Therapeutic NOTCH3 cysteine correction in CADASIL using exon skipping: in vitro proof of concept.

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy, or CADASIL, is a hereditary cerebral small vessel disease caused by characteristic cysteine altering missense mutations in the NOTCH3 gene. NOTCH3 mutations in CADASIL result in an uneven number of cysteine residues in one of the 34 epidermal growth factor like-repeat (EGFr) domains of the NOTCH3 protein. The consequence of an unpaired cysteine residue in an EGFr domain is an increased multimerization tendency of mutant NOTCH3, leading to toxic accumulation of the protein in the (cerebro)vasculature, and ultimately reduced cerebral blood flow, recurrent stroke and vascular dementia. There is no therapy to delay or alleviate symptoms in CADASIL. We hypothesized that exclusion of the mutant EGFr domain from NOTCH3 would abolish the detrimental effect of the unpaired cysteine and thus prevent toxic NOTCH3 accumulation and the negative cascade of events leading to CADASIL. To accomplish this NOTCH3 cysteine correction by EGFr domain exclusion, we used pre-mRNA antisense-mediated skipping of specific NOTCH3 exons. Selection of these exons was achieved using in silico studies and based on the criterion that skipping of a particular exon or exon pair would modulate the protein in such a way that the mutant EGFr domain is eliminated, without otherwise corrupting NOTCH3 structure and function. Remarkably, we found that this strategy closely mimics evolutionary events, where the elimination and fusion of NOTCH EGFr domains led to the generation of four functional NOTCH homologues. We modelled a selection of exon skip strategies using cDNA constructs and show that the skip proteins retain normal protein processing, can bind ligand and be activated by ligand. We then determined the technical feasibility of targeted NOTCH3 exon skipping, by designing antisense oligonucleotides targeting exons 2-3, 4-5 and 6, which together harbour the majority of distinct CADASIL-causing mutations. Transfection of these antisense oligonucleotides into CADASIL patient-derived cerebral vascular smooth muscle cells resulted in successful exon skipping, without abrogating NOTCH3 signalling. Combined, these data provide proof of concept for this novel application of exon skipping, and are a first step towards the development of a rational therapeutic approach applicable to up to 94% of CADASIL-causing mutations.

[1]  W. V. van Roon-Mom,et al.  Antisense oligonucleotides in therapy for neurodegenerative disorders. , 2015, Advanced drug delivery reviews.

[2]  K. Davies,et al.  Functional correction in mouse models of muscular dystrophy using exon-skipping tricyclo-DNA oligomers , 2015, Nature Medicine.

[3]  U. Lendahl,et al.  Notch3 Is Necessary for Blood Vessel Integrity in the Central Nervous System , 2015, Arteriosclerosis, thrombosis, and vascular biology.

[4]  V. Ricotti,et al.  Safety and efficacy of drisapersen for the treatment of Duchenne muscular dystrophy (DEMAND II): an exploratory, randomised, placebo-controlled phase 2 study , 2014, The Lancet Neurology.

[5]  S. Iannaccone,et al.  Spinal Muscular Atrophy: Therapeutic Strategies , 2014, Current Treatment Options in Neurology.

[6]  G. Terwindt,et al.  Interpretation of NOTCH3 mutations in the diagnosis of CADASIL , 2014, Expert review of molecular diagnostics.

[7]  J. Dauwerse,et al.  Hypomorphic NOTCH3 Alleles Do Not Cause CADASIL in Humans , 2013, Human mutation.

[8]  A. Joutel,et al.  Abnormal recruitment of extracellular matrix proteins by excess Notch3 ECD: a new pathomechanism in CADASIL. , 2013, Brain : a journal of neurology.

[9]  K. McKeage,et al.  Mipomersen Sodium: First Global Approval , 2013, Drugs.

[10]  E. Kremmer,et al.  Co-aggregate formation of CADASIL-mutant NOTCH3: a single-particle analysis. , 2011, Human molecular genetics.

[11]  M. Dichgans,et al.  CADASIL , 2009, The Lancet Neurology.

[12]  M. Baumann,et al.  Evolutionary scenarios of Notch proteins. , 2009, Molecular biology and evolution.

[13]  G. van Ommen,et al.  Theoretic applicability of antisense‐mediated exon skipping for Duchenne muscular dystrophy mutations , 2009, Human mutation.

[14]  A. Moorman,et al.  Amplification efficiency: linking baseline and bias in the analysis of quantitative PCR data , 2009, Nucleic acids research.

[15]  P. Handford,et al.  A conserved face of the Jagged/Serrate DSL domain is involved in Notch trans-activation and cis-inhibition , 2008, Nature Structural &Molecular Biology.

[16]  R. Kole,et al.  Efficient and persistent splice switching by systemically delivered LNA oligonucleotides in mice. , 2006, Molecular therapy : the journal of the American Society of Gene Therapy.

[17]  S. Saiki,et al.  Varicose veins associated with CADASIL result from a novel mutation in the Notch3 gene , 2006, Neurology.

[18]  T. Tabira,et al.  CADASIL-causing mutations do not alter Notch3 receptor processing and activation , 2006, Neuroreport.

[19]  G. van Ommen,et al.  Functional analysis of 114 exon-internal AONs for targeted DMD exon skipping: indication for steric hindrance of SR protein binding sites. , 2005, Oligonucleotides.

[20]  Tan Xiao-mei,et al.  CADASIL , 2005 .

[21]  B. Klonjkowski,et al.  Notch3 is required for arterial identity and maturation of vascular smooth muscle cells. , 2004, Genes & development.

[22]  M. Dichgans,et al.  CADASIL-associated Notch3 mutations have differential effects both on ligand binding and ligand-induced Notch3 receptor signaling through RBP-Jk. , 2004, Experimental cell research.

[23]  Lawrence J Marnett,et al.  A Novel Mechanism of Cyclooxygenase-2 Inhibition Involving Interactions with Ser-530 and Tyr-385* , 2003, Journal of Biological Chemistry.

[24]  U. Lendahl,et al.  A CADASIL-mutated Notch 3 receptor exhibits impaired intracellular trafficking and maturation but normal ligand-induced signaling , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[25]  M. Reiser,et al.  Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy: decrease in regional cerebral blood volume in hyperintense subcortical lesions inversely correlates with disability and cognitive performance. , 2001, AJNR. American journal of neuroradiology.

[26]  M. Dichgans,et al.  Reduced Cerebrovascular CO2 Reactivity in CADASIL: A Transcranial Doppler Sonography Study , 2001, Stroke.

[27]  G. Weinmaster,et al.  Fringe differentially modulates Jagged1 and Delta1 signalling through Notch1 and Notch2 , 2000, Nature Cell Biology.

[28]  F. Chapon,et al.  The ectodomain of the Notch3 receptor accumulates within the cerebrovasculature of CADASIL patients. , 2000, The Journal of clinical investigation.

[29]  J. Weissenbach,et al.  Strong clustering and stereotyped nature of Notch3 mutations in CADASIL patients , 1997, The Lancet.

[30]  J. Weissenbach,et al.  Notch3 mutations in CADASIL, a hereditary adult-onset condition causing stroke and dementia , 1996, Nature.

[31]  David W. Banner,et al.  The crystal structure of the complex of blood coagulation factor VIIa with soluble tissue factor , 1996, Nature.

[32]  U. Lendahl,et al.  The novel Notch homologue mouse Notch 3 lacks specific epidermal growth factor-repeats and is expressed in proliferating neuroepithelium , 1994, Mechanisms of Development.

[33]  G Vriend,et al.  WHAT IF: a molecular modeling and drug design program. , 1990, Journal of molecular graphics.

[34]  Annemieke Aartsma-Rus,et al.  Overview on applications of antisense-mediated exon skipping. , 2012, Methods in molecular biology.

[35]  D. Leys,et al.  Systemic vascular smooth muscle cell impairment in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy , 2004, Acta Neuropathologica.

[36]  K. Kosik,et al.  Distribution pattern of Notch3 mutations suggests a gain-of-function mechanism for CADASIL. , 2004, Genomics.