Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 309
Filter
1.
Sci Rep ; 14(1): 18283, 2024 08 07.
Article in English | MEDLINE | ID: mdl-39112499

ABSTRACT

Renal fibrosis (RF) represents the most widespread pathological condition in chronic kidney disease (CKD). Recently, protein prenylation has been implicated in the fibrosis's progression. The research examined the renoprotective effect of zoledronic acid (ZA) (50 µg/kg/week) in a rat model of carbon tetrachloride (CCl4)-induced RF through targeting protein prenylation. Forty Wistar male rats were split up into the control group, vehicle-treated group, model-RF group, and RF-ZA group. Mean arterial blood pressure (MBP), BUN, serum creatinine, and urine albumin-creatinine ratio (uACR), protein levels of farnesyl pyrophosphate (FPP), tumour necrosis factor-alpha (TNF-α), transforming growth factor-ß (TGF-ß), and malondialdehyde (MDA), and catalase and gene expression of farnesyl pyrophosphate synthase (FPPS) and nuclear factor-kB (NF-κB) were measured. Immunohistochemical staining for renal interleukin-6 (IL-6), α-smooth muscle actin (α-SMA), and caspase-3, as well as histopathological alterations, were assessed. ZA considerably ceased the reduction in MBP, markedly reduced uACR, serum creatinine, BUN, and expression of FPPS, FPP, NF-κB, TGF-ß, TNF-α, and MDA, and significantly increased catalase levels compared to the model-RF rats. ZA ameliorated the CCl4-induced histopathological alterations and suppressed the expression of caspase-3, α-SMA, and IL-6. In conclusion, ZA preserved renal function and prevented renal fibrosis in a rat model. These were achieved through targeting protein prenylation mainly by inhibiting FPPS.


Subject(s)
Fibrosis , Geranyltranstransferase , Kidney , Protein Prenylation , Rats, Wistar , Zoledronic Acid , Animals , Zoledronic Acid/pharmacology , Male , Rats , Protein Prenylation/drug effects , Geranyltranstransferase/metabolism , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Carbon Tetrachloride , Polyisoprenyl Phosphates/metabolism , Polyisoprenyl Phosphates/pharmacology , Disease Models, Animal , Transforming Growth Factor beta/metabolism , Sesquiterpenes/pharmacology , Sesquiterpenes/therapeutic use , Tumor Necrosis Factor-alpha/metabolism
2.
Proc Natl Acad Sci U S A ; 121(29): e2315310121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38990944

ABSTRACT

Bacitracin is a macrocyclic peptide antibiotic that is widely used as a topical treatment for infections caused by gram-positive bacteria. Mechanistically, bacitracin targets bacteria by specifically binding to the phospholipid undecaprenyl pyrophosphate (C55PP), which plays a key role in the bacterial lipid II cycle. Recent crystallographic studies have shown that when bound to C55PP, bacitracin adopts a highly ordered amphipathic conformation. In doing so, all hydrophobic side chains align on one face of the bacitracin-C55PP complex, presumably interacting with the bacterial cell membrane. These insights led us to undertake structure-activity investigations into the individual contribution of the nonpolar amino acids found in bacitracin. To achieve this we designed, synthesized, and evaluated a series of bacitracin analogues, a number of which were found to exhibit significantly enhanced antibacterial activity against clinically relevant, drug-resistant pathogens. As for the natural product, these next-generation bacitracins were found to form stable complexes with C55PP. The structure-activity insights thus obtained serve to inform the design of C55PP-targeting antibiotics, a key and underexploited antibacterial strategy.


Subject(s)
Anti-Bacterial Agents , Bacitracin , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Bacitracin/pharmacology , Bacitracin/chemistry , Structure-Activity Relationship , Drug Resistance, Bacterial/drug effects , Vancomycin/pharmacology , Vancomycin/chemistry , Vancomycin/analogs & derivatives , Drug Design , Polyisoprenyl Phosphates/metabolism , Polyisoprenyl Phosphates/chemistry , Polyisoprenyl Phosphates/pharmacology
3.
Mol Metab ; 85: 101964, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38823776

ABSTRACT

OBJECTIVE: Cancer cells must maintain lipid supplies for their proliferation and do so by upregulating lipogenic gene programs. The sterol regulatory element-binding proteins (SREBPs) act as modulators of lipid homeostasis by acting as transcriptional activators of genes required for fatty acid and cholesterol synthesis and uptake. SREBPs have been recognized as chemotherapeutic targets in multiple cancers, however it is not well understood which SREBP target genes are essential for tumorigenesis. In this study, we examined the requirement of SREBP target genes for pancreatic ductal adenocarcinoma (PDAC) tumor growth. METHODS: Here we constructed a custom CRISPR knockout library containing known SREBP target genes and performed in vitro 2D culture and in vivo orthotopic xenograft CRISPR screens using a patient-derived PDAC cell line. In vitro, we grew cells in medium supplemented with 10% fetal bovine serum (FBS) or 10% lipoprotein-deficient serum (LPDS) to examine differences in gene essentiality in different lipid environments. In vivo, we injected cells into the pancreata of nude mice and collected tumors after 4 weeks. RESULTS: We identified terpenoid backbone biosynthesis genes as essential for PDAC tumor development. Specifically, we identified the non-sterol isoprenoid product of the mevalonate pathway, geranylgeranyl diphosphate (GGPP), as an essential lipid for tumor growth. Mechanistically, we observed that restricting mevalonate pathway activity using statins and SREBP inhibitors synergistically induced apoptosis and caused disruptions in small G protein prenylation that have pleiotropic effects on cellular signaling pathways. Finally, we demonstrated that geranylgeranyl diphosphate synthase 1 (GGPS1) knockdown significantly reduces tumor burden in an orthotopic xenograft mouse model. CONCLUSIONS: These findings indicate that PDAC tumors selectively require GGPP over other lipids such as cholesterol and fatty acids and that this is a targetable vulnerability of pancreatic cancer cells.


Subject(s)
Cell Proliferation , Mice, Nude , Pancreatic Neoplasms , Polyisoprenyl Phosphates , Humans , Animals , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Mice , Cell Line, Tumor , Cell Proliferation/drug effects , Polyisoprenyl Phosphates/metabolism , Polyisoprenyl Phosphates/pharmacology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Sterol Regulatory Element Binding Proteins/metabolism , Sterol Regulatory Element Binding Proteins/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics
4.
Am J Physiol Endocrinol Metab ; 327(1): E55-E68, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38717364

ABSTRACT

Statins are used to treat hypercholesterolemia and function by inhibiting the production of the rate-limiting metabolite mevalonate. As such, statin treatment not only inhibits de novo synthesis of cholesterol but also isoprenoids that are involved in prenylation, the posttranslational lipid modification of proteins. The immunomodulatory effects of statins are broad and often conflicting. Previous work demonstrated that statins increased survival and inhibited myeloid cell trafficking in a murine model of sepsis, but the exact mechanisms underlying this phenomenon were unclear. Herein, we investigated the role of prenylation in chemoattractant responses. We found that simvastatin treatment abolished chemoattractant responses induced by stimulation by C5a and FMLP. The inhibitory effect of simvastatin treatment was unaffected by the addition of either farnesyl pyrophosphate (FPP) or squalene but was reversed by restoring geranylgeranyl pyrophosphate (GGPP). Treatment with prenyltransferase inhibitors showed that the chemoattractant response to both chemoattractants was dependent on geranylgeranylation. Proteomic analysis of C15AlkOPP-prenylated proteins identified several geranylgeranylated proteins involved in chemoattractant responses, including RHOA, RAC1, CDC42, and GNG2. Chemoattractant responses in THP-1 human macrophages were also geranylgeranylation dependent. These studies provide data that help clarify paradoxical findings on the immunomodulatory effects of statins. Furthermore, they establish the role of geranylgeranylation in mediating the morphological response to chemoattractant C5a.NEW & NOTEWORTHY The immunomodulatory effect of prenylation is ill-defined. We investigated the role of prenylation on the chemoattractant response to C5a. Simvastatin treatment inhibits the cytoskeletal remodeling associated with a chemotactic response. We showed that the chemoattractant response to C5a was dependent on geranylgeranylation, and proteomic analysis identified several geranylgeranylated proteins that are involved in C5a receptor signaling and cytoskeletal remodeling. Furthermore, they establish the role of geranylgeranylation in mediating the response to chemoattractant C5a.


Subject(s)
Polyisoprenyl Phosphates , Polyisoprenyl Phosphates/pharmacology , Polyisoprenyl Phosphates/metabolism , Humans , Simvastatin/pharmacology , Chemotactic Factors/pharmacology , Chemotactic Factors/metabolism , Phagocytes/drug effects , Phagocytes/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Complement C5a/metabolism , Protein Prenylation/drug effects , Animals , Mice , Sesquiterpenes
5.
J Genet Genomics ; 48(4): 300-311, 2021 04 20.
Article in English | MEDLINE | ID: mdl-34049800

ABSTRACT

Vascular development is essential for the establishment of the circulatory system during embryonic development and requires the proliferation of endothelial cells. However, the underpinning regulatory mechanisms are not well understood. Here, we report that geranylgeranyl pyrophosphate (GGPP), a metabolite involved in protein geranylgeranylation, plays an indispensable role in embryonic vascular development. GGPP is synthesized by geranylgeranyl pyrophosphate synthase (GGPPS) in the mevalonate pathway. The selective knockout of Ggpps in endothelial cells led to aberrant vascular development and embryonic lethality, resulting from the decreased proliferation and enhanced apoptosis of endothelial cells during vasculogenesis. The defect in protein geranylgeranylation induced by GGPP depletion inhibited the membrane localization of RhoA and enhanced yes-associated protein (YAP) phosphorylation, thereby prohibiting the entry of YAP into the nucleus and the expression of YAP target genes related to cell proliferation and the antiapoptosis process. Moreover, inhibition of the mevalonate pathway by simvastatin induced endothelial cell proliferation defects and apoptosis, which were ameliorated by GGPP. Geranylgeraniol (GGOH), a precursor of GGPP, ameliorated the harmful effects of simvastatin on vascular development of developing fetuses in pregnant mice. These results indicate that GGPP-mediated protein geranylgeranylation is essential for endothelial cell proliferation and the antiapoptosis process during embryonic vascular development.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Embryonic Development/genetics , Farnesyltranstransferase/genetics , Multienzyme Complexes/genetics , Animals , Cell Differentiation/drug effects , Embryo, Mammalian , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Mice , Morphogenesis/genetics , Polyisoprenyl Phosphates/metabolism , Polyisoprenyl Phosphates/pharmacology , Pregnancy , Protein Prenylation/drug effects , Protein Prenylation/genetics , YAP-Signaling Proteins/genetics , rhoA GTP-Binding Protein/genetics
6.
PLoS Biol ; 19(4): e3001134, 2021 04.
Article in English | MEDLINE | ID: mdl-33901180

ABSTRACT

Cell death is a vital event in life. Infections and injuries cause lytic cell death, which gives rise to danger signals that can further induce cell death, inflammation, and tissue damage. The mevalonate (MVA) pathway is an essential, highly conserved and dynamic metabolic pathway. Here, we discover that farnesyl pyrophosphate (FPP), a metabolic intermediate of the MVA pathway, functions as a newly identified danger signal to trigger acute cell death leading to neuron loss in stroke. Harboring both a hydrophobic 15-carbon isoprenyl chain and a heavily charged pyrophosphate head, FPP leads to acute cell death independent of its downstream metabolic pathways. Mechanistically, extracellular calcium influx and the cation channel transient receptor potential melastatin 2 (TRPM2) exhibit essential roles in FPP-induced cell death. FPP activates TRPM2 opening for ion influx. Furthermore, in terms of a mouse model constructing by middle cerebral artery occlusion (MCAO), FPP accumulates in the brain, which indicates the function of the FPP and TRPM2 danger signal axis in ischemic injury. Overall, our data have revealed a novel function of the MVA pathway intermediate metabolite FPP as a danger signal via transient receptor potential cation channels.


Subject(s)
Cell Death/drug effects , Polyisoprenyl Phosphates/pharmacology , Sesquiterpenes/pharmacology , Animals , Barium/pharmacology , Calcium/pharmacology , Cell Death/genetics , Cells, Cultured , Embryo, Mammalian , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Polyisoprenyl Phosphates/metabolism , Rats , Rats, Sprague-Dawley , Sesquiterpenes/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Strontium/pharmacology
7.
Arch Immunol Ther Exp (Warsz) ; 69(1): 10, 2021 Apr 03.
Article in English | MEDLINE | ID: mdl-33811524

ABSTRACT

The review discusses a new approach to the prevention and treatment of viral infections based on the use of pine needles polyprenyl phosphate (PPP) and associated with the infringement of prenylation process-the attachment of farnesol or geranyl geraniol to the viral protein. Currently, prenylation has been detected in type 1 adenovirus, hepatitis C virus, several herpes viruses, influenza virus, HIV. However, this list is far from complete, given that prenylated proteins play an extremely important role in the activity of the virus. We assume that the interferon produced in response to PPP may suppress expression of the SREBP2 transcription factor. As a result, the mevalonic acid pathway is violated and, as a result, the formation of early polyprenols precursors (geraniol, geranyl geraniol, farnesol), which are necessary for the prenylation of viral proteins, is blocked and the formation of mature, virulent virus particles is broken. As a consequence, the maturation of viral particles is inhibited, and defective particles are formed. Polyprenol was extracted from greenery (pine, fir and spruce needles, mulberry leaves, etc.), purified by chromatography, phosphorylated and identified by HPLC and NMR. Obtained PPP was used as antiviral in some experimental models in vitro and in vivo. During numerous studies, it was found that PPP manifested versatile antiviral effects, both in vitro and in vivo. The maximum effect was observed with viruses in which the presence of prenylated proteins was established, namely influenza A virus, HIV-1, tick-borne encephalitis virus, hepatitis A and C viruses, herpes simplex viruses type 1 and 2, some coronavirus. The available data obtained both in the experimental conditions and during clinical trials allow us to regard PPPs as safe and effective medicine for prevention and treatment of viral diseases.


Subject(s)
Antiviral Agents/pharmacology , Pinus/chemistry , Polyisoprenyl Phosphates/pharmacology , Protein Prenylation/drug effects , Virus Diseases/drug therapy , Animals , Antiviral Agents/therapeutic use , Clinical Trials as Topic , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Interferons/metabolism , Microscopy, Electron , Polyisoprenyl Phosphates/therapeutic use , Sterol Regulatory Element Binding Protein 2/metabolism , Treatment Outcome , Viral Proteins/metabolism , Virion/drug effects , Virion/ultrastructure , Virus Diseases/immunology , Virus Diseases/prevention & control , Virus Replication/drug effects , Virus Replication/immunology
8.
Cell Biol Toxicol ; 37(3): 441-460, 2021 06.
Article in English | MEDLINE | ID: mdl-33034787

ABSTRACT

Myopathy is the major adverse effect of statins. However, the underlying mechanism of statin-induced skeletal muscle atrophy, one of statin-induced myopathy, remains to be elucidated. Myostatin is a negative regulator of skeletal muscle mass and functions. Whether myostatin is involved in statin-induced skeletal muscle atrophy remains unknown. In this study, we uncovered that simvastatin administration increased serum myostatin levels in mice. Inhibition of myostatin with follistatin, an antagonist of myostatin, improved simvastatin-induced skeletal muscle atrophy. Simvastatin induced myostatin expression not only in skeletal muscle but also in brown adipose tissue (BAT). Mechanistically, simvastatin inhibited the phosphorylation of forkhead box protein O1 (FOXO1) in C2C12 myotubes, promoting the nuclear translocation of FOXO1 and thereby stimulating the transcription of myostatin. In differentiated brown adipocytes, simvastatin promoted myostatin expression mainly by inhibiting the expression of interferon regulatory factor 4 (IRF4). Moreover, the stimulative effect of simvastatin on myostatin expression was blunted by geranylgeranyl diphosphate (GGPP) supplementation in both myotubes and brown adipocytes, suggesting that GGPP depletion was attributed to simvastatin-induced myostatin expression. Besides, the capacities of statins on stimulating myostatin expression were positively correlated with the lipophilicity of statins. Our findings provide new insights into statin-induced skeletal muscle atrophy. Graphical headlights 1. Simvastatin induces skeletal muscle atrophy via increasing serum myostatin levels in mice; 2. Simvastatin promotes myostatin expression in both skeletal muscle and brown adipose tissue through inhibiting GGPP production; 3. The stimulating effect of statins on myostatin expression is positively correlated with the lipophilicity of statins.


Subject(s)
Forkhead Box Protein O1/genetics , Interferon Regulatory Factors/genetics , Muscular Atrophy/genetics , Myostatin/blood , Simvastatin/adverse effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Animals , Gene Expression Regulation/drug effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/chemically induced , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Diseases/chemically induced , Muscular Diseases/genetics , Muscular Diseases/pathology , Myostatin/genetics , Polyisoprenyl Phosphates/pharmacology , Simvastatin/pharmacology
9.
Leukemia ; 35(3): 796-808, 2021 03.
Article in English | MEDLINE | ID: mdl-32665698

ABSTRACT

Multiple myeloma (MM) is a plasma cell malignancy that is often driven by chromosomal translocations. In particular, patients with t(4;14)-positive disease have worse prognosis compared to other MM subtypes. Herein, we demonstrated that t(4;14)-positive cells are highly dependent on the mevalonate (MVA) pathway for survival. Moreover, we showed that this metabolic vulnerability is immediately actionable, as inhibiting the MVA pathway with a statin preferentially induced apoptosis in t(4;14)-positive cells. In response to statin treatment, t(4;14)-positive cells activated the integrated stress response (ISR), which was augmented by co-treatment with bortezomib, a proteasome inhibitor. We identified that t(4;14)-positive cells depend on the MVA pathway for the synthesis of geranylgeranyl pyrophosphate (GGPP), as exogenous GGPP fully rescued statin-induced ISR activation and apoptosis. Inhibiting protein geranylgeranylation similarly induced the ISR in t(4;14)-positive cells, suggesting that this subtype of MM depends on GGPP, at least in part, for protein geranylgeranylation. Notably, fluvastatin treatment synergized with bortezomib to induce apoptosis in t(4;14)-positive cells and potentiated the anti-tumor activity of bortezomib in vivo. Our data implicate the t(4;14) translocation as a biomarker of statin sensitivity and warrant further clinical evaluation of a statin in combination with bortezomib for the treatment of t(4;14)-positive disease.


Subject(s)
Bortezomib/pharmacology , Fluvastatin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mevalonic Acid/metabolism , Multiple Myeloma/drug therapy , Polyisoprenyl Phosphates/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 4 , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Translocation, Genetic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Bull Exp Biol Med ; 170(2): 219-222, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33269450

ABSTRACT

The immunomodulatory properties of immunobiological drugs Glutoxim and Phosprenyl we well as vesicular stomatitis virus and inactivated tick-borne encephalitis vaccine virus were studied using human diploid fibroblast cell line from the collection of M. P. Chumakov Federal Research Center for Research and Development of Immunobiological Products. All tested preparations exhibited immunomodulatory activity in human diploid fibroblast cell line. Glutoxim in doses of 0.1 and 0.25 µg/ml stimulated production of IL-6 and IL-10 during 24-48 h of culturing, but did not stimulate production of IL-1ß. Phosprenyl, on the contrary, increased production of IL-1ß and the levels of IL-6 and IL-10. Vesicular stomatitis virus stimulated the production of IL-1ß, IL-6, and IL-10, while inactivated tick-borne encephalitis vaccine virus stimulated the production of cytokines IL-8 and IL-18. Immunomodulatory activity of inactivated tick-borne encephalitis vaccine virus was first demonstrated in the in vitro system.


Subject(s)
Drug Evaluation, Preclinical/methods , Fibroblasts/metabolism , Animals , Cell Line , Diploidy , Encephalitis Viruses, Tick-Borne/metabolism , Fibroblasts/virology , Humans , Immunologic Factors/pharmacology , Inflammation/drug therapy , Interleukin-10/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Muscles/metabolism , Polyisoprenyl Phosphates/pharmacology , Skin/metabolism , Ticks , Time Factors , Vesicular stomatitis Indiana virus
11.
BMC Cancer ; 20(1): 703, 2020 Jul 29.
Article in English | MEDLINE | ID: mdl-32727400

ABSTRACT

BACKGROUND: Ovarian cancer remains the most fatal gynecological malignancy. Current therapeutic options are limited due to late diagnosis in the majority of the cases, metastatic spread to the peritoneal cavity and the onset of chemo-resistance. Thus, novel therapeutic approaches are required. Statins and amino-bisphosphonates are inhibitors of the mevalonate pathway, which is a fundamental pathway of cellular metabolism, essential for cholesterol production and posttranslational protein farnesylation and geranylgeranylation. While this pathway has emerged as a promising treatment target in several human malignancies, its potential as a therapeutic approach in ovarian cancer is still not fully understood. METHODS: Human ovarian cancer cell lines (IGROV-1, A2780, A2780cis) were treated with increasing concentrations (0.5-100 µM) of statins (simvastatin, atorvastatin, rosuvastatin) and zoledronic acid. Effects on cell vitality and apoptosis were assessed using Cell Titer Blue®, Caspase 3/7 Glo®, clonogenic assays as well as cleaved poly (ADP-ribose) polymerase (cPARP) detection. The inhibition of the mevalonate pathway was confirmed using Western Blot of unprenylated Ras and Rap1a proteins. Quantitative real-time PCR and ELISA were used to analyze modulations on several key regulators of ovarian cancer tumorigenesis. RESULTS: The treatment of IGROV-1 and A2780 cells with statins and zoledronic acid reduced vitality (by up to 80%; p < 0.001) and induced apoptosis by up to 8-folds (p < 0.001) in a dose-dependent fashion. Rescue experiments using farnesyl pyrophosphate or geranylgeranyl pyrophosphate evidenced that blocked geranylgeranylation is the major underlying mechanism of the pro-apoptotic effects. Gene expression of the tumor-promoting cytokines and mediators, such as transforming growth factor (TGF)-ß1, vascular endothelial growth factor (VEGF), interleukin (IL)-8, and IL-6 were significantly suppressed by statins and zoledronic acid by up to 90% (p < 0.001). For all readouts, simvastatin was most potent of all agents used. Cisplatin-resistant A2780cis cells showed a relative resistance to statins and zoledronic acid. However, similar to the effects in A2780 cells, simvastatin and zoledronic acid significantly induced caspase 3/7 activation (6-folds; p < 0.001). CONCLUSION: Our in vitro findings point to promising anti-tumor effects of statins and zoledronic acid in ovarian cancer and warrant additional validation in preclinical and clinical settings.


Subject(s)
Cell Survival/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mevalonic Acid/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Apoptosis/drug effects , Atorvastatin/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Gene Expression/drug effects , Humans , Interleukin-6/genetics , Interleukin-8/drug effects , Interleukin-8/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Polyisoprenyl Phosphates/pharmacology , Prenylation/drug effects , Rosuvastatin Calcium/pharmacology , Sesquiterpenes/pharmacology , Simvastatin/pharmacology , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/genetics , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/genetics , Zoledronic Acid/pharmacology
12.
FASEB J ; 34(3): 4684-4701, 2020 03.
Article in English | MEDLINE | ID: mdl-32030813

ABSTRACT

Statins, the most prescribed class of drugs for the treatment of hypercholesterolemia, can cause muscle-related adverse effects. It has been shown that the glucocorticoid-induced leucine zipper (GILZ) plays a key role in the anti-myogenic action of dexamethasone. In the present study, we aimed to evaluate the role of GILZ in statin-induced myopathy. Statins induced GILZ expression in C2C12 cells, primary murine myoblasts/myotubes, primary human myoblasts, and in vivo in zebrafish embryos and human quadriceps femoris muscle. Gilz induction was mediated by FOXO3 activation and binding to the Gilz promoter, and could be reversed by the addition of geranylgeranyl, but not farnesyl, pyrophosphate. Atorvastatin decreased Akt phosphorylation and increased cleaved caspase-3 levels in myoblasts. This effect was reversed in myoblasts from GILZ knockout mice. Similarly, myofibers isolated from knockout animals were more resistant toward statin-induced cell death than their wild-type counterparts. Statins also impaired myoblast differentiation, and this effect was accompanied by GILZ induction. The in vivo relevance of our findings was supported by the observation that gilz overexpression in zebrafish embryos led to impaired embryonic muscle development. Taken together, our data point toward GILZ as an essential mediator of the molecular mechanisms leading to statin-induced muscle damage.


Subject(s)
Glucocorticoids/pharmacology , Leucine Zippers/physiology , Muscles/metabolism , Muscles/pathology , Animals , Blotting, Western , Cell Line , Cells, Cultured , Chromatin Immunoprecipitation , Fluorescent Antibody Technique , Humans , In Situ Hybridization , Lentivirus/genetics , Mice , Mice, Inbred C57BL , Muscles/drug effects , Polyisoprenyl Phosphates/pharmacology , Zebrafish
13.
Diabetes ; 69(1): 48-59, 2020 01.
Article in English | MEDLINE | ID: mdl-31649162

ABSTRACT

Statins are cholesterol-lowering agents that increase the incidence of diabetes and impair glucose tolerance via their detrimental effects on nonhepatic tissues, such as pancreatic islets, but the underlying mechanism has not been determined. In atorvastatin (ator)-treated high-fat diet-fed mice, we found reduced pancreatic ß-cell size and ß-cell mass, fewer mature insulin granules, and reduced insulin secretion and glucose tolerance. Transcriptome profiling of primary pancreatic islets showed that ator inhibited the expression of pancreatic transcription factor, mechanistic target of rapamycin (mTOR) signaling, and small G protein (sGP) genes. Supplementation of the mevalonate pathway intermediate geranylgeranyl pyrophosphate (GGPP), which is produced by 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase, significantly restored the attenuated mTOR activity, v-maf musculoaponeurotic fibrosarcoma oncogene homolog A (MafA) expression, and ß-cell function after ator, lovastatin, rosuvastatin, and fluvastatin treatment; this effect was potentially mediated by sGP prenylation. Rab5a, the sGP in pancreatic islets most affected by ator treatment, was found to positively regulate mTOR signaling and ß-cell function. Rab5a knockdown mimicked the effect of ator treatment on ß-cells. Thus, ator impairs ß-cell function by regulating sGPs, for example, Rab5a, which subsequently attenuates islet mTOR signaling and reduces functional ß-cell mass. GGPP supplementation could constitute a new approach for preventing statin-induced hyperglycemia.


Subject(s)
Atorvastatin/pharmacology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mevalonic Acid/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Count , Cells, Cultured , Female , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/physiology , Islets of Langerhans/growth & development , Male , Metabolic Networks and Pathways/genetics , Mice , Mice, Inbred C57BL , Organ Size/genetics , Polyisoprenyl Phosphates/pharmacology , Signal Transduction/genetics
14.
Pflugers Arch ; 471(10): 1273-1289, 2019 10.
Article in English | MEDLINE | ID: mdl-31612282

ABSTRACT

TRPV3, a member of the thermosensitive Ca2+-permeable TRPV channel subfamily expressed in skin and sensory nerves, is also activated by chemical agonists such as 2-aminoethyl diphenylborinate (2-APB). Repetitive stimuli induce sensitization of TRPV3 activation, characterized by the cumulative increase in current amplitude and linearization of current-voltage relation (I/V curve). Through genomic analysis of various populations, we found non-rare TRPV3 mutation (p.A628T) in East Asian people with an allele frequency of 0.249 while 0.007 in Caucasian. Slope conductance of unitary channel was not different between WT and p.A628T. Whole-cell patch clamp study of wildtype TRPV3 (WT) and p.A628T overexpressed in HEK293T cells showed similar sensitization by the repetitive increase in temperature from 23 to 37 °C, while slightly higher sensitization to 43 °C in p.A628T. In contrast, the repetitive application of 2-APB (10 µM) or carvacrol (100 µM) induced faster sensitization in p.A628T than WT. However, 1 µM farnesyl pyrophosphate, an intrinsic lipid metabolite agonist, induced similar level of slow activations in WT and p.A628T. In Fura-2 microspectrofluorimetry, the 2-APB pulses induced a faster increase of [Ca2+]c in p.A628T than WT. In terms of ionic selectivity of channels, WT and p.A628T showed similar Ca2+ permeability (PCa/PNa) calculated from the reversal potential of I/V curves. Taken together, p.A628T shows faster sensitization to chemical agonists that are reflected as higher [Ca2+]c signaling. Based on the intriguing pharmacological sensitivity, the physiological implications of p.A628T in the East Asian population require further investigation.


Subject(s)
Mutation, Missense , Polymorphism, Single Nucleotide , TRPV Cation Channels/genetics , Asian People/genetics , Boron Compounds/pharmacology , Calcium Signaling , Cymenes/pharmacology , HEK293 Cells , Humans , Ion Channel Gating , Polyisoprenyl Phosphates/pharmacology , Sesquiterpenes/pharmacology , TRPV Cation Channels/agonists
15.
Sci Rep ; 9(1): 10009, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31292513

ABSTRACT

Statins have been reported to increase the plasma concentration of arachidonic acid (AA), an omega-6 long chain polyunsaturated fatty acid (LCPUFA) in several clinical studies indicating that statins affect the endogenous synthesis of LCUFAs. In the present study, we investigated the roles of the intrinsic mevalonate cascade and Rho-dependent pathway in LCPUFA synthesis, especially focusing on fatty acid desaturases (Fads) 2, using the human hepatocellular carcinoma cell line HepG2. Cell number and the activity of caspase-3 and 7 (caspase-3/7) was measured using a commercial kit. Gene expression was analyzed by quantitative real-time PCR. Protein expression was detected by Western blot analysis. Atorvastatin decreased cell viability and increased caspase-3/7 activity in a dose-dependent manner. At lower concentrations, atorvastatin stimulated both mRNA and protein expression of Fads2, and increased mRNA expression of FADS1 and ELVOL5. Both mevalonate and geranylgeranyl-pyrophosphate (GGPP), but not cholesterol, fully reversed atorvastatin-induced upregulation of Fads2, and mevalonate-effected reversal was inhibited by treatment with the Rho-associated protein kinase inhibitor Y-27632. These data clearly demonstrated that in human HepG2 cells, statins affect the endogenous synthesis of LCPUFAs by regulation of not only Fads2, but also Fads1 and Elovl5, through the GGPP-dependent Rho kinase pathway.


Subject(s)
Atorvastatin/pharmacology , Fatty Acid Desaturases/genetics , Fatty Acid Desaturases/metabolism , Polyisoprenyl Phosphates/pharmacology , Signal Transduction/drug effects , Amides/pharmacology , Cell Survival/drug effects , Delta-5 Fatty Acid Desaturase , Dose-Response Relationship, Drug , Eicosanoic Acids/metabolism , Fatty Acid Elongases/genetics , Gene Expression Regulation, Enzymologic/drug effects , Hep G2 Cells , Humans , Mevalonic Acid/pharmacology , Pyridines/pharmacology , Up-Regulation , rho-Associated Kinases/metabolism
16.
Cell Death Dis ; 9(12): 1170, 2018 12 05.
Article in English | MEDLINE | ID: mdl-30518913

ABSTRACT

UbiA prenyltransferase domain-containing protein 1 (UBIAD1) plays a key role in biosynthesis of vitamin K2 and coenzyme Q10 using geranylgeranyl diphosphate (GGPP). However, the mechanism by which UBIAD1 participates in tumorigenesis remains unknown. This study show that UBIAD1 interacts with H-Ras, retains H-Ras in the Golgi apparatus, prevents H-Ras trafficking from the Golgi apparatus to the plasma membrane, blocks the aberrant activation of Ras/MAPK signaling, and inhibits the proliferation of bladder cancer cells. In addition, GGPP was required to maintain the function of UBIAD1 in regulating the Ras/ERK signaling pathway. A Drosophila model was employed to confirm the function of UBIAD1/HEIX in vivo. The activation of Ras/ERK signaling at the plasma membrane induced melanotic masses in Drosophila larvae. Our study suggests that UBIAD1 serves as a tumor suppressor in cancer and tentatively reveals the underlying mechanism of melanotic mass formation in Drosophila.


Subject(s)
Dimethylallyltranstransferase/genetics , Drosophila Proteins/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Golgi Apparatus/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Proliferation/drug effects , Dimethylallyltranstransferase/chemistry , Dimethylallyltranstransferase/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Epithelial Cells/drug effects , Epithelial Cells/pathology , Genes, Reporter , Golgi Apparatus/drug effects , Golgi Apparatus/ultrastructure , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Larva/cytology , Larva/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Plasmids/chemistry , Plasmids/metabolism , Polyisoprenyl Phosphates/pharmacology , Protein Binding , Protein Domains , Protein Transport/drug effects , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Transfection , Red Fluorescent Protein
17.
Dokl Biochem Biophys ; 482(1): 261-263, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30397888

ABSTRACT

The search for new adjuvants remains the critical task for the creation of hepatitis C vaccines due to the weak immunogenicity of biotechnological products. When immunizing mice with the recombinant proteins NS3 and NS5B of the hepatitis C virus (HCV), the adjuvant activity of three immunomodulators was compared. Phosprenyl® on the basis of polyprenyl phosphate (PPP), chemically synthesized analogue of the bacterial cell wall glucosaminyl muramyl dipeptide (GMDP), and IFN-α recombinant protein were tested. GMDP increased the activity of IgG1 antibodies 4-6 times but did not stimulate the production of IFN-γ; IFN-α has not shown any adjuvant properties. The introduction of recombinant HCV proteins together with PPP in low doses increased the activity of IgG2a isotype antibodies 4-7 times and increased IFN-γ secretion 3 times. Thus, it was first shown that PPP polarizes the immune response to Th1-type and is a promising adjuvant for the development of a vaccine against hepatitis C.


Subject(s)
Adjuvants, Immunologic , Hepacivirus/drug effects , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Polyisoprenyl Phosphates/pharmacology , Vaccines/therapeutic use , Animals , Immunoglobulin G/metabolism , Immunologic Factors/classification , Immunologic Factors/pharmacology , Mice , Recombinant Proteins , Virus Replication
18.
Oncol Rep ; 40(4): 2171-2182, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30106444

ABSTRACT

Mutant KRAS and BRAF are associated with primary EGFR inhibitor resistance in colorectal cancer (CRC). However, other biomarkers that could predict EGFR inhibitor resistance remain elusive. In the present study, immunoblotting and cell proliferation results revealed that yes­associated protein (YAP), a downstream effector of the Hippo pathway, was positively associated with primary cetuximab resistance in CRC cells. YAP knockdown enhanced the cytotoxicity of cetuximab in CRC cells. Simvastatin, a 3­hydroxy­3­methylglutaryl­coenzyme A (HMG­CoA) reductase inhibitor of the mevalonate pathway that inhibits YAP bioactivity through nuclear translocation and total YAP expression, increased the cytotoxicity of EGFR inhibitors (cetuximab and gefitinib) against CRC cells. The combination of simvastatin and EGFR inhibitors inhibited YAP and EGFR signaling more markedly than each agent alone. Adding back geranylgeranyl pyrophosphate (GGPP), a key product of the mevalonate pathway, reversed the YAP bioactivity inhibition induced by simvastatin and the cell proliferation inhibition induced by the combination of simvastatin and EGFR inhibitors. Collectively, these results revealed that YAP may be useful in identifying cetuximab resistance in CRC and indicated that targeting of both YAP and EGFR signals may present a promising therapeutic approach for CRC.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Phosphoproteins/antagonists & inhibitors , Polyisoprenyl Phosphates/pharmacology , Protein Kinase Inhibitors/pharmacology , Simvastatin/pharmacology , Animals , Anticholesteremic Agents/pharmacology , Apoptosis , Cell Proliferation , Cetuximab/pharmacology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Combinations , ErbB Receptors/metabolism , Female , Gefitinib , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Quinazolines/pharmacology , Signal Transduction , Transcription Factors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , YAP-Signaling Proteins
19.
Biomed Pharmacother ; 105: 274-281, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29860219

ABSTRACT

BACKGROUND: Curcumin is a natural polyphenol with beneficial effects on NAFLD patients and NAFLD is accompanied by metabolism decompensation. METHODS: This study was focused on the effect of curcumin on the relationship between endogenous bile acids metabolism pathway and exogenous xenobiotics metabolism pathway in C57BL/6 mice of non-alcoholic fatty liver disease induced by high-fat and high-fructose diet (HFHFr) and in cultured mice hepatocytes. RESULTS: Our results showed curcumin treatment apparently attenuated the hepatic steatosis and reversed the abnormalities of serum biochemical parameters in HFHFr-fed mice. Curcumin effectively reversed the expression of CYP3A and CYP7A in fatty liver status to restore metabolism capability. In the meantime, lipid synthesis has been controlled by curcumin, evidenced by the expression of CD36, SREBP-1c and FAS. Further, FXR, SHP and Nrf2 expressions were remarkably dropped in HFHFr-fed mice and LXRα expression was significantly enhanced, while curcumin treatment was quite effective to restore this pathway. In addition, LXRα antagonist GGPP pretreatment weakened the curcumin effects on CYP3A, CYP7A and SREBP-1c. CONCLUSIONS: These findings indicate that the Nrf2/FXR/LXRα pathway might synergistically regulate both endogenous and exogenous metabolism in NAFLD mice and LXRα may be a novel therapeutic target of curcumin for the prevention and treatment of NAFLD.


Subject(s)
Curcumin/pharmacology , Liver X Receptors/metabolism , NF-E2-Related Factor 2/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Animals , Cytochrome P-450 Enzyme System/metabolism , Diet, High-Fat , Fructose , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Lipids/biosynthesis , Liver/drug effects , Liver/enzymology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/pathology , Polyisoprenyl Phosphates/pharmacology , Protective Agents/pharmacology , Protective Agents/therapeutic use , Sterol Regulatory Element Binding Protein 1/metabolism
20.
PLoS One ; 13(5): e0196387, 2018.
Article in English | MEDLINE | ID: mdl-29738536

ABSTRACT

Statins inhibit HMG-CoA reductase, the rate-limiting enzyme in the cholesterol biosynthesis pathway (CBP), and are used for the prevention of cardiovascular disease. The anti-inflammatory effects of statins may also provide therapeutic benefits and have led to their use in clinical trials for preeclampsia, a pregnancy-associated inflammatory condition, despite their current classification as category X (i.e. contraindicated during pregnancy). In the developing neocortex, products of the CBP play essential roles in proliferation and differentiation of neural stem-progenitor cells (NSPCs). To understand how statins could impact the developing brain, we studied effects of pravastatin and simvastatin on primary embryonic NSPC survival, proliferation, global transcription, and cell fate in vitro. We found that statins dose dependently decrease NSPC expansion by promoting cell death and autophagy of NSPCs progressing through the G1 phase of the cell cycle. Genome-wide transcriptome analysis demonstrates an increase in expression of CBP genes following pravastatin treatment, through activation of the SREBP2 transcription factor. Co-treatment with farnesyl pyrophosphate (FPP), a CBP metabolite downstream of HMG-CoA reductase, reduces SREBP2 activation and pravastatin-induced PARP cleavage. Finally, pravastatin and simvastatin differentially alter NSPC cell fate and mRNA expression during differentiation, through a non-CBP dependent pathway.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Animals , Autophagy/drug effects , Biosynthetic Pathways/drug effects , Cell Cycle/drug effects , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cholesterol/biosynthesis , Female , Male , Mice , Mouse Embryonic Stem Cells/metabolism , Neural Stem Cells/metabolism , Polyisoprenyl Phosphates/pharmacology , Pravastatin/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sesquiterpenes/pharmacology , Simvastatin/pharmacology , Sterol Regulatory Element Binding Protein 2/genetics , Transcriptome/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL