Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
ACS Cent Sci ; 7(8): 1292-1294, 2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34471672
2.
Elife ; 102021 02 02.
Article in English | MEDLINE | ID: mdl-33526168

ABSTRACT

A farnesylated and methylated form of prelamin A called progerin causes Hutchinson-Gilford progeria syndrome (HGPS). Inhibiting progerin methylation by inactivating the isoprenylcysteine carboxylmethyltransferase (ICMT) gene stimulates proliferation of HGPS cells and improves survival of Zmpste24-deficient mice. However, we don't know whether Icmt inactivation improves phenotypes in an authentic HGPS mouse model. Moreover, it is unknown whether pharmacologic targeting of ICMT would be tolerated by cells and produce similar cellular effects as genetic inactivation. Here, we show that knockout of Icmt improves survival of HGPS mice and restores vascular smooth muscle cell numbers in the aorta. We also synthesized a potent ICMT inhibitor called C75 and found that it delays senescence and stimulates proliferation of late-passage HGPS cells and Zmpste24-deficient mouse fibroblasts. Importantly, C75 did not influence proliferation of wild-type human cells or Zmpste24-deficient mouse cells lacking Icmt, indicating drug specificity. These results raise hopes that ICMT inhibitors could be useful for treating children with HGPS.


Subject(s)
Cellular Senescence/drug effects , Progeria/drug therapy , Protein Methyltransferases/drug effects , Pyrans/pharmacology , Animals , Aorta/pathology , Cell Line , Cell Proliferation/drug effects , Disease Models, Animal , Humans , Lamin Type A/metabolism , Mice , Mice, Knockout , Myocytes, Smooth Muscle , Progeria/genetics , Progeria/pathology , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism
3.
Antioxidants (Basel) ; 10(2)2021 Feb 04.
Article in English | MEDLINE | ID: mdl-33557356

ABSTRACT

Dietary antioxidants and supplements are widely used to protect against cancer, even though it is now clear that antioxidants can promote tumor progression by helping cancer cells to overcome barriers of oxidative stress. Although recent studies have, in great detail, explored the role of antioxidants in lung and skin tumors driven by RAS and RAF mutations, little is known about the impact of antioxidant supplementation on other cancers, including Wnt-driven tumors originating from the gut. Here, we show that supplementation with the antioxidants N-acetylcysteine (NAC) and vitamin E promotes intestinal tumor progression in the ApcMin mouse model for familial adenomatous polyposis, a hereditary form of colorectal cancer, driven by Wnt signaling. Both antioxidants increased tumor size in early neoplasias and tumor grades in more advanced lesions without any impact on tumor initiation. Importantly, NAC treatment accelerated tumor progression at plasma concentrations comparable to those obtained in human subjects after prescription doses of the drug. These results demonstrate that antioxidants play an important role in the progression of intestinal tumors, which may have implications for patients with or predisposed to colorectal cancer.

4.
Antioxidants (Basel) ; 10(2)2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33499262

ABSTRACT

Cancer cells produce high levels of mitochondria-associated reactive oxygen species (ROS) that can damage macromolecules, but also promote cell signaling and proliferation. Therefore, mitochondria-targeted antioxidants have been suggested to be useful in anti-cancer therapy, but no studies have convincingly addressed this question. Here, we administered the mitochondria-targeted antioxidants MitoQ and MitoTEMPO to mice with BRAF-induced malignant melanoma and KRAS-induced lung cancer, and found that these compounds had no impact on the number of primary tumors and metastases; and did not influence mitochondrial and nuclear DNA damage levels. Moreover, MitoQ and MitoTEMPO did not influence proliferation of human melanoma and lung cancer cell lines. MitoQ and its control substance dTPP, but not MitoTEMPO, increased glycolytic rates and reduced respiration in melanoma cells; whereas only dTPP produced this effect in lung cancer cells. Our results do not support the use of mitochondria-targeted antioxidants for anti-cancer monotherapy, at least not in malignant melanoma and lung cancer.

6.
Aging Cell ; 19(8): e13200, 2020 08.
Article in English | MEDLINE | ID: mdl-32910507

ABSTRACT

Several progeroid disorders are caused by deficiency in the endoprotease ZMPSTE24 which leads to accumulation of prelamin A at the nuclear envelope. ZMPSTE24 cleaves prelamin A twice: at the third carboxyl-terminal amino acid following farnesylation of a -CSIM motif; and 15 residues upstream to produce mature lamin A. The carboxyl-terminal cleavage can also be performed by RAS-converting enzyme 1 (RCE1) but little is known about the importance of this cleavage for the ability of prelamin A to cause disease. Here, we found that knockout of RCE1 delayed senescence and increased proliferation of ZMPSTE24-deficient fibroblasts from a patient with non-classical Hutchinson-Gilford progeria syndrome (HGPS), but did not influence proliferation of classical LMNA-mutant HGPS cells. Knockout of Rce1 in Zmpste24-deficient mice at postnatal week 4-5 increased body weight and doubled the median survival time. The absence of Rce1 in Zmpste24-deficient fibroblasts did not influence nuclear shape but reduced an interaction between prelamin A and AKT which activated AKT-mTOR signaling and was required for the increased proliferation. Prelamin A levels increased in Rce1-deficient cells due to a slower turnover rate but its localization at the nuclear rim was unaffected. These results strengthen the idea that the presence of misshapen nuclei does not prevent phenotype improvement and suggest that targeting RCE1 might be useful for treating the rare progeroid disorders associated with ZMPSTE24 deficiency.


Subject(s)
Genes, ras/genetics , Membrane Proteins/deficiency , Metalloendopeptidases/deficiency , Progeria/genetics , Animals , Disease Models, Animal , Humans , Mice , Mice, Knockout , Phenotype
7.
Nat Commun ; 10(1): 3975, 2019 09 04.
Article in English | MEDLINE | ID: mdl-31484924

ABSTRACT

Rho family proteins are prenylated by geranylgeranyltransferase type I (GGTase-I), which normally target proteins to membranes for GTP-loading. However, conditional deletion of GGTase-I in mouse macrophages increases GTP-loading of Rho proteins, leading to enhanced inflammatory responses and severe rheumatoid arthritis. Here we show that heterozygous deletion of the Rho family gene Rac1, but not Rhoa and Cdc42, reverses inflammation and arthritis in GGTase-I-deficient mice. Non-prenylated Rac1 has a high affinity for the adaptor protein Ras GTPase-activating-like protein 1 (Iqgap1), which facilitates both GTP exchange and ubiquitination-mediated degradation of Rac1. Consistently, inactivating Iqgap1 normalizes Rac1 GTP-loading, and reduces inflammation and arthritis in GGTase-I-deficient mice, as well as prevents statins from increasing Rac1 GTP-loading and cytokine production in macrophages. We conclude that blocking prenylation stimulates Rac1 effector interactions and unleashes proinflammatory signaling. Our results thus suggest that prenylation normally restrains innate immune responses by preventing Rac1 effector interactions.


Subject(s)
Immunity, Innate/genetics , Protein Prenylation , Signal Transduction/genetics , rac1 GTP-Binding Protein/genetics , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Animals , Cytokines/metabolism , Macrophages/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Binding , RAW 264.7 Cells , rac1 GTP-Binding Protein/metabolism , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
8.
Cell ; 178(2): 330-345.e22, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31257027

ABSTRACT

For tumors to progress efficiently, cancer cells must overcome barriers of oxidative stress. Although dietary antioxidant supplementation or activation of endogenous antioxidants by NRF2 reduces oxidative stress and promotes early lung tumor progression, little is known about its effect on lung cancer metastasis. Here, we show that long-term supplementation with the antioxidants N-acetylcysteine and vitamin E promotes KRAS-driven lung cancer metastasis. The antioxidants stimulate metastasis by reducing levels of free heme and stabilizing the transcription factor BACH1. BACH1 activates transcription of Hexokinase 2 and Gapdh and increases glucose uptake, glycolysis rates, and lactate secretion, thereby stimulating glycolysis-dependent metastasis of mouse and human lung cancer cells. Targeting BACH1 normalized glycolysis and prevented antioxidant-induced metastasis, while increasing endogenous BACH1 expression stimulated glycolysis and promoted metastasis, also in the absence of antioxidants. We conclude that BACH1 stimulates glycolysis-dependent lung cancer metastasis and that BACH1 is activated under conditions of reduced oxidative stress.


Subject(s)
Antioxidants/pharmacology , Basic-Leucine Zipper Transcription Factors/metabolism , Glycolysis/drug effects , Lung Neoplasms/pathology , Animals , Antioxidants/administration & dosage , Basic-Leucine Zipper Transcription Factors/genetics , Cell Movement/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Heme/metabolism , Hexokinase/antagonists & inhibitors , Hexokinase/genetics , Hexokinase/metabolism , Humans , Kaplan-Meier Estimate , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , NF-E2-Related Factor 2/metabolism , Neoplasm Metastasis , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism
9.
Sci Transl Med ; 7(308): 308re8, 2015 Oct 07.
Article in English | MEDLINE | ID: mdl-26446958

ABSTRACT

Antioxidants in the diet and supplements are widely used to protect against cancer, but clinical trials with antioxidants do not support this concept. Some trials show that antioxidants actually increase cancer risk and a study in mice showed that antioxidants accelerate the progression of primary lung tumors. However, little is known about the impact of antioxidant supplementation on the progression of other types of cancer, including malignant melanoma. We show that administration of N-acetylcysteine (NAC) increases lymph node metastases in an endogenous mouse model of malignant melanoma but has no impact on the number and size of primary tumors. Similarly, NAC and the soluble vitamin E analog Trolox markedly increased the migration and invasive properties of human malignant melanoma cells but did not affect their proliferation. Both antioxidants increased the ratio between reduced and oxidized glutathione in melanoma cells and in lymph node metastases, and the increased migration depended on new glutathione synthesis. Furthermore, both NAC and Trolox increased the activation of the small guanosine triphosphatase (GTPase) RHOA, and blocking downstream RHOA signaling abolished antioxidant-induced migration. These results demonstrate that antioxidants and the glutathione system play a previously unappreciated role in malignant melanoma progression.


Subject(s)
Antioxidants/pharmacology , Melanoma/chemically induced , Acetylcysteine/adverse effects , Acetylcysteine/pharmacology , Animals , Antioxidants/adverse effects , Cell Line, Tumor , Chromans/adverse effects , Chromans/pharmacology , Dietary Supplements/adverse effects , Disease Models, Animal , Glutathione/metabolism , Humans , Male , Melanoma/pathology , Mice , Neoplasm Metastasis/pathology
10.
Circ Res ; 115(9): 781-9, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25212213

ABSTRACT

RATIONALE: Cell proliferation and cell cycle control mechanisms are thought to play central roles in the pathogenesis of atherosclerosis. The transcription factor Zinc finger protein 148 (Zfp148) was shown recently to maintain cell proliferation under oxidative conditions by suppressing p53, a checkpoint protein that arrests proliferation in response to various stressors. It is established that inactivation of p53 accelerates atherosclerosis, but whether increased p53 activation confers protection against the disease remains to be determined. OBJECTIVE: We aimed to test the hypothesis that Zfp148 deficiency reduces atherosclerosis by unleashing p53 activity. METHODS AND RESULTS: Mice harboring a gene-trap mutation in the Zfp148 locus (Zfp148(gt/+)) were bred onto the apolipoprotein E (Apoe)(-/-) genetic background and fed a high-fat or chow diet. Loss of 1 copy of Zfp148 markedly reduced atherosclerosis without affecting lipid metabolism. Bone marrow transplantation experiments revealed that the effector cell is of hematopoietic origin. Peritoneal macrophages and atherosclerotic lesions from Zfp148(gt/+)Apoe(-/-) mice showed increased levels of phosphorylated p53 compared with controls, and atherosclerotic lesions contained fewer proliferating macrophages. Zfp148(gt/+)Apoe(-/-) mice were further crossed with p53-null mice (Trp53(-/-) [the gene encoding p53]). There was no difference in atherosclerosis between Zfp148(gt/+)Apoe(-/-) mice and controls on a Trp53(+/-) genetic background, and there was no difference in levels of phosphorylated p53 or cell proliferation. CONCLUSIONS: Zfp148 deficiency increases p53 activity and protects against atherosclerosis by causing proliferation arrest of lesional macrophages, suggesting that drugs targeting macrophage proliferation may be useful in the treatment of atherosclerosis.


Subject(s)
Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Cell Cycle Checkpoints , Cell Proliferation , DNA-Binding Proteins/deficiency , Macrophages, Peritoneal/metabolism , Transcription Factors/deficiency , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Animals , Aortic Diseases/etiology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/etiology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Bone Marrow Transplantation , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diet, High-Fat , Disease Models, Animal , Humans , Macrophages, Peritoneal/pathology , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Plaque, Atherosclerotic , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics
11.
Sci Transl Med ; 6(221): 221ra15, 2014 Jan 29.
Article in English | MEDLINE | ID: mdl-24477002

ABSTRACT

Antioxidants are widely used to protect cells from damage induced by reactive oxygen species (ROS). The concept that antioxidants can help fight cancer is deeply rooted in the general population, promoted by the food supplement industry, and supported by some scientific studies. However, clinical trials have reported inconsistent results. We show that supplementing the diet with the antioxidants N-acetylcysteine (NAC) and vitamin E markedly increases tumor progression and reduces survival in mouse models of B-RAF- and K-RAS-induced lung cancer. RNA sequencing revealed that NAC and vitamin E, which are structurally unrelated, produce highly coordinated changes in tumor transcriptome profiles, dominated by reduced expression of endogenous antioxidant genes. NAC and vitamin E increase tumor cell proliferation by reducing ROS, DNA damage, and p53 expression in mouse and human lung tumor cells. Inactivation of p53 increases tumor growth to a similar degree as antioxidants and abolishes the antioxidant effect. Thus, antioxidants accelerate tumor growth by disrupting the ROS-p53 axis. Because somatic mutations in p53 occur late in tumor progression, antioxidants may accelerate the growth of early tumors or precancerous lesions in high-risk populations such as smokers and patients with chronic obstructive pulmonary disease who receive NAC to relieve mucus production.


Subject(s)
Antioxidants/adverse effects , Disease Progression , Lung Neoplasms/pathology , Acetylcysteine/adverse effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Mice , Reactive Oxygen Species/metabolism , Solubility , Tumor Suppressor Protein p53/metabolism , Vitamin E/adverse effects , Vitamin E/analogs & derivatives
12.
Science ; 340(6138): 1330-3, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23686339

ABSTRACT

Several progeroid disorders, including Hutchinson-Gilford progeria syndrome (HGPS) and restrictive dermopathy (ZMPSTE24 deficiency), arise when a farnesylated and methylated form of prelamin A accumulates at the nuclear envelope. Here, we found that a hypomorphic allele of isoprenylcysteine carboxyl methyltransferase (ICMT) increased body weight, normalized grip strength, and prevented bone fractures and death in Zmpste24-deficient mice. The reduced ICMT activity caused prelamin A mislocalization within the nucleus and triggered prelamin A-dependent activation of AKT-mammalian target of rapamycin (mTOR) signaling, which abolished the premature senescence of Zmpste24-deficient fibroblasts. ICMT inhibition increased AKT-mTOR signaling and proliferation and delayed senescence in human HGPS fibroblasts but did not reduce the levels of misshapen nuclei in mouse and human cells. Thus, targeting ICMT might be useful for treating prelamin A-associated progeroid disorders.


Subject(s)
Gene Knockout Techniques , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Progeria/therapy , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism , Animals , Cell Nucleus/metabolism , Cellular Senescence/genetics , Disease Models, Animal , Fibroblasts/metabolism , Hand Strength , Humans , Lamin Type A , Membrane Proteins/genetics , Metalloendopeptidases/genetics , Methylation , Mice , Mice, Mutant Strains , Nuclear Proteins/metabolism , Progeria/physiopathology , Protein Precursors/metabolism , TOR Serine-Threonine Kinases/metabolism , Weight Gain/genetics
13.
PLoS One ; 8(2): e55720, 2013.
Article in English | MEDLINE | ID: mdl-23405202

ABSTRACT

The transcription factor Zfp148 (Zbp-89, BFCOL, BERF1, htß) interacts physically with the tumor suppressor p53 and is implicated in cell cycle control, but the physiological role of Zfp148 remains unknown. Here we show that Zfp148 deficiency leads to respiratory distress and lethality in newborn mice. Zfp148 deficiency prevented structural maturation of the prenatal lung without affecting type II cell differentiation or surfactant production. BrdU analyses revealed that Zfp148 deficiency caused proliferation arrest of pulmonary cells at E18.5-19.5. Similarly, Zfp148-deficient fibroblasts exhibited proliferative arrest that was dependent on p53, raising the possibility that cell stress is part of the underlying mechanism. Indeed, Zfp148 deficiency lowered the threshold for activation of p53 under oxidative conditions. Moreover, both in vivo and cellular phenotypes were rescued on Trp53(+/-) or Trp53(-/-) backgrounds and by antioxidant treatment. Thus, Zfp148 prevents respiratory distress and lethality in newborn mice by attenuating oxidative stress-dependent p53-activity during the saccular stage of lung development. Our results establish Zfp148 as a novel player in mammalian lung maturation and demonstrate that Zfp148 is critical for cell cycle progression in vivo.


Subject(s)
Antioxidants/pharmacology , DNA-Binding Proteins/physiology , Gene Deletion , Genes, Lethal , Lung/embryology , Oxidative Stress/drug effects , Transcription Factors/physiology , Tumor Suppressor Protein p53/genetics , Animals , Animals, Newborn , Apoptosis , Blotting, Southern , Blotting, Western , Cell Cycle , Cell Proliferation , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Immunoenzyme Techniques , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Respiratory Tract Diseases/genetics , Respiratory Tract Diseases/pathology , Respiratory Tract Diseases/prevention & control , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/deficiency
14.
Mol Cancer ; 11: 50, 2012 Aug 02.
Article in English | MEDLINE | ID: mdl-22857000

ABSTRACT

BACKGROUND: Many human cancer cells express filamin A (FLNA), an actin-binding structural protein that interacts with a diverse set of cell signaling proteins, but little is known about the biological importance of FLNA in tumor development. FLNA is also expressed in endothelial cells, which may be important for tumor angiogenesis. In this study, we defined the impact of targeting Flna in cancer and endothelial cells on the development of tumors in vivo and on the proliferation of fibroblasts in vitro. METHODS: First, we used a Cre-adenovirus to simultaneously activate the expression of oncogenic K-RAS and inactivate the expression of Flna in the lung and in fibroblasts. Second, we subcutaneously injected mouse fibrosarcoma cells into mice lacking Flna in endothelial cells. RESULTS: Knockout of Flna significantly reduced K-RAS-induced lung tumor formation and the proliferation of oncogenic K-RAS-expressing fibroblasts, and attenuated the activation of the downstream signaling molecules ERK and AKT. Genetic deletion of endothelial FLNA in mice did not impact cardiovascular development; however, knockout of Flna in endothelial cells reduced subcutaneous fibrosarcoma growth and vascularity within tumors. CONCLUSIONS: We conclude that FLNA is important for lung tumor growth and that endothelial Flna impacts local tumor growth. The data shed new light on the biological importance of FLNA and suggest that targeting this protein might be useful in cancer therapeutics.


Subject(s)
Adenocarcinoma/genetics , Contractile Proteins/genetics , Endothelial Cells/metabolism , Genes, ras , Lung Neoplasms/genetics , Microfilament Proteins/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Cell Movement/genetics , Cell Proliferation , Endothelial Cells/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosarcoma/genetics , Filamins , Gene Expression Regulation, Neoplastic , Gene Order , Gene Targeting , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Tumor Burden/genetics
15.
J Clin Invest ; 121(2): 628-39, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21266780

ABSTRACT

RHO family proteins are important for the function of inflammatory cells. They are modified with a 20-carbon geranylgeranyl lipid in a process catalyzed by protein geranylgeranyltransferase type I (GGTase-I). Geranylgeranylation is viewed as essential for the membrane targeting and activity of RHO proteins. Consequently, inhibiting GGTase-I to interfere with RHO protein activity has been proposed as a strategy to treat inflammatory disorders. However, here we show that mice lacking GGTase-I in macrophages develop severe joint inflammation resembling erosive rheumatoid arthritis. The disease was initiated by the GGTase-I-deficient macrophages and was transplantable and reversible in bone marrow transplantation experiments. The cells accumulated high levels of active GTP-bound RAC1, CDC42, and RHOA, and RAC1 remained associated with the plasma membrane. Moreover, GGTase-I deficiency activated p38 and NF-κB and increased the production of proinflammatory cytokines. The results challenge the view that geranylgeranylation is essential for the activity and localization of RHO family proteins and suggest that reduced geranylgeranylation in macrophages can initiate erosive arthritis.


Subject(s)
Alkyl and Aryl Transferases/deficiency , Arthritis/immunology , Arthritis/pathology , Macrophages/immunology , Alkyl and Aryl Transferases/genetics , Animals , Cytokines/immunology , Macrophages/cytology , Macrophages/enzymology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
16.
Proc Natl Acad Sci U S A ; 107(14): 6471-6, 2010 Apr 06.
Article in English | MEDLINE | ID: mdl-20308544

ABSTRACT

RAS and RHO proteins, which contribute to tumorigenesis and metastasis, undergo posttranslational modification with an isoprenyl lipid by protein farnesyltransferase (FTase) or protein geranylgeranyltransferase-I (GGTase-I). Inhibitors of FTase and GGTase-I were developed to block RAS-induced malignancies, but their utility has been difficult to evaluate because of off-target effects, drug resistance, and toxicity. Moreover, the impact of FTase deficiency and combined FTase/GGTase-I deficiency has not been evaluated with genetic approaches. We found that inactivation of FTase eliminated farnesylation of HDJ2 and H-RAS, prevented H-RAS targeting to the plasma membrane, and blocked proliferation of primary and K-RAS(G12D)-expressing fibroblasts. FTase inactivation in mice with K-RAS-induced lung cancer reduced tumor growth and improved survival, similar to results obtained previously with inactivation of GGTase-I. Simultaneous inactivation of FTase and GGTase-I markedly reduced lung tumors and improved survival without apparent pulmonary toxicity. These data shed light on the biochemical and therapeutic importance of FTase and suggest that simultaneous inhibition of FTase and GGTase-I could be useful in cancer therapeutics.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Dimethylallyltranstransferase/metabolism , Lung Neoplasms/enzymology , Proto-Oncogene Proteins p21(ras)/metabolism , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Alleles , Animals , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Dimethylallyltranstransferase/deficiency , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Mice , Mice, Knockout , Mutation , Proto-Oncogene Proteins p21(ras)/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...