenesis in mammalian cells and that H-Ras and ErbB2 cooperate with PKCι for loss of polarization and dysplasia. The identification of a PKCι inhibitor that can restore polarized morphogenesis has implications for the treatment of Ras and ErbB2 driven malignancies.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.