1
|
Stephens JQ, Blas-Machado U, Sherrill C, Caudell D, Kock N, Davis AM, Whitfield JM, Hart B, Kavanagh K. Male mice treated with combined anti-fibrotic therapeutics, IPW5371 and tadalafil, are predisposed to adverse cardiovascular events. Front Pharmacol 2025; 16:1537494. [PMID: 40242438 PMCID: PMC12000068 DOI: 10.3389/fphar.2025.1537494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/17/2025] [Indexed: 04/18/2025] Open
Abstract
Fibrosis is a pathological process with few therapeutic options. Experimental molecules are being developed to counteract the fibrotic effects through TGFβ receptor inhibition. Additionally, phosphodiesterase 5 (PDE5) inhibitors also have anti-fibrotic effects; however, the mechanism of action remains unresolved. IPW5371 is an example of an experimental TGFβ-mediated anti-fibrotic compound, and tadalafil is an example of a PDE5 inhibitor. Irradiation increases the frequency of fibrotic lesions, driven by the activation of the TGFβ pathway. We hypothesized that the TGFβ receptor and PDE5 inhibitor agents would be additive in their ability to prevent fibrosis development in tissues in a sub-lethal whole-body irradiation mouse model. However, the combined use of anti-fibrotic agents, tadalafil and IPW5371, caused increased male mouse mortality associated with ascending and thoracic aortic rupture compared to mice that only received one of the drugs. Following histopathological analysis of the mouse hearts, we also observed that irradiation protected against lesions caused by the combination therapy as non-irradiated male mice had significantly worse outcomes as compared to irradiated male mice, substantiating the drug-drug interaction independent of the radiation effects. This important drug interaction needs further investigation as these agents are developed for anti-fibrosis therapy, and PDE5 inhibitors are commonly prescribed to male patients.
Collapse
Affiliation(s)
- Jazz Q. Stephens
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC, United States
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
- Department of Population Health and Pathobiology, North Carolina College of Veterinary Medicine, Raleigh, NC, United States
- Division of Comparative Medicine and Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Uriel Blas-Machado
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
- StageBio, Mt. Jackson, VA, United States
| | - Chrissy Sherrill
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - David Caudell
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Nancy Kock
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Ashley M. Davis
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Jordyn M. Whitfield
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC, United States
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Barry Hart
- Innovation Pathways, Palo Alto, CA, United States
| | - Kylie Kavanagh
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC, United States
- College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
2
|
Curtis L. Erectile dysfunction drugs are essential and probably life-saving and should be provided to all men who need them just as birth control should be provided to all women who need them. Int J Impot Res 2024; 36:537-538. [PMID: 37443301 DOI: 10.1038/s41443-023-00736-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023]
Affiliation(s)
- Luke Curtis
- East Carolina University, Hazelwood, MO, USA.
| |
Collapse
|
3
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarise the recent developments in trial readiness, natural history studies, and interventional clinical trials for Becker muscular dystrophy (BMD). RECENT FINDINGS As several treatment concepts have claimed to convert patients with Duchenne muscular dystrophy (DMD) into a BMD phenotype, BMD itself has moved into the focus of clinical research. Natural history studies have helped to better characterize patients with BMD and the disease is now a target for interventional trials. In parallel, there have been advances in diagnostics and in the development of preclinical models. SUMMARY Despite increased collaborative efforts to improve trial readiness amongst patients with BMD, there is still a lack of long-term natural history data, and the broad spectrum of disease severity remains a challenge for well designed clinical trials.
Collapse
Affiliation(s)
- Volker Straub
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | |
Collapse
|
4
|
Samidurai A, Xi L, Das A, Kukreja RC. Beyond Erectile Dysfunction: cGMP-Specific Phosphodiesterase 5 Inhibitors for Other Clinical Disorders. Annu Rev Pharmacol Toxicol 2023; 63:585-615. [PMID: 36206989 DOI: 10.1146/annurev-pharmtox-040122-034745] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cyclic guanosine monophosphate (cGMP), an important intracellular second messenger, mediates cellular functional responses in all vital organs. Phosphodiesterase 5 (PDE5) is one of the 11 members of the cyclic nucleotide phosphodiesterase (PDE) family that specifically targets cGMP generated by nitric oxide-driven activation of the soluble guanylyl cyclase. PDE5 inhibitors, including sildenafil and tadalafil, are widely used for the treatment of erectile dysfunction, pulmonary arterial hypertension, and certain urological disorders. Preclinical studies have shown promising effects of PDE5 inhibitors in the treatment of myocardial infarction, cardiac hypertrophy, heart failure, cancer and anticancer-drug-associated cardiotoxicity, diabetes, Duchenne muscular dystrophy, Alzheimer's disease, and other aging-related conditions. Many clinical trials with PDE5 inhibitors have focused on the potential cardiovascular, anticancer, and neurological benefits. In this review, we provide an overview of the current state of knowledge on PDE5 inhibitors and their potential therapeutic indications for various clinical disorders beyond erectile dysfunction.
Collapse
Affiliation(s)
- Arun Samidurai
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA;
| | - Lei Xi
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA;
| | - Anindita Das
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA;
| | - Rakesh C Kukreja
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA;
| |
Collapse
|
5
|
Świątkowska‐Flis B, Zdolińska‐Malinowska I, Sługocka D, Boruczkowski D. The use of umbilical cord-derived mesenchymal stem cells in patients with muscular dystrophies: Results from compassionate use in real-life settings. Stem Cells Transl Med 2021; 10:1372-1383. [PMID: 34313400 PMCID: PMC8459640 DOI: 10.1002/sctm.21-0027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/10/2021] [Accepted: 06/16/2021] [Indexed: 12/15/2022] Open
Abstract
Muscular dystrophies are genetically determined progressive diseases with no cause-related treatment and limited supportive treatment. Although stem cells cannot resolve the underlying genetic conditions, their wide-ranging therapeutic properties may ameliorate the consequences of the involved mutations (oxidative stress, inflammation, mitochondrial dysfunction, necrosis). In this study, we administered advanced therapy medicinal product containing umbilical cord-derived mesenchymal stem cells (UC-MSCs) to 22 patients with muscular dystrophies. Patients received one to five intravenous and/or intrathecal injections per treatment course in up to two courses every 2 months. Four standard doses of 10, 20, 30, or 40 × 106 UC-MSCs per injection were used; the approximate dose per kilogram was 1 × 106 UC-MSCs. Muscle strength was measured with a set of CQ Dynamometer computerized force meters (CQ Elektronik System, Czernica, Poland). Statistical analysis of muscle strength in the whole group showed significant improvement in the right upper limb (+4.0 N); left hip straightening (+4.5 N) and adduction (+0.5 N); right hip straightening (+1.0 N), bending (+7.5 N), and adduction (+2.5 N); right knee straightening (+8.5 N); left shoulder revocation (+13.0 N), straightening (+5.5 N), and bending (+6.5 N); right shoulder adduction (+3.0 N), revocation (+10.5 N), and bending (+5 N); and right elbow straightening (+9.5 N); all these differences were statistically significant. In six patients (27.3%) these changes led to improvement in gait analysis or movement scale result. Only one patient experienced transient headache and lower back pain after the last administration. In conclusion, UC-MSC therapy may be considered as a therapeutic option for these patients.
Collapse
Affiliation(s)
- Beata Świątkowska‐Flis
- Polish Center of Cell Therapy and Immunotherapy in Częstochowa, CM KlaraCzęstochowaPoland
- Faculty of Health SciencesJan Długosz University of Humanities and Life SciencesCzęstochowaPoland
| | | | - Dominika Sługocka
- Polish Center of Cell Therapy and Immunotherapy in Częstochowa, CM KlaraCzęstochowaPoland
| | | |
Collapse
|
6
|
Assessing the Use of the sGC Stimulator BAY-747, as a Potential Treatment for Duchenne Muscular Dystrophy. Int J Mol Sci 2021; 22:ijms22158016. [PMID: 34360780 PMCID: PMC8347633 DOI: 10.3390/ijms22158016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/09/2021] [Accepted: 07/17/2021] [Indexed: 12/12/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe and progressive muscle wasting disorder, affecting one in 3500 to 5000 boys worldwide. The NO-sGC-cGMP pathway plays an important role in skeletal muscle function, primarily by improving blood flow and oxygen supply to the muscles during exercise. In fact, PDE5 inhibitors have previously been investigated as a potential therapy for DMD, however, a large-scale Phase III clinical trial did not meet its primary endpoint. Since the efficacy of PDE5i is dependent on sufficient endogenous NO production, which might be impaired in DMD, we investigated if NO-independent sGC stimulators, could have therapeutic benefits in a mouse model of DMD. Male mdx/mTRG2 mice aged six weeks were given food supplemented with the sGC stimulator, BAY-747 (150 mg/kg of food) or food alone (untreated) ad libitum for 16 weeks. Untreated C57BL6/J mice were used as wild type (WT) controls. Assessments of the four-limb hang, grip strength, running wheel and serum creatine kinase (CK) levels showed that mdx/mTRG2 mice had significantly reduced skeletal muscle function and severe muscle damage compared to WT mice. Treatment with BAY-747 improved grip strength and running speed, and these mice also had reduced CK levels compared to untreated mdx/mTRG2 mice. We also observed increased inflammation and fibrosis in the skeletal muscle of mdx/mTRG2 mice compared to WT. While gene expression of pro-inflammatory cytokines and some pro-fibrotic markers in the skeletal muscle was reduced following BAY-747 treatment, there was no reduction in infiltration of myeloid immune cells nor collagen deposition. In conclusion, treatment with BAY-747 significantly improves several functional and pathological parameters of the skeletal muscle in mdx/mTRG2 mice. However, the effect size was moderate and therefore, more studies are needed to fully understand the potential treatment benefit of sGC stimulators in DMD.
Collapse
|
7
|
Baligand C, Hirschler L, Veeger TTJ, Václavů L, Franklin SL, van Osch MJP, Kan HE. A split-label design for simultaneous measurements of perfusion in distant slices by pulsed arterial spin labeling. Magn Reson Med 2021; 86:2441-2453. [PMID: 34105189 PMCID: PMC8596809 DOI: 10.1002/mrm.28879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/16/2022]
Abstract
Purpose Multislice arterial spin labeling (ASL) MRI acquisitions are currently challenging in skeletal muscle because of long transit times, translating into low‐perfusion SNR in distal slices when large spatial coverage is required. However, fiber type and oxidative capacity vary along the length of healthy muscles, calling for multislice acquisitions in clinical studies. We propose a new variant of flow alternating inversion recovery (FAIR) that generates sufficient ASL signal to monitor exercise‐induced perfusion changes in muscle in two distant slices. Methods Label around and between two 7‐cm distant slices was created by applying the presaturation/postsaturation and selective inversion modules selectively to each slice (split‐label multislice FAIR). Images were acquired using simultaneous multislice EPI. We validated our approach in the brain to take advantage of the high resting‐state perfusion, and applied it in the lower leg muscle during and after exercise, interleaved with a single‐slice FAIR as a reference. Results We show that standard multislice FAIR leads to an underestimation of perfusion, while the proposed split‐label multislice approach shows good agreement with separate single‐slice FAIR acquisitions in brain, as well as in muscle following exercise. Conclusion Split‐label FAIR allows measuring muscle perfusion in two distant slices simultaneously without losing sensitivity in the distal slice.
Collapse
Affiliation(s)
- Celine Baligand
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Lydiane Hirschler
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Thom T J Veeger
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Lena Václavů
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Suzanne L Franklin
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands.,Center for image sciences, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Matthias J P van Osch
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands.,Leiden Institute for Brain and Cognition, Leiden University, Leiden, the Netherlands
| | - Hermien E Kan
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands.,Duchenne Center, Leiden, the Netherlands
| |
Collapse
|
8
|
Lindsay A, Kemp B, Larson AA, Baumann CW, McCourt PM, Holm J, Karachunski P, Lowe DA, Ervasti JM. Tetrahydrobiopterin synthesis and metabolism is impaired in dystrophin-deficient mdx mice and humans. Acta Physiol (Oxf) 2021; 231:e13627. [PMID: 33580591 DOI: 10.1111/apha.13627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/20/2022]
Abstract
AIM Loss of dystrophin causes oxidative stress and affects nitric oxide synthase-mediated vascular function in striated muscle. Because tetrahydrobiopterin is an antioxidant and co-factor for nitric oxide synthase, we tested the hypothesis that tetrahydrobiopterin would be low in mdx mice and humans deficient for dystrophin. METHODS Tetrahydrobiopterin and its metabolites were measured at rest and in response to exercise in Duchenne and Becker muscular dystrophy patients, age-matched male controls as well as wild-type, mdx and mdx mice transgenically overexpressing skeletal muscle-specific dystrophins. Mdx mice were also supplemented with tetrahydrobiopterin and pathophysiology was assessed. RESULTS Duchenne muscular dystrophy patients had lower urinary dihydrobiopterin + tetrahydrobiopterin/specific gravity1.020 compared to unaffected age-matched males and Becker muscular dystrophy patients. Mdx mice had low urinary and skeletal muscle dihydrobiopterin + tetrahydrobiopterin compared to wild-type mice. Overexpression of dystrophins that localize neuronal nitric oxide synthase restored dihydrobiopterin + tetrahydrobiopterin in mdx mice to wild-type levels while utrophin overexpression did not. Mdx mice and Duchenne muscular dystrophy patients did not increase tetrahydrobiopterin during exercise and in mdx mice tetrahydrobiopterin deficiency was likely because of lower levels of sepiapterin reductase in skeletal muscle. Tetrahydrobiopterin supplementation improved skeletal muscle strength, resistance to fatiguing and injurious contractions in vivo, increased utrophin and capillary density of skeletal muscle and lowered cardiac muscle fibrosis and left ventricular wall thickness in mdx mice. CONCLUSION These data demonstrate that impaired tetrahydrobiopterin synthesis is associated with dystrophin loss and treatment with tetrahydrobiopterin improves striated muscle histopathology and skeletal muscle function in mdx mice.
Collapse
Affiliation(s)
- Angus Lindsay
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Bailey Kemp
- Lillehei Heart Institute, Cancer and Cardiovascular Research Center, University of Minnesota, Minneapolis, MN, USA
| | - Alexie A Larson
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Cory W Baumann
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Preston M McCourt
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - John Holm
- Lillehei Heart Institute, Cancer and Cardiovascular Research Center, University of Minnesota, Minneapolis, MN, USA
| | - Peter Karachunski
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | - Dawn A Lowe
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
9
|
Verma M, Shimizu-Motohashi Y, Asakura Y, Ennen JP, Bosco J, Zhou Z, Fong GH, Josiah S, Keefe D, Asakura A. Inhibition of FLT1 ameliorates muscular dystrophy phenotype by increased vasculature in a mouse model of Duchenne muscular dystrophy. PLoS Genet 2019; 15:e1008468. [PMID: 31877123 PMCID: PMC6932757 DOI: 10.1371/journal.pgen.1008468] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 10/08/2019] [Indexed: 12/17/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive genetic disease in which the dystrophin coding for a membrane stabilizing protein is mutated. Recently, the vasculature has also shown to be perturbed in DMD and DMD model mdx mice. Recent DMD transcriptomics revealed the defects were correlated to a vascular endothelial growth factor (VEGF) signaling pathway. To reveal the relationship between DMD and VEGF signaling, mdx mice were crossed with constitutive (CAGCreERTM:Flt1LoxP/LoxP) and endothelial cell-specific conditional gene knockout mice (Cdh5CreERT2:Flt1LoxP/LoxP) for Flt1 (VEGFR1) which is a decoy receptor for VEGF. Here, we showed that while constitutive deletion of Flt1 is detrimental to the skeletal muscle function, endothelial cell-specific Flt1 deletion resulted in increased vascular density, increased satellite cell number and improvement in the DMD-associated phenotype in the mdx mice. These decreases in pathology, including improved muscle histology and function, were recapitulated in mdx mice given anti-FLT1 peptides or monoclonal antibodies, which blocked VEGF-FLT1 binding. The histological and functional improvement of dystrophic muscle by FLT1 blockade provides a novel pharmacological strategy for the potential treatment of DMD. Duchenne muscular dystrophy (DMD) is a devastating muscle disease affecting one in 5,000 newborn males, in which the gene encoding the dystrophin protein is mutated. It is a progressive muscle degenerative disease with death by either respiratory insufficiency or cardiac failure in their 20s. Recently, the vasculature has also shown to be perturbed in DMD and DMD model mdx mice with the defects correlated to a vascular endothelial growth factor (VEGF) signaling pathway. To reveal the relationship between DMD and VEGF signaling, mdx mice were crossed with mice carrying mutated a decoy receptor gene (Flt1) for VEGF. Here, we showed that Flt1 deletion resulted in increased vascular density and improvement in the DMD-associated skeletal muscle phenotype in the mdx mice. These decreases in pathology, including improved muscle histology and function, were recapitulated in mdx mice given anti-FLT1 peptides or monoclonal antibodies, which blocked VEGF-FLT1 binding. The histological and functional improvement of dystrophic muscle by FLT1 blockade provides a novel pharmacological strategy for the potential treatment of DMD.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/pharmacology
- Disease Models, Animal
- Endothelial Cells/metabolism
- Gene Knockout Techniques
- Male
- Mice
- Mice, Inbred mdx
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiopathology
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/physiopathology
- Organ Specificity
- Peptides/administration & dosage
- Peptides/pharmacology
- Signal Transduction/drug effects
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors
- Vascular Endothelial Growth Factor Receptor-1/genetics
Collapse
Affiliation(s)
- Mayank Verma
- Medical Scientist Training Program, University of Minnesota Medical School, Minneapolis, MN, United States of America
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, United States of America
- Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, United States of America
- Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Yuko Shimizu-Motohashi
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, United States of America
- Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, United States of America
- Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Yoko Asakura
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, United States of America
- Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, United States of America
- Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - James P. Ennen
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, United States of America
- Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, United States of America
- Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Jennifer Bosco
- Shire Human Genetic Therapies, Inc., a member of the Takeda group of companies, Lexington, MA, United States of America
| | - Zhiwei Zhou
- Shire Human Genetic Therapies, Inc., a member of the Takeda group of companies, Lexington, MA, United States of America
| | - Guo-Hua Fong
- Center for Vascular Biology, University of Connecticut Health Center, University of Connecticut School of Medicine, Farmington, CT, United States of America
| | - Serene Josiah
- Shire Human Genetic Therapies, Inc., a member of the Takeda group of companies, Lexington, MA, United States of America
| | - Dennis Keefe
- Shire Human Genetic Therapies, Inc., a member of the Takeda group of companies, Lexington, MA, United States of America
| | - Atsushi Asakura
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, United States of America
- Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, United States of America
- Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, United States of America
- * E-mail:
| |
Collapse
|
10
|
Dietz AR, Connolly A, Dori A, Zaidman CM. Intramuscular blood flow in Duchenne and Becker Muscular Dystrophy: Quantitative power Doppler sonography relates to disease severity. Clin Neurophysiol 2019; 131:1-5. [PMID: 31751835 DOI: 10.1016/j.clinph.2019.09.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 08/02/2019] [Accepted: 09/11/2019] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Absent or truncated dystrophin in Duchenne (DMD) and Becker (BMD) muscular dystrophies results in impaired vasodilatory pathways and exercise induced muscle ischemia. Here, we used power Doppler sonography to quantify changes in intramuscular blood flow immediately following exercise in boys with D/BMD. METHOD We quantified changes in intramuscular blood flow following exercise using power Doppler sonography in 14 boys with D/BMD and compared changes in muscle blood flow to disease severity and to historic controls. RESULT Post exercise blood flow change in the anterior forearm muscles is lower in (1) DMD (median 0.25%; range -0.47 to 2.19%) than BMD (2.46%; 2.02-3.38%, p < 0.05) and historical controls (6.59%; 2.16-12.40%, p < 0.01); (2) in non-ambulatory (0.04%; -0.47 to 0.10%) than ambulatory DMD boys (0.71%; 0.07-2.19%, p < 0.05); and (3) in muscle with higher echointensity (rs = -0.7253, p = 0.005). The tibialis anterior showed similar findings. We estimate that a single sample clinical trial would require 19 subjects to detect a doubling of blood flow to the anterior forearm after the intervention. CONCLUSION Post-exercise blood flow is reduced in D/BMD and relates to disease severity. SIGNIFICANCE Our protocol for quantifying post-exercise intramuscular blood flow is feasible for clinical trials in D/BMD.
Collapse
Affiliation(s)
- Alexander R Dietz
- Blue Sky Neurology, Englewood, CO, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne Connolly
- Department of Pediatrics, Division of Neurology, Nationwide Children's Hospital, Columbus OH, USA
| | - Amir Dori
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Department of Neurology, Talpiot Medical Leadership Program, Chaim Sheba Medical Center, Tel HaShomer, and Joseph Sagol Neuroscience Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Craig M Zaidman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
11
|
Baillie GS, Tejeda GS, Kelly MP. Therapeutic targeting of 3',5'-cyclic nucleotide phosphodiesterases: inhibition and beyond. Nat Rev Drug Discov 2019; 18:770-796. [PMID: 31388135 PMCID: PMC6773486 DOI: 10.1038/s41573-019-0033-4] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2019] [Indexed: 01/24/2023]
Abstract
Phosphodiesterases (PDEs), enzymes that degrade 3',5'-cyclic nucleotides, are being pursued as therapeutic targets for several diseases, including those affecting the nervous system, the cardiovascular system, fertility, immunity, cancer and metabolism. Clinical development programmes have focused exclusively on catalytic inhibition, which continues to be a strong focus of ongoing drug discovery efforts. However, emerging evidence supports novel strategies to therapeutically target PDE function, including enhancing catalytic activity, normalizing altered compartmentalization and modulating post-translational modifications, as well as the potential use of PDEs as disease biomarkers. Importantly, a more refined appreciation of the intramolecular mechanisms regulating PDE function and trafficking is emerging, making these pioneering drug discovery efforts tractable.
Collapse
Affiliation(s)
- George S Baillie
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| | - Gonzalo S Tejeda
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| | - Michy P Kelly
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA.
| |
Collapse
|
12
|
Boehler JF, Ricotti V, Gonzalez JP, Soustek-Kramer M, Such L, Brown KJ, Schneider JS, Morris CA. Membrane recruitment of nNOSµ in microdystrophin gene transfer to enhance durability. Neuromuscul Disord 2019; 29:735-741. [PMID: 31521486 DOI: 10.1016/j.nmd.2019.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/23/2019] [Accepted: 07/31/2019] [Indexed: 12/25/2022]
Abstract
Several gene transfer clinical trials are currently ongoing with the common aim of delivering a shortened version of dystrophin, termed a microdystrophin, for the treatment of Duchenne muscular dystrophy (DMD). However, one of the main differences between these trials is the microdystrophin protein produced following treatment. Each gene transfer product is based on different selections of dystrophin domain combinations to assemble microdystrophin transgenes that maintain functional dystrophin domains and fit within the packaging limits of an adeno-associated virus (AAV) vector. While domains involved in mechanical function, such as the actin-binding domain and β-dystroglycan binding domain, have been identified for many years and included in microdystrophin constructs, more recently the neuronal nitric oxide synthase (nNOS) domain has also been identified due to its role in enhancing nNOS membrane localization. As nNOS membrane localization has been established as an important requirement for prevention of functional ischemia in skeletal muscle, inclusion of the nNOS domain into a microdystrophin construct represents an important consideration. The aim of this mini review is to highlight what is currently known about the nNOS domain of dystrophin and to describe potential implications of this domain in a microdystrophin gene transfer clinical trial.
Collapse
Affiliation(s)
- Jessica F Boehler
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | - Valeria Ricotti
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | - J Patrick Gonzalez
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | | | - Lauren Such
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | - Kristy J Brown
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | - Joel S Schneider
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | - Carl A Morris
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States.
| |
Collapse
|
13
|
Meyers TA, Townsend D. Cardiac Pathophysiology and the Future of Cardiac Therapies in Duchenne Muscular Dystrophy. Int J Mol Sci 2019; 20:E4098. [PMID: 31443395 PMCID: PMC6747383 DOI: 10.3390/ijms20174098] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/12/2019] [Accepted: 08/19/2019] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating disease featuring skeletal muscle wasting, respiratory insufficiency, and cardiomyopathy. Historically, respiratory failure has been the leading cause of mortality in DMD, but recent improvements in symptomatic respiratory management have extended the life expectancy of DMD patients. With increased longevity, the clinical relevance of heart disease in DMD is growing, as virtually all DMD patients over 18 year of age display signs of cardiomyopathy. This review will focus on the pathophysiological basis of DMD in the heart and discuss the therapeutic approaches currently in use and those in development to treat dystrophic cardiomyopathy. The first section will describe the aspects of the DMD that result in the loss of cardiac tissue and accumulation of fibrosis. The second section will discuss cardiac small molecule therapies currently used to treat heart disease in DMD, with a focus on the evidence supporting the use of each drug in dystrophic patients. The final section will outline the strengths and limitations of approaches directed at correcting the genetic defect through dystrophin gene replacement, modification, or repair. There are several new and promising therapeutic approaches that may protect the dystrophic heart, but their limitations suggest that future management of dystrophic cardiomyopathy may benefit from combining gene-targeted therapies with small molecule therapies. Understanding the mechanistic basis of dystrophic heart disease and the effects of current and emerging therapies will be critical for their success in the treatment of patients with DMD.
Collapse
Affiliation(s)
- Tatyana A Meyers
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, MN 55455, USA
| | - DeWayne Townsend
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
14
|
Zhao J, Yang HT, Wasala L, Zhang K, Yue Y, Duan D, Lai Y. Dystrophin R16/17 protein therapy restores sarcolemmal nNOS in trans and improves muscle perfusion and function. Mol Med 2019; 25:31. [PMID: 31266455 PMCID: PMC6607532 DOI: 10.1186/s10020-019-0101-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 06/20/2019] [Indexed: 01/08/2023] Open
Abstract
Background Delocalization of neuronal nitric oxide synthase (nNOS) from the sarcolemma leads to functional muscle ischemia. This contributes to the pathogenesis in cachexia, aging and muscular dystrophy. Mutations in the gene encoding dystrophin result in Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD). In many BMD patients and DMD patients that have been converted to BMD by gene therapy, sarcolemmal nNOS is missing due to the lack of dystrophin nNOS-binding domain. Methods Dystrophin spectrin-like repeats 16 and 17 (R16/17) is the sarcolemmal nNOS localization domain. Here we explored whether R16/17 protein therapy can restore nNOS to the sarcolemma and prevent functional ischemia in transgenic mice which expressed an R16/17-deleted human micro-dystrophin gene in the dystrophic muscle. The palmitoylated R16/17.GFP fusion protein was conjugated to various cell-penetrating peptides and produced in the baculovirus-insect cell system. The best fusion protein was delivered to the transgenic mice and functional muscle ischemia was quantified. Results Among five candidate cell-penetrating peptides, the mutant HIV trans-acting activator of transcription (TAT) protein transduction domain (mTAT) was the best in transferring the R16/17.GFP protein to the muscle. Systemic delivery of the mTAT.R16/17.GFP protein to micro-dystrophin transgenic mice successfully restored sarcolemmal nNOS without inducing T cell infiltration. More importantly, R16/17 protein therapy effectively prevented treadmill challenge-induced force loss and improved muscle perfusion during contraction. Conclusions Our results suggest that R16/17 protein delivery is a highly promising therapy for muscle diseases involving sarcolemmal nNOS delocalizaton. Electronic supplementary material The online version of this article (10.1186/s10020-019-0101-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Junling Zhao
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Hsiao Tung Yang
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65212, USA
| | - Lakmini Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA. .,Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65212, USA. .,Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA. .,Department of Bioengineering, University of Missouri, Columbia, MO, 65212, USA.
| | - Yi Lai
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA.
| |
Collapse
|
15
|
Goldberg EJ, Schmidt CA, Green TD, Karnekar R, Yamaguchi DJ, Spangenberg EE, McClung JM. Temporal Association Between Ischemic Muscle Perfusion Recovery and the Restoration of Muscle Contractile Function After Hindlimb Ischemia. Front Physiol 2019; 10:804. [PMID: 31316393 PMCID: PMC6611152 DOI: 10.3389/fphys.2019.00804] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/06/2019] [Indexed: 12/15/2022] Open
Abstract
During incomplete skeletal muscle recovery from ischemia, such as that occurs with critical limb ischemia, the temporal relationship between recovery of muscle capillary perfusion and contractile function is poorly defined. We examined this relationship in BALB/cJ mice (N = 24) following unilateral hindlimb ischemia (HLI), which pre-clinically mimics the myopathy observed in critical limb ischemia patients. Specifically, we examined this relationship in two phenotypically distinct muscles (i.e., "oxidative" soleus - Sol and "glycolytic" extensor digitorum longus - EDL) 14- or 56-days after HLI. Although overall limb blood flow (LDPI) reached its' recovery peak (48% of control) by HLI d14, the capillary networks in both the Sol and EDL (whole mount confocal imaging) were disrupted and competent muscle capillary perfusion (perfused lectin+μm2/muscle μm2) remained reduced. Interestingly, both Sol and EDL muscles recovered their distinct capillary structures and perfusion (Con Sol; 0.056 ± 0.02 lectin+μm2/muscle μm2, and Con EDL; 0.039 ± 0.005 lectin+μm2/muscle μm2) by HLI d56 (Sol; 0.062 ± 0.011 lectin+μm2/muscle μm2 and EDL; 0.0035 ± 0.005 lectin+μm2/muscle μm2), despite no further improvement in limb blood flow (LDPI). Both muscles suffered severe myopathy, indicated by loss of dystrophin positive immunostaining and the absence of stimulation induced isometric force production at HLI d14. Dystrophin immunofluorescence returned at HLI d56, although neither myofiber CSA (μm2) nor isometric force production (58 and 28% sustained deficits, Sol and EDL, respectively) recovered completely in either muscle. In summary, we reveal that the temporal relationship between the restoration of muscle capillary perfusion and functional ischemic skeletal muscle regeneration favors competent muscle capillary perfusion recovery in BALB/c mice in a phenotypically non-distinct manner.
Collapse
Affiliation(s)
- Emma J Goldberg
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States.,East Carolina Diabetes and Obesity Institute, East Carolina Heart Institute, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Cameron A Schmidt
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States.,East Carolina Diabetes and Obesity Institute, East Carolina Heart Institute, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - T D Green
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States.,East Carolina Diabetes and Obesity Institute, East Carolina Heart Institute, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - R Karnekar
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States.,East Carolina Diabetes and Obesity Institute, East Carolina Heart Institute, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - D J Yamaguchi
- Department of Cardiovascular Sciences, Brody School of Medicine, East Carolina University, Greenville, NC, United States.,Division of Surgery, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - E E Spangenberg
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States.,East Carolina Diabetes and Obesity Institute, East Carolina Heart Institute, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Joseph M McClung
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States.,East Carolina Diabetes and Obesity Institute, East Carolina Heart Institute, Brody School of Medicine, East Carolina University, Greenville, NC, United States.,Department of Cardiovascular Sciences, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| |
Collapse
|
16
|
|
17
|
Balke JE, Zhang L, Percival JM. Neuronal nitric oxide synthase (nNOS) splice variant function: Insights into nitric oxide signaling from skeletal muscle. Nitric Oxide 2018; 82:35-47. [PMID: 30503614 DOI: 10.1016/j.niox.2018.11.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 11/19/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023]
Abstract
Defects in neuronal nitric oxide synthase (nNOS) splice variant localization and signaling in skeletal muscle are a firmly established pathogenic characteristic of many neuromuscular diseases, including Duchenne and Becker muscular dystrophy (DMD and BMD, respectively). Therefore, substantial efforts have been made to understand and therapeutically target skeletal muscle nNOS isoform signaling. The purpose of this review is to summarize recent salient advances in understanding of the regulation, targeting, and function of nNOSμ and nNOSβ splice variants in normal and dystrophic skeletal muscle, primarily using findings from mouse models. The first focus of this review is how the differential targeting of nNOS splice variants creates spatially and functionally distinct nitric oxide (NO) signaling compartments at the sarcolemma, Golgi complex, and cytoplasm. Particular attention is given to the functions of sarcolemmal nNOSμ and limitations of current nNOS knockout models. The second major focus is to review current understanding of cGMP-mediated nNOS signaling in skeletal muscle and its emergence as a therapeutic target in DMD and BMD. Accordingly, we address the preclinical and clinical successes and setbacks with the testing of phosphodiesterase 5 inhibitors to redress nNOS signaling defects in DMD and BMD. In summary, this review of nNOS function in normal and dystrophic muscle aims to advance understanding how the messenger NO is harnessed for cellular signaling from a skeletal muscle perspective.
Collapse
Affiliation(s)
- Jordan E Balke
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine Miami, Florida, 33101, USA
| | - Ling Zhang
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine Miami, Florida, 33101, USA
| | - Justin M Percival
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine Miami, Florida, 33101, USA.
| |
Collapse
|
18
|
Kodippili K, Hakim CH, Yang HT, Pan X, Yang NN, Laughlin MH, Terjung RL, Duan D. Nitric oxide-dependent attenuation of noradrenaline-induced vasoconstriction is impaired in the canine model of Duchenne muscular dystrophy. J Physiol 2018; 596:5199-5216. [PMID: 30152022 DOI: 10.1113/jp275672] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/08/2018] [Indexed: 12/31/2022] Open
Abstract
KEY POINTS We developed a novel method to study sympatholysis in dogs. We showed abolishment of sarcolemmal nNOS, and reduction of total nNOS and total eNOS in the canine Duchenne muscular dystrophy (DMD) model. We showed sympatholysis in dogs involving both nNOS-derived NO-dependent and NO-independent mechanisms. We showed that the loss of sarcolemmal nNOS compromised sympatholysis in the canine DMD model. We showed that NO-independent sympatholysis was not affected in the canine DMD model. ABSTRACT The absence of dystrophin in Duchenne muscular dystrophy (DMD) leads to the delocalization of neuronal nitric oxide synthase (nNOS) from the sarcolemma. Sarcolemmal nNOS plays an important role in sympatholysis, a process of attenuating reflex sympathetic vasoconstriction during exercise to ensure blood perfusion in working muscle. Delocalization of nNOS compromises sympatholysis resulting in functional ischaemia and muscle damage in DMD patients and mouse models. Little is known about the contribution of membrane-associated nNOS to blood flow regulation in dystrophin-deficient DMD dogs. We tested the hypothesis that the loss of sarcolemmal nNOS abolishes protective sympatholysis in contracting muscle of affected dogs. Haemodynamic responses to noradrenaline in the brachial artery were evaluated at rest and during contraction in the absence and presence of NOS inhibitors. We found sympatholysis was significantly compromised in DMD dogs, as well as in normal dogs treated with a selective nNOS inhibitor, suggesting that the absence of sarcolemmal nNOS underlies defective sympatholysis in the canine DMD model. Surprisingly, inhibition of all NOS isoforms did not completely abolish sympatholysis in normal dogs, suggesting sympatholysis in canine muscle also involves NO-independent mechanism(s). Our study established a foundation for using the dog model to test therapies aimed at restoring nNOS homeostasis in DMD.
Collapse
Affiliation(s)
- Kasun Kodippili
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA
| | - Chady H Hakim
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA.,National Center for Advancing Translational Sciences (NCATS), Bethesda, MD, USA
| | - Hsiao T Yang
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA.,Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA
| | - N Nora Yang
- National Center for Advancing Translational Sciences (NCATS), Bethesda, MD, USA
| | - Maurice H Laughlin
- Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Ronald L Terjung
- Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA.,Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA.,Department of Bioengineering, University of Missouri, Columbia, MO, USA
| |
Collapse
|
19
|
Dombernowsky NW, Ölmestig JNE, Witting N, Kruuse C. Role of neuronal nitric oxide synthase (nNOS) in Duchenne and Becker muscular dystrophies - Still a possible treatment modality? Neuromuscul Disord 2018; 28:914-926. [PMID: 30352768 DOI: 10.1016/j.nmd.2018.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/07/2018] [Accepted: 09/05/2018] [Indexed: 02/08/2023]
Abstract
Neuronal nitric oxide synthase (nNOS) is involved in nitric oxide (NO) production and suggested to play a crucial role in blood flow regulation of skeletal muscle. During activation of the muscle, NO helps attenuate the sympathetic vasoconstriction to accommodate increased metabolic demands, a phenomenon known as functional sympatholysis. In inherited myopathies such as the dystrophinopathies Duchenne and Becker muscle dystrophies (DMD and BMD), nNOS is lost from the sarcolemma. The loss of nNOS may cause functional ischemia contributing to skeletal and cardiac muscle cell injury. Effects of NO is augmented by inhibiting degradation of the second messenger cyclic guanosine monophosphate (cGMP) using sildenafil and tadalafil, both of which inhibit the enzyme phosphodiesterase 5 (PDE5). In animal models of DMD, PDE5-inhibitors prevent functional ischemia, reduce post-exercise skeletal muscle pathology and fatigue, show amelioration of cardiac muscle cell damage and increase cardiac performance. However, effect on clinical outcomes in DMD and BMD patients have been disappointing with minor effects on upper limb performance and none on ambulation. This review aims to summarize the current knowledge of nNOS function related to functional sympatholysis in skeletal muscle and studies on PDE5-inhibitor treatment in nNOS-deficient animal models and patients.
Collapse
Affiliation(s)
- Nanna W Dombernowsky
- Department of Neurology, Rigshospitalet Glostrup, University of Copenhagen, Denmark
| | - Joakim N E Ölmestig
- Department of Neurology, Neurovascular Research Unit, Herlev Gentofte Hospital, University of Copenhagen, Denmark
| | - Nanna Witting
- Department of Neurology, Rigshospitalet Glostrup, University of Copenhagen, Denmark
| | - Christina Kruuse
- Department of Neurology, Neurovascular Research Unit, Herlev Gentofte Hospital, University of Copenhagen, Denmark; PDE Research Group, Lundbeck Foundation Center for Neurovascular Research (LUCENS), Denmark.
| |
Collapse
|
20
|
Effects of single and combined metformin and L-citrulline supplementation on L-arginine-related pathways in Becker muscular dystrophy patients: possible biochemical and clinical implications. Amino Acids 2018; 50:1391-1406. [PMID: 30003335 DOI: 10.1007/s00726-018-2614-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/03/2018] [Indexed: 12/21/2022]
Abstract
The L-arginine/nitric oxide synthase (NOS) pathway is considered to be altered in muscular dystrophy such as Becker muscular dystrophy (BMD). We investigated two pharmacological options aimed to increase nitric oxide (NO) synthesis in 20 male BMD patients (age range 21-44 years): (1) supplementation with L-citrulline (3 × 5 g/d), the precursor of L-arginine which is the substrate of neuronal NO synthase (nNOS); and (2) treatment with the antidiabetic drug metformin (3 × 500 mg/d) which activates nNOS in human skeletal muscle. We also investigated the combined use of L-citrulline (3 × 5 g/d) and metformin (3 × 500 mg/d). Before and after treatment, we measured in serum and urine samples the concentration of amino acids and metabolites of L-arginine-related pathways and the oxidative stress biomarker malondialdehyde (MDA). Compared to healthy subjects, BMD patients have altered NOS, arginine:glycine amidinotransferase (AGAT) and guanidinoacetate methyltransferase (GAMT) pathways. Metformin treatment resulted in concentration decrease of arginine and MDA in serum, and of homoarginine (hArg) and guanidinoacetate (GAA) in serum and urine. L-Citrulline supplementation resulted in considerable increase of the concentrations of amino acids and creatinine in the serum, and in their urinary excretion rates. Combined use of metformin and L-citrulline attenuated the effects obtained from their single administrations. Metformin, L-citrulline or their combination did not alter serum nitrite and nitrate concentrations and their urinary excretion rates. In conclusion, metformin or L-citrulline supplementation to BMD patients results in remarkable antidromic changes of the AGAT and GAMT pathways. In combination, metformin and L-citrulline at the doses used in the present study seem to abolish the biochemical effects of the single drugs in slight favor of L-citrulline.
Collapse
|
21
|
Ramalho TC, de Castro AA, Tavares TS, Silva MC, Silva DR, Cesar PH, Santos LA, da Cunha EFF, Nepovimova E, Kuca K. Insights into the pharmaceuticals and mechanisms of neurological orphan diseases: Current Status and future expectations. Prog Neurobiol 2018; 169:135-157. [PMID: 29981392 DOI: 10.1016/j.pneurobio.2018.06.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 06/30/2018] [Indexed: 12/20/2022]
Abstract
Several rare or orphan diseases have been characterized that singly affect low numbers of people, but cumulatively reach ∼6%-10% of the population in Europe and in the United States. Human genetics has shown to be broadly effective when evaluating subjacent genetic defects such as orphan genetic diseases, but on the other hand, a modest progress has been achieved toward comprehending the molecular pathologies and designing new therapies. Chemical genetics, placed at the interface of chemistry and genetics, could be employed to understand the molecular mechanisms of subjacent illnesses and for the discovery of new remediation processes. This review debates current progress in chemical genetics, and how a variety of compounds and reaction mechanisms can be used to study and ultimately treat rare genetic diseases. We focus here on a study involving Amyotrophic lateral sclerosis (ALS), Duchenne Muscular Dystrophy (DMD), Spinal muscular atrophy (SMA) and Familial Amyloid Polyneuropathy (FAP), approaching different treatment methods and the reaction mechanisms of several compounds, trying to elucidate new routes capable of assisting in the treatment profile.
Collapse
Affiliation(s)
- Teodorico C Ramalho
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil; Center for Basic and Applied Research, Faculty of Informatics and Management, University of Hradec Kralove, Hradec Kralove, Czech Republic.
| | | | - Tássia S Tavares
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Maria C Silva
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Daniela R Silva
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Pedro H Cesar
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Lucas A Santos
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Elaine F F da Cunha
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic.
| |
Collapse
|
22
|
Lindberg U, Kruuse C, Witting N, Jørgensen SL, Vissing J, Rostrup E, Larsson HBW. Altered somatosensory neurovascular response in patients with Becker muscular dystrophy. Brain Behav 2018; 8:e00985. [PMID: 30106246 PMCID: PMC5991560 DOI: 10.1002/brb3.985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 02/12/2018] [Accepted: 03/31/2018] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Patients with dystrophinopathies show low levels of neuronal nitric oxide synthase (nNOS), due to reduced or absent dystrophin expression, as nNOS is attached to the dystrophin-associated protein complex. Deficient nNOS function leads to functional ischemia during muscle activity. Dystrophin-like proteins with nNOS attached have also been identified in the brain. This suggests that a mechanism of cerebral functional ischemia with attenuation of normal activation-related vascular response may cause changes in brain function. METHODS The aim of this study was to investigate whether the brain response of patients with Becker muscular dystrophy (BMD) is dysfunctional compared to that of healthy controls. To investigate a potential change in brain activation response in patients with BMD, median nerve somatosensory evoked stimulation, with stimulation durations of 2, 4, and 10 s, was performed while recording electroencephalography and blood oxygen level-dependent (BOLD) functional magnetic resonance imaging. RESULTS Results in 14 male patients with BMD (36.2 ± 9.9 years) were compared with those of 10 healthy controls (34.4 ± 10.9 years). Compared to controls, the patients with BMD showed sustained cortical electrical activity and a significant smaller BOLD activation in contralateral primary somatosensory cortex and bilaterally in secondary somatosensory cortex. In addition, significant activation differences were found after long duration (10 s) stimuli in thalamus. CONCLUSION An altered neurovascular response in patients with BMD may increase our understanding of neurovascular coupling and the pathogenesis related to dystrophinopathy and nNOS.
Collapse
Affiliation(s)
- Ulrich Lindberg
- Functional Imaging UnitDepartment of Clinical PhysiologyNuclear Medicine and PETRigshospitalet GlostrupUniversity of CopenhagenGlostrupDenmark
- Lundbeck Foundation Center for Neurovascular signalling (LUCENS)Rigshospitalet GlostrupUniversity of CopenhagenGlostrupDenmark
| | - Christina Kruuse
- Lundbeck Foundation Center for Neurovascular signalling (LUCENS)Rigshospitalet GlostrupUniversity of CopenhagenGlostrupDenmark
- Neurovascular Research UnitDepartment of NeurologyHerlev Gentofte HospitalUniversity of CopenhagenHerlevDenmark
| | - Nanna Witting
- Copenhagen Neuromuscular CenterDepartment of NeurologyRigshospitaletUniversity of CopenhagenDenmark
| | - Stine Lundgaard Jørgensen
- Lundbeck Foundation Center for Neurovascular signalling (LUCENS)Rigshospitalet GlostrupUniversity of CopenhagenGlostrupDenmark
- Neurovascular Research UnitDepartment of NeurologyHerlev Gentofte HospitalUniversity of CopenhagenHerlevDenmark
| | - John Vissing
- Copenhagen Neuromuscular CenterDepartment of NeurologyRigshospitaletUniversity of CopenhagenDenmark
| | - Egill Rostrup
- Functional Imaging UnitDepartment of Clinical PhysiologyNuclear Medicine and PETRigshospitalet GlostrupUniversity of CopenhagenGlostrupDenmark
| | - Henrik Bo Wiberg Larsson
- Functional Imaging UnitDepartment of Clinical PhysiologyNuclear Medicine and PETRigshospitalet GlostrupUniversity of CopenhagenGlostrupDenmark
- Lundbeck Foundation Center for Neurovascular signalling (LUCENS)Rigshospitalet GlostrupUniversity of CopenhagenGlostrupDenmark
| |
Collapse
|
23
|
Fallon JR, McNally EM. Non-Glycanated Biglycan and LTBP4: Leveraging the extracellular matrix for Duchenne Muscular Dystrophy therapeutics. Matrix Biol 2018; 68-69:616-627. [PMID: 29481844 DOI: 10.1016/j.matbio.2018.02.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 02/18/2018] [Accepted: 02/19/2018] [Indexed: 12/15/2022]
Abstract
The extracellular matrix (ECM) plays key roles in normal and diseased skeletal and cardiac muscle. In healthy muscle the ECM is essential for transmitting contractile force, maintaining myofiber integrity and orchestrating cellular signaling. Duchenne Muscular Dystrophy (DMD) is caused by loss of dystrophin, a cytosolic protein that anchors a transmembrane complex and serves as a vital link between the actin cytoskeleton and the basal lamina. Loss of dystrophin leads to membrane fragility and impaired signaling, resulting in myofiber death and cycles of inflammation and regeneration. Fibrosis is also a cardinal feature of DMD. In this review, we will focus on two cases where understanding the normal function and regulation of ECM in muscle has led to the discovery of candidate therapeutics for DMD. Biglycan is a small leucine rich repeat ECM protein present as two glycoforms in muscle that have dramatically different functions. One widely expressed form is biglycan proteoglycan (PG) that bears two chondroitin sulfate GAG chains (typically chondroitin sulfate) and two N-linked carbohydrates. The second glycoform, referred to as 'NG' (non-glycanated) biglycan, lacks the GAG side chains. NG, but not PG biglycan recruits utrophin, an autosomal paralog of dystrophin, and an NOS-containing signaling complex to the muscle cell membrane. Recombinant NG biglycan can be systemically delivered to dystrophic mice where it upregulates utrophin at the membrane and improves muscle health and function. An optimized version of NG biglycan, 'TVN-102', is under development as a candidate therapeutic for DMD. A second matrix-embedded protein being evaluated for therapeutic potential is latent TGFβ binding protein 4 (LTBP4). Identified in a genomic screen for modifiers of muscular dystrophy, LTBP4 binds both TGFβ and myostatin. Genetic studies identified the hinge region of LTBP4 as linked to TGFβ release and contributing to the "hyper-TGFβ" signaling state that promotes fibrosis in muscular dystrophy. This hinge region can be stabilized by antibodies directed towards this domain. Stabilizing the hinge region of LTBP4 is expected to reduce latent TGFβ release and thus reduce fibrosis.
Collapse
Affiliation(s)
- Justin R Fallon
- Dept. of Neuroscience, Brown University, Providence, RI 02912, United States.
| | - Elizabeth M McNally
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| |
Collapse
|
24
|
Baghdadi MB, Tajbakhsh S. Regulation and phylogeny of skeletal muscle regeneration. Dev Biol 2018; 433:200-209. [DOI: 10.1016/j.ydbio.2017.07.026] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/30/2017] [Accepted: 07/31/2017] [Indexed: 12/31/2022]
|
25
|
Chao HT, Liu L, Bellen HJ. Building dialogues between clinical and biomedical research through cross-species collaborations. Semin Cell Dev Biol 2017; 70:49-57. [PMID: 28579453 PMCID: PMC5623622 DOI: 10.1016/j.semcdb.2017.05.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 05/16/2017] [Accepted: 05/23/2017] [Indexed: 10/19/2022]
Abstract
Today, biomedical science is equipped with an impressive array of technologies and genetic resources that bolster our basic understanding of fundamental biology and enhance the practice of modern medicine by providing clinicians with a diverse toolkit to diagnose, prognosticate, and treat a plethora of conditions. Many significant advances in our understanding of disease mechanisms and therapeutic interventions have arisen from fruitful dialogues between clinicians and biomedical research scientists. However, the increasingly specialized scientific and medical disciplines, globalization of science and technology, and complex datasets often hinder the development of effective interdisciplinary collaborations between clinical medicine and biomedical research. The goal of this review is to provide examples of diverse strategies to enhance communication and collaboration across diverse disciplines. First, we discuss examples of efforts to foster interdisciplinary collaborations at institutional and multi-institutional levels. Second, we explore resources and tools for clinicians and research scientists to facilitate effective bi-directional dialogues. Third, we use our experiences in neurobiology and human genetics to highlight how communication between clinical medicine and biomedical research lead to effective implementation of cross-species model organism approaches to uncover the biological underpinnings of health and disease.
Collapse
Affiliation(s)
- Hsiao-Tuan Chao
- Department of Pediatrics, Section of Child Neurology, Baylor College of Medicine, Houston, TX 77030, United States; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, United States.
| | - Lucy Liu
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, United States; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, United States
| | - Hugo J Bellen
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, United States; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, United States; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, United States; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, United States; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, United States.
| |
Collapse
|
26
|
Victor RG, Sweeney HL, Finkel R, McDonald CM, Byrne B, Eagle M, Goemans N, Vandenborne K, Dubrovsky AL, Topaloglu H, Miceli MC, Furlong P, Landry J, Elashoff R, Cox D. A phase 3 randomized placebo-controlled trial of tadalafil for Duchenne muscular dystrophy. Neurology 2017; 89:1811-1820. [PMID: 28972192 PMCID: PMC5664308 DOI: 10.1212/wnl.0000000000004570] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/28/2017] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To conduct a randomized trial to test the primary hypothesis that once-daily tadalafil, administered orally for 48 weeks, lessens the decline in ambulatory ability in boys with Duchenne muscular dystrophy (DMD). METHODS Three hundred thirty-one participants with DMD 7 to 14 years of age taking glucocorticoids were randomized to tadalafil 0.3 mg·kg-1·d-1, tadalafil 0.6 mg·kg-1·d-1, or placebo. The primary efficacy measure was 6-minute walk distance (6MWD) after 48 weeks. Secondary efficacy measures included North Star Ambulatory Assessment and timed function tests. Performance of Upper Limb (PUL) was a prespecified exploratory outcome. RESULTS Tadalafil had no effect on the primary outcome: 48-week declines in 6MWD were 51.0 ± 9.3 m with placebo, 64.7 ± 9.8 m with low-dose tadalafil (p = 0.307 vs placebo), and 59.1 ± 9.4 m with high-dose tadalafil (p = 0.538 vs placebo). Tadalafil also had no effect on secondary outcomes. In boys >10 years of age, total PUL score and shoulder subscore declined less with low-dose tadalafil than placebo. Adverse events were consistent with the known safety profile of tadalafil and the DMD disease state. CONCLUSIONS Tadalafil did not lessen the decline in ambulatory ability in boys with DMD. Further studies should be considered to confirm the hypothesis-generating upper limb data and to determine whether ambulatory decline can be slowed by initiation of tadalafil before 7 years of age. CLINICALTRIALSGOV IDENTIFIER NCT01865084. CLASSIFICATION OF EVIDENCE This study provides Class I evidence that tadalafil does not slow ambulatory decline in 7- to 14-year-old boys with Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Ronald G Victor
- From the Cedars-Sinai Medical Center (R.G.V.), Los Angeles, CA; University of Florida (H.L.S., B.B., K.V.), Gainesville; Nemours Children's Hospital (R.F.), Orlando, FL; University of California at Davis (C.M.M.), Sacramento; Newcastle University (M.E.), Newcastle Upon Tyne, UK; University Hospitals Leuven (N.G.), Belgium; Instituto de Neurociencias-Fundacion Favaloro (A.L.D.), Buenos Aires, Argentina; Hacettepe University School of Medicine (H.T.), Ankara, Turkey; UCLA (M.C.M., R.E.), Los Angeles, CA; Parent Project Muscular Dystrophy (P.F.), Hackensack, NJ; Eli Lilly Canada, Eli Lilly and Company, Toronto, ON (J.L.); and Eli Lilly and Company (D.C.), Indianapolis, IN.
| | - H Lee Sweeney
- From the Cedars-Sinai Medical Center (R.G.V.), Los Angeles, CA; University of Florida (H.L.S., B.B., K.V.), Gainesville; Nemours Children's Hospital (R.F.), Orlando, FL; University of California at Davis (C.M.M.), Sacramento; Newcastle University (M.E.), Newcastle Upon Tyne, UK; University Hospitals Leuven (N.G.), Belgium; Instituto de Neurociencias-Fundacion Favaloro (A.L.D.), Buenos Aires, Argentina; Hacettepe University School of Medicine (H.T.), Ankara, Turkey; UCLA (M.C.M., R.E.), Los Angeles, CA; Parent Project Muscular Dystrophy (P.F.), Hackensack, NJ; Eli Lilly Canada, Eli Lilly and Company, Toronto, ON (J.L.); and Eli Lilly and Company (D.C.), Indianapolis, IN
| | - Richard Finkel
- From the Cedars-Sinai Medical Center (R.G.V.), Los Angeles, CA; University of Florida (H.L.S., B.B., K.V.), Gainesville; Nemours Children's Hospital (R.F.), Orlando, FL; University of California at Davis (C.M.M.), Sacramento; Newcastle University (M.E.), Newcastle Upon Tyne, UK; University Hospitals Leuven (N.G.), Belgium; Instituto de Neurociencias-Fundacion Favaloro (A.L.D.), Buenos Aires, Argentina; Hacettepe University School of Medicine (H.T.), Ankara, Turkey; UCLA (M.C.M., R.E.), Los Angeles, CA; Parent Project Muscular Dystrophy (P.F.), Hackensack, NJ; Eli Lilly Canada, Eli Lilly and Company, Toronto, ON (J.L.); and Eli Lilly and Company (D.C.), Indianapolis, IN
| | - Craig M McDonald
- From the Cedars-Sinai Medical Center (R.G.V.), Los Angeles, CA; University of Florida (H.L.S., B.B., K.V.), Gainesville; Nemours Children's Hospital (R.F.), Orlando, FL; University of California at Davis (C.M.M.), Sacramento; Newcastle University (M.E.), Newcastle Upon Tyne, UK; University Hospitals Leuven (N.G.), Belgium; Instituto de Neurociencias-Fundacion Favaloro (A.L.D.), Buenos Aires, Argentina; Hacettepe University School of Medicine (H.T.), Ankara, Turkey; UCLA (M.C.M., R.E.), Los Angeles, CA; Parent Project Muscular Dystrophy (P.F.), Hackensack, NJ; Eli Lilly Canada, Eli Lilly and Company, Toronto, ON (J.L.); and Eli Lilly and Company (D.C.), Indianapolis, IN
| | - Barry Byrne
- From the Cedars-Sinai Medical Center (R.G.V.), Los Angeles, CA; University of Florida (H.L.S., B.B., K.V.), Gainesville; Nemours Children's Hospital (R.F.), Orlando, FL; University of California at Davis (C.M.M.), Sacramento; Newcastle University (M.E.), Newcastle Upon Tyne, UK; University Hospitals Leuven (N.G.), Belgium; Instituto de Neurociencias-Fundacion Favaloro (A.L.D.), Buenos Aires, Argentina; Hacettepe University School of Medicine (H.T.), Ankara, Turkey; UCLA (M.C.M., R.E.), Los Angeles, CA; Parent Project Muscular Dystrophy (P.F.), Hackensack, NJ; Eli Lilly Canada, Eli Lilly and Company, Toronto, ON (J.L.); and Eli Lilly and Company (D.C.), Indianapolis, IN
| | - Michelle Eagle
- From the Cedars-Sinai Medical Center (R.G.V.), Los Angeles, CA; University of Florida (H.L.S., B.B., K.V.), Gainesville; Nemours Children's Hospital (R.F.), Orlando, FL; University of California at Davis (C.M.M.), Sacramento; Newcastle University (M.E.), Newcastle Upon Tyne, UK; University Hospitals Leuven (N.G.), Belgium; Instituto de Neurociencias-Fundacion Favaloro (A.L.D.), Buenos Aires, Argentina; Hacettepe University School of Medicine (H.T.), Ankara, Turkey; UCLA (M.C.M., R.E.), Los Angeles, CA; Parent Project Muscular Dystrophy (P.F.), Hackensack, NJ; Eli Lilly Canada, Eli Lilly and Company, Toronto, ON (J.L.); and Eli Lilly and Company (D.C.), Indianapolis, IN
| | - Nathalie Goemans
- From the Cedars-Sinai Medical Center (R.G.V.), Los Angeles, CA; University of Florida (H.L.S., B.B., K.V.), Gainesville; Nemours Children's Hospital (R.F.), Orlando, FL; University of California at Davis (C.M.M.), Sacramento; Newcastle University (M.E.), Newcastle Upon Tyne, UK; University Hospitals Leuven (N.G.), Belgium; Instituto de Neurociencias-Fundacion Favaloro (A.L.D.), Buenos Aires, Argentina; Hacettepe University School of Medicine (H.T.), Ankara, Turkey; UCLA (M.C.M., R.E.), Los Angeles, CA; Parent Project Muscular Dystrophy (P.F.), Hackensack, NJ; Eli Lilly Canada, Eli Lilly and Company, Toronto, ON (J.L.); and Eli Lilly and Company (D.C.), Indianapolis, IN
| | - Krista Vandenborne
- From the Cedars-Sinai Medical Center (R.G.V.), Los Angeles, CA; University of Florida (H.L.S., B.B., K.V.), Gainesville; Nemours Children's Hospital (R.F.), Orlando, FL; University of California at Davis (C.M.M.), Sacramento; Newcastle University (M.E.), Newcastle Upon Tyne, UK; University Hospitals Leuven (N.G.), Belgium; Instituto de Neurociencias-Fundacion Favaloro (A.L.D.), Buenos Aires, Argentina; Hacettepe University School of Medicine (H.T.), Ankara, Turkey; UCLA (M.C.M., R.E.), Los Angeles, CA; Parent Project Muscular Dystrophy (P.F.), Hackensack, NJ; Eli Lilly Canada, Eli Lilly and Company, Toronto, ON (J.L.); and Eli Lilly and Company (D.C.), Indianapolis, IN
| | - Alberto L Dubrovsky
- From the Cedars-Sinai Medical Center (R.G.V.), Los Angeles, CA; University of Florida (H.L.S., B.B., K.V.), Gainesville; Nemours Children's Hospital (R.F.), Orlando, FL; University of California at Davis (C.M.M.), Sacramento; Newcastle University (M.E.), Newcastle Upon Tyne, UK; University Hospitals Leuven (N.G.), Belgium; Instituto de Neurociencias-Fundacion Favaloro (A.L.D.), Buenos Aires, Argentina; Hacettepe University School of Medicine (H.T.), Ankara, Turkey; UCLA (M.C.M., R.E.), Los Angeles, CA; Parent Project Muscular Dystrophy (P.F.), Hackensack, NJ; Eli Lilly Canada, Eli Lilly and Company, Toronto, ON (J.L.); and Eli Lilly and Company (D.C.), Indianapolis, IN
| | - Haluk Topaloglu
- From the Cedars-Sinai Medical Center (R.G.V.), Los Angeles, CA; University of Florida (H.L.S., B.B., K.V.), Gainesville; Nemours Children's Hospital (R.F.), Orlando, FL; University of California at Davis (C.M.M.), Sacramento; Newcastle University (M.E.), Newcastle Upon Tyne, UK; University Hospitals Leuven (N.G.), Belgium; Instituto de Neurociencias-Fundacion Favaloro (A.L.D.), Buenos Aires, Argentina; Hacettepe University School of Medicine (H.T.), Ankara, Turkey; UCLA (M.C.M., R.E.), Los Angeles, CA; Parent Project Muscular Dystrophy (P.F.), Hackensack, NJ; Eli Lilly Canada, Eli Lilly and Company, Toronto, ON (J.L.); and Eli Lilly and Company (D.C.), Indianapolis, IN
| | - M Carrie Miceli
- From the Cedars-Sinai Medical Center (R.G.V.), Los Angeles, CA; University of Florida (H.L.S., B.B., K.V.), Gainesville; Nemours Children's Hospital (R.F.), Orlando, FL; University of California at Davis (C.M.M.), Sacramento; Newcastle University (M.E.), Newcastle Upon Tyne, UK; University Hospitals Leuven (N.G.), Belgium; Instituto de Neurociencias-Fundacion Favaloro (A.L.D.), Buenos Aires, Argentina; Hacettepe University School of Medicine (H.T.), Ankara, Turkey; UCLA (M.C.M., R.E.), Los Angeles, CA; Parent Project Muscular Dystrophy (P.F.), Hackensack, NJ; Eli Lilly Canada, Eli Lilly and Company, Toronto, ON (J.L.); and Eli Lilly and Company (D.C.), Indianapolis, IN
| | - Pat Furlong
- From the Cedars-Sinai Medical Center (R.G.V.), Los Angeles, CA; University of Florida (H.L.S., B.B., K.V.), Gainesville; Nemours Children's Hospital (R.F.), Orlando, FL; University of California at Davis (C.M.M.), Sacramento; Newcastle University (M.E.), Newcastle Upon Tyne, UK; University Hospitals Leuven (N.G.), Belgium; Instituto de Neurociencias-Fundacion Favaloro (A.L.D.), Buenos Aires, Argentina; Hacettepe University School of Medicine (H.T.), Ankara, Turkey; UCLA (M.C.M., R.E.), Los Angeles, CA; Parent Project Muscular Dystrophy (P.F.), Hackensack, NJ; Eli Lilly Canada, Eli Lilly and Company, Toronto, ON (J.L.); and Eli Lilly and Company (D.C.), Indianapolis, IN
| | - John Landry
- From the Cedars-Sinai Medical Center (R.G.V.), Los Angeles, CA; University of Florida (H.L.S., B.B., K.V.), Gainesville; Nemours Children's Hospital (R.F.), Orlando, FL; University of California at Davis (C.M.M.), Sacramento; Newcastle University (M.E.), Newcastle Upon Tyne, UK; University Hospitals Leuven (N.G.), Belgium; Instituto de Neurociencias-Fundacion Favaloro (A.L.D.), Buenos Aires, Argentina; Hacettepe University School of Medicine (H.T.), Ankara, Turkey; UCLA (M.C.M., R.E.), Los Angeles, CA; Parent Project Muscular Dystrophy (P.F.), Hackensack, NJ; Eli Lilly Canada, Eli Lilly and Company, Toronto, ON (J.L.); and Eli Lilly and Company (D.C.), Indianapolis, IN
| | - Robert Elashoff
- From the Cedars-Sinai Medical Center (R.G.V.), Los Angeles, CA; University of Florida (H.L.S., B.B., K.V.), Gainesville; Nemours Children's Hospital (R.F.), Orlando, FL; University of California at Davis (C.M.M.), Sacramento; Newcastle University (M.E.), Newcastle Upon Tyne, UK; University Hospitals Leuven (N.G.), Belgium; Instituto de Neurociencias-Fundacion Favaloro (A.L.D.), Buenos Aires, Argentina; Hacettepe University School of Medicine (H.T.), Ankara, Turkey; UCLA (M.C.M., R.E.), Los Angeles, CA; Parent Project Muscular Dystrophy (P.F.), Hackensack, NJ; Eli Lilly Canada, Eli Lilly and Company, Toronto, ON (J.L.); and Eli Lilly and Company (D.C.), Indianapolis, IN
| | - David Cox
- From the Cedars-Sinai Medical Center (R.G.V.), Los Angeles, CA; University of Florida (H.L.S., B.B., K.V.), Gainesville; Nemours Children's Hospital (R.F.), Orlando, FL; University of California at Davis (C.M.M.), Sacramento; Newcastle University (M.E.), Newcastle Upon Tyne, UK; University Hospitals Leuven (N.G.), Belgium; Instituto de Neurociencias-Fundacion Favaloro (A.L.D.), Buenos Aires, Argentina; Hacettepe University School of Medicine (H.T.), Ankara, Turkey; UCLA (M.C.M., R.E.), Los Angeles, CA; Parent Project Muscular Dystrophy (P.F.), Hackensack, NJ; Eli Lilly Canada, Eli Lilly and Company, Toronto, ON (J.L.); and Eli Lilly and Company (D.C.), Indianapolis, IN
| | | |
Collapse
|
27
|
Nio Y, Tanaka M, Hirozane Y, Muraki Y, Okawara M, Hazama M, Matsuo T. Phosphodiesterase 4 inhibitor and phosphodiesterase 5 inhibitor combination therapy has antifibrotic and anti‐inflammatory effects in mdx mice with Duchenne muscular dystrophy. FASEB J 2017; 31:5307-5320. [DOI: 10.1096/fj.201700249r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/25/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Yasunori Nio
- Extra Value Generation and General Medicine Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Masayuki Tanaka
- Inflammation Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Yoshihiko Hirozane
- Biomolecular Research LaboratoriesPharmaceutical Research DivisionTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Yo Muraki
- Extra Value Generation and General Medicine Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Mitsugi Okawara
- Extra Value Generation and General Medicine Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Masatoshi Hazama
- Extra Value Generation and General Medicine Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Takanori Matsuo
- Extra Value Generation and General Medicine Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| |
Collapse
|
28
|
Moon Y, Balke JE, Madorma D, Siegel MP, Knowels G, Brouckaert P, Buys ES, Marcinek DJ, Percival JM. Nitric Oxide Regulates Skeletal Muscle Fatigue, Fiber Type, Microtubule Organization, and Mitochondrial ATP Synthesis Efficiency Through cGMP-Dependent Mechanisms. Antioxid Redox Signal 2017; 26:966-985. [PMID: 27393340 PMCID: PMC5467110 DOI: 10.1089/ars.2016.6630] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIM Skeletal muscle nitric oxide-cyclic guanosine monophosphate (NO-cGMP) pathways are impaired in Duchenne and Becker muscular dystrophy partly because of reduced nNOSμ and soluble guanylate cyclase (GC) activity. However, GC function and the consequences of reduced GC activity in skeletal muscle are unknown. In this study, we explore the functions of GC and NO-cGMP signaling in skeletal muscle. RESULTS GC1, but not GC2, expression was higher in oxidative than glycolytic muscles. GC1 was found in a complex with nNOSμ and targeted to nNOS compartments at the Golgi complex and neuromuscular junction. Baseline GC activity and GC agonist responsiveness was reduced in the absence of nNOS. Structural analyses revealed aberrant microtubule directionality in GC1-/- muscle. Functional analyses of GC1-/- muscles revealed reduced fatigue resistance and postexercise force recovery that were not due to shifts in type IIA-IIX fiber balance. Force deficits in GC1-/- muscles were also not driven by defects in resting mitochondrial adenosine triphosphate (ATP) synthesis. However, increasing muscle cGMP with sildenafil decreased ATP synthesis efficiency and capacity, without impacting mitochondrial content or ultrastructure. INNOVATION GC may represent a new target for alleviating muscle fatigue and that NO-cGMP signaling may play important roles in muscle structure, contractility, and bioenergetics. CONCLUSIONS These findings suggest that GC activity is nNOS dependent and that muscle-specific control of GC expression and differential GC targeting may facilitate NO-cGMP signaling diversity. They suggest that nNOS regulates muscle fiber type, microtubule organization, fatigability, and postexercise force recovery partly through GC1 and suggest that NO-cGMP pathways may modulate mitochondrial ATP synthesis efficiency. Antioxid. Redox Signal. 26, 966-985.
Collapse
Affiliation(s)
- Younghye Moon
- 1 Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine , Miami, Florida
| | - Jordan E Balke
- 1 Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine , Miami, Florida
| | - Derik Madorma
- 1 Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine , Miami, Florida
| | - Michael P Siegel
- 2 Department of Bioengineering, University of Washington , Seattle, Washington
| | - Gary Knowels
- 2 Department of Bioengineering, University of Washington , Seattle, Washington
| | - Peter Brouckaert
- 3 Department for Molecular Biomedical Research and Biomedical Molecular Biology, Ghent University , Ghent, Belgium
| | - Emmanuel S Buys
- 4 Department of Anesthesia, Critical Care and Pain Medicine, Anesthesia Center for Critical Care Research , Massachusetts General Hospital, Boston, Massachusetts
| | - David J Marcinek
- 2 Department of Bioengineering, University of Washington , Seattle, Washington.,5 Department of Radiology, University of Washington , Seattle, Washington
| | - Justin M Percival
- 1 Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine , Miami, Florida
| |
Collapse
|
29
|
Timpani CA, Hayes A, Rybalka E. Therapeutic strategies to address neuronal nitric oxide synthase deficiency and the loss of nitric oxide bioavailability in Duchenne Muscular Dystrophy. Orphanet J Rare Dis 2017; 12:100. [PMID: 28545481 PMCID: PMC5445371 DOI: 10.1186/s13023-017-0652-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 05/12/2017] [Indexed: 12/25/2022] Open
Abstract
Duchenne Muscular Dystrophy is a rare and fatal neuromuscular disease in which the absence of dystrophin from the muscle membrane induces a secondary loss of neuronal nitric oxide synthase and the muscles capacity for endogenous nitric oxide synthesis. Since nitric oxide is a potent regulator of skeletal muscle metabolism, mass, function and regeneration, the loss of nitric oxide bioavailability is likely a key contributor to the chronic pathological wasting evident in Duchenne Muscular Dystrophy. As such, various therapeutic interventions to re-establish either the neuronal nitric oxide synthase protein deficit or the consequential loss of nitric oxide synthesis and bioavailability have been investigated in both animal models of Duchenne Muscular Dystrophy and in human clinical trials. Notably, the efficacy of these interventions are varied and not always translatable from animal model to human patients, highlighting a complex interplay of factors which determine the downstream modulatory effects of nitric oxide. We review these studies herein.
Collapse
Affiliation(s)
- Cara A Timpani
- College of Health & Biomedicine, Victoria University, PO Box 14428, Melbourne, Victoria, Australia, 8001.,Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, Victoria, 3021, Australia
| | - Alan Hayes
- College of Health & Biomedicine, Victoria University, PO Box 14428, Melbourne, Victoria, Australia, 8001.,Institute of Sport, Exercise & Active Living (ISEAL), Victoria University, Melbourne, Victoria, 8001, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, Victoria, 3021, Australia
| | - Emma Rybalka
- College of Health & Biomedicine, Victoria University, PO Box 14428, Melbourne, Victoria, Australia, 8001. .,Institute of Sport, Exercise & Active Living (ISEAL), Victoria University, Melbourne, Victoria, 8001, Australia. .,Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, Victoria, 3021, Australia.
| |
Collapse
|
30
|
Johnstone VPA, Viola HM, Hool LC. Dystrophic Cardiomyopathy-Potential Role of Calcium in Pathogenesis, Treatment and Novel Therapies. Genes (Basel) 2017; 8:genes8040108. [PMID: 28338606 PMCID: PMC5406855 DOI: 10.3390/genes8040108] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 03/16/2017] [Accepted: 03/21/2017] [Indexed: 01/06/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by defects in the DMD gene and results in progressive wasting of skeletal and cardiac muscle due to an absence of functional dystrophin. Cardiomyopathy is prominent in DMD patients, and contributes significantly to mortality. This is particularly true following respiratory interventions that reduce death rate and increase ambulation and consequently cardiac load. Cardiomyopathy shows an increasing prevalence with age and disease progression, and over 95% of patients exhibit dilated cardiomyopathy by the time they reach adulthood. Development of the myopathy is complex, and elevations in intracellular calcium, functional muscle ischemia, and mitochondrial dysfunction characterise the pathophysiology. Current therapies are limited to treating symptoms of the disease and there is therefore an urgent need to treat the underlying genetic defect. Several novel therapies are outlined here, and the unprecedented success of phosphorodiamidate morpholino oligomers (PMOs) in preclinical and clinical studies is overviewed.
Collapse
Affiliation(s)
- Victoria P A Johnstone
- School of Human Sciences, The University of Western Australia, Crawley, WA 6009, Australia.
| | - Helena M Viola
- School of Human Sciences, The University of Western Australia, Crawley, WA 6009, Australia.
| | - Livia C Hool
- School of Human Sciences, The University of Western Australia, Crawley, WA 6009, Australia.
- Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia.
| |
Collapse
|
31
|
Guiraud S, Migeon T, Ferry A, Chen Z, Ouchelouche S, Verpont MC, Sado Y, Allamand V, Ronco P, Plaisier E. HANAC Col4a1 Mutation in Mice Leads to Skeletal Muscle Alterations due to a Primary Vascular Defect. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:505-516. [PMID: 28056338 DOI: 10.1016/j.ajpath.2016.10.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 10/24/2016] [Accepted: 10/25/2016] [Indexed: 12/29/2022]
Abstract
Collagen IV is a major component of basement membranes (BMs). The α1(IV) chain, encoded by the COL4A1 gene, is expressed ubiquitously and associates with the α2(IV) chain to form the α1α1α2(IV) heterotrimer. Several COL4A1 mutations affecting a conformational domain containing integrin-binding sites are responsible for the systemic syndrome of hereditary angiopathy, nephropathy, aneurysms, and cramps (HANAC). To analyze the pathophysiology of HANAC, Col4a1 mutant mice bearing the p.Gly498Val mutation were generated. Analysis of the skeletal muscles of Col4a1G498V mutant animals showed morphologic characteristics of a muscular dystrophy phenotype with myofiber atrophy, centronucleation, focal inflammatory infiltrates, and fibrosis. Abnormal ultrastructural aspects of muscle BMs was associated with reduced extracellular secretion of the mutant α1α1α2(IV) trimer. In addition to muscular dystrophic features, endothelial cell defects of the muscle capillaries were observed, with intracytoplasmic accumulation of the mutant α1α1α2(IV) molecules, endoplasmic reticulum cisternae dilation, and up-regulation of endoplasmic reticulum stress markers. Induction of the unfolded protein response in Col4a1 mutant muscle tissue resulted in an excess of apoptosis in endothelial cells. HANAC mutant animals also presented with a muscular functional impairment and increased serum creatine kinase levels reflecting altered muscle fiber sarcolemma. This extensive description of the muscular phenotype of the Col4a1 HANAC murine model suggests a potential contribution of primary endothelial cell defects, together with muscle BM alterations, to the development of COL4A1-related myopathy.
Collapse
Affiliation(s)
- Simon Guiraud
- Mixed Research Unit S1155, INSERM, Paris, France; University Pierre and Marie Curie Paris 06, Sorbonne University, Paris, France; Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom
| | - Tiffany Migeon
- Mixed Research Unit S1155, INSERM, Paris, France; University Pierre and Marie Curie Paris 06, Sorbonne University, Paris, France
| | - Arnaud Ferry
- Research Center in Myology, Institut de Myologie, the Inserm UMRS974, CNRS FRE3617, Pitié-Salpêtrière Hospital Group, University Pierre and Marie Curie Paris 06, Paris Descartes University, The Sorbonne University, Paris, France
| | - Zhiyong Chen
- Mixed Research Unit S1155, INSERM, Paris, France
| | - Souhila Ouchelouche
- Mixed Research Unit S1155, INSERM, Paris, France; University Pierre and Marie Curie Paris 06, Sorbonne University, Paris, France
| | - Marie-Christine Verpont
- Mixed Research Unit S1155, INSERM, Paris, France; University Pierre and Marie Curie Paris 06, Sorbonne University, Paris, France
| | | | - Valérie Allamand
- Research Center in Myology, Institut de Myologie, the Inserm UMRS974, CNRS FRE3617, Pitié-Salpêtrière Hospital Group, University Pierre and Marie Curie Paris 06, Paris Descartes University, The Sorbonne University, Paris, France
| | - Pierre Ronco
- Mixed Research Unit S1155, INSERM, Paris, France; University Pierre and Marie Curie Paris 06, Sorbonne University, Paris, France; Department of Nephrology and Dialysis, Assistance Publique-Hôpitaux de Paris, Tenon Hospital, Paris, France
| | - Emmanuelle Plaisier
- Mixed Research Unit S1155, INSERM, Paris, France; University Pierre and Marie Curie Paris 06, Sorbonne University, Paris, France; Department of Nephrology and Dialysis, Assistance Publique-Hôpitaux de Paris, Tenon Hospital, Paris, France.
| |
Collapse
|
32
|
Lindberg U, Witting N, Jørgensen SL, Vissing J, Rostrup E, Larsson HBW, Kruuse C. Effects of Sildenafil on Cerebrovascular Reactivity in Patients with Becker Muscular Dystrophy. Neurotherapeutics 2017; 14:182-190. [PMID: 27485237 PMCID: PMC5233618 DOI: 10.1007/s13311-016-0467-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Patients suffering from Becker muscular dystrophy (BMD) have dysfunctional dystrophin proteins and are deficient in neuronal nitric oxide synthase (nNOS) in muscles. This causes functional ischemia and contributes to muscle wasting. Similar functional ischemia may be present in brains of patients with BMD, who often have mild cognitive impairment, and nNOS may be important for the regulation of the microvascular circulation in the brain. We hypothesized that treatment with sildenafil, a phosphodiesterase type 5 inhibitor that potentiates nitric oxide responses, would augment both the blood oxygen level-dependent (BOLD) response and cerebral blood flow (CBF) in patients with BMD. Seventeen patients (mean ± SD age 38.5 ± 10.8 years) with BMD were included in this randomized, double-blind, placebo-controlled, crossover trial. Twelve patients completed the entire study. Effects of sildenafil were assessed by 3 T magnetic resonance (MR) scanning, evoked potentials, somatosensory task-induced BOLD functional MR imaging, regional and global perfusion, and angiography before and after 4 weeks of sildenafil, 20 mg (Revatio in gelatine capsules, oral, 3 times daily), or placebo treatment. Sildenafil increased the event-related sensory and visual BOLD response compared with placebo (p < 0.01). However, sildenafil did not alter CBF, measured by MR phase contrast mapping, or the arterial diameter of the middle cerebral artery, measured by MR angiography. We conclude that nNOS may play a role in event-related neurovascular responses. Further studies in patients with BMD may help clarify the roles of dystrophin and nNOS in neurovascular coupling in general, and in patients with BMD in particular.
Collapse
Affiliation(s)
- Ulrich Lindberg
- Functional Imaging Unit, Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet Glostrup, University of Copenhagen, Glostrup, Denmark
- Lundbeck Foundation Center for Neurovascular signalling (LUCENS), Rigshospitalet Glostrup, University of Copenhagen, Glostrup, Denmark
| | - Nanna Witting
- Copenhagen Neuromuscular Center and Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Stine Lundgaard Jørgensen
- Lundbeck Foundation Center for Neurovascular signalling (LUCENS), Rigshospitalet Glostrup, University of Copenhagen, Glostrup, Denmark
| | - John Vissing
- Copenhagen Neuromuscular Center and Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Egill Rostrup
- Functional Imaging Unit, Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet Glostrup, University of Copenhagen, Glostrup, Denmark
| | - Henrik Bo Wiberg Larsson
- Functional Imaging Unit, Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet Glostrup, University of Copenhagen, Glostrup, Denmark
- Lundbeck Foundation Center for Neurovascular signalling (LUCENS), Rigshospitalet Glostrup, University of Copenhagen, Glostrup, Denmark
| | - Christina Kruuse
- Lundbeck Foundation Center for Neurovascular signalling (LUCENS), Rigshospitalet Glostrup, University of Copenhagen, Glostrup, Denmark.
- Neurovascular Research Unit, Department of Neurology, Herlev Gentofte Hospital, University of Copenhagen, Herlev, Denmark.
| |
Collapse
|
33
|
Spinazzola JM, Kunkel LM. Pharmacological therapeutics targeting the secondary defects and downstream pathology of Duchenne muscular dystrophy. Expert Opin Orphan Drugs 2016; 4:1179-1194. [PMID: 28670506 DOI: 10.1080/21678707.2016.1240613] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Since the identification of the dystrophin gene in 1986, a cure for Duchenne muscular dystrophy (DMD) has yet to be discovered. Presently, there are a number of genetic-based therapies in development aimed at restoration and/or repair of the primary defect. However, growing understanding of the pathophysiological consequences of dystrophin absence has revealed several promising downstream targets for the development of therapeutics. AREAS COVERED In this review, we discuss various strategies for DMD therapy targeting downstream consequences of dystrophin absence including loss of muscle mass, inflammation, fibrosis, calcium overload, oxidative stress, and ischemia. The rationale of each approach and the efficacy of drugs in preclinical and clinical studies are discussed. EXPERT OPINION For the last 30 years, effective DMD drug therapy has been limited to corticosteroids, which are associated with a number of negative side effects. Our knowledge of the consequences of dystrophin absence that contribute to DMD pathology has revealed several potential therapeutic targets. Some of these approaches may have potential to improve or slow disease progression independently or in combination with genetic-based approaches. The applicability of these pharmacological therapies to DMD patients irrespective of their genetic mutation, as well as the potential benefits even for advanced stage patients warrants their continued investigation.
Collapse
Affiliation(s)
- Janelle M Spinazzola
- Boston Children's Hospital, Division of Genetics and Genomics, Boston, MA 02115.,Harvard Medical School, Departments of Pediatrics and Genetics, Boston, MA 02115
| | - Louis M Kunkel
- Boston Children's Hospital, Division of Genetics and Genomics, Boston, MA 02115.,Harvard Medical School, Departments of Pediatrics and Genetics, Boston, MA 02115.,The Stem Cell Program at Boston Children's Hospital, Boston, MA 02115.,The Manton Center for Orphan Diseases, Boston, MA 02115.,Harvard Stem Cell Institute, Cambridge, MA 02138
| |
Collapse
|
34
|
Alexander MS, Gasperini MJ, Tsai PT, Gibbs DE, Spinazzola JM, Marshall JL, Feyder MJ, Pletcher MT, Chekler ELP, Morris CA, Sahin M, Harms JF, Schmidt CJ, Kleiman RJ, Kunkel LM. Reversal of neurobehavioral social deficits in dystrophic mice using inhibitors of phosphodiesterases PDE5A and PDE9A. Transl Psychiatry 2016; 6:e901. [PMID: 27676442 PMCID: PMC5048211 DOI: 10.1038/tp.2016.174] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 07/18/2016] [Indexed: 12/31/2022] Open
Abstract
Duchenne muscular dystrophy is caused by mutations in the DYSTROPHIN gene. Although primarily associated with muscle wasting, a significant portion of patients (approximately 25%) are also diagnosed with autism spectrum disorder. We describe social behavioral deficits in dystrophin-deficient mice and present evidence of cerebellar deficits in cGMP production. We demonstrate therapeutic potential for selective inhibitors of the cGMP-specific PDE5A and PDE9A enzymes to restore social behaviors in dystrophin-deficient mice.
Collapse
Affiliation(s)
- M S Alexander
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- Departments of Pediatrics and Genetics, Harvard Medical School, Boston, MA, USA
- The Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - M J Gasperini
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - P T Tsai
- The F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - D E Gibbs
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - J M Spinazzola
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- Departments of Pediatrics and Genetics, Harvard Medical School, Boston, MA, USA
| | - J L Marshall
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - M J Feyder
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - M T Pletcher
- Rare Disease Research Unit, Pfizer, Cambridge, MA, USA
| | - E L P Chekler
- Rare Disease Research Unit, Pfizer, Cambridge, MA, USA
| | - C A Morris
- Rare Disease Research Unit, Pfizer, Cambridge, MA, USA
| | - M Sahin
- The F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - J F Harms
- Neuroscience Research Unit, Pfizer Global Research and Development, Cambridge, MA, USA
| | - C J Schmidt
- Neuroscience Research Unit, Pfizer Global Research and Development, Cambridge, MA, USA
| | - R J Kleiman
- The F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - L M Kunkel
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- Departments of Pediatrics and Genetics, Harvard Medical School, Boston, MA, USA
- The Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
- The Manton Center for Orphan Diseases, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| |
Collapse
|
35
|
Hammers DW, Sleeper MM, Forbes SC, Shima A, Walter GA, Sweeney HL. Tadalafil Treatment Delays the Onset of Cardiomyopathy in Dystrophin-Deficient Hearts. J Am Heart Assoc 2016; 5:JAHA.116.003911. [PMID: 27506543 PMCID: PMC5015305 DOI: 10.1161/jaha.116.003911] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Cardiomyopathy is a leading cause of mortality among Duchenne muscular dystrophy patients and lacks effective therapies. Phosphodiesterase type 5 is implicated in dystrophic pathology, and the phosphodiesterase type 5 inhibitor tadalafil has recently been studied in a clinical trial for Duchenne muscular dystrophy. Methods and Results Tadalafil was evaluated for the prevention of cardiomyopathy in the mdx mouse and golden retriever muscular dystrophy dog models of Duchenne muscular dystrophy. Tadalafil blunted the adrenergic response in mdx hearts during a 30‐minute dobutamine challenge, which coincided with cardioprotective signaling, reduced induction of μ‐calpain levels, and decreased sarcomeric protein proteolysis. Dogs with golden retriever muscular dystrophy began daily tadalafil treatment prior to detectable cardiomyopathy and demonstrated preserved cardiac function, as assessed by echocardiography and magnetic resonance imaging at ages 18, 21, and 25 months. Tadalafil treatment improved golden retriever muscular dystrophy histopathological features, decreased levels of the cation channel TRPC6, increased total threonine phosphorylation status of TRPC6, decreased m‐calpain levels and indicators of calpain target proteolysis, and elevated levels of utrophin. In addition, we showed that Duchenne muscular dystrophy patient myocardium exhibited increased TRPC6, m‐calpain, and calpain cleavage products compared with control human myocardium. Conclusions Prophylactic use of tadalafil delays the onset of dystrophic cardiomyopathy, which is likely attributed to modulation of TRPC6 levels and permeability and inhibition of protease content and activity. Consequently, phosphodiesterase type 5 inhibition is a candidate therapy for slowing the development of cardiomyopathy in Duchenne muscular dystrophy patients.
Collapse
Affiliation(s)
- David W Hammers
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL Myology Institute, University of Florida College of Medicine, Gainesville, FL
| | - Margaret M Sleeper
- Myology Institute, University of Florida College of Medicine, Gainesville, FL Clinical Studies, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA Small Animal Clinical Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL
| | - Sean C Forbes
- Myology Institute, University of Florida College of Medicine, Gainesville, FL Physical Therapy, University of Florida, Gainesville, FL
| | - Ai Shima
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Glenn A Walter
- Myology Institute, University of Florida College of Medicine, Gainesville, FL Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL
| | - H Lee Sweeney
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL Myology Institute, University of Florida College of Medicine, Gainesville, FL
| |
Collapse
|
36
|
Sanges S, Launay D, Rhee RL, Sitbon O, Hachulla É, Mouthon L, Guillevin L, Rottat L, Montani D, De Groote P, Cottin V, Magro P, Prévot G, Bauer F, Bergot E, Chabanne C, Reynaud-Gaubert M, Leroy S, Canuet M, Sanchez O, Gut-Gobert C, Dauphin C, Pison C, Boissin C, Habib G, Clerson P, Conesa F, Cordier JF, Kawut SM, Simonneau G, Humbert M. A prospective study of the 6 min walk test as a surrogate marker for haemodynamics in two independent cohorts of treatment-naïve systemic sclerosis-associated pulmonary arterial hypertension. Ann Rheum Dis 2016; 75:1457-65. [PMID: 26324844 DOI: 10.1136/annrheumdis-2015-207336] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 08/12/2015] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Despite the wide use of the 6 min walk distance (6MWD), no study has ever assessed its validity as a surrogate marker for haemodynamics and predictor of outcome in isolated pulmonary arterial hypertension associated with systemic sclerosis (SSc-PAH). We designed this work to address this issue. METHODS Treatment-naïve patients with SSc-PAH were prospectively included from two sources: the French PAH Network (a prospective epidemiological cohort) (n=83) and randomised clinical trials submitted for drug approval (Food and Drug Administration) (n=332). Correlations between absolute values of the 6MWD and haemodynamics at baseline, as well as between variations of 6MWD and haemodynamics during follow-up, were studied in both populations. RESULTS In the French cohort, baseline cardiac output (CO) (R(2)=0.19, p=0.001) and New York Heart Association class (R(2)=0.10, p<0.001) were significantly and independently correlated with baseline 6MWD in multivariate analysis. A significant, independent, but weaker, correlation with CO was also found in the Food and Drug Administration sample (R(2)=0.04, p<0.001). During follow-up, there was no association between the changes in 6MWD and haemodynamic parameters in patients under PAH-specific treatments. CONCLUSIONS In SSc-PAH, CO independently correlates with 6MWD at baseline, but accounts for a small amount of the variance of 6MWD in both study samples. This suggests that other non-haemodynamic factors could have an impact on the walk distance. Moreover, variations of 6MWD do not reflect changes in haemodynamics among treated patients. Our results suggest that 6MWD is not an accurate surrogate marker for haemodynamic severity, nor an appropriate outcome measure to assess changes in haemodynamics during follow-up in treated SSc-PAH.
Collapse
Affiliation(s)
- Sébastien Sanges
- Université de Lille, UFR Médecine, Lille, France Département de Médecine Interne et Immunologie Clinique, CHRU Lille, Pôle Spécialités Médicales et Gérontologie, Lille Cedex, France Centre National de Référence Maladies Systémiques et Auto-immunes Rares (Sclérodermie Systémique), Lille Cedex, France LIRIC, INSERM UMR 995, EA2686, Lille, France
| | - David Launay
- Université de Lille, UFR Médecine, Lille, France Département de Médecine Interne et Immunologie Clinique, CHRU Lille, Pôle Spécialités Médicales et Gérontologie, Lille Cedex, France Centre National de Référence Maladies Systémiques et Auto-immunes Rares (Sclérodermie Systémique), Lille Cedex, France LIRIC, INSERM UMR 995, EA2686, Lille, France
| | - Rennie L Rhee
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Olivier Sitbon
- Faculté de Médecine, Université Paris-Sud, Le Kremlin-Bicêtre, France AP-HP, Service de Pneumologie, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France INSERM U999, Centre Chirurgical Marie-Lannelongue, LabEx LERMIT, Le Plessis-Robinson, France
| | - Éric Hachulla
- Université de Lille, UFR Médecine, Lille, France Département de Médecine Interne et Immunologie Clinique, CHRU Lille, Pôle Spécialités Médicales et Gérontologie, Lille Cedex, France Centre National de Référence Maladies Systémiques et Auto-immunes Rares (Sclérodermie Systémique), Lille Cedex, France LIRIC, INSERM UMR 995, EA2686, Lille, France
| | - Luc Mouthon
- Service de Médecine Interne, Centre de Référence des Vascularites Nécrosantes et de la Sclérodermie Systémique, Université Paris Descartes, Hôpital Cochin, Paris, France
| | - Loïc Guillevin
- Service de Médecine Interne, Centre de Référence des Vascularites Nécrosantes et de la Sclérodermie Systémique, Université Paris Descartes, Hôpital Cochin, Paris, France
| | - Laurence Rottat
- Faculté de Médecine, Université Paris-Sud, Le Kremlin-Bicêtre, France AP-HP, Service de Pneumologie, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France INSERM U999, Centre Chirurgical Marie-Lannelongue, LabEx LERMIT, Le Plessis-Robinson, France
| | - David Montani
- Faculté de Médecine, Université Paris-Sud, Le Kremlin-Bicêtre, France AP-HP, Service de Pneumologie, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France INSERM U999, Centre Chirurgical Marie-Lannelongue, LabEx LERMIT, Le Plessis-Robinson, France
| | - Pascal De Groote
- Université de Lille, UFR Médecine, Lille, France Pôle Cardio-Vasculaire et Pulmonaire, Clinique de Cardiologie, CHRU de Lille, Lille, France
| | - Vincent Cottin
- Service de Pneumologie, Hospices Civils de Lyon, Centre de Compétence de l'Hypertension Pulmonaire, Centre de Référence des Maladies Pulmonaires Rares, Lyon, France
| | - Pascal Magro
- Service de Pneumologie, Centre Hospitalier Régional Universitaire, Tours, France
| | - Grégoire Prévot
- Pôle des Voies Respiratoires, Hôpital Larrey, Centre Hospitalier Universitaire, Toulouse, France
| | - Fabrice Bauer
- Service de Cardiologie, Hôpital Charles Nicolle, Centre Hospitalier Universitaire, Rouen, France
| | - Emmanuel Bergot
- Service de Pneumologie, Centre Hospitalier Universitaire Côte-de-Nacre, Caen, France
| | - Céline Chabanne
- Service de Chirurgie Thoracique et Cardiovasculaire, Centre Hospitalier Universitaire Pontchaillou, Université de Rennes I, Rennes, France
| | - Martine Reynaud-Gaubert
- Service de Pneumologie, Centre Hospitalier Universitaire Nord, APHM, Université de la Méditerranée, Marseille, France
| | - Sylvie Leroy
- Service de Pneumologie, Hôpital Pasteur, Centre Hospitalier Universitaire, Université de Nice Sophia Antipolis, Nice, France
| | - Matthieu Canuet
- Service de Pneumologie, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Strasbourg, France
| | - Olivier Sanchez
- AP-HP, Service de Pneumologie et Soins Intensifs, Université Paris Descartes, Hôpital Européen Georges-Pompidou, INSERM UMR-S 1140, Paris, France
| | - Christophe Gut-Gobert
- Service de Médecine Interne et Pneumologie, Centre Hospitalier Universitaire La Cavale Blanche, Brest, France
| | - Claire Dauphin
- Service de Cardiologie et Maladies Vasculaires, Hôpital Gabriel Montpied, Centre Hospitalier Universitaire, Clermont-Ferrand, France
| | - Christophe Pison
- Clinique Universitaire de Pneumologie, Centre Hospitalier Universitaire, Grenoble, France Université Joseph Fourier, Grenoble, France
| | - Clément Boissin
- Service des Maladies Respiratoires, Hôpital Arnaud-de-Villeneuve, Centre Hospitalier Universitaire, Montpellier, France
| | - Gilbert Habib
- Service de Cardiologie, Hôpital de la Timone, Centre Hospitalier Universitaire, Marseille, France
| | - Pierre Clerson
- Soladis Clinical Studies, Biostatistics, Roubaix, France
| | | | - Jean-François Cordier
- Pôle Cardio-Vasculaire et Pulmonaire, Clinique de Cardiologie, CHRU de Lille, Lille, France
| | - Steven M Kawut
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gerald Simonneau
- Faculté de Médecine, Université Paris-Sud, Le Kremlin-Bicêtre, France AP-HP, Service de Pneumologie, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France INSERM U999, Centre Chirurgical Marie-Lannelongue, LabEx LERMIT, Le Plessis-Robinson, France
| | - Marc Humbert
- Faculté de Médecine, Université Paris-Sud, Le Kremlin-Bicêtre, France AP-HP, Service de Pneumologie, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France INSERM U999, Centre Chirurgical Marie-Lannelongue, LabEx LERMIT, Le Plessis-Robinson, France
| |
Collapse
|
37
|
Cea LA, Puebla C, Cisterna BA, Escamilla R, Vargas AA, Frank M, Martínez-Montero P, Prior C, Molano J, Esteban-Rodríguez I, Pascual I, Gallano P, Lorenzo G, Pian H, Barrio LC, Willecke K, Sáez JC. Fast skeletal myofibers of mdx mouse, model of Duchenne muscular dystrophy, express connexin hemichannels that lead to apoptosis. Cell Mol Life Sci 2016; 73:2583-99. [PMID: 26803842 PMCID: PMC11108387 DOI: 10.1007/s00018-016-2132-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 12/15/2015] [Accepted: 01/07/2016] [Indexed: 02/07/2023]
Abstract
Skeletal muscles of patients with Duchenne muscular dystrophy (DMD) show numerous alterations including inflammation, apoptosis, and necrosis of myofibers. However, the molecular mechanism that explains these changes remains largely unknown. Here, the involvement of hemichannels formed by connexins (Cx HCs) was evaluated in skeletal muscle of mdx mouse model of DMD. Fast myofibers of mdx mice were found to express three connexins (39, 43 and 45) and high sarcolemma permeability, which was absent in myofibers of mdx Cx43(fl/fl)Cx45(fl/fl):Myo-Cre mice (deficient in skeletal muscle Cx43/Cx45 expression). These myofibers did not show elevated basal intracellular free Ca(2+) levels, immunoreactivity to phosphorylated p65 (active NF-κB), eNOS and annexin V/active Caspase 3 (marker of apoptosis) but presented dystrophin immunoreactivity. Moreover, muscles of mdx Cx43(fl/fl)Cx45(fl/fl):Myo-Cre mice exhibited partial decrease of necrotic features (big cells and high creatine kinase levels). Accordingly, these muscles showed similar macrophage infiltration as control mdx muscles. Nonetheless, the hanging test performance of mdx Cx43(fl/fl)Cx45(fl/fl):Myo-Cre mice was significantly better than that of control mdx Cx43(fl/fl)Cx45(fl/fl) mice. All three Cxs found in skeletal muscles of mdx mice were also detected in fast myofibers of biopsy specimens from patients with muscular dystrophy. Thus, reduction of Cx expression and/or function of Cx HCs may be potential therapeutic approaches to abrogate myofiber apoptosis in DMD.
Collapse
Affiliation(s)
- Luis A Cea
- Present: Program of Anatomy and Developmental Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile.
| | - Carlos Puebla
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile
- Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso, Chile
| | - Bruno A Cisterna
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile
- Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso, Chile
| | - Rosalba Escamilla
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile
- Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso, Chile
| | - Aníbal A Vargas
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile
| | - Marina Frank
- Division of Molecular Genetics, Life and Medical Sciences Institute, University of Bonn, 53115, Bonn, Germany
| | | | - Carmen Prior
- Unidad de Genética Molecular-INGEMM, Hospital Universitario La Paz-IdIPAZ, Madrid, Spain
| | - Jesús Molano
- Unidad de Genética Molecular-INGEMM, Hospital Universitario La Paz-IdIPAZ, Madrid, Spain
| | | | - Ignacio Pascual
- Servicio de Neuropediatría, Hospital Universitario La Paz-IdIPAZ, Madrid, Spain
| | - Pía Gallano
- Servicio de Genética, Hospital Santa Creu i Sant Pablo-CIBERER, Barcelona, Spain
| | - Gustavo Lorenzo
- Servicio de Pediatria, "Ramón y Cajal" Hospital-IRYCIS, Madrid, Spain
| | - Héctor Pian
- Servicio de Anatomía Patológica, "Ramón y Cajal" Hospital-IRYCIS, Madrid, Spain
| | - Luis C Barrio
- Unidad de Neurología Experimental, "Ramón y Cajal" Hospital-IRYCIS, Madrid, Spain
| | - Klaus Willecke
- Division of Molecular Genetics, Life and Medical Sciences Institute, University of Bonn, 53115, Bonn, Germany
| | - Juan C Sáez
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile.
- Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso, Chile.
| |
Collapse
|
38
|
McCammon JM, Sive H. Challenges in understanding psychiatric disorders and developing therapeutics: a role for zebrafish. Dis Model Mech 2016; 8:647-56. [PMID: 26092527 PMCID: PMC4486859 DOI: 10.1242/dmm.019620] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The treatment of psychiatric disorders presents three major challenges to the research and clinical community: defining a genotype associated with a disorder, characterizing the molecular pathology of each disorder and developing new therapies. This Review addresses how cellular and animal systems can help to meet these challenges, with an emphasis on the role of the zebrafish. Genetic changes account for a large proportion of psychiatric disorders and, as gene variants that predispose to psychiatric disease are beginning to be identified in patients, these are tractable for study in cellular and animal systems. Defining cellular and molecular criteria associated with each disorder will help to uncover causal physiological changes in patients and will lead to more objective diagnostic criteria. These criteria should also define co-morbid pathologies within the nervous system or in other organ systems. The definition of genotypes and of any associated pathophysiology is integral to the development of new therapies. Cell culture-based approaches can address these challenges by identifying cellular pathology and by high-throughput screening of gene variants and potential therapeutics. Whole-animal systems can define the broadest function of disorder-associated gene variants and the organismal impact of candidate medications. Given its evolutionary conservation with humans and its experimental tractability, the zebrafish offers several advantages to psychiatric disorder research. These include assays ranging from molecular to behavioural, and capability for chemical screening. There is optimism that the multiple approaches discussed here will link together effectively to provide new diagnostics and treatments for psychiatric patients. Summary: In this review, we discuss strengths and limitations of prevalent laboratory models that are used for understanding psychiatric disorders and developing therapeutics, with emphasis on the zebrafish.
Collapse
Affiliation(s)
- Jasmine M McCammon
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA 02142, USA
| | - Hazel Sive
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA 02142, USA Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
39
|
2015 William Allan Award. Am J Hum Genet 2016; 98:419-426. [PMID: 26942278 DOI: 10.1016/j.ajhg.2016.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Indexed: 11/21/2022] Open
|
40
|
Allen DG, Whitehead NP, Froehner SC. Absence of Dystrophin Disrupts Skeletal Muscle Signaling: Roles of Ca2+, Reactive Oxygen Species, and Nitric Oxide in the Development of Muscular Dystrophy. Physiol Rev 2016; 96:253-305. [PMID: 26676145 DOI: 10.1152/physrev.00007.2015] [Citation(s) in RCA: 308] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Dystrophin is a long rod-shaped protein that connects the subsarcolemmal cytoskeleton to a complex of proteins in the surface membrane (dystrophin protein complex, DPC), with further connections via laminin to other extracellular matrix proteins. Initially considered a structural complex that protected the sarcolemma from mechanical damage, the DPC is now known to serve as a scaffold for numerous signaling proteins. Absence or reduced expression of dystrophin or many of the DPC components cause the muscular dystrophies, a group of inherited diseases in which repeated bouts of muscle damage lead to atrophy and fibrosis, and eventually muscle degeneration. The normal function of dystrophin is poorly defined. In its absence a complex series of changes occur with multiple muscle proteins showing reduced or increased expression or being modified in various ways. In this review, we will consider the various proteins whose expression and function is changed in muscular dystrophies, focusing on Ca(2+)-permeable channels, nitric oxide synthase, NADPH oxidase, and caveolins. Excessive Ca(2+) entry, increased membrane permeability, disordered caveolar function, and increased levels of reactive oxygen species are early changes in the disease, and the hypotheses for these phenomena will be critically considered. The aim of the review is to define the early damage pathways in muscular dystrophy which might be appropriate targets for therapy designed to minimize the muscle degeneration and slow the progression of the disease.
Collapse
Affiliation(s)
- David G Allen
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| | - Nicholas P Whitehead
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| | - Stanley C Froehner
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| |
Collapse
|
41
|
Stelter Z, Strakova J, Yellamilli A, Fischer K, Sharpe K, Townsend D. Hypoxia-induced cardiac injury in dystrophic mice. Am J Physiol Heart Circ Physiol 2016; 310:H938-48. [PMID: 26851247 DOI: 10.1152/ajpheart.00917.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/02/2016] [Indexed: 02/07/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a disease of progressive destruction of striated muscle, resulting in muscle weakness with progressive respiratory and cardiac failure. Respiratory and cardiac disease are the leading causes of death in DMD patients. Previous studies have suggested an important link between cardiac dysfunction and hypoxia in the dystrophic heart; these studies aim to understand the mechanism underlying this connection. Here we demonstrate that anesthetized dystrophic mice display significant mortality following acute exposure to hypoxia. This increased mortality is associated with a significant metabolic acidosis, despite having significantly higher levels of arterial Po2 Chronic hypoxia does not result in mortality, but rather is characterized by marked cardiac fibrosis. Studies in isolated hearts reveal that the contractile function of dystrophic hearts is highly susceptible to short bouts of ischemia, but these hearts tolerate prolonged acidosis better than wild-type hearts, indicating an increased sensitivity of the dystrophic heart to hypoxia. Dystrophic hearts display decreased cardiac efficiency and oxygen extraction. Isolated dystrophic cardiomyocytes and hearts have normal levels of FCCP-induced oxygen consumption, and mitochondrial morphology and content are normal in the dystrophic heart. These studies demonstrate reductions in cardiac efficiency and oxygen extraction of the dystrophic heart. The underlying cause of this reduced oxygen extraction is not clear; however, the current studies suggest that large disruptions of mitochondrial respiratory function or coronary flow regulation are not responsible. This finding is significant, as hypoxia is a common and largely preventable component of DMD that may contribute to the progression of the cardiac disease in DMD patients.
Collapse
Affiliation(s)
- Zachary Stelter
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Jana Strakova
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Amritha Yellamilli
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Kaleb Fischer
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Katharine Sharpe
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - DeWayne Townsend
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
42
|
De Arcangelis V, Strimpakos G, Gabanella F, Corbi N, Luvisetto S, Magrelli A, Onori A, Passananti C, Pisani C, Rome S, Severini C, Naro F, Mattei E, Di Certo MG, Monaco L. Pathways Implicated in Tadalafil Amelioration of Duchenne Muscular Dystrophy. J Cell Physiol 2016; 231:224-32. [PMID: 26097015 DOI: 10.1002/jcp.25075] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 06/08/2015] [Indexed: 12/15/2022]
Abstract
Numerous therapeutic approaches for Duchenne and Becker Muscular Dystrophy (DMD and BMD), the most common X-linked muscle degenerative disease, have been proposed. So far, the only one showing a clear beneficial effect is the use of corticosteroids. Recent evidence indicates an improvement of dystrophic cardiac and skeletal muscles in the presence of sustained cGMP levels secondary to a blocking of their degradation by phosphodiesterase five (PDE5). Due to these data, we performed a study to investigate the effect of the specific PDE5 inhibitor, tadalafil, on dystrophic skeletal muscle function. Chronic pharmacological treatment with tadalafil has been carried out in mdx mice. Behavioral and physiological tests, as well as histological and biochemical analyses, confirmed the efficacy of the therapy. We then performed a microarray-based genomic analysis to assess the pattern of gene expression in muscle samples obtained from the different cohorts of animals treated with tadalafil. This scrutiny allowed us to identify several classes of modulated genes. Our results show that PDE5 inhibition can ameliorate dystrophy by acting at different levels. Tadalafil can lead to (1) increased lipid metabolism; (2) a switch towards slow oxidative fibers driven by the up-regulation of PGC-1α; (3) an increased protein synthesis efficiency; (4) a better actin network organization at Z-disk.
Collapse
Affiliation(s)
- Valeria De Arcangelis
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | | | | | - Nicoletta Corbi
- CNR-IBPM, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Armando Magrelli
- National Centre for Rare Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Annalisa Onori
- CNR-IBPM, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Claudio Passananti
- CNR-IBPM, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Cinzia Pisani
- CNR-IBPM, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Sophie Rome
- CarMen Laboratory (INSERM 1060, INRA 1362, INSA), University of Lyon, Lyon, France
| | - Cinzia Severini
- CNR-IBCN, Rome, Italy.,European Brain Research Institute, Rome, Italy
| | - Fabio Naro
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | | | | | - Lucia Monaco
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
43
|
Damon BM, Li K, Bryant ND. Magnetic resonance imaging of skeletal muscle disease. HANDBOOK OF CLINICAL NEUROLOGY 2016; 136:827-42. [PMID: 27430444 DOI: 10.1016/b978-0-444-53486-6.00041-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Neuromuscular diseases often exhibit a temporally varying, spatially heterogeneous, and multifaceted pathology. The goals of this chapter are to describe and evaluate the use of quantitative magnetic resonance imaging (MRI) methods to characterize muscle pathology. The following criteria are used for this evaluation: objective measurement of continuously distributed variables; clear and well-understood relationship to the pathology of interest; sensitivity to improvement or worsening of clinical status; and the measurement properties of accuracy and precision. Two major classes of MRI methods meet all of these criteria: (1) MRI methods for measuring muscle contractile volume or cross-sectional area by combining structural MRI and quantitative fat-water MRI; and (2) an MRI method for characterizing the edema caused by inflammation, the measurement of the transverse relaxation time constant (T2). These methods are evaluated with respect to the four criteria listed above and examples from neuromuscular disorders are provided. Finally, these methods are summarized and synthesized and recommendations for additional quantitative MRI developments are made.
Collapse
Affiliation(s)
- Bruce M Damon
- Vanderbilt University Institute of Imaging Science and the Department of Radiology and Radiological Sciences, Vanderbilt University, Nashville, TN, USA; Departments of Biomedical Engineering and Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| | - Ke Li
- Vanderbilt University Institute of Imaging Science and the Department of Radiology and Radiological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Nathan D Bryant
- Vanderbilt University Institute of Imaging Science and the Department of Radiology and Radiological Sciences, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
44
|
Abstract
Zebrafish cancer models have provided critical insight into understanding the link between aberrant developmental pathways and tumorigenesis. The unique strengths of zebrafish as compared to other vertebrate model systems include the combination of fecundity, readily available and efficient transgenesis techniques, transparency that facilitates in vivo cell lineage tracing, and amenability for high-throughput applications. In addition to early embryo readouts, zebrafish can develop tumors at ages ranging from 2 weeks old to adulthood. Tumorigenesis is driven by genetically introducing oncogenes using selected promoter/tissue-specific expression, with either mosaic expression or with the generation of a stable transgenic line. Here, we detail a research pipeline to facilitate the study of human oncogenes in zebrafish systems. The goals of this approach are to identify conserved developmental pathways that may be critical for tumor development and to create platforms for testing novel therapies.
Collapse
|
45
|
Ryan TE, Schmidt CA, Green TD, Brown DA, Neufer PD, McClung JM. Mitochondrial Regulation of the Muscle Microenvironment in Critical Limb Ischemia. Front Physiol 2015; 6:336. [PMID: 26635622 PMCID: PMC4649016 DOI: 10.3389/fphys.2015.00336] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/02/2015] [Indexed: 01/11/2023] Open
Abstract
Critical limb ischemia (CLI) is the most severe clinical presentation of peripheral arterial disease and manifests as chronic limb pain at rest and/or tissue necrosis. Current clinical interventions are largely ineffective and therapeutic angiogenesis based trials have shown little efficacy, highlighting the dire need for new ideas and novel therapeutic approaches. Despite a decade of research related to skeletal muscle as a determinant of morbidity and mortality outcomes in CLI, very little progress has been made toward an effective therapy aimed directly at the muscle myopathies of this disease. Within the muscle cell, mitochondria are well positioned to modulate the ischemic cellular response, as they are the principal sites of cellular energy production and the major regulators of cellular redox charge and cell death. In this mini review, we update the crucial importance of skeletal muscle to CLI pathology and examine the evolving influence of muscle and endothelial cell mitochondria in the complex ischemic microenvironment. Finally, we discuss the novelty of muscle mitochondria as a therapeutic target for ischemic pathology in the context of the complex co-morbidities often associated with CLI.
Collapse
Affiliation(s)
- Terence E Ryan
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Cameron A Schmidt
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Tom D Green
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - David A Brown
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - P Darrell Neufer
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Joseph M McClung
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| |
Collapse
|
46
|
Nelson MD, Rosenberry R, Barresi R, Tsimerinov EI, Rader F, Tang X, Mason O, Schwartz A, Stabler T, Shidban S, Mobaligh N, Hogan S, Elashoff R, Allen JD, Victor RG. Sodium nitrate alleviates functional muscle ischaemia in patients with Becker muscular dystrophy. J Physiol 2015; 593:5183-200. [PMID: 26437761 DOI: 10.1113/jp271252] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/21/2015] [Indexed: 12/26/2022] Open
Abstract
Becker muscular dystrophy (BMD) is a progressive X-linked muscle wasting disease for which there is no treatment. BMD is caused by in-frame mutations in the gene encoding dystrophin, a structural cytoskeletal protein that also targets other proteins to the sarcolemma. Among these is neuronal nitric oxide synthase mu (nNOSμ), which requires specific spectrin-like repeats (SR16/17) in dystrophin's rod domain and the adaptor protein α-syntrophin for sarcolemmal targeting. When healthy skeletal muscle is exercised, sarcolemmal nNOSμ-derived nitric oxide (NO) attenuates α-adrenergic vasoconstriction, thus optimizing perfusion. In the mdx mouse model of dystrophinopathy, this protective mechanism (functional sympatholysis) is defective, resulting in functional muscle ischaemia. Treatment with a NO-donating non-steroidal anti-inflammatory drug (NSAID) alleviates this ischaemia and improves the murine dystrophic phenotype. In the present study, we report that, in 13 men with BMD, sympatholysis is defective mainly in patients whose mutations disrupt sarcolemmal targeting of nNOSμ, with the vasoconstrictor response measured as a decrease in muscle oxygenation (near infrared spectroscopy) to reflex sympathetic activation. Then, in a single-arm, open-label trial in 11 BMD patients and a double-blind, placebo-controlled cross-over trial in six patients, we show that acute treatment with oral sodium nitrate, an inorganic NO donor without a NSIAD moiety, restores sympatholysis and improves post-exercise hyperaemia (Doppler ultrasound). By contrast, sodium nitrate improves neither sympatholysis, nor hyperaemia in healthy controls. Thus, a simple NO donor recapitulates the vasoregulatory actions of sarcolemmal nNOS in BMD patients, and constitutes a putative novel therapy for this disease.
Collapse
Affiliation(s)
- Michael D Nelson
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ryan Rosenberry
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Rita Barresi
- NSCT Diagnostic & Advisory Service for Rare Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle upon Tyne, UK
| | | | - Florian Rader
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Xiu Tang
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - O'Neil Mason
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Avery Schwartz
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Thomas Stabler
- Duke Institute of Molecular Physiology, Duke University Medical Centre, Durham, NC, USA
| | - Sarah Shidban
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Neigena Mobaligh
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shomari Hogan
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Robert Elashoff
- Department of Biomathematics, University of California Los Angeles, Los Angeles, CA, USA
| | - Jason D Allen
- Clinical Exercise Science Program, Institute of Sport, Exercise and Active Living (ISEAL), Victoria University, Melbourne, VIC, Australia
| | - Ronald G Victor
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
47
|
|
48
|
Swiderski K, Lynch GS. Therapeutic potential of orphan drugs for the rare skeletal muscle diseases. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1085858] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
49
|
McCammon JM, Sive H. Addressing the Genetics of Human Mental Health Disorders in Model Organisms. Annu Rev Genomics Hum Genet 2015; 16:173-97. [DOI: 10.1146/annurev-genom-090314-050048] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Jasmine M. McCammon
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142;
| | - Hazel Sive
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142;
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| |
Collapse
|
50
|
Guiraud S, Aartsma-Rus A, Vieira NM, Davies KE, van Ommen GJB, Kunkel LM. The Pathogenesis and Therapy of Muscular Dystrophies. Annu Rev Genomics Hum Genet 2015; 16:281-308. [DOI: 10.1146/annurev-genom-090314-025003] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Simon Guiraud
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy, and Genetics, University of Oxford, OX1 3PT Oxford, United Kingdom; ,
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; ,
| | - Natassia M. Vieira
- Division of Genetics and Genomics and Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts 02115
- Departments of Pediatrics and Genetics, Harvard Medical School, Boston, Massachusetts 02115; ,
| | - Kay E. Davies
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy, and Genetics, University of Oxford, OX1 3PT Oxford, United Kingdom; ,
| | - Gert-Jan B. van Ommen
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; ,
| | - Louis M. Kunkel
- Division of Genetics and Genomics and Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts 02115
- Departments of Pediatrics and Genetics, Harvard Medical School, Boston, Massachusetts 02115; ,
| |
Collapse
|