Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Zarife Sahenk is active.

Publication


Featured researches published by Zarife Sahenk.


Journal of Clinical Investigation | 2007

Interplay of IKK/NF-κB signaling in macrophages and myofibers promotes muscle degeneration in Duchenne muscular dystrophy

Swarnali Acharyya; S. Armando Villalta; Nadine Bakkar; Tepmanas Bupha-Intr; Paul M. L. Janssen; Micheal Carathers; Zhi-Wei Li; Amer A. Beg; Sankar Ghosh; Zarife Sahenk; Michael Weinstein; Katherine L. Gardner; Jill A. Rafael-Fortney; Michael Karin; James G. Tidball; Albert S. Baldwin; Denis C. Guttridge

Duchenne muscular dystrophy (DMD) is a lethal X-linked disorder associated with dystrophin deficiency that results in chronic inflammation and severe skeletal muscle degeneration. In DMD mouse models and patients, we find that IkappaB kinase/NF-kappaB (IKK/NF-kappaB) signaling is persistently elevated in immune cells and regenerative muscle fibers. Ablation of 1 allele of the p65 subunit of NF-kappaB was sufficient to improve pathology in mdx mice, a model of DMD. In addition, conditional deletion of IKKbeta in mdx mice elucidated that NF-kappaB functions in activated macrophages to promote inflammation and muscle necrosis and in skeletal muscle fibers to limit regeneration through the inhibition of muscle progenitor cells. Furthermore, specific pharmacological inhibition of IKK resulted in improved pathology and muscle function in mdx mice. Collectively, these results underscore the critical role of NF-kappaB in the progression of muscular dystrophy and suggest the IKK/NF-kappaB signaling pathway as a potential therapeutic target for DMD.


Annals of Neurology | 2010

Gentamicin-induced readthrough of stop codons in duchenne muscular dystrophy

Vinod Malik; L. Rodino-Klapac; Laurence Viollet; Cheryl Wall; Wendy M. King; Roula al-Dahhak; Sarah Lewis; C. Shilling; Janaiah Kota; Carmen Serrano-Munuera; John R. Hayes; John D. Mahan; Katherine J. Campbell; Brenda Banwell; Majed Dasouki; Victoria Watts; Kumaraswamy Sivakumar; Ricardo Bien-Willner; Kevin M. Flanigan; Zarife Sahenk; Richard J. Barohn; Christopher M. Walker

The objective of this study was to establish the feasibility of long‐term gentamicin dosing to achieve stop codon readthrough and produce full‐length dystrophin. Mutation suppression of stop codons, successfully achieved in the mdx mouse using gentamicin, represents an important evolving treatment strategy in Duchenne muscular dystrophy (DMD).


Neurology | 1995

Inclusion body myositis: Explanation for poor response to immunosuppressive therapy

Richard J. Barohn; Anthony A. Amato; Zarife Sahenk; John T. Kissel

We treated eight patients who had inclusion body myositis (IBM) with oral prednisone therapy, and we performed muscle biopsies before and after treatment. We documented the patients clinical response to therapy and changes in serum CK. Although the serum CK level fell, muscle strength worsened after prednisone treatment. In addition, while inflammation decreased in the muscle biopsy specimens, the number of vacuolated and amyloid-positive fibers increased after oral prednisone therapy. These observations indicate that the inflammatory response in IBM may play a secondary role in the pathogenesis of IBM. The unique findings of intracellular amyloid deposits and rimmed vacuoles distinguishing IBM from other inflammatory myopathies, and recognition that suppression of inflammation has no effect on the clinical course, suggest that IBM may represent a degenerative muscle disorder.


Neurology | 1993

Polyneuropathy complicating bone marrow and solid organ transplantation

Anthony A. Amato; Richard J. Barohn; Zarife Sahenk; P. J. Tutschka

We report a generalized polyneuropathy coincident with the occurrence of graft-versus-host disease in four patients undergoing bone marrow transplantation and accompanying solid organ rejection (heart and kidney) in two patients. The neuropathy affected proximal and distal muscles, demonstrated hyporeflexia or areflexia, and usually had elevated CSF protein. Electrophysiologic studies did not meet strict criteria for demyelination. The signs of neuropathy improved after immunosuppressive treatment or simultaneous to the resolution of graft-versus-host disease or tissue rejection. Polyneuropathy must be considered as a potential complication of tissue transplantation.


Neuroscience Letters | 2012

Gene Therapy for Muscular Dystrophy: Lessons Learned and Path Forward

L. Rodino-Klapac; Zarife Sahenk; Vinod Malik; Brian K. Kaspar; Christopher M. Walker; K. Reed Clark

Our Translational Gene Therapy Center has used small molecules for exon skipping and mutation suppression and gene transfer to replace or provide surrogate genes as tools for molecular-based approaches for the treatment of muscular dystrophies. Exon skipping is targeted at the pre-mRNA level allowing one or more exons to be omitted to restore the reading frame. In Duchenne Muscular Dystrophy (DMD), clinical trials have been performed with two different oligomers, a 2O-methyl-ribo-oligonucleoside-phosphorothioate (2OMe) and a phosphorodiamidate morpholino (PMO). Both have demonstrated early evidence of efficacy. A second molecular approach involves suppression of stop codons to promote readthrough of the DMD gene. We have been able to establish proof of principle for mutation suppression using the aminoglycoside, gentamicin. A safer, orally administered, alternative agent referred to as Ataluren (PTC124) has been used in clinical trials and is currently under consideration for approval by the FDA. Using a gene therapy approach, we have completed two trials and have initiated a third. For DMD, we used a mini-dystrophin transferred in adeno-associated virus (AAV). In this trial an immune response was seen directed against transgene product, a quite unexpected outcome that will help guide further studies. For limb girdle muscular dystrophy 2D (alpha-sarcoglycan deficiency), the transgene was again transferred using AAV but in this study, a muscle specific creatine kinase promoter controlled gene expression that persisted for six months. A third gene therapy trial has been initiated with transfer of the follistatin gene in AAV directly to the quadriceps muscle. Two diseases with selective quadriceps muscle weakness are undergoing gene transfer including sporadic inclusion body myositis (sIBM) and Becker muscular dystrophy (BMD). Increasing the size and strength of the muscle is the goal of this study. Most importantly, no adverse events have been encountered in any of these clinical trials.


Molecular Therapy | 2010

Persistent Expression of FLAG-tagged Micro dystrophin in Nonhuman Primates Following Intramuscular and Vascular Delivery

L. Rodino-Klapac; Chrystal L. Montgomery; William G. Bremer; K. Shontz; Vinod Malik; Nancy Davis; Spencer Sprinkle; Katherine J. Campbell; Zarife Sahenk; K. Reed Clark; Christopher M. Walker; Louis G. Chicoine

Animal models for Duchenne muscular dystrophy (DMD) have species limitations related to assessing function, immune response, and distribution of micro- or mini-dystrophins. Nonhuman primates (NHPs) provide the ideal model to optimize vector delivery across a vascular barrier and provide accurate dose estimates for widespread transduction. To address vascular delivery and dosing in rhesus macaques, we have generated a fusion construct that encodes an eight amino-acid FLAG epitope at the C-terminus of micro-dystrophin to facilitate translational studies targeting DMD. Intramuscular (IM) injection of AAV8.MCK.micro-dys.FLAG in the tibialis anterior (TA) of macaques demonstrated robust gene expression, with muscle transduction (50–79%) persisting for up to 5 months. Success by IM injection was followed by targeted vascular delivery studies using a fluoroscopy-guided catheter threaded through the femoral artery. Three months after gene transfer, >80% of muscle fibers showed gene expression in the targeted muscle. No cellular immune response to AAV8 capsid, micro-dystrophin, or the FLAG tag was detected by interferon-γ (IFN-γ) enzyme-linked immunosorbent spot (ELISpot) at any time point with either route. In summary, an epitope-tagged micro-dystrophin cassette enhances the ability to evaluate site-specific localization and distribution of gene expression in the NHP in preparation for vascular delivery clinical trials.


Molecular Therapy | 2014

Plasmapheresis Eliminates the Negative Impact of AAV Antibodies on Microdystrophin Gene Expression Following Vascular Delivery

Louis G. Chicoine; Chrystal L. Montgomery; William G. Bremer; Kimberly M. Shontz; Danielle A. Griffin; Kristin N. Heller; Sarah Lewis; Vinod Malik; William Grose; Cj Shilling; Katherine J. Campbell; Thomas J. Preston; Brian D. Coley; Pt Martin; Christopher M. Walker; Kelly Reed Clark; Zarife Sahenk; L. Rodino-Klapac

Duchenne muscular dystrophy is a monogenic disease potentially treatable by gene replacement. Use of recombinant adeno-associated virus (AAV) will ultimately require a vascular approach to broadly transduce muscle cells. We tested the impact of preexisting AAV antibodies on microdystrophin expression following vascular delivery to nonhuman primates. Rhesus macaques were treated by isolated limb perfusion using a fluoroscopically guided catheter. In addition to serostatus stratification, the animals were placed into one of the three immune suppression groups: no immune suppression, prednisone, and triple immune suppression (prednisone, tacrolimus, and mycophenolate mofetil). The animals were analyzed for transgene expression at 3 or 6 months. Microdystrophin expression was visualized in AAV, rhesus serotype 74 sero-negative animals (mean: 48.0u2009±u200920.8%) that was attenuated in sero-positive animals (19.6u2009±u200918.7%). Immunosuppression did not affect transgene expression. Importantly, removal of AAV binding antibodies by plasmapheresis in AAV sero-positive animals resulted in high-level transduction (60.8u2009±u200918.0%), which is comparable with that of AAV sero-negative animals (53.7u2009±u20097.6%), whereas non-pheresed sero-positive animals demonstrated significantly lower transduction levels (10.1u2009±u20096.0%). These data support the hypothesis that removal of AAV binding antibodies by plasmapheresis permits successful and sustained gene transfer in the presence of preexisting immunity (natural infection) to AAV.


Current Neurology and Neuroscience Reports | 2013

Update on the Treatment of Duchenne Muscular Dystrophy

L. Rodino-Klapac; Zarife Sahenk

Duchenne muscular dystrophy is the most severe childhood form of muscular dystrophy caused by mutations in the gene responsible for dystrophin production. There is no cure, and treatment is limited to glucocorticoids that prolong ambulation and drugs to treat the cardiomyopathy. Multiple treatment strategies are under investigation and have shown promise for Duchenne muscular dystrophy. Use of molecular-based therapies that replace or correct the missing or nonfunctional dystrophin protein has gained momentum. These strategies include gene replacement with adeno-associated virus, exon skipping with antisense oligonucleotides, and mutation suppression with compounds that “read through” stop codon mutations. Other strategies include cell therapy and surrogate gene products to compensate for the loss of dystrophin. All of these approaches are discussed in this review, with particular emphasis on the most recent advances made in each therapeutic discipline. The advantages of each approach and challenges in translation are outlined in detail. Individually or in combination, all of these therapeutic strategies hold great promise for treatment of this devastating childhood disease.


Molecular Therapy | 2014

Vascular Delivery of rAAVrh74.MCK.GALGT2 to the Gastrocnemius Muscle of the Rhesus Macaque Stimulates the Expression of Dystrophin and Laminin α2 Surrogates

Louis G. Chicoine; L. Rodino-Klapac; Guohong Shao; Rui Xu; William G. Bremer; Marybeth Camboni; Bethannie Golden; Chrystal L. Montgomery; Kimberly M. Shontz; Kristin N. Heller; Danielle A. Griffin; Sarah Lewis; Brian D. Coley; Christopher M. Walker; K. Reed Clark; Zarife Sahenk; Pt Martin

Overexpression of GALGT2 in skeletal muscle can stimulate the glycosylation of α dystroglycan and the upregulation of normally synaptic dystroglycan-binding proteins, some of which are dystrophin and laminin α2 surrogates known to be therapeutic for several forms of muscular dystrophy. This article describes the vascular delivery of GALGT2 gene therapy in a large animal model, the rhesus macaque. Recombinant adeno-associated virus, rhesus serotype 74 (rAAVrh74), was used to deliver GALGT2 via the femoral artery to the gastrocnemius muscle using an isolated focal limb perfusion method. GALGT2 expression averaged 44u2009±u20094% of myofibers after treatment in macaques with low preexisting anti-rAAVrh74 serum antibodies, and expression was reduced to 9u2009±u20094% of myofibers in macaques with high preexisting rAAVrh74 immunity (P < 0.001; n = 12 per group). This was the case regardless of the addition of immunosuppressants, including prednisolone, tacrolimus, and mycophenolate mofetil. GALGT2-treated macaque muscles showed increased glycosylation of α dystroglycan and increased expression of dystrophin and laminin α2 surrogate proteins, including utrophin, plectin1, agrin, and laminin α5. These experiments demonstrate successful transduction of rhesus macaque muscle with rAAVrh74.MCK.GALGT2 after vascular delivery and induction of molecular changes thought to be therapeutic in several forms of muscular dystrophy.


Molecular Genetics and Metabolism | 2013

Molecular and clinical characterization of the myopathic form of mitochondrial DNA depletion syndrome caused by mutations in the thymidine kinase (TK2) gene

Sirisak Chanprasert; Jing Wang; Shao Wen Weng; Gregory M. Enns; Daniel R. Boué; Brenda Wong; Deborah Perry; Zarife Sahenk; William J. Craigen; Francisco J Climent Alcala; Juan M. Pascual; Serge Melançon; Victor Wei Zhang; Fernando Scaglia; Lee-Jun C. Wong

Mitochondrial DNA (mtDNA) depletion syndromes (MDSs) are a clinically and molecularly heterogeneous group of mitochondrial cytopathies characterized by severe mtDNA copy number reduction in affected tissues. Clinically, MDSs are mainly categorized as myopathic, encephalomyopathic, hepatocerebral, or multi-systemic forms. To date, the myopathic form of MDS is mainly caused by mutations in the TK2 gene, which encodes thymidine kinase 2, the first and rate limiting step enzyme in the phosphorylation of pyrimidine nucleosides. We analyzed 9 unrelated families with 11 affected subjects exhibiting the myopathic form of MDS, by sequencing the TK2 gene. Twelve mutations including 4 novel mutations were detected in 9 families. Skeletal muscle specimens were available from 7 out of 11 subjects. Respiratory chain enzymatic activities in skeletal muscle were measured in 6 subjects, and enzymatic activities were reduced in 3 subjects. Quantitative analysis of mtDNA content in skeletal muscle was performed in 5 subjects, and marked mtDNA content reduction was observed in each. In addition, we outline the molecular and clinical characteristics of this syndrome in a total of 52 patients including those previously reported, and a total of 36 TK2 mutations are summarized. Clinically, hypotonia and proximal muscle weakness are the major phenotypes present in all subjects. In summary, our study expands the molecular and clinical spectrum associated with TK2 deficiency.

Collaboration


Dive into the Zarife Sahenk's collaboration.

Top Co-Authors

Avatar

L. Rodino-Klapac

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Sarah Lewis

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Vinod Malik

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Kevin M. Flanigan

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Linda Lowes

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Lindsay Alfano

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

C. Shilling

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

K. Berry

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

K. Shontz

Nationwide Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge