32 results on '"Osman EY"'
Search Results
2. Efficacy of the cardiac glycoside digoxin as an adjunct to csDMARDs in rheumatoid arthritis patients: a randomized, double-blind, placebo-controlled trial.
- Author
-
El-Mahdy NA, Tadros MG, El-Masry TA, Binsaleh AY, Alsubaie N, Alrossies A, Abd Elhamid MI, Osman EY, Shalaby HM, and Saif DS
- Abstract
Background: Inflammation and angiogenesis are two main mechanisms that act as mutual pathways in rheumatoid arthritis (RA). This work aimed to study the efficacy of digoxin as an adjunct therapy to conventional synthetic disease-modifying anti-rheumatic drugs (csDMARDs) in active RA patients., Methods: In a randomized, double-blinded, placebo-controlled study, 60 adult patients with active RA received a placebo or digoxin (0.25 mg every other day) combined with csDMARDs for 6 months. The American College of Rheumatology (ACR) 20, ACR50, and ACR70 response rates and the disease activity score (DAS28) were assessed for patients. Flow cytometric analysis of Th17 cells and serum concentrations of IL-17A, IL-23, HIF-1α, and VEGF were evaluated before and after three and 6 months of therapy., Results: Following three and 6 months of digoxin therapy combined with csDMARDs, significant differences were detected in laboratory and clinical parameters relative to the control group. After 6 months, 83.3% of patients in the digoxin group, compared to 56.7% in the control group, achieved an ACR20 response ( p = 0.024). The digoxin group had a significantly higher percentage of patients who achieved DAS28 remission after 6 months ( p = 0.024). Notable improvements in the Health Assessment Questionnaire Disability Index, ACR50, and ACR70 were detected in the digoxin group., Conclusion: Digoxin was well tolerated and exerted profound immunomodulatory and anti-inflammatory effects in RA patients, and may also exhibit anti-angiogenic properties, indicating that it might be an effective adjunct to csDMARDs in treating RA., Clinical Trial Registration: clinicaltrials.gov, identifier NCT04834557., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 El-Mahdy, Tadros, El-Masry, Binsaleh, Alsubaie, Alrossies, Abd Elhamid, Osman, Shalaby and Saif.)
- Published
- 2024
- Full Text
- View/download PDF
3. Hepatoprotective Effect of Camel Thorn Polyphenols in Concanavalin A-Induced Hepatitis in Mice.
- Author
-
El-Mahdy NA, El-Masry TA, El-Tarahony AM, Alherz FA, and Osman EY
- Subjects
- Animals, Mice, Oxidative Stress drug effects, Male, Protective Agents pharmacology, Chemical and Drug Induced Liver Injury drug therapy, Chemical and Drug Induced Liver Injury pathology, Chemical and Drug Induced Liver Injury prevention & control, Apoptosis drug effects, Hepatitis drug therapy, Hepatitis pathology, Antioxidants pharmacology, CD4-Positive T-Lymphocytes drug effects, Inflammation Mediators metabolism, Concanavalin A, Polyphenols pharmacology, Liver pathology, Liver drug effects, Plant Extracts pharmacology, Plant Extracts therapeutic use, Camelus
- Abstract
Objectives: To explore the prophylactic and therapeutic effects of Alhagi maurorum ethanolic extract (AME) in concanavalin A (Con A)-induced hepatitis (CIH) as well as possible underlying mechanisms., Methods: Polyphenols in AME were characterized using high performance liquid chromatography (HPLC). Swiss albino mice were divided into 4 groups. Normal group received intravenous phosphate-buffered saline (PBS); Con A group received 40 mg/kg intravenous Con A. Prophylaxis group administered 300 mg/(kg·d) AME orally for 5 days before Con A intervention. Treatment group received intravenous Con A then administered 300 mg/kg AME at 30 min and 3 h after Con A intervention. After 24 h of Con A injection, hepatic injury, oxidative stress, and inflammatory mediators were assessed. Histopathological examination and markers of apoptosis, inflammation, and CD4
+ cell infiltration were also investigated., Results: HPLC analysis revealed that AME contains abundant polyphenols with pharmacological constituents, such as ellagic acid, gallic acid, ferulic acid, methylgallate, and naringenin. AME alleviated Con A-induced hepatic injury, as manifested by a significant reduction in alanine aminotransferase, aspartate aminotransferase and alkaline phosphatase (P<0.01). Additionally, the antioxidant effect of AME was revealed by a significant reduction in oxidative stress markers (nitric oxide and malondialdehyde) and restored glutathione (P<0.01). The levels of proinflammatory cytokines (tumor necrosis factor-α, interferon-γ, and interleukin-6) and c-Jun N-terminal kinase (JNK) activity were reduced (P<0.01). Histopathological examination of liver tissue showed that AME significantly ameliorated necrotic and inflammatory lesions induced by Con A (P<0.01). Moreover, AME reduced the expression of nuclear factor kappa B, pro-apoptotic protein (Bax), caspase-3, and CD4+ T cell hepatic infiltration (P<0.01). The expression of anti-apoptotic protein Bcl-2 was increased (P<0.01)., Conclusion: AME has hepatoprotective and ameliorative effects in CIH mice. These beneficial effects are likely due to the anti-inflammatory, antioxidant, and anti-apoptotic effects of the clinically important polyphenolic content. AME could be a novel and promising hepatoprotective agent for managing immune-mediated hepatitis., Competing Interests: Conflict of Interest. The authors declare that there are no competing interests regarding the publication of this paper., (© 2024. The Chinese Journal of Integrated Traditional and Western Medicine Press and Springer-Verlag GmbH Germany, part of Springer Nature.)- Published
- 2024
- Full Text
- View/download PDF
4. TLR4 inhibitors through inhibiting (MYD88-TRIF) pathway, protect against experimentally-induced intestinal (I/R) injury.
- Author
-
Osman EY, Abdelghafar HI, and Elsisi AE
- Subjects
- Animals, Rats, Male, Intestines drug effects, Intestines pathology, Toll-Like Receptor 4 metabolism, Toll-Like Receptor 4 antagonists & inhibitors, Myeloid Differentiation Factor 88 metabolism, Myeloid Differentiation Factor 88 antagonists & inhibitors, Reperfusion Injury drug therapy, Reperfusion Injury metabolism, Adaptor Proteins, Vesicular Transport metabolism, Signal Transduction drug effects
- Abstract
Intestinal ischemia/reperfusion (I/R) injury is a serious condition that causes intestinal dysfunction and can be fatal. Previous research has shown that toll-like receptor 4 (TLR4) inhibitors have a protective effect against this injury. This study aimed to investigate the protective effects of TLR4 inhibitors, specifically cyclobenzaprine, ketotifen, amitriptyline, and naltrexone, in rats with intestinal (I/R) injury. Albino rats were divided into seven groups: vehicle control, sham-operated, I/R injury, I/R-cyclobenzaprine (10 mg/kg body weight), I/R-ketotifen (1 mg/kg body weight), I/R-amitriptyline (10 mg/kg body weight), and I/R-naltrexone (4 mg/kg body weight) groups. Anesthetized rats (urethane 1.8 g/kg) underwent 30 min of intestinal ischemia by occluding the superior mesenteric artery (SMA), followed by 2 h of reperfusion. Intestinal tissue samples were collected to measure various parameters, including malondialdehyde (MDA), nitric oxide synthase (NO), myeloperoxidase (MPO), superoxide dismutase (SOD), TLR4, intercellular adhesion molecule-1 (ICAM-1), nuclear factor kappa bp65 (NF-ĸBP65), monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-α (TNF-α), macrophages CD68, myeloid differentiation factor 88 (MYD88), and toll interleukin receptor-domain-containing adaptor-inducing interferon β (TRIF). The use of TLR4 inhibitors significantly reduced MDA, MPO, and NO levels, while increasing SOD activity. Furthermore, it significantly decreased TLR4, ICAM-1, TNF-α, MCP-1, MYD88, and TRIF levels. These drugs also showed partial restoration of normal cellular structure with reduced inflammation. Additionally, there was a decrease in NF-ĸBP65 and macrophages CD68 staining compared to rats in the I/R groups. This study focuses on how TLR4 inhibitors enhance intestinal function and protect against intestinal (I/R) injury by influencing macrophages CD86 through (MYD88-TRIF) pathway, as well as their effects on oxidation and inflammation., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
5. Potential Antitumor Activity of Combined Lycopene and Sorafenib against Solid Ehrlich Carcinoma via Targeting Autophagy and Apoptosis and Suppressing Proliferation.
- Author
-
El-Masry TA, El-Nagar MMF, El Mahdy NA, Alherz FA, Taher R, and Osman EY
- Abstract
An FDA-approved kinase inhibitor called sorafenib (SOR) is used to treat primary kidney and liver cancer as well as to stop the spread of advanced breast cancer. Side effects from SOR, such as palmar-plantar erythrodysesthesia syndrome, can negatively impact an individual's quality of life. There are a lot of data supporting the importance of lycopene (LYC) in preventing cancer. The antitumor properties of the combination of sorafenib and lycopene were examined in this study. A viability test against MDA-MB-231 was used to assess the anticancer efficacy of sorafenib, lycopene, and their combination in vitro. Moreover, a cell cycle analysis and Annexin-V/PI double staining were performed by using flow cytometry. In addition, the protein level of JNK-1, ERK-1, Beclin-1, P38, and P53 of the MDA-MB-231 cell line was estimated using ELISA kits. In addition, mice with SEC were divided into four equal groups at random ( n = 10) to investigate the possible processes underlying the in vivo antitumor effect. Group IV (SEC-SOR-LYC) received SOR (30 mg/kg/day, p.o.) and LYC (20 mg/kg/day, p.o.); Group I received the SEC control; Group II received SEC-SOR (30 mg/kg/day, p.o.); and Group III received SEC-LYC (20 mg/kg/day, p.o.). The findings demonstrated that the combination of sorafenib and lycopene was superior to sorafenib and lycopene alone in causing early cell cycle arrest, suppressing the viability of cancer cells, and increasing cell apoptosis and autophagy. Likewise, the combination of sorafenib and lycopene demonstrated inhibition of the levels of Bcl-2, Ki-67, VEGF, IL-1β, and TNF-α protein. Otherwise, the quantities of the proteins BAX, P53, and caspase 3 were amplified. Furthermore, the combined treatment led to a substantial increase in TNF-α , caspase 3 , and VEGF gene expression compared to the equivalent dosages of monotherapy. The combination of sorafenib and lycopene enhanced apoptosis and reduced inflammation, as seen by the tumor's decreased weight and volume, hence demonstrating its potential anticancer effect.
- Published
- 2024
- Full Text
- View/download PDF
6. Quantitative Attribution of the Protective Effects of Aminosterols against Protein Aggregates to Their Chemical Structures and Ability to Modulate Biological Membranes.
- Author
-
Errico S, Lucchesi G, Odino D, Osman EY, Cascella R, Neri L, Capitini C, Calamai M, Bemporad F, Cecchi C, Kinney WA, Barbut D, Relini A, Canale C, Caminati G, Limbocker R, Vendruscolo M, Zasloff M, and Chiti F
- Subjects
- Humans, Cell Membrane metabolism, Amyloidogenic Proteins chemistry, Lipids, Lipid Bilayers metabolism, Amyloid beta-Peptides metabolism, Protein Aggregates, Neurodegenerative Diseases metabolism
- Abstract
Natural aminosterols are promising drug candidates against neurodegenerative diseases, like Alzheimer and Parkinson, and one relevant protective mechanism occurs via their binding to biological membranes and displacement or binding inhibition of amyloidogenic proteins and their cytotoxic oligomers. We compared three chemically different aminosterols, finding that they exhibited different (i) binding affinities, (ii) charge neutralizations, (iii) mechanical reinforcements, and (iv) key lipid redistributions within membranes of reconstituted liposomes. They also had different potencies (EC
50 ) in protecting cultured cell membranes against amyloid-β oligomers. A global fitting analysis led to an analytical equation describing quantitatively the protective effects of aminosterols as a function of their concentration and relevant membrane effects. The analysis correlates aminosterol-mediated protection with well-defined chemical moieties, including the polyamine group inducing a partial membrane-neutralizing effect (79 ± 7%) and the cholestane-like tail causing lipid redistribution and bilayer mechanical resistance (21 ± 7%), linking quantitatively their chemistry to their protective effects on biological membranes.- Published
- 2023
- Full Text
- View/download PDF
7. Motor unit recovery following Smn restoration in mouse models of spinal muscular atrophy.
- Author
-
Comley LH, Kline RA, Thomson AK, Woschitz V, Landeros EV, Osman EY, Lorson CL, and Murray LM
- Subjects
- Animals, Disease Models, Animal, Mice, Motor Neurons metabolism, Oligonucleotides pharmacology, Oligonucleotides, Antisense pharmacology, Survival of Motor Neuron 1 Protein genetics, Survival of Motor Neuron 1 Protein metabolism, Muscular Atrophy, Spinal genetics, Muscular Atrophy, Spinal metabolism, Muscular Atrophy, Spinal therapy, Tumor Suppressor Protein p53 metabolism
- Abstract
Spinal muscular atrophy (SMA) is a childhood motor neuron disease caused by anomalies in the SMN1 gene. Although therapeutics have been approved for the treatment of SMA, there is a therapeutic time window, after which efficacy is reduced. Hallmarks of motor unit pathology in SMA include loss of motor-neurons and neuromuscular junction (NMJs). Following an increase in Smn levels, it is unclear how much damage can be repaired and the degree to which normal connections are re-established. Here, we perform a detailed analysis of motor unit pathology before and after restoration of Smn levels. Using a Smn-inducible mouse model of SMA, we show that genetic restoration of Smn results in a dramatic reduction in NMJ pathology, with restoration of innervation patterns, preservation of axon and endplate number and normalized expression of P53-associated transcripts. Notably, presynaptic swelling and elevated Pmaip levels remained. We analysed the effect of either early or delayed treated of an antisense oligonucleotide (ASO) targeting SMN2 on a range of differentially vulnerable muscles. Following ASO administration, the majority of endplates appeared fully occupied. However, there was an underlying loss of axons and endplates, which was more prevalent following a delay in treatment. There was an increase in average motor unit size following both early and delayed treatment. Together this work demonstrates the remarkably regenerative capacity of the motor neuron following Smn restoration, but highlights that recovery is incomplete. This work suggests that there is an opportunity to enhance neuromuscular junction recovery following administration of Smn-enhancing therapeutics., (© The Author(s) 2022. Published by Oxford University Press.)
- Published
- 2022
- Full Text
- View/download PDF
8. Protective effect of cilostazol and verapamil against thioacetamide-induced hepatotoxicity in rats may involve Nrf2/GSK-3β/NF-κB signaling pathway.
- Author
-
Elsisi AE, Elmarhoumy EH, and Osman EY
- Abstract
Background: Verapamil (VER) and cilostazol (Cilo) are mostly used as cardiovascular drugs; they have beneficial effects on different organs toxicities., Aim: we investigated whether the Nuclear factor erythroid 2-related factor 2 (Nrf2), Glycogen synthase kinase-3β (GSK-3β), and Nuclear factor-kappa B (NF-κB) pathway involved in the protective role of these drugs against Thioacetamide (TAA) induced hepatotoxicity., Method: male rats were randomized divided into five groups, each group ( n = 10): control, TAA, VER+TAA, Cilo+TAA, and VER+Cilo+TAA groups. Hepatotoxicity induced in rats by TAA injection once on the 7th day of the experiment., Results: TAA-induced hepatotoxicity indicated by a significant elevated in serum markers (Alanine aminotransferases (ALT), Aspartate aminotransferases (AST), and bilirubin), oxidative stress markers (Malondialdehyde (MDA), and Nitric oxide (NO)), and protein levels markers (NF-κB, and S100 calcium-binding protein A4 (S100A4)). Also, TAA decreased Nrf2, and increased GSK-3β genes expression. Histopathological alterations in the liver also appeared as a response to TAA injection. On the other hand VER and/or Cilo significantly prevented TAA-induced hepatotoxicity in rats through significantly decreased in ALT, AST, bilirubin, MDA, NO, NF-κB, and S100A4 protein levels. Also, they increased Nrf2 and decreased GSK-3β genes expression which caused improvement in the histopathological changes of the liver., Conclusion: the addition of verapamil to cilostazol potentiated the hepatoprotective activity, and inhibited the progression of hepatotoxicity caused by TAA through the Nrf2/GSK-3β/NF-κBpathway and their activity on oxidative stress, inflammation, and NF-κB protein expression., (© The Author(s) 2022. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
9. Dexamethasone and losartan combination treatment protected cigarette smoke-induced COPD in rats.
- Author
-
Sokar SS, Afify EH, and Osman EY
- Subjects
- Angiotensin II Type 1 Receptor Blockers pharmacology, Animals, Anti-Inflammatory Agents pharmacology, Bronchoalveolar Lavage Fluid chemistry, Bronchoalveolar Lavage Fluid cytology, C-Reactive Protein analysis, Cell Count, Dexamethasone pharmacology, Drug Therapy, Combination, Intercellular Adhesion Molecule-1 metabolism, Losartan pharmacology, Lung drug effects, Lung metabolism, Lung pathology, Male, Malondialdehyde metabolism, Matrix Metalloproteinase 9 metabolism, NF-kappa B metabolism, Oxidative Stress drug effects, Pulmonary Disease, Chronic Obstructive pathology, Rats, Sprague-Dawley, Smoke, Superoxide Dismutase metabolism, Nicotiana, Rats, Angiotensin II Type 1 Receptor Blockers therapeutic use, Anti-Inflammatory Agents therapeutic use, Dexamethasone therapeutic use, Losartan therapeutic use, Pulmonary Disease, Chronic Obstructive drug therapy
- Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a dangerous prevalent smoking-related disease characterized by abnormal inflammation and oxidative stress and expected to be the third cause of death in the world next decade. Corticosteroids have low effects in decreasing numbers of inflammatory mediators specifically in long-term use. Our study designed to investigate the possible protective effects of combined dexamethasone (Dex) (2mg/kg) and losartan (Los) (30mg/kg angiotensin receptor blocker, it possesses antioxidant and anti-inflammatory properties in lung injury in mice) against cigarette -smoke (CS) induced COPD in rats compared with dexamethasone and losartan. Male Sprague Dawley rats (N = 40) divided into five groups (n = 8): control group, CS group, Dex group, Los group, and Dex +Los group. COPD induced in rats by CS exposure twice daily for 10 weeks. After the specified treatment period, bronchoalveolar lavage fluid (BALF) and lung tissue were collected for measurement of SOD, NO, MDA, ICAM-, MMP-9, CRP, NF-κB and histopathology scoring. Our results indicated that Los+Dex significantly prevent CS-induced COPD emphysema, congested alveoli, and elevation of lung injury parameters in BALF. They also showed a significant decrease in MDA, ICAM-1, MMP-9, CRP, and NF-κB and a significant increase in SOD and NO. In conclusion, adding Los to Dex potentiating their activity in inhibition the progression of COPD based on its activity on oxidative stress, inflammation, and NF-κB protein expression.
- Published
- 2021
- Full Text
- View/download PDF
10. Polymeric nanoencapsulation of zaleplon into PLGA nanoparticles for enhanced pharmacokinetics and pharmacological activity.
- Author
-
Haggag YA, Abosalha AK, Tambuwala MM, Osman EY, El-Gizawy SA, Essa EA, and Donia AA
- Subjects
- Acetamides pharmacokinetics, Acetamides pharmacology, Animals, Biological Availability, Hypnotics and Sedatives pharmacology, Male, Pyrimidines pharmacokinetics, Pyrimidines pharmacology, Rabbits, Rats, gamma-Aminobutyric Acid blood, Acetamides chemistry, Anticonvulsants chemistry, Nanoparticles chemistry, Polylactic Acid-Polyglycolic Acid Copolymer chemistry, Pyrimidines chemistry
- Abstract
Zaleplon (ZP) is a sedative and hypnotic drug used for the treatment of insomnia. Despite its potent anticonvulsant activity, ZP is not commonly used for the treatment of convulsion since ZP is characterized by its low oral bioavailability as a result of poor solubility and extensive liver metabolism. The following study aimed to formulate specifically controlled release nano-vehicles for oral and parenteral delivery of ZP to enhance its oral bioavailability and biological activity. A modified single emulsification-solvent evaporation method of sonication force was adopted to optimize the inclusion of ZP into biodegradable nanoparticles (NPs) using poly (dl-lactic-co-glycolic acid) (PLGA). The impacts of various formulation variables on the physicochemical characteristics of the ZP-PLGA-NPs and drug release profiles were investigated. Pharmacokinetics and pharmacological activity of ZP-PLGA-NPs were studied using experimental animals and were compared with generic ZP tablets. Assessment of gamma-aminobutyric acid (GABA) level in plasma after oral administration was conducted using enzyme-linked immunosorbent assay. The maximal electroshock-induced seizures model evaluated anticonvulsant activity after the parenteral administration of ZP-loaded NPs. The prepared ZP-PLGA NPs were negatively charged spherical particles with an average size of 120-300 nm. Optimized ZP-PLGA NPs showed higher plasma GABA levels, longer sedative, hypnotic effects, and a 3.42-fold augmentation in oral drug bioavailability in comparison to ZP-marketed products. Moreover, parenteral administration of ZP-NPs showed higher anticonvulsant activity compared to free drug. Oral administration of ZP-PLGA NPs achieved a significant improvement in the drug bioavailability, and parenteral administration showed a pronounced anticonvulsant activity., (© 2021 The Authors. Biopharmaceutics & Drug Disposition published by John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
11. Short-duration splice promoting compound enables a tunable mouse model of spinal muscular atrophy.
- Author
-
Rietz A, Hodgetts KJ, Lusic H, Quist KM, Osman EY, Lorson CL, and Androphy EJ
- Subjects
- Animals, Animals, Newborn, Cell Survival drug effects, Disease Models, Animal, Dose-Response Relationship, Drug, Kaplan-Meier Estimate, Mice, Mice, Transgenic, Motor Neurons metabolism, Muscular Atrophy, Spinal metabolism, Phenotype, Survival of Motor Neuron 2 Protein genetics, Survival of Motor Neuron 2 Protein metabolism, Time-to-Treatment, Muscular Atrophy, Spinal drug therapy, Muscular Atrophy, Spinal genetics, Piperidines administration & dosage, Pyrazoles administration & dosage, Pyridazines administration & dosage, RNA Splicing drug effects
- Abstract
Spinal muscular atrophy (SMA) is a motor neuron disease and the leading genetic cause of infant mortality. SMA results from insufficient survival motor neuron (SMN) protein due to alternative splicing. Antisense oligonucleotides, gene therapy and splicing modifiers recently received FDA approval. Although severe SMA transgenic mouse models have been beneficial for testing therapeutic efficacy, models mimicking milder cases that manifest post-infancy have proven challenging to develop. We established a titratable model of mild and moderate SMA using the splicing compound NVS-SM2. Administration for 30 d prevented development of the SMA phenotype in severe SMA mice, which typically show rapid weakness and succumb by postnatal day 11. Furthermore, administration at day eight resulted in phenotypic recovery. Remarkably, acute dosing limited to the first 3 d of life significantly enhanced survival in two severe SMA mice models, easing the burden on neonates and demonstrating the compound as suitable for evaluation of follow-on therapies without potential drug-drug interactions. This pharmacologically tunable SMA model represents a useful tool to investigate cellular and molecular pathogenesis at different stages of disease., (© 2020 Rietz et al.)
- Published
- 2020
- Full Text
- View/download PDF
12. A snapshot of Plasmodium falciparum malaria drug resistance markers in Sudan: a pilot study.
- Author
-
Mohamed NS, Abdelbagi H, Osman HA, Ahmed AE, Yousif AM, Edris YB, Osman EY, Elsadig AR, Siddig EE, Mustafa M, Mohammed AA, Ali Y, Osman MM, Ali MS, Omer RA, Ahmed A, and Sibley CH
- Subjects
- Drug Combinations, Drug Resistance genetics, Humans, Pilot Projects, Plasmodium falciparum genetics, Protozoan Proteins genetics, Pyrimethamine therapeutic use, Sudan, Sulfadoxine therapeutic use, Antimalarials pharmacology, Antimalarials therapeutic use, Malaria, Falciparum drug therapy, Malaria, Falciparum epidemiology
- Abstract
Objectives: Malaria infection is still known to be a worldwide public health problem, especially in tropical and sub-tropical African countries like Sudan. A pilot study conducted to describe the trend of P. falciparum drug resistance markers in 2017-2018 in comparison to CQ and AS/SP eras in Sudan. The Pfcrt, Pfmdr-1, Pfdhfr, and Pfdhps genes were investigated. Data deposited by the worldwide antimalarial resistance network was consulted, and the molecular markers previously reported from Sudan were analyzed., Results: Drug molecular markers analysis was successfully done on 20 P. falciparum isolates. The Pfcrt K76 showed high frequency; 16 (80%). For the Pfmdr-1, 9 (45%) isolates were carrying the N86 allele, and 11 (55%) were 86Y allele. While the Y184F of the Pfmdr-1 showed a higher frequency of 184F compared to Y184; 16 (80%) and 4 (20%), respectively. In the Pfdhfr, 51I allele showed higher frequency compared to N51; 18 (90%) and 2 (10%), respectively. For S108N, 18 (90%) were 108 N and 2 (10%) were S108. In the Pfdhps, all isolates were carrying the mutant alleles; 437G and 540E. The frequency distribution of the Pfcrt, Pfmdr-1, Pfdhfr, Pfdhps was significantly different across the whole years in Sudan.
- Published
- 2020
- Full Text
- View/download PDF
13. Minor snRNA gene delivery improves the loss of proprioceptive synapses on SMA motor neurons.
- Author
-
Osman EY, Van Alstyne M, Yen PF, Lotti F, Feng Z, Ling KK, Ko CP, Pellizzoni L, and Lorson CL
- Subjects
- Animals, Disease Models, Animal, Mice, Muscular Atrophy, Spinal pathology, RNA Splicing genetics, RNA, Messenger metabolism, RNA, Small Nuclear metabolism, Spinal Cord metabolism, Motor Neurons metabolism, Muscular Atrophy, Spinal genetics, RNA, Small Nuclear genetics, Synapses metabolism
- Abstract
Spinal muscular atrophy (SMA) is an inherited neuromuscular disorder caused by reduced expression of the survival motor neuron (SMN) protein. SMN has key functions in multiple RNA pathways, including the biogenesis of small nuclear ribonucleoproteins that are essential components of both major (U2-dependent) and minor (U12-dependent) spliceosomes. Here we investigated the specific contribution of U12 splicing dysfunction to SMA pathology through selective restoration of this RNA pathway in mouse models of varying phenotypic severity. We show that virus-mediated delivery of minor snRNA genes specifically improves select U12 splicing defects induced by SMN deficiency in cultured mammalian cells, as well as in the spinal cord and dorsal root ganglia of SMA mice without increasing SMN expression. This approach resulted in a moderate amelioration of several parameters of the disease phenotype in SMA mice, including survival, weight gain, and motor function. Importantly, minor snRNA gene delivery improved aberrant splicing of the U12 intron-containing gene Stasimon and rescued the severe loss of proprioceptive sensory synapses on SMA motor neurons, which are early signatures of motor circuit dysfunction in mouse models. Taken together, these findings establish the direct contribution of U12 splicing dysfunction to synaptic deafferentation and motor circuit pathology in SMA.
- Published
- 2020
- Full Text
- View/download PDF
14. AAV9-Stathmin1 gene delivery improves disease phenotype in an intermediate mouse model of spinal muscular atrophy.
- Author
-
Villalón E, Kline RA, Smith CE, Lorson ZC, Osman EY, O'Day S, Murray LM, and Lorson CL
- Subjects
- Animals, Dependovirus genetics, Disease Models, Animal, Female, Gene Transfer Techniques, Genetic Therapy methods, Genetic Vectors genetics, Infusions, Intraventricular, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Microtubules metabolism, Motor Neurons metabolism, Muscular Atrophy, Spinal physiopathology, Phenotype, Stathmin metabolism, Survival of Motor Neuron 1 Protein metabolism, Muscular Atrophy, Spinal genetics, Stathmin genetics, Survival of Motor Neuron 1 Protein genetics
- Abstract
Spinal muscular atrophy (SMA) is a devastating infantile genetic disorder caused by the loss of survival motor neuron (SMN) protein that leads to premature death due to loss of motor neurons and muscle atrophy. The approval of an antisense oligonucleotide therapy for SMA was an important milestone in SMA research; however, effective next-generation therapeutics will likely require combinatorial SMN-dependent therapeutics and SMN-independent disease modifiers. A recent cross-disease transcriptomic analysis identified Stathmin-1 (STMN1), a tubulin-depolymerizing protein, as a potential disease modifier across different motor neuron diseases, including SMA. Here, we investigated whether viral-based delivery of STMN1 decreased disease severity in a well-characterized SMA mouse model. Intracerebroventricular delivery of scAAV9-STMN1 in SMA mice at P2 significantly increased survival and weight gain compared to untreated SMA mice without elevating Smn levels. scAAV9-STMN1 improved important hallmarks of disease, including motor function, NMJ pathology and motor neuron cell preservation. Furthermore, scAAV9-STMN1 treatment restored microtubule networks and tubulin expression without affecting tubulin stability. Our results show that scAAV9-STMN1 treatment improves SMA pathology possibly by increasing microtubule turnover leading to restored levels of stable microtubules. Overall, these data demonstrate that STMN1 can significantly reduce the SMA phenotype independent of restoring SMN protein and highlight the importance of developing SMN-independent therapeutics for the treatment of SMA., (© The Author(s) 2019. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
15. Functional characterization of SMN evolution in mouse models of SMA.
- Author
-
Osman EY, Bolding MR, Villalón E, Kaifer KA, Lorson ZC, Tisdale S, Hao Y, Conant GC, Pires JC, Pellizzoni L, and Lorson CL
- Subjects
- Animals, Caenorhabditis elegans, Disease Models, Animal, Drosophila melanogaster, Evolution, Molecular, Mice, Mice, Knockout, Muscular Atrophy, Spinal genetics, Schizosaccharomyces, Survival of Motor Neuron 1 Protein genetics, Xenopus Proteins genetics, Xenopus Proteins metabolism, Xenopus laevis, Zebrafish genetics, Zebrafish Proteins genetics, Zebrafish Proteins metabolism, Muscular Atrophy, Spinal metabolism, Survival of Motor Neuron 1 Protein metabolism
- Abstract
Spinal Muscular Atrophy (SMA) is a monogenic neurodegenerative disorder and the leading genetic cause of infantile mortality. While several functions have been ascribed to the SMN (survival motor neuron) protein, their specific contribution to the disease has yet to be fully elucidated. We hypothesized that some, but not all, SMN homologues would rescue the SMA phenotype in mouse models, thereby identifying disease-relevant domains. Using AAV9 to deliver Smn homologs to SMA mice, we identified a conservation threshold that marks the boundary at which homologs can rescue the SMA phenotype. Smn from Danio rerio and Xenopus laevis significantly prevent disease, whereas Smn from Drosophila melanogaster, Caenorhabditis elegans, and Schizosaccharomyces pombe was significantly less efficacious. This phenotypic rescue correlated with correction of RNA processing defects induced by SMN deficiency and neuromuscular junction pathology. Based upon the sequence conservation in the rescuing homologs, a minimal SMN construct was designed consisting of exons 2, 3, and 6, which showed a partial rescue of the SMA phenotype. While a significant extension in survival was observed, the absence of a complete rescue suggests that while the core conserved region is essential, additional sequences contribute to the overall ability of the SMN protein to rescue disease pathology.
- Published
- 2019
- Full Text
- View/download PDF
16. Intraperitoneal delivery of a novel drug-like compound improves disease severity in severe and intermediate mouse models of Spinal Muscular Atrophy.
- Author
-
Osman EY, Rietz A, Kline RA, Cherry JJ, Hodgetts KJ, Lorson CL, and Androphy EJ
- Subjects
- Animals, Brain drug effects, Brain metabolism, Disease Models, Animal, Injections, Intraperitoneal, Mice, Knockout, Muscular Atrophy, Spinal mortality, Neuromuscular Junction drug effects, Severity of Illness Index, Spinal Cord drug effects, Spinal Cord metabolism, Survival of Motor Neuron 1 Protein genetics, Muscular Atrophy, Spinal drug therapy, Muscular Atrophy, Spinal etiology, Survival of Motor Neuron 1 Protein metabolism
- Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive neurodegenerative disorder that causes progressive muscle weakness and is the leading genetic cause of infant mortality worldwide. SMA is caused by the loss of survival motor neuron 1 (SMN1). In humans, a nearly identical copy gene is present, called SMN2. Although SMN2 maintains the same coding sequence, this gene cannot compensate for the loss of SMN1 because of a single silent nucleotide difference in SMN2 exon 7. SMN2 primarily produces an alternatively spliced isoform lacking exon 7, which is critical for protein function. SMN2 is an important disease modifier that makes for an excellent target for therapeutic intervention because all SMA patients retain SMN2. Therefore, compounds and small molecules that can increase SMN2 exon 7 inclusion, transcription and SMN protein stability have great potential for SMA therapeutics. Previously, we performed a high throughput screen and established a class of compounds that increase SMN protein in various cellular contexts. In this study, a novel compound was identified that increased SMN protein levels in vivo and ameliorated the disease phenotype in severe and intermediate mouse models of SMA.
- Published
- 2019
- Full Text
- View/download PDF
17. Comparison of independent screens on differentially vulnerable motor neurons reveals alpha-synuclein as a common modifier in motor neuron diseases.
- Author
-
Kline RA, Kaifer KA, Osman EY, Carella F, Tiberi A, Ross J, Pennetta G, Lorson CL, and Murray LM
- Subjects
- Amyotrophic Lateral Sclerosis pathology, Animals, Axons metabolism, Axons pathology, Disease Models, Animal, Drosophila melanogaster genetics, Gene Expression Regulation genetics, Humans, Mice, Motor Neuron Disease pathology, Motor Neurons pathology, Muscle, Skeletal metabolism, Muscle, Skeletal pathology, Neuromuscular Junction genetics, Neuromuscular Junction pathology, Phenotype, Rats, Transcriptome genetics, alpha-Synuclein biosynthesis, Amyotrophic Lateral Sclerosis genetics, Motor Neuron Disease genetics, Motor Neurons metabolism, alpha-Synuclein genetics
- Abstract
The term "motor neuron disease" encompasses a spectrum of disorders in which motor neurons are the primary pathological target. However, in both patients and animal models of these diseases, not all motor neurons are equally vulnerable, in that while some motor neurons are lost very early in disease, others remain comparatively intact, even at late stages. This creates a valuable system to investigate the factors that regulate motor neuron vulnerability. In this study, we aim to use this experimental paradigm to identify potential transcriptional modifiers. We have compared the transcriptome of motor neurons from healthy wild-type mice, which are differentially vulnerable in the childhood motor neuron disease Spinal Muscular Atrophy (SMA), and have identified 910 transcriptional changes. We have compared this data set with published microarray data sets on other differentially vulnerable motor neurons. These neurons were differentially vulnerable in the adult onset motor neuron disease Amyotrophic Lateral Sclerosis (ALS), but the screen was performed on the equivalent population of neurons from neurologically normal human, rat and mouse. This cross species comparison has generated a refined list of differentially expressed genes, including CELF5, Col5a2, PGEMN1, SNCA, Stmn1 and HOXa5, alongside a further enrichment for synaptic and axonal transcripts. As an in vivo validation, we demonstrate that the manipulation of a significant number of these transcripts can modify the neurodegenerative phenotype observed in a Drosophila line carrying an ALS causing mutation. Finally, we demonstrate that vector-mediated expression of alpha-synuclein (SNCA), a transcript decreased in selectively vulnerable motor neurons in all four screens, can extend life span, increase weight and decrease neuromuscular junction pathology in a mouse model of SMA. In summary, we have combined multiple data sets to identify transcripts, which are strong candidates for being phenotypic modifiers, and demonstrated SNCA is a modifier of pathology in motor neuron disease.
- Published
- 2017
- Full Text
- View/download PDF
18. Plastin-3 extends survival and reduces severity in mouse models of spinal muscular atrophy.
- Author
-
Kaifer KA, Villalón E, Osman EY, Glascock JJ, Arnold LL, Cornelison DDW, and Lorson CL
- Subjects
- Animals, Dependovirus genetics, Disease Models, Animal, Genetic Vectors, Humans, Membrane Glycoproteins genetics, Mice, Mice, Knockout, Microfilament Proteins genetics, Motor Neurons physiology, Muscle Fibers, Skeletal pathology, Survival Analysis, Survival of Motor Neuron 1 Protein genetics, Membrane Glycoproteins physiology, Microfilament Proteins physiology, Muscular Atrophy, Spinal pathology
- Abstract
Spinal muscular atrophy (SMA) is a leading genetic cause of infantile death and is caused by the loss of survival motor neuron-1 ( SMN1 ). Importantly, a nearly identical gene is present called SMN2 ; however, the majority of SMN2 -derived transcripts are alternatively spliced and encode a truncated, dysfunctional protein. Recently, several compounds designed to increase SMN protein have entered clinical trials, including antisense oligonucleotides (ASOs), traditional small molecules, and gene therapy. Expanding beyond SMN-centric therapeutics is important, as it is likely that the breadth of the patient spectrum and the inherent complexity of the disease will be difficult to address with a single therapeutic strategy. Several SMN-independent pathways that could impinge upon the SMA phenotype have been examined with varied success. To identify disease-modifying pathways that could serve as stand-alone therapeutic targets or could be used in combination with an SMN-inducing compound, we investigated adeno-associated virus-mediated (AAV-mediated) gene therapy using plastin-3 ( PLS3 ). Here, we report that AAV9- PLS3 extends survival in an intermediate model of SMA mice as well as in a pharmacologically induced model of SMA using a splice-switching ASO that increases SMN production. PLS3 coadministration improves the phenotype beyond the ASO, demonstrating the potential utility of combinatorial therapeutics in SMA that target SMN-independent and SMN-dependent pathways., Competing Interests: Conflict of interest: The authors have declared that no conflict of interest exists.
- Published
- 2017
- Full Text
- View/download PDF
19. Analysis of Azithromycin Monohydrate as a Single or a Combinatorial Therapy in a Mouse Model of Severe Spinal Muscular Atrophy.
- Author
-
Osman EY, Washington CW, Simon ME, Megiddo D, Greif H, and Lorson CL
- Subjects
- Animals, Brain pathology, Disease Models, Animal, Mice, Mice, Transgenic, Oligonucleotides, Antisense pharmacology, Spinal Cord drug effects, Spinal Cord pathology, Azithromycin pharmacology, Brain drug effects, Muscular Atrophy, Spinal pathology, Neuroprotective Agents pharmacology
- Abstract
Background: Spinal muscular atrophy (SMA) is a neurodegenerative autosomal recessive disorder characterized by the loss of α-motor neurons. A variety of molecular pathways are being investigated to elevate SMN protein expression in SMA models and in the clinic. One of these approaches involves stabilizing the SMNΔ7 protein by inducing translational read-through. Previous studies have demonstrated that functionality and stability are partially restored to the otherwise unstable SMNΔ7 by the addition of non-specific C-terminal peptide sequences, or by inducing a similar molecular event through the use of read-through inducing compounds such as aminoglycosides., Objective: The objective was to determine the efficacy of the macrolide Azithromycin (AZM), an FDA approved read-through-inducing compound, in the well-established severe mouse model of SMA., Methods: Initially, dosing regimen following ICV administrations of AZM at different post-natal days and concentrations was determined by their impact on SMN levels in disease-relevant tissues. Selected dose was then tested for phenotypic parameters changes as compared to the appropriate controls and in conjugation to another therapy., Results: AZM increases SMN protein in disease relevant tissues, however, this did not translate into similar improvements in the SMA phenotype in a severe mouse model of SMA. Co-administration of AZM and a previously developed antisense oligonucleotide that increases SMN2 splicing, resulted in an improvement in the SMA phenotype beyond either AZM or ASO alone, including a highly significant extension in survival with improvement in body weight and movement., Conclusions: It is important to explore various approaches for SMA therapeutics, hence compounds that specifically induce SMNΔ7 read-through, without having prohibitive toxicity, may provide an alternative platform for a combinatorial treatment. Here we established that AZM activity at a low dose can increase SMN protein in disease-relevant animal model and can impact disease severity.
- Published
- 2017
- Full Text
- View/download PDF
20. Optimization of Morpholino Antisense Oligonucleotides Targeting the Intronic Repressor Element1 in Spinal Muscular Atrophy.
- Author
-
Osman EY, Washington CW 3rd, Kaifer KA, Mazzasette C, Patitucci TN, Florea KM, Simon ME, Ko CP, Ebert AD, and Lorson CL
- Subjects
- Animals, Disease Models, Animal, Gene Expression Regulation, Gene Targeting, Humans, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, Mice, Mice, Knockout, Muscular Atrophy, Spinal metabolism, Muscular Atrophy, Spinal mortality, Mutation, Prognosis, RNA, Messenger genetics, RNA, Messenger metabolism, Survival of Motor Neuron 1 Protein genetics, Survival of Motor Neuron 1 Protein metabolism, Transcription, Genetic, Introns, Morpholinos genetics, Muscular Atrophy, Spinal genetics, Response Elements
- Abstract
Loss of Survival Motor Neuron-1 (SMN1) causes Spinal Muscular Atrophy, a devastating neurodegenerative disease. SMN2 is a nearly identical copy gene; however SMN2 cannot prevent disease development in the absence of SMN1 since the majority of SMN2-derived transcripts are alternatively spliced, encoding a truncated, unstable protein lacking exon 7. Nevertheless, SMN2 retains the ability to produce low levels of functional protein. Previously we have described a splice-switching Morpholino antisense oligonucleotide (ASO) sequence that targets a potent intronic repressor, Element1 (E1), located upstream of SMN2 exon 7. In this study, we have assessed a novel panel of Morpholino ASOs with the goal of optimizing E1 ASO activity. Screening for efficacy in the SMNΔ7 mouse model, a single ASO variant was more active in vivo compared with the original E1(MO)-ASO. Sequence variant eleven (E1(MOv11)) consistently showed greater efficacy by increasing the lifespan of severe Spinal Muscular Atrophy mice after a single intracerebroventricular injection in the central nervous system, exhibited a strong dose-response across an order of magnitude, and demonstrated excellent target engagement by partially reversing the pathogenic SMN2 splicing event. We conclude that Morpholino modified ASOs are effective in modifying SMN2 splicing and have the potential for future Spinal Muscular Atrophy clinical applications.
- Published
- 2016
- Full Text
- View/download PDF
21. Celecoxib and omega-3 fatty acids alone and in combination with risperidone affect the behavior and brain biochemistry in amphetamine-induced model of schizophrenia.
- Author
-
El-Sayed El-Sisi A, Sokkar SS, El-Sayed El-Sayad M, Sayed Ramadan E, and Osman EY
- Subjects
- Amphetamine, Animals, Brain drug effects, Celecoxib pharmacology, Disease Models, Animal, Drug Therapy, Combination, Fatty Acids, Omega-3 pharmacology, Male, Malondialdehyde metabolism, Maze Learning drug effects, Rats, Risperidone pharmacology, Tumor Necrosis Factor-alpha metabolism, Behavior, Animal drug effects, Brain metabolism, Celecoxib therapeutic use, Fatty Acids, Omega-3 therapeutic use, Risperidone therapeutic use, Schizophrenia chemically induced, Schizophrenia drug therapy
- Abstract
The implications of oxidative stress and neuro-inflammation in the pathogenesis of schizophrenia have been elucidated. Despite their effectiveness against positive symptoms of schizophrenia, antipsychotics have limited effectiveness against negative and cognitive symptoms and are associated with remarkable adverse effects. The use of celecoxib or omega-3 in schizophrenia may have beneficial effects. This study aimed to evaluate the possible efficacies of celecoxib, omega-3 or the combination of celecoxib+risperidone and omega-3+ risperidone compared to risperidone on the behavior and brain biochemistry in rats. In the present study, an amphetamine-induced model of schizophrenia in adult male rats was used to evaluate the effects of celecoxib, omega-3, celecoxib+risperidone and omega-3+ risperidone on the behavior of animals and on brain lipid peroxidation or tumor necrosis factor-alpha. In the water maze task, celecoxib, omega-3, celecoxib+risperidone, omega-3+ risperidone significantly decreased the latency time compared to amphetamine-treated group. Celecoxib, omega-3, celecoxib+risperidone, omega-3+risperidone also significantly reversed the decreased spontaneous alternation induced by amphetamine in the Y-maze task. In the social interaction task, groups treated with celecoxib, omega-3, celecoxib+risperidone, omega-3+ risperidone spent less time to recognize foreign animals than animals in the amphetamine-treated group. Increased brain MDA and TNF-α levels due to amphetamine were significantly reduced in groups treated with celecoxib+risperidone or omega-3+ risperidone. The present findings showed that celecoxib or omega-3 can attenuate amphetamine- induced behavioral impairment and these effects may be associated with their ability to decrease lipid peroxidation and cytokine release. Celecoxib or omega-3 may be promising candidates as adjuvant therapy for schizophrenia., (Copyright © 2016 Elsevier Masson SAS. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
22. Rescue of a Mouse Model of Spinal Muscular Atrophy With Respiratory Distress Type 1 by AAV9-IGHMBP2 Is Dose Dependent.
- Author
-
Shababi M, Feng Z, Villalon E, Sibigtroth CM, Osman EY, Miller MR, Williams-Simon PA, Lombardi A, Sass TH, Atkinson AK, Garcia ML, Ko CP, and Lorson CL
- Subjects
- Animals, Body Weight, Dependovirus genetics, Disease Models, Animal, Dose-Response Relationship, Drug, Female, Humans, Male, Mice, Muscular Atrophy, Spinal genetics, Mutation, Respiratory Distress Syndrome, Newborn genetics, Survival Analysis, DNA-Binding Proteins genetics, Genetic Therapy, Genetic Vectors administration & dosage, Muscular Atrophy, Spinal therapy, Respiratory Distress Syndrome, Newborn therapy, Transcription Factors genetics
- Abstract
Spinal muscular atrophy with respiratory distress type 1 (SMARD1) is an autosomal recessive disease occurring during childhood. The gene responsible for disease development is a ubiquitously expressed protein, IGHMBP2. Mutations in IGHMBP2 result in the loss of α-motor neurons leading to muscle atrophy in the distal limbs accompanied by respiratory complications. Although genetically and clinically distinct, proximal SMA is also caused by the loss of a ubiquitously expressed gene (SMN). Significant preclinical success has been achieved in proximal SMA using viral-based gene replacement strategies. We leveraged the technologies employed in SMA to demonstrate gene replacement efficacy in an SMARD1 animal model. Intracerebroventricular (ICV) injection of single-stranded AAV9 expressing the full-length cDNA of IGHMBP2 in a low dose led to a significant level of rescue in treated SMARD1 animals. Consistent with drastically increased survival, weight gain, and strength, the rescued animals demonstrated a significant improvement in muscle, NMJ, motor neurons, and axonal pathology. In addition, increased levels of IGHMBP2 in lumbar motor neurons verified the efficacy of the virus to transduce the target tissues. Our results indicate that AAV9-based gene replacement is a viable strategy for SMARD1, although dosing effects and potential negative impacts of high dose and ICV injection should be thoroughly investigated.
- Published
- 2016
- Full Text
- View/download PDF
23. Placental development in a mouse model of spinal muscular atrophy.
- Author
-
Van Gronigen Caesar G, Dale JM, Osman EY, Garcia ML, Lorson CL, and Schulz LC
- Subjects
- Animals, Female, Male, Mice, Pregnancy, Muscular Atrophy, Spinal pathology, Muscular Atrophy, Spinal physiopathology, Placentation, SMN Complex Proteins metabolism, Trophoblasts metabolism, Trophoblasts pathology
- Abstract
Spinal Muscular Atrophy (SMA) is an autosomal recessive disorder, leading to fatal loss of motor neurons. It is caused by loss of function of the SMN gene, which is expressed throughout the body, and there is increasing evidence of dysfunction in non-neuronal tissues. Birthweight is one of most powerful prognostic factors for infants born with SMA, and intrauterine growth restriction is common. In the SMNΔ7 mouse model of SMA, pups with the disease lived 25% longer when their mothers were fed a higher fat, "breeder" diet. The placenta is responsible for transport of nutrients from mother to fetus, and is a major determinant of fetal growth. Thus, the present study tested the hypothesis that placental development is impaired in SMNΔ7 conceptuses. Detailed morphological characterization revealed no defects in SMNΔ7 placental development, and expression of key transcription factors regulating mouse placental development was unaffected. The intrauterine growth restriction observed in SMA infants likely does not result from impaired placental development., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
24. Detection of Helicobacter pylori glmM gene in bovine milk using Nested polymerase chain reaction.
- Author
-
Osman EY, El-Eragi AM, Musa AM, El-Magboul SB, A/Rahman MB, and Abdo AE
- Abstract
Aim: The aim was to detect the glmM gene of Helicobacter pylori (H. pylori) in cow's milk from different dairy farms in Khartoum State using Nested polymerase chain reaction (PCR)., Materials and Methods: A total of 50 milk samples were collected from different dairy farms in Khartoum State (13 from Khartoum, 24 Khartoum North, and 13 from Omdurman Provinces)., Results: The generated results showed that 11/50 (22%) were harboring the investigated H. pylori glmM gene in Khartoum State (1/13 [7.7%] Khartoum, 9/24 [37.5%] Khartoum North, and 1/13 [7.7%] Omdurman provinces, respectively)., Conclusion: To the best of our knowledge, this was the first report on the detection of H. pylori glmM gene in cattle milk in Khartoum State. Nonetheless, the high percentages of H. pylori DNA detection in milk opened new avenues toward exploring the risk of human infection with H. pylori through the consumption of raw milk.
- Published
- 2015
- Full Text
- View/download PDF
25. Morpholino antisense oligonucleotides targeting intronic repressor Element1 improve phenotype in SMA mouse models.
- Author
-
Osman EY, Miller MR, Robbins KL, Lombardi AM, Atkinson AK, Brehm AJ, and Lorson CL
- Subjects
- Animals, Disease Models, Animal, Humans, Introns, Mice, Muscular Atrophy, Spinal genetics, Muscular Atrophy, Spinal pathology, Survival Rate, Survival of Motor Neuron 2 Protein genetics, Weight Gain, Genetic Therapy methods, Morpholinos administration & dosage, Muscular Atrophy, Spinal therapy, Regulatory Sequences, Nucleic Acid, Survival of Motor Neuron 2 Protein metabolism
- Abstract
Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by the loss of Survival Motor Neuron-1 (SMN1). In all SMA patients, a nearly identical copy gene called SMN2 is present, which produces low levels of functional protein owing to an alternative splicing event. To prevent exon-skipping, we have targeted an intronic repressor, Element1 (E1), located upstream of SMN2 exon 7 using Morpholino-based antisense oligonucleotides (E1(MO)-ASOs). A single intracerebroventricular injection in the relatively severe mouse model of SMA (SMNΔ7 mouse model) elicited a robust induction of SMN protein, and mean life span was extended from an average survival of 13 to 54 days following a single dose, consistent with large weight gains and a correction of the neuronal pathology. Additionally, E1(MO)-ASO treatment in an intermediate SMA mouse (SMN(RT) mouse model) significantly extended life span by ∼700% and weight gain was comparable with the unaffected animals. While a number of experimental therapeutics have targeted the ISS-N1 element of SMN2 pre-mRNA, the development of E1 ASOs provides a new molecular target for SMA therapeutics that dramatically extends survival in two important pre-clinical models of disease., (© The Author 2014. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2014
- Full Text
- View/download PDF
26. Defining the therapeutic window in a severe animal model of spinal muscular atrophy.
- Author
-
Robbins KL, Glascock JJ, Osman EY, Miller MR, and Lorson CL
- Subjects
- Animals, Body Weight, Chickens, Dependovirus genetics, Disease Models, Animal, Genetic Therapy, Genetic Vectors administration & dosage, HEK293 Cells, Humans, Injections, Intraventricular, Mice, Motor Activity, Muscle, Skeletal pathology, Muscle, Skeletal physiopathology, Muscular Atrophy, Spinal pathology, Muscular Atrophy, Spinal physiopathology, Neuromuscular Junction pathology, Neuromuscular Junction physiopathology, Phenotype, Survival Analysis, Survival of Motor Neuron 1 Protein genetics, Survival of Motor Neuron 1 Protein therapeutic use, Time Factors, Muscular Atrophy, Spinal therapy
- Abstract
Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by the loss of a single gene, Survival Motor Neuron-1 (SMN1). Administration of a self-complementary Adeno-Associated Virus vector expressing full-length SMN cDNA (scAAV-SMN) has proven an effective means to rescue the SMA phenotype in SMA mice, either by intravenous (IV) or intracerebroventricular (ICV) administration at very early time points. We have recently shown that ICV delivery of scAAV9-SMN is more effective than a similar dose of vector administered via an IV injection, thereby providing an important mechanism to examine a timeline for rescuing the disease and determining the therapeutic window in a severe model of SMA. In this report, we utilized a relatively severe mouse model of SMA, SMNΔ7. Animals were injected with scAAV9-SMN vector via ICV injection on a single day, from P2 through P8. At each delivery point from P2 through P8, scAAV9-SMN decreased disease severity. A near complete rescue was obtained following P2 injection while a P8 injection produced a ∼ 40% extension in survival. Analysis of the underlying neuromuscular junction (NMJ) pathology revealed that late-stage delivery of the vector failed to provide protection from NMJ defects despite robust SMN expression in the central nervous system. While our study demonstrates that a maximal benefit is obtained when treatment is delivered during pre-symptomatic stages, significant therapeutic benefit can still be achieved after the onset of disease symptoms., (© The Author 2014. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2014
- Full Text
- View/download PDF
27. Enhancement of SMN protein levels in a mouse model of spinal muscular atrophy using novel drug-like compounds.
- Author
-
Cherry JJ, Osman EY, Evans MC, Choi S, Xing X, Cuny GD, Glicksman MA, Lorson CL, and Androphy EJ
- Subjects
- Animals, Cells, Cultured, Fibroblasts drug effects, Fibroblasts metabolism, Fibroblasts pathology, High-Throughput Screening Assays, Humans, Mice, Muscular Atrophy, Spinal genetics, Muscular Atrophy, Spinal physiopathology, RNA, Messenger genetics, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology, Survival of Motor Neuron 1 Protein analysis, Survival of Motor Neuron 1 Protein genetics, Survival of Motor Neuron 2 Protein analysis, Drug Discovery, Muscular Atrophy, Spinal drug therapy, Small Molecule Libraries therapeutic use, Survival of Motor Neuron 2 Protein genetics, Up-Regulation drug effects
- Abstract
Spinal muscular atrophy (SMA) is a neurodegenerative disease that causes progressive muscle weakness, which primarily targets proximal muscles. About 95% of SMA cases are caused by the loss of both copies of the SMN1 gene. SMN2 is a nearly identical copy of SMN1, which expresses much less functional SMN protein. SMN2 is unable to fully compensate for the loss of SMN1 in motor neurons but does provide an excellent target for therapeutic intervention. Increased expression of functional full-length SMN protein from the endogenous SMN2 gene should lessen disease severity. We have developed and implemented a new high-throughput screening assay to identify small molecules that increase the expression of full-length SMN from a SMN2 reporter gene. Here, we characterize two novel compounds that increased SMN protein levels in both reporter cells and SMA fibroblasts and show that one increases lifespan, motor function, and SMN protein levels in a severe mouse model of SMA., (© 2013 The Authors. Published by John Wiley and Sons, Ltd on behalf of EMBO.)
- Published
- 2013
- Full Text
- View/download PDF
28. Development and characterization of an SMN2-based intermediate mouse model of Spinal Muscular Atrophy.
- Author
-
Cobb MS, Rose FF, Rindt H, Glascock JJ, Shababi M, Miller MR, Osman EY, Yen PF, Garcia ML, Martin BR, Wetz MJ, Mazzasette C, Feng Z, Ko CP, and Lorson CL
- Subjects
- Animals, Body Weight, Brain metabolism, Exons, Gene Expression Regulation, Humans, Longevity, Mice, Mice, Inbred C57BL, Mice, Transgenic, Muscular Atrophy, Spinal pathology, Phenotype, Promoter Regions, Genetic, RNA genetics, RNA Splicing, Spinal Cord metabolism, Survival of Motor Neuron 1 Protein genetics, Disease Models, Animal, Muscular Atrophy, Spinal genetics, Survival of Motor Neuron 2 Protein genetics
- Abstract
Spinal Muscular Atrophy (SMA) is due to the loss of the survival motor neuron gene 1 (SMN1), resulting in motor neuron (MN) degeneration, muscle atrophy and loss of motor function. While SMN2 encodes a protein identical to SMN1, a single nucleotide difference in exon 7 causes most of the SMN2-derived transcripts to be alternatively spliced resulting in a truncated and unstable protein (SMNΔ7). SMA patients retain at least one SMN2 copy, making it an important target for therapeutics. Many of the existing SMA models are very severe, with animals typically living less than 2 weeks. Here, we present a novel intermediate mouse model of SMA based upon the human genomic SMN2 gene. Genetically, this model is similar to the well-characterized SMNΔ7 model; however, we have manipulated the SMNΔ7 transgene to encode a modestly more functional protein referred to as SMN read-through (SMN(RT)). By introducing the SMN(RT) transgene onto the background of a severe mouse model of SMA (SMN2(+/+);Smn(-/-)), disease severity was significantly decreased based upon a battery of phenotypic parameters, including MN pathology and a significant extension in survival. Importantly, there is not a full phenotypic correction, allowing for the examination of a broad range of therapeutics, including SMN2-dependent and SMN-independent pathways. This novel animal model serves as an important biological and therapeutic model for less severe forms of SMA and provides an in vivo validation of the SMN(RT) protein.
- Published
- 2013
- Full Text
- View/download PDF
29. Decreasing disease severity in symptomatic, Smn(-/-);SMN2(+/+), spinal muscular atrophy mice following scAAV9-SMN delivery.
- Author
-
Glascock JJ, Osman EY, Wetz MJ, Krogman MM, Shababi M, and Lorson CL
- Subjects
- Animals, Disease Models, Animal, Genetic Therapy, Genetic Vectors, Mice, Mice, Transgenic, Muscular Atrophy, Spinal genetics, Muscular Atrophy, Spinal metabolism, SMN Complex Proteins metabolism, Severity of Illness Index, Dependovirus genetics, Muscular Atrophy, Spinal therapy, SMN Complex Proteins genetics
- Abstract
Spinal muscular atrophy (SMA), an autosomal recessive neuromuscular disorder, is the leading genetic cause of infant mortality. SMA is caused by the homozygous loss of Survival Motor Neuron-1 (SMN1). In humans, a nearly identical copy gene is present, SMN2. SMN2 is retained in all SMA patients and encodes the same protein as SMN1. However, SMN1 and SMN2 differ by a silent C-to-T transition at the 5' end of exon 7, causing alternative splicing of SMN2 transcripts and low levels of full-length SMN. SMA is monogenic and therefore well suited for gene-replacement strategies. Recently, self-complementary adeno-associated virus (scAAV) vectors have been used to deliver the SMN cDNA to an animal model of disease, the SMNΔ7 mouse. In this study, we examine a severe model of SMA, Smn(-/-);SMN2(+/+), to determine whether gene replacement is viable in a model in which disease development begins in utero. Using two delivery paradigms, intracerebroventricular injections and intravenous injections, we delivered scAAV9-SMN and demonstrated a two to four fold increase in survival, in addition to improving many of the phenotypic parameters of the model. This represents the longest extension in survival for this severe model for any therapeutic intervention and suggests that postsymptomatic treatment of SMA may lead to significant improvement of disease severity.
- Published
- 2012
- Full Text
- View/download PDF
30. Bifunctional RNAs targeting the intronic splicing silencer N1 increase SMN levels and reduce disease severity in an animal model of spinal muscular atrophy.
- Author
-
Osman EY, Yen PF, and Lorson CL
- Subjects
- Animals, Base Sequence, Disease Models, Animal, Gene Order, Gene Targeting, Genetic Therapy, Mice, Mice, Knockout, Motor Activity genetics, Muscular Atrophy, Spinal genetics, Muscular Atrophy, Spinal mortality, SMN Complex Proteins metabolism, Weight Gain genetics, Introns, Muscular Atrophy, Spinal therapy, Oligoribonucleotides, Antisense administration & dosage, RNA Splicing, Regulatory Sequences, Ribonucleic Acid, SMN Complex Proteins genetics
- Abstract
Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by loss of survival motor neuron-1 (SMN1). A nearly identical copy gene, SMN2, is present in all SMA patients. Although the SMN2 coding sequence has the potential to produce full-length SMN, nearly 90% of SMN2-derived transcripts are alternatively spliced and encode a truncated protein. SMN2, however, is an excellent therapeutic target. Previously, we developed antisense-based oligonucleotides (bifunctional RNAs) that specifically recruit SR/SR-like splicing factors and target a negative regulator of SMN2 exon-7 inclusion within intron-6. As a means to optimize the antisense sequence of the bifunctional RNAs, we chose to target a potent intronic repressor downstream of SMN2 exon 7, called intronic splicing silencer N1 (ISS-N1). We developed RNAs that specifically target ISS-N1 and concurrently recruit the modular SR proteins SF2/ASF or hTra2β1. RNAs were directly injected in the brains of SMA mice. Bifunctional RNA injections were able to elicit robust induction of SMN protein in the brain and spinal column of neonatal SMA mice. Importantly, hTra2β1-ISS-N1 and SF2/ASF-ISS-N1 bifunctional RNAs significantly extended lifespan and increased weight in the SMNΔ7 mice. This technology has direct implications for SMA therapy and provides similar therapeutic strategies for other diseases caused by aberrant splicing.
- Published
- 2012
- Full Text
- View/download PDF
31. Delivery of therapeutic agents through intracerebroventricular (ICV) and intravenous (IV) injection in mice.
- Author
-
Glascock JJ, Osman EY, Coady TH, Rose FF, Shababi M, and Lorson CL
- Subjects
- Animals, Animals, Newborn, Blood-Brain Barrier metabolism, Injections, Intravenous instrumentation, Injections, Intraventricular instrumentation, Mice, Injections, Intravenous methods, Injections, Intravenous veterinary, Injections, Intraventricular methods, Injections, Intraventricular veterinary, Pharmaceutical Preparations administration & dosage
- Abstract
Despite the protective role that blood brain barrier plays in shielding the brain, it limits the access to the central nervous system (CNS) which most often results in failure of potential therapeutics designed for neurodegenerative disorders. Neurodegenerative diseases such as Spinal Muscular Atrophy (SMA), in which the lower motor neurons are affected, can benefit greatly from introducing the therapeutic agents into the CNS. The purpose of this video is to demonstrate two different injection paradigms to deliver therapeutic materials into neonatal mice soon after birth. One of these methods is injecting directly into cerebral lateral ventricles (Intracerebroventricular) which results in delivery of materials into the CNS through the cerebrospinal fluid. The second method is a temporal vein injection (intravenous) that can introduce different therapeutics into the circulatory system, leading to systemic delivery including the CNS. Widespread transduction of the CNS is achievable if an appropriate viral vector and viral serotype is utilized. Visualization and utilization of the temporal vein for injection is feasible up to postnatal day 6. However, if the delivered material is intended to reach the CNS, these injections should take place while the blood brain barrier is more permeable due to its immature status, preferably prior to postnatal day 2. The fully developed blood brain barrier greatly limits the effectiveness of intravenous delivery. Both delivery systems are simple and effective once the surgical aptitude is achieved. They do not require any extensive surgical devices and can be performed by a single person. However, these techniques are not without challenges. The small size of postnatal day 2 pups and the subsequent small target areas can make the injections difficult to perform and initially challenging to replicate.
- Published
- 2011
- Full Text
- View/download PDF
32. Delivery of bifunctional RNAs that target an intronic repressor and increase SMN levels in an animal model of spinal muscular atrophy.
- Author
-
Baughan TD, Dickson A, Osman EY, and Lorson CL
- Subjects
- Animals, Cells, Cultured, Disease Models, Animal, Fibroblasts metabolism, Humans, Mice, Mice, Transgenic, Muscular Atrophy, Spinal genetics, RNA, Antisense chemistry, RNA, Antisense genetics, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Survival of Motor Neuron 2 Protein metabolism, Genetic Therapy, Introns, Muscular Atrophy, Spinal metabolism, Muscular Atrophy, Spinal therapy, RNA, Antisense therapeutic use, Regulatory Sequences, Nucleic Acid, Survival of Motor Neuron 2 Protein genetics
- Abstract
Spinal muscular atrophy (SMA) is a motor neuron disease caused by the loss of survival motor neuron-1 (SMN1). A nearly identical copy gene, SMN2, is present in all SMA patients, which produces low levels of functional protein. Although the SMN2 coding sequence has the potential to produce normal, full-length SMN, approximately 90% of SMN2-derived transcripts are alternatively spliced and encode a truncated protein lacking the final coding exon (exon 7). SMN2, however, is an excellent therapeutic target. Previously, we developed bifunctional RNAs that bound SMN exon 7 and modulated SMN2 splicing. To optimize the efficiency of the bifunctional RNAs, a different antisense target was required. To this end, we genetically verified the identity of a putative intronic repressor and developed bifunctional RNAs that target this sequence. Consequently, there is a 2-fold mechanism of SMN induction: inhibition of the intronic repressor and recruitment of SR proteins via the SR recruitment sequence of the bifunctional RNA. The bifunctional RNAs effectively increased SMN in human primary SMA fibroblasts. Lead candidates were synthesized as 2'-O-methyl RNAs and were directly injected in the central nervous system of SMA mice. Single-RNA injections were able to illicit a robust induction of SMN protein in the brain and throughout the spinal column of neonatal SMA mice. In a severe model of SMA, mean life span was extended following the delivery of bifunctional RNAs. This technology has direct implications for the development of an SMA therapy, but also lends itself to a multitude of diseases caused by aberrant pre-mRNA splicing.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.