12
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Combined HMG-COA reductase and prenylation inhibition in treatment of CCM

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Significance

          Cerebral cavernous malformations (CCMs) are common vascular anomalies of the central nervous system that can lead to seizures, focal neurological deficits, and brain hemorrhage. Clinical options are limited mainly to treatment of symptoms or surgical resection, and targeted pharmacological therapy is lacking. Here we undertake a high-throughput screen and identify fluvastatin and zoledronate, two drugs already approved for clinical use for other indications, which act synergistically to reverse outcomes of CCM3 loss. Used in combination, fluvastatin and zoledronate effectively attenuate neural and vascular deficits in mouse models of CCM in vivo, significantly reducing formation of lesions and extending longevity. Our studies suggest that combined therapy targeting the mevalonate pathway might have therapeutic effects in CCM disease.

          Abstract

          Cerebral cavernous malformations (CCMs) are common vascular anomalies that develop in the central nervous system and, more rarely, the retina. The lesions can cause headache, seizures, focal neurological deficits, and hemorrhagic stroke. Symptomatic lesions are treated according to their presentation; however, targeted pharmacological therapies that improve the outcome of CCM disease are currently lacking. We performed a high-throughput screen to identify Food and Drug Administration-approved drugs or other bioactive compounds that could effectively suppress hyperproliferation of mouse brain primary astrocytes deficient for CCM3. We demonstrate that fluvastatin, an inhibitor of 3-hydroxy-3-methyl-glutaryl (HMG)-CoA reductase and the N-bisphosphonate zoledronic acid monohydrate, an inhibitor of protein prenylation, act synergistically to reverse outcomes of CCM3 loss in cultured mouse primary astrocytes and in Drosophila glial cells in vivo. Further, the two drugs effectively attenuate neural and vascular deficits in chronic and acute mouse models of CCM3 loss in vivo, significantly reducing lesion burden and extending longevity. Sustained inhibition of the mevalonate pathway represents a potential pharmacological treatment option and suggests advantages of combination therapy for CCM disease.

          Related collections

          Most cited references28

          • Record: found
          • Abstract: found
          • Article: not found

          Collective migration of an epithelial monolayer in response to a model wound.

          Using an original microfabrication-based technique, we experimentally study situations in which a virgin surface is presented to a confluent epithelium with no damage made to the cells. Although inspired by wound-healing experiments, the situation is markedly different from classical scratch wounding because it focuses on the influence of the free surface and uncouples it from the other possible contributions such as cell damage and/or permeabilization. Dealing with Madin-Darby canine kidney cells on various surfaces, we found that a sudden release of the available surface is sufficient to trigger collective motility. This migration is independent of the proliferation of the cells that mainly takes place on the fraction of the surface initially covered. We find that this motility is characterized by a duality between collective and individual behaviors. On the one hand, the velocity fields within the monolayer are very long range and involve many cells in a coordinated way. On the other hand, we have identified very active "leader cells" that precede a small cohort and destabilize the border by a fingering instability. The sides of the fingers reveal a pluricellular actin "belt" that may be at the origin of a mechanical signaling between the leader and the followers. Experiments performed with autocrine cells constitutively expressing hepatocyte growth factor (HGF) or in the presence of exogenous HGF show a higher average velocity of the border and no leader.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Protein prenylation: molecular mechanisms and functional consequences.

            Prenylation is a class of lipid modification involving covalent addition of either farnesyl (15-carbon) or geranylgeranyl (20-carbon) isoprenoids to conserved cysteine residues at or near the C-terminus of proteins. Known prenylated proteins include fungal mating factors, nuclear lamins, Ras and Ras-related GTP-binding proteins (G proteins), the subunits of trimeric G proteins, protein kinases, and at least one viral protein. Prenylation promotes membrane interactions of most of these proteins, which is not surprising given the hydrophobicity of the lipids involved. In addition, however, prenylation appears to play a major role in several protein-protein interactions involving these species. The emphasis in this review is on the enzymology of prenyl protein processing and the functional significance of prenylation in cellular events. Several other recent reviews provide more detailed coverage of aspects of prenylation that receive limited attention here owing to length restrictions (1-4).
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Combined treatment with statins and aminobisphosphonates extends longevity in a mouse model of human premature aging.

              Several human progerias, including Hutchinson-Gilford progeria syndrome (HGPS), are caused by the accumulation at the nuclear envelope of farnesylated forms of truncated prelamin A, a protein that is also altered during normal aging. Previous studies in cells from individuals with HGPS have shown that farnesyltransferase inhibitors (FTIs) improve nuclear abnormalities associated with prelamin A accumulation, suggesting that these compounds could represent a therapeutic approach for this devastating progeroid syndrome. We show herein that both prelamin A and its truncated form progerin/LADelta50 undergo alternative prenylation by geranylgeranyltransferase in the setting of farnesyltransferase inhibition, which could explain the low efficiency of FTIs in ameliorating the phenotypes of progeroid mouse models. We also show that a combination of statins and aminobisphosphonates efficiently inhibits both farnesylation and geranylgeranylation of progerin and prelamin A and markedly improves the aging-like phenotypes of mice deficient in the metalloproteinase Zmpste24, including growth retardation, loss of weight, lipodystrophy, hair loss and bone defects. Likewise, the longevity of these mice is substantially extended. These findings open a new therapeutic approach for human progeroid syndromes associated with nuclear-envelope abnormalities.
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc. Natl. Acad. Sci. U.S.A
                pnas
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                23 May 2017
                12 May 2017
                12 May 2017
                : 114
                : 21
                : 5503-5508
                Affiliations
                [1] aDepartment of Neurosurgery, Yale School of Medicine , New Haven, CT 06520;
                [2] bDepartment of Biomedical Engineering, Yale University , New Haven, CT 06520;
                [3] cYale Systems Biology Institute, Yale University , West Haven, CT 06516;
                [4] dYale Center for Molecular Discovery, Yale University , West Haven, CT 06516;
                [5] eDepartment of Drug Discovery, Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center , Tampa, FL 33612;
                [6] fDepartment of Oncologic Sciences, Morsani College of Medicine, University of South Florida , Tampa, FL 33612;
                [7] gDepartment of Neuroscience, Program on Neurogenetics, Yale School of Medicine , New Haven, CT 06520;
                [8] hDepartment of Genetics, Yale School of Medicine , New Haven, CT 06520
                Author notes
                2To whom correspondence may be addressed. Email: angeliki.louvi@ 123456yale.edu or murat.gunel@ 123456yale.edu .

                Edited by Jeremy Nathans, Johns Hopkins University, Baltimore, MD, and approved April 13, 2017 (received for review February 20, 2017)

                Author contributions: S.N., K.M.-G., J.P., A. Levchenko, A. Louvi, and M.G. designed research; S.N., K.M.-G., and J.P. performed research; Y.V.S. assisted with the high-throughput screen; S.M.S. contributed new reagents; A. Levchenko supervised research; S.N., K.M.-G., J.P., and A. Louvi analyzed data; S.N., K.M.-G., and A. Louvi wrote the paper; and A.Louvi and M.G. revised the paper.

                1S.N. and K.M.-G. contributed equally to this work.

                Article
                PMC5448170 PMC5448170 5448170 201702942
                10.1073/pnas.1702942114
                5448170
                28500274
                7291d703-44f5-4f78-85df-bf218e40c227

                Freely available online through the PNAS open access option.

                History
                Page count
                Pages: 6
                Funding
                Funded by: HHS | NIH | National Institute of Neurological Disorders and Stroke (NINDS) 100000065
                Award ID: NS046521
                Funded by: Yale Program on Neurogenetics
                Award ID: N/A
                Categories
                Biological Sciences
                Medical Sciences

                cerebral cavernous malformations,fluvastatin,zoledronic acid,mevalonate pathway,high-throughput screen

                Comments

                Comment on this article