82
Views
23
CrossRef citations to date
0
Altmetric
Review

Emerging chemotherapeutic strategies for Huntington’s disease

&
Pages 345-363 | Published online: 10 May 2005

Bibliography

  • KREMER BP, GOLDBERG S, ANDREW J et al.: A worldwide study of the Huntington's disease mutation — the sensitivity and specificity of measuring GAG repeats. N Engl. J. Med. (1994) 330:1401–1406.
  • MYERS R, MACDONALD M, KOROSHETZ W et al.: De novo expansion of a (CAG)n repeat in sporadic Huntington's disease. Nat. Genet. (1993) 5:168–173.
  • PAULSEN JS, ZHAO H, STOUT JC et al.: Clinical markers of early disease in persons near onset of Huntington's disease. Neurology (2001) 57:658–662.
  • HELDER DI, KAPTEIN AA, VAN KEMPEN GM, VAN HOUWELINGEN JC, ROOS RA: Impact of Huntington's disease on quality of life. Mov. Disord. (2001) 16:325–330.
  • FERRANTE RJ, KOWALL NW, BEAL MF et al.: Selective sparing of a class of striatal neurons in Huntington's disease. Science (1985) 230:561–563.
  • HERSCH SM, ROSAS HD, FERRANTE RJ: Neuropathology and pathophysiology of Huntington's disease, in movement disorders. In: Neurologic Principles and Practice. Koller W (Ed.), McGraw-Hill, New York, USA (2004):503–526.
  • FERRANTE RJ, KOWALL NW, RI CHARDSON EP Jr : Proliferative and degenerative changes in striatal spiny neurons in Huntington's disease: a combined study using the section-Golgi method and calbindin D28k immunocytochemistry. J. Neurosci. (1991) 11:3877–3887.
  • GRAVELAND GA, WILLIAMS RS, DIFIGLIA MA: Evidence for degenerative and regenerative changes in neostriatal spiny neurons in Huntington's disease. Science (1985) 227:770–773.
  • HUNTINGTON'S DISEASE COLLABORATIVE RESEARCH GROUP: A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington's disease chromosomes. Cell (1993)72:971–983.
  • SZEBENYI G, MORFINI GA, BABCOCK A et al.: Neuropathogenic forms of huntingtin and androgen receptor inhibit fast axonal transport. Neuron (2003) 40:41–52.
  • TRUSHINA E, HELDEBRANT MP, PEREZ-TERZIC CM et al.: Microtubule destabilization and nuclear entry are sequential steps leading to toxicity in Huntington's disease. Proc. NatL Acad. Sci. USA (2003) 100:12171–12176.
  • GAUTHIER LR, CHARRIN BC, BORRELL-PAGES M et al.: Huntingtin controls neurotrophic support and survival of neurons by enhancing BDNF vesicular transport along microtubules. Cell (2004) 118:127–138.
  • ROSS CA: Huntington's disease: new paths to pathogenesis. Cell (2004) 118(1):4–7.
  • ROBITAILLE Y, LOPES-CENDES I, BECHER M, ROULEAU G, CLARK AW: The neuropathology of CAG repeat diseases: review and update of genetic and molecular features. Brain PathoL (1997) 7:901–926.
  • ROSS CA: Intranuclear neuronal inclusions: a common pathogenic mechanism for glutamine-repeat neurodegenerative diseases? Neuron (1997) 19:1147–1150.
  • ROSENBLATT A, RANEN N, NANCE M, PAULSEN J: A physician's guide to the management of Huntington's Disease (2nd edition), Huntington's Disease Society of America, New York, USA (1999).
  • GROUP HS: Unified Huntington's disease rating scale: reliability and consistency. Huntington Study Group. Mov. Disord. (1996) 11:136–142.
  • JENKINS BG, KOROSHETZ WJ, BEAL MF, ROSEN BR: Evidence for impairment of energy metabolism in vivo in Huntington's disease using localized 1H NMR spectroscopy. Neurology (1993) 43:2689–2695.
  • KIEBURTZ K, FEIGIN A, MCDERMOTT M et al.: A controlled trial of remacemide hydrochloride in Huntington's disease. Mov. Disord. (1996) 11:273–277.
  • GROUP HS: Safety and tolerability of thefree-radical scavenger OPC-14117 in Huntington's disease. Neurology (1998) 50:1366–1373.
  • GROUP HS: A randomized, placebo-controlled trial of coenzyme Q10 and remacemide in Huntington's disease. Neurology (2001) 57:397–404.
  • PEYSER CE, FOLSTEIN M, CHASE GA et al.: Trial of D-alpha-tocopherol in Huntington's disease. Am. J. Psychiatry (1995) 152:1771–1775.
  • RANEN NG, PEYSER CE, COYLE JT et al.: A controlled trial of idebenone in Huntington's disease. Mov. Disord. (1996) 11:549–554.
  • KREMER B, CLARK C, HARDY M, ALMQVIST E, RAYMOND L, HAYDEN M: Lamotrigine does not retard the progression of Huntington's disease. WFN Working Group on Huntington's Disease, (1997):34.
  • ROSAS HD, KOROSHETZ WJ, JENKINS BG et al.: Riluzole therapy in Huntington's disease (HD). Mov. Disord. (1999) 14:326–330.
  • SMITH RA, KELSO GF, JAMES AM, MURPHY MP: Targeting coenzyme Q derivatives to mitochondria. Methods Enqmol. (2004) 382:45–67.
  • SHULTS CW, BEAL MF, SONG D, FONTAINE D: Pilot trial of high dosages of coenzyme Q10 in patients with Parkinson's disease. Exp. NeuroL (2004) 188:491–494.
  • YAMAMOTO A. LUCAS JJ, HEN R: Reversal of neuropathology and motor dysfunction in a conditional model of Huntington's disease. Cell (2000) 101:57–66.
  • BEAL MF, FERRANTE RJ: Experimentaltherapeutics in transgenic mouse models of Huntington's disease. Nat. Rev. Neurosci. (2004) 5:373–384.
  • HERSCH SM, FERRANTE RJ: Translating therapies for Huntington's disease from genetic animal models to clinical trials. Neurokc (2004) 1:298–306.
  • PERSICHETTI F, AMBROSE CM, GE P et al.: Normal and expanded Huntington's disease gene alleles produce distinguishable proteins due to translation across the CAG repeat. Mol. Med. (1995) 1:374–383.
  • FERRANTE RJ, GUTEKUNST CA, PERSICHETTI F et al.: Heterogeneous topographic and cellular distribution of huntingtin expression in the normal human neostriatum. J. Neurosci. (1997) 17: 3052–3063.
  • DIFIGLIA M, SAPP E, CHASE KO et al.:Huntingtin is a cytoplasmic protein associated with vesicles in human and rat brain neurons. Neuron (1995) 14:1075–1081.
  • ZUCCATO C, CIAMMOLA A, RIGAMONTI D et al.: Loss of huntingtin-mediated BDNF gene transcription in Huntington's disease. Science (2001) 293:493–498.
  • ZHANG Y, LI M, DROZDA M et al.: Depletion of wild-type huntingtin in mouse models of neurologic diseases. J. Neurochem. (2003) 87:102–106.
  • WELLINGTON CL, BRINKMAN RR, O'KUSKY JR, HAYDEN MR: Toward understanding the molecular pathology of Huntington's disease. Brain PathoL (1997) 7:979–1002.
  • PAULSON HL: Protein fate in neurodegenerative proteinopathies: polyglutamine diseases join the (mis)fold. Am. J. Hum. Genet. (1999) 64:339–345.
  • JANA NR, ZEMSKOV EA, WANG G, NUKINA N: Altered proteasomal function due to the expression of polyglutamine-expanded truncated N-terminal huntingtin induces apoptosis by caspase activation through mitochondrial cytochrome c release. Hum. MoL Genet. (2001) 10:1049–1059.
  • JANA NR, TANAKA M, WANG G, NUKINA N: Polyglutamine length-dependent interaction of Hsp40 and Hsp70 family chaperones with truncated N-terminal huntingtin: their role in suppression of aggregation and cellular toxicity. Hum. MoL Genet. (2000) 9:2009–2018.
  • CHAT Y, KOPPENHAFER SL, SHOESMITH SJ, PEREZ MK, PAULSON HL: Evidence for proteasome involvement in polyglutamine disease: localization to nuclear inclusions in SCA3/ MJD and suppression of polyglutamine aggregation in vitro. Hum. MoL Genet. (1999) 8:673–682.
  • CHA JH: Transcriptional dysregulation in Huntington's disease. Trends Neurosci. (2000) 23(9):387–392.
  • LUTHI-CARTER R, APOSTOL BL, DUNAH AW et al.: Complex alteration of NMDA receptors in transgenic Huntington's disease mouse brain: analysis of mRNA and protein expression, plasma membrane association, interacting proteins, and phosphorylation. NeurobioL Dis. (2003) 14:624–636.
  • PREISINGER E, JORDAN BM, KAZANTSEV A, HOUSMAN D: Evidence for a recruitment and sequestration mechanism in Huntington's disease. Philos. Trans. R. Soc. Pond. B. Biol. Sci. (1999) 354:1029–1034.
  • DIFIGLIA M, SAPP E, CHASE KO et al.: Aggregation of huntingtin in neuronal intranuclear inclusions and dystrophic neurites in brain. Science (1997) 277:1990–1993.
  • GUTEKUNST C, LI SH, YI H et al.: Nuclear and neuropil aggregates in Huntington's disease: relationship to neuropathology. J. Neurosci. (1999) 19:2522–2534.
  • KUEMMERLE S, GUTEKUNST CA, KLEIN AM et al.: Huntington aggregates may not predict neuronal death in Huntington's disease. Ann. NeuroL (1999) 46:842–849.
  • COOPER JK, SCHILLING G, PETERS MF et al.: Truncated N-terminal fragments of huntingtin with expanded glutamine repeats form nuclear and cytoplasmic aggregates in cell culture. Hum. MoL Genet. (1998) 7:783–790.
  • DAVIES SW, BEARDSALL K, TURMAINE M, DIFIGLIA M, ARONIN N, BATES GP: Are neuronal intranuclear inclusions the common neuropathology of triplet-repeat disorders with polyglutamine-repeat expansions? Lancet (1998) 351:131–133.
  • MEADE CA, DENG YP, FUSCO FR et al.: Cellular localization and development of neuronal intranuclear inclusions in striatal and cortical neurons in R6/2 transgenic mice. J. Comp. NeuroL (2002) 449:241–269.
  • ORDWAY JM, TALLAKSEN-GREENE S, GUTEKUNST CA et al.: Ectopically expressed GAG repeats cause intranuclear inclusions and a progressive late onset neurological phenotype in the mouse. Cell (1997) 91:753–763.
  • KLEMENT IA, SKINNER PJ, KAYTOR MD et al.: Ataxin-1 nuclear localization and aggregation: role in polyglutamine-induced disease in SCA1 transgenic mice. Cell (1998) 95:41–53.
  • KIM M, LEE HS, LAFORET G et al.: Mutant huntingtin expression in clonal striatal cells: dissociation of inclusion formation and neuronal survival by caspase inhibition. J. Neurosci. (1999) 19:964–973.
  • SAUDOU F, FINKBEINER S, DEVYS D, GREENBERG ME: Huntingtin acts in the nucleus to induce apoptosis but death does not correlate with the formation of intranuclear inclusions. Cell (1998) 95: 55–66.
  • SISODIA SS: Nuclear inclusions in glutamine repeat disorders: are they pernicious, coincidental, or beneficial? Cell (1998) 95:1–4.
  • ARRASATE M, MITRA S, SCHWEITZER ES, SEGAL MR, FINKBEINER S: Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death. Nature (2004) 431:805–810.
  • DEDEOGLU A, KUBILUS JK, JEITNER TM et al.: Therapeutic effects of cystamine in a murine model of Huntington's disease. J. Neurosci. (2002) 22:8942–8950.
  • DEDEOGLU A, KUBILUS JK, YANG L et al.: Creatine therapy provides neuroprotection after onset of clinical symptoms in Huntington's disease transgenic mice. J. Neurochem. (2003) 85:1359–1367.
  • FERRANTE RJ, ANDREASSEN OA, JENKINS BG et al.: Neuroprotective effects of creatine in a transgenic mouse model of Huntington's disease. J. Neurosci. (2000) 20:4389–4397.
  • ANDREASSEN OA, DEDEOGLU A, FERRANTE RJ et al.: Creatine increase survival and delays motor symptoms in a transgenic animal model of Huntington's disease. NeurobioL Dis. (2001) 8:479–491.
  • FERRANTE RJ, ANDREASSEN OA, DEDEOGLU A et al.: Therapeutic effects of Coenzyme Q10 and remacemide in transgenic models of Huntington's disease. J. Neurosci. (2002) 22:1592–1599.
  • NUCIFORA FC Jr, SASAKI M, PETERS MF et al.: Interference by huntingtin and atrophin-1 with CBP-mediated transcription leading to cellular toxicity. Science (2001) 291:2423–2428.
  • BENCE NF, SAMPAT RM, KOPITO RR: Impairment of the ubiquitin-proteasome system by protein aggregation. Science (2001) 292:1552–1555.
  • PANOV AV, GUTEKUNST CA, LEAVITT BR et al.: Early mitochondrial calcium defects in Huntington's disease are a direct effect of polyglutamines. Nat. Neurosci. (2002) 5:731–736.
  • JIANG H, NUCIFORA FC, ROSS CA. DEFRANCO DB: Cell death triggered by polyglutamine-expanded huntingtin in a neuronal cell line is associated with degradation of CREB-binding protein. Hum. Mol. Genet. (2003) 12:1–12.
  • BEAL MF: Oxidative metabolism. Ann. NYAcad. Sci. (2000) 924:164–169.
  • DESAGHER S, MARTINOU JC: Mitochondria as the central control point of apoptosis. Trends Cell Biol. (2000) 10:369–377.
  • GREEN DR, REED JC: Mitochondria and apoptosis. Science (1998) 281: 1309–1312.
  • ROY S, NICHOLSON DW: Cross-talk in cell death signaling. J. Exp. Med. (2000) 192:21–26.
  • KIECHLE T, DEDEOGLU A, KUBILUS J et al.: Cytochrome C and caspase-9 expression in Huntington's disease. Neuromolecular Med. (2002) 1:183–195.
  • FERRANTE RJ, KUBILUS JK, LEE J et al.: Histone deacetylase inhibition by sodium butyrate chemotherapy ameliorates the neurodegenerative phenotype in Huntington's disease mice. J. Neurosci. (2003) 23:9418–9427.
  • FERRANTE RJ, RYU H, KUBILUS JK et al.: Chemotherapy for the brain: the antitumor antibiotic mithramycin prolongs survival in a mouse model of Huntington's disease. J. Neurosci. (2004) 24:10335–10342.
  • HOCKLY E, RICHON VM, WOODMAN B et al.: Suberoylanilide hydroxamic acid, a histone deacetylase inhibitor, ameliorates motor deficits in a mouse model of Huntington's disease. Proc. NatL Acad. Sci. USA (2003) 100:2041–2046.
  • GARDIAN G, BROWNE SE, CHOI DK et al.: Neuroprotective effects of phenylbutyrate in the N171-82Q transgenic mouse model of Huntington's disease. J. Biol. Chem. (2005) 280(1):556–563.
  • LORAND L, STEERING GE JR, LOWE-KRENTZ L: Formation of gamma-glutamyl-epsilon-lysine bridges between membrane proteins by a Ca2*-regulated enzyme in intact erythrocytes. J. SupramoL Struct. (1978) 9:427–440.
  • KAHLEM P, GREEN H, DJIAN P: Transglutaminase action imitates Huntington's disease: selective polymerization of huntingtin containing expanded polyglutamine. MoL Cell (1998) 1: 595–601.
  • IGARASHI S, KOIDE R, SHIMOHATA T et al.: Suppression of aggregate formation and apoptosis by transglutaminase inhibitors in cells expressing truncated DRPLA protein with an expanded polyglutamine stretch. Nat. Genet. (1998) 18:111-117. Expert Op/n. Emerging Drugs (2005) 10(2)
  • LESORT M, CHUN W, JOHNSON GVW, FERRANTE RJ: Tissue transglutaminase is increased in Huntington's disease brain. J. Neurochem. (1999) 73:2018–2027.
  • JEITNER TM, BOGDANOV MB, MATSON WR et al.: N-(y-L-glutamyfi-L-lysine is increased in cerebrospinal fluid of patients with Huntington's disease. J. Neurochem. (2001) 79:1109–1112.
  • REVESZ L, MODIG H: Cysteamine-induced increase of cellular glutathione-level: a new hypothesis of the radioprotective mechanism. Nature (1965) 207:430–431.
  • ROBITAILLE K, DAVIAU A, TUCHOLSKI J, JOHNSON GV, RANCOURT C, BLOUIN R: Tissue transglutaminase triggers oligomerization and activation of dual leucine zipper-bearing kinase in calphostin C-treated cells to facilitate apoptosis. Cell Death Differ. (2004) 11:542–549.
  • PALKOVITS M, BROWNSTEIN MJ, EIDEN LE et al.: Selective depletion of somatostatin in rat brain by cysteamine. Brain Res. (1982) 240:178–180.
  • MCDONNELL NB, DE GUZMAN RN, RICE WG, TURPIN JA, SUMMERS MF: Zinc ejection as a new rationale for the use of cystamine and related disulfide-containing antiviral agents in the treatment of AIDS. J. Med. Chem. (1997) 40: 1969–1976.
  • KARPUJ MV BECHER MW, SPRINGER JE et al.: Prolonged survival and decreased abnormal movements in transgenic model of Huntington's disease, with administration of the transglutaminase inhibitor cystamine. Nat. Med. (2002) 8:143–149.
  • FOX JH, BARBER DS, SINGH B et al.:Cystamine increases L-cysteine levels in Huntington's disease transgenic mouse brain and in a PC12 model of polyglutamine aggregation. J. Neurochem. (2004) 91:413–422.
  • BAILEY CD, JOHNSON GV: Tissue transglutaminase contributes to disease progression in the R6/2 Huntington's disease mouse model via aggregate-independent mechanisms. J. Neurochem. (2005) 92:83–92.
  • PRESCOTT LF, NEWTON RW, SWAINSON CP, WRIGHT N, FORREST AR, MATTHEW H: Successful treatment of severe paracetamol overdosage with cysteamine. Lancet (1974) 1:588–592.
  • GM-IL WA, THOENE JG, SCHNEIDER JA: Cystinosis. N Engl Med. (2002) 347(2):111–121.
  • CORDEN BJ, SCHULMAN JD, SCHNEIDER JA, THOENE JG: Adverse reactions to oral cysteamine use in nephropathic cystinosis. Dev. Pharmacol Ther. (1981) 3:25–30.
  • FRIEDLANDER RM: Apoptosis and caspases in neurodegenerative diseases. N Engl. J. Med. (2003) 348:1365–1375.
  • YRJANHEIKKI J, KEINANEN R, PELLIKKA M, HOKFELT T, KOISTINAHO J: Tetracyclines inhibit microglial activation and are neuroprotective in global brain ischemia. Proc. Nati Acad. Sci. USA (1998) 95:15769–15774.
  • CHEN M, ONA VO, LI M et al.: Minocycline inhibits caspase-1 and caspase-3 expression and delays mortality in a transgenic mouse model of Huntington's disease. Nat. Med. (2000) 6:797–801.
  • SANCHEZ MEJIA RO, ONA VO, LI M, FRIEDLANDER RM: Minocycline reduces traumatic brain injury-mediated caspase-1 activation, tissue damage, and neurological dysfunction. Neurosurgery (2001) 48:1393–1401.
  • TIKKA T, FIEBICH BL, GOLDSTEINS G, KEINANEN R, KOISTINAHO J: Minocycline, a tetracycline derivative, is neuroprotective against excitotoxicity by inhibiting activation and proliferation of microglia. Neurosci. (2001) 21:2580–2588.
  • WU DC, JACKSON-LEWIS V, VILA M et al.: Blockade of microglial activation is neuroprotective in the 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine mouse model of Parkinson's disease. J. Neurosci. (2002) 22:1763–1771.
  • DU Y, MA Z, LIN S et al.: Minocycline prevents nigrostriatal dopaminergic neurodegeneration in the MPTP model of Parkinson's disease. Proc. Nati Acad. Sci. USA (2001) 98:14669–14674.
  • ZHU S, STAVROVSKAYA IG, DROZDA M et al.: Minocycline inhibits cytochrome c release and delays progression of ALS in mice. Nature (2002) 417:72–78.
  • WANG X, ZHU S, DROZDA M et al.: Minocycline inhibits caspase-independent and -dependent mitochondrial cell death pathways in models of Huntington's disease. Proc. Natl Acad. Sci. USA (2003) 100:10483–10487.
  • SCHERZINGER E, LURZ R, TURMAINE M et al.: Huntingtin-encoded polyglutamine expansions form amyloid-like protein aggregates in vitro and in vivo. Cell (1997) 90(3):549–558.
  • SMITH DL, WOODMAN B, MAHAL A et al.: Minocycline and doxycycline are not beneficial in a model of Huntington's disease. Ann. NeuroL (2003) 54:186–196.
  • HERSCH S, FINK K, VONSATTEL JP, FRIEDLANDER RM: Minocycline is protective in a mouse model of Huntington's disease. Ann. Neurol (2003) 54:841; author reply 842–843.
  • BANTUBUNGI K, JACQUARD C, GRECO A et al.: Minocycline in phenotypic models of Huntington's disease. Neurobiol Dis. (2005) 18:206–217.
  • GONI-ALLO B, RAMOS M, JORDAN J, AGUIRRE N: In vivo studies on the protective role of minocycline against excitotoxicity caused by malonate or N-methyl-D-aspartate. Exp. Neurol (2005) 191:326–330.
  • CARRERAS I, KUBILUS JK, SMITH K et al.: Combination therapy using minocycline and coenzyme Q10 in R6/2 transgenic HD mice. Program No. 208.10. Abstract Viewer/Itinerary Planner. Society for Neuroscience, Washington, DC (2003).
  • HUNTINGTON STUDY GROUP: Minocycline safety and tolerability in Huntington's disease. Neurology (2004) 63:547–549.
  • THOMAS M, ASHIZAWA T, JANKOVIC J: Minocycline in Huntington's disease: a pilot study. Mov. Disord. (2004) 19:692–695.
  • BONELLI RM, HODL AK, HOFMANN P, KAPFHAMMER HP: Neuroprotection in Huntington's disease: a 2-year study on minocycline. Int. Clin. Psychopharmacol (2004) 19:337–342.
  • DENOVAN-WRIGHT EM, DEVARAJAN S, DURSUN SM, ROBERTSON HA: Maintained improvement with minocycline of a patient with advanced Huntington's disease. Psychopharmacol (2002) 16:393–394.
  • BLUM D, CHTARTO A, TENENBAUM L, BROTCHI J, LEVI VIER M: Clinical potential of minocycline for neurodegenerative disorders. NeurobioL Dis. (2004) 17:359–366.
  • YONG VW, WELLS J, GIULIANI F, CASHA S, POWER C, METZ LM: The promise of minocycline in neurology Lancet NeuroL (2004) 3:744–751.
  • TAN HH: Antibacterial therapy for acne: a guide to selection and use of systemic agents. Am. J. Clin. DermatoL (2003) 4:307–314.
  • EMERY P, SUAREZ-ALMAZOR M: Rheumatoid arthritis. Clin. Evid. (2003) 9:1349–1371.
  • KNOWLES SR, SHAPIRO L, SHEAR NH: Serious adverse reactions induced by minocycline. Report of 13 patients and review of the literature. Arch. Dermatol. (1996) 132:934–939.
  • STEFFAN JS, BODAI L, PALLOS J et al.: Histone deacetylase inhibitors arrest polyglutamine-dependent neurodegeneration in Drosophila. Nature (2001) 413:739–743.
  • HAKE SB, MAO A, ALLIS CD: Linking the epigenetic 'language' of covalent histone modifications to cancer. Br. J. Cancer (2004) 90:761–769.
  • BLANCO G, FU H, MENDEZ C, KHOSLA C, SALAS JA: Deciphering the biosynthetic origin of the aglycone of the aureolic acid group of anti-tumor agents. Chem. Biol. (1996) 3:193–196.
  • RALSTON SH: Pathogenesis and management of cancer associated hypercalcaemia. Cancer Surv. (1994) 21:179–196.
  • CHAKRABARTI S, BHATTACHARYYA D, DASGUPTA D: Structural basis of DNA recognition by anticancer antibiotics, chromomycin A3, and mithramycin: roles of minor groove width and ligand flexibility. Biopolymers (2001) 56:85–95.
  • HESCOCK H Jr, PARKER M, WANG TY, BALLINGER R, BALDUCCI L: Metastatic carcinoma of unknown primary: complete response to second-line treatment with plicamycin. Am. J. Med. Sci. (1989) 298:34–37.
  • PRADO L, LOMBO F, BRANA AF, MENDEZ C, ROHR J, SALAS JA: Analysis of two chromosomal region adjacent to genes for a Type II polyketide synthase involved in the biosynthesis of the antitumor polyketide mithramycin in Streptomyces argillaceus. Mol. Gen. Genet. (1999) 261:216–225.
  • RYAN WG: Treatment of Paget's disease of bone with mithramycin. Clin. Orthop. (1977) 127:106–110.
  • KENNEDY BJ: Mithramycin therapy in testicular cancer. J. Urol. (1972) 107:429–432.
  • RANSOHOFF J, MARTIN BF, MEDREK TJ, HARRIS MN, GOLOMB FM, WRIGHT JC: Preliminary clinical study of mithramycin (nsc-24559) in primary tumors of the central nervous system. Cancer Chemother. Rep. (1965) 49:51–57.
  • HAGEN G, DENNIG J, PREISS A, BEATO M, SUSKE G: Functional analyses of the transcription factor Sp4 reveal properties distinct from Spl and Sp3. J. Biol. Chem. (1995) 270:24989–24994.
  • MAJELLO B, DE LUCA P, SUSKE G, LANIA L: Differential transcriptional regulation of c-myc promoter through the same DNA binding sites targeted by Spl-like proteins. Oncogene (1995) 10:1841–1848.
  • CHATTERJEE S, ZAMAN K, RYU H, CONFORTO A, RATAN RR: Sequence-selective DNA binding drugs mithramycin A and chromomycin A3 are potent inhibitors of neuronal apoptosis induced by oxidative stress and DNA damage in cortical neurons. Ann. NeuroL (2001) 49:345–354.
  • DUNAH AW, JEONG H, GRIFFIN A et al.: Sp 1 and TAFII130 transcriptional activity disrupted in early Huntington's disease. Science (2002) 296:2238–2243.
  • LI SH, CHENG AL, ZHOU H et al.: Interaction of Huntington's disease protein with transcriptional activator Spl. MoL Cell Biol. (2002) 22:1277–1287.
  • SISSI C, MORO S, RICHTER S et al.: DNA-interactive anticancer aza-anthrapyrazoles: biophysical and biochemical studies relevant to the mechanism of action. MoL Pharmacol. (2001) 59:96–103.
  • MARKS PA, RICHON VM, RIFKIND RA. Nucleotide, protein histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. J. Need Cancer Inst. (2000) 92:1210–1216.
  • CONLEY BA, EGORIN MJ, TAIT N et al.: Phase I study of the orally administered butyrate prodrug, tributyrin, in patients with solid tumors. Clin. Cancer Res. (1998) 4:629–634.
  • SHARMA SK, MICHAELIS C, LEE KY, WANG JH, WEEKS G: Binding and catalytic properties of the Cdc2 and Crp proteins of Dictyostelium. Eur. j Biochem. (1999) 260:603–608.
  • RICHON VM, ZHOU X, RIFKIND RA, MARKS Pk Histone deacetylase inhibitors: development of suberoylanilide hydroxamic acid (SAHA) for the treatment of cancers. Blood Cells MoL Dis. (2001) 27:260–264.
  • GORIN NC, ESTEY E, JONES RJ, LEVITSKY HI, BORRELLO I, SLAVIN S: New developments in the therapy of acute myelocytic leukemia. Hematology (Am. Soc. HematoL Educ. Program) (2000) 179:69–89.
  • CHANG JG, HSIEH-LI HM, JONG YJ et al.: Treatment of spinal muscular atrophy by sodium butyrate. Proc. Nail. Acad. Sci. USA (2001) 98:9808–9813.
  • EGORIN MJ, YUAN ZM, SENTZ DL, PLAISANCE K, EISEMAN JL: Plasma pharmacokinetics of butyrate after intravenous administration of sodium butyrate or oral administration of tributyrin or sodium butyrate to mice and rats. Cancer Chemother. PharmacoL (1999) 43:445–453.
  • SEALY L, CHALKLEY R: The effect of sodium butyrate on histone modification. Cell (1978) 14: 115–121.
  • CANDIDO EP, REEVES R, DAVIE JR: Sodium butyrate inhibits histone deacetylation in cultured cells. Cell (1978) 14:105–113.
  • COLLINS AF, PEARSON HA, GIARDINA P, MCDONAGH KT, BRUSILOW SW, DOVER GJ: Oral sodium phenylbutyrate therapy in homozygous beta thalassemia: a clinical trial. Blood (1995) 85:43–49.
  • SHER GD, GINDER GD, LITTLE J, YANG S, DOVER GJ, OLIVIERI NF: Extended therapy with intravenous arginine butyrate in patients with beta-hemoglobinopathies. New EngL J. Med. (1995) 332:1606–1610.
  • BATSHAW ML, MACARTHUR RB, TUCHMAN M: Alternative pathway therapy for urea cycle disorders: twenty years later. J. Pediatr. (2001) 138:S46–S55.
  • BERGS, SERABE B, ALEKSIC A et al.: Pharmacokinetics and cerebrospinal fluid penetration of phenylacetate and phenylbutyrate in the nonhuman primate. Cancer Chemother. PharmacoL (2001) 47:385–390.
  • BURLINA AB, OGIER H, KORALL H, TREFZ FK: Long-term treatment with Expert Op/n. Emerging Drugs (2005) 10(2) sodium phenylbutyrate in ornithine transcarbamylase-deficient patients. Mol. Genet. Metab. (2001) 72:351–355.
  • GILBERT J, BAKER SD, BOWLING MK et al.: A Phase I dose escalation and bioavailability study of oral sodium phenylbutyrate in patients with refractory solid tumor malignancies. Clin. Cancer Res. (2001) 7:2292–2300.
  • SAMID D: Therapeutic targeting of transcription in acute promyelocytic leukemia by use of an inhibitor of histone deacetylase. J. Natl. Cancer Inst. (1999) 91:475–476.
  • MARKS PA, RICHON VM, KELLY WK, CHIAO JH, MILLER T: Histone deacetylase inhibitors: development as cancer therapy. Novartis Found. Symp. (2004) 259:269–281.
  • MCGUINNESS MC, ZHANG HP, SMITH KD: Evaluation of pharmacological induction of fatty acid beta-oxidation in X-linked adrenoleukodystrophy. MoL Genet. Metab. (2001) 74:256–263
  • WEI H, KEMP S, MCGUINNESS MC, MOSER AB, SMITH KD: Pharmacological induction of peroxisomes in peroxisome biogenesis disorders. Ann. NeuroL (2000) 47:286–296.
  • KEMP S, WEI HM, LU JF et al.: Gene redundancy and pharmacological gene therapy: implications for X-linked adrenoleukodystrophy. Nat. Med. (1998) 4:1261–1268.
  • DOVER GJ, BRUSILOW S, CHARACHE S: Induction of fetal hemoglobin production in subjects with sickle cell anemia by oral sodium phenylbutyrate. Blood (1994) 84:339–343.
  • DOVER GJ: Hemoglobin switching protocols in thalassemia. Experience with sodium phenylbutyrate and hydroxyurea. Ann. IVY Acad. Sci. (1998) 850:80–86.
  • BROUILLET E, HANTRAYE P, FERRANTE RJ et al.: Chronic mitochondrial energy impairment produces selective striatal degeneration and abnormal choreiform movements in primates. Proc. Nail. Acad. Sci. USA (1995) 92:7105–7109.
  • BROWNE SE, BOWLING AC, MACGARVEY U et al.: Oxidative damage and metabolic dysfunction in Huntington's disease: selective vulnerability of the basal ganglia. Ann. NeuroL (1997) 41:646–653.
  • GUM, GASH MT, MANN VM, JAVOY-AGID F, COOPER JM, SCHAPIRA Al-TV: Mitochondrial defect in Huntington's disease caudate nucleus. Ann. NeuroL (1996) 39:385–389.
  • JENKINS B, KOROSHETZ W, BEAL MF, ROSEN B: Evidence for an energy metabolism defect in Huntington's disease using localized proton spectroscopy. Neurology (1993) 43:2689–2695.
  • KOROSHETZ WJ, JENKINS BG, ROSEN BR, BEAL MF: Energy metabolism defects in Huntington's disease and possible therapy with coenzyme Q10. Ann. NeuroL (1997) 41: 160–165.
  • BESSMAN SP, GEIGER PJ: Transport of energy in muscle: the phosphorylcreatine shuttle. Science (1981) 211:448–452.
  • MATTHEWS RT, YANG L, JENKINS BG et al.: Neuroprotective effects of creatine and cyclocreatine in animal models of Huntington's disease. Neurosci. (1998) 18: 156–163.
  • CARTER AJ, MULLER RE, PSCHORN U, STRANSKY W: Preincubation with creatine enhances levels of creatine phosphate and prevents anoxic damage in rat hippocampal slices. Neurochem. (1995) 64:2691–2699.
  • MATTHEWS RT, FERRANTE RJ, KLIVENYI P et al.: Creatine and cyclocreatine attenuate MPTP neurotoxicity. Exp. NeuroL (1999) 157:142–149.
  • KLIVENYI P, FERRANTE RJ, MATTHEWS RT et al.: Neuroprotective effects of creatine in a transgenic animal model of ALS. Nat. Med. (1999) 5:347–350.
  • VERBESSEM P, LEMIERE J, EIJNDE B et al.: Creatine supplementation in Huntington's disease: a placebo-controlled pilot trial. Neurology (2003) 61:925–930.
  • TABRIZI SJ, BLAMIRE AM, MANNERS DN et al.: Creatine therapy for Huntington's disease: clinical and MRS findings in a 1-year pilot study. Neurology (2003) 61:141–142.
  • BENDER A, AUER DP, MERL T et al.: Creatine supplementation lowers brain glutamate levels in Huntington's disease. Neurol. (2005) 252:36–41.
  • LAWLER JM, BARNES WS, WU G, SONG W, DEMAREE S: Direct antioxidant properties of creatine. Biochem. Biophys. Res. Comm. (2002) 290:47–52.
  • BENZI G: Is there a rationale for the use of creatine either as nutritional supplementation or drug administration in humans participating in sport? PharmacoL Res. (2000) 41:255–264.
  • COOKE WH, GRANDJEAN PW, BARNES WS. Effect of oral creatine supplementation on power output and fatigue during bicycle ergometry. J. AppL Physiol. (1995) 78:670–673.
  • TARNOPOLSKY MA, BEAL MF: Potential for creatine and other therapies targeting cellular energy dysfunction in neurological disorders. Ann. NeuroL (2001) 49:561–574.
  • WYSS M, SCHULZE A: Health implications of creatine: can oral creatine supplementation protect against neurological and atherosclerotic disease? Neurosci. (2002) 112:243–260.
  • MERTSCHENK B, GLOXHUBER C, WALLIMANN T: Health assessment of creatine as a dietary supplement. Dtsch. Lebensm. Rundsch. (2001) 97:250–257.
  • BEYER RE: An analysis of the role of coenzyme Q in free radical generation and as an antioxidant. Biochem. Cell Biol. (1992) 70:390–403.
  • NOACK H, KUBE U, AUGUSTIN W: Relations between tocopherol depletion and coenzyme Q during lipid peroxidation in rat liver mitochondria. Free. Radic. Res. (1994) 20:375–386.
  • KAGAN V, SERBINOVA E, PACKER L: Antioxidant effects of ubiquinones in microsomes and mitochondria are mediated by tocopherol recycling. Biochem. Biophys. Res. Commun. (1990) 169:851–857.
  • ECHTAY KS, ROUSSEL D, ST-PIERRE J et al.: Superoxide activates mitochondrial uncoupling proteins. Nature (2002) 415:96–99.
  • CASTEILLA L, RIGOULET M, PENICAUD L: Mitochondrial ROS metabolism: modulation by uncoupling proteins. IUBMB Life (2001) 52:181–188.
  • NISHIKAWA Y, TAKAHASHI M, YORIFUJI S et al.: Long-term coenzyme Q„ therapy for a mitochondrial encephalomyopathy with cytochrome c oxidase deficiency: A31P NMR study. Neurology (1989) 39:399–403.
  • IHARA Y, NAMBA R, KURODA S, SATO T, SHIRABE T: Mitochondrial encephalomyopathy (MELAS): pathological study and successful therapy with coenzyme Q„ and idebenone. J. NeuroL Sci. (1989) 90:263–271.
  • CHARIOT P, BRUGIERES P, ELIEZER-VANEROT MC, GENY C, BINAGHI M, CESARO P: Choreic movements and MRI abnormalities in the subthalamic nuclei reversible after administration of coenzyme Q„ and multiple vitamins in a patient with bilateral optic neuropathy. Mov. Disord. (1999) 14:855–859.
  • ABE K, FUJIMURA H, NISHIKAWA Y et al.: Marked reduction in CSF lactate and pyruvate levels after CoQ therapy in a patient with mitochondrial myopathy, encephalopathy, lactic acidosis and stroke-like episodes (MELAS). Acta Neurol. Scand. (1991) 83:356–359.
  • TOMASETTI M, LITTARRU GP, STOCKER R, ALLEVA R: Coenzyme Q10 enrichment decreases oxidative DNA damage in human lymphocytes. Free Radic. Biol. Med. (1999) 27:1027–1032.
  • CHAN A, REICHMANN H, KOGEL A et al.: Metabolic changes in patients with mitochondrial myopathies and effects of coenzyme Q„ therapy. J. Neurol. (1998) 245:681–685.
  • SCHILLING G, SAVONENKO AV, COONFIELD ML et al.: Environmental, pharmacological, and genetic modulation of the HD phenotype in transgenic mice. Exp. Neurol. (2004) 187:137–149.
  • BEAL MF, HENSHAW R, JENKINS BG, ROSEN BR, SCHULZ JB: Coenzyme Q10 and nicotinamide block striatal lesions produced by the mitochondrial toxin malonate. Ann. Neurol. (1994) 36:882–888.
  • LASS A, FORSTER MJ, SOHAL RS: Effects of coenzyme Q„ and a-tocopherol administration on their tissue levels in the mouse: elevation of mitochondrial alpha tocopherol by coenzyme Q. Free Radic. Biol. Med. (1999) 26:1375–1382.
  • SHULTS CW, OAKES D, KIEBURTZ K et al.: Effects of coenzyme Q10 in early Parkinson's disease: evidence of slowing of the functional decline. Arch Neurol. (2002) 59:1541–1550.
  • SMITH K, MATSON S, CORMIER K et al.: Therapeutic effects of high dose administration of coenzyme Q10 in the R6/2 model of Huntington's disease. Abstract Viewer/Itinerary Planner. Society for Neuroscience, Washington, DC, USA (2004).
  • OKAMOTO T, FUKUI K, NAKAMOTO M et al.: Serum levels of coenzyme Q10 and lipids in patients during total parenteral nutrition. J. Num. Sci. VitaminoL (1986) 32: 1–12.
  • MOLYNEUX S, FLORKOWSKI C, LEVER M, GEORGE P: The bioavailability of coenzyme Q10 supplements available in New Zealand differs markedly. NZ Med. J. (2004) 117(1203):U1108.
  • WEBER C, SEJERSGARD JAKOBSEN T, MORTENSEN SA, PAULSEN G, HOLMER G: Antioxidative effect of dietary coenzyme Q_„ in human blood plasma. Int. J. Vitam. Num. Res. (1994) 64:311–315.
  • MATTHEWS RT, YANG L, BROWNE S, BAIK M, BEAL MF: Coenzyme Q„ administration increases brain mitochondrial concentrations and exerts neuroprotective effects. Proc. Nail. Acad. Sci. USA (1998) 95:8892–8897.
  • HUNTINGTON STUDY GROUP: A randomized, placebo-controlled trial of coenzyme Q10 and remacemide in Huntington's disease. Neurology (2001) 57:397–404.
  • MUSUMECI O, NAINI A, SLONIM AE et al.: Familial cerebellar ataxia with muscle coenzyme Q10 deficiency. Neurology (2001) 56:849–855.

Websites

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.