Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
Pediatr Neurol ; 146: 16-20, 2023 09.
Article in English | MEDLINE | ID: mdl-37392669

ABSTRACT

Myosin-binding protein C1 (MYBPC1) encodes myosin-binding protein C, slow type (sMyBP-C), an accessory protein that regulates actomyosin cross-linking, stabilizes thick filaments, and modulates contractility in muscle sarcomeres and has recently been linked to myopathy with tremor. The clinical features of MYBPC1 mutations manifesting in early childhood bear some similarities to those of spinal muscular atrophy (SMA), such as hypotonia, involuntary movement of the tongue and limbs, and delayed motor development. The development of novel therapies for SMA has necessitated the importance of differentiating SMA from other diseases in the early infancy period. We report the characteristic tongue movements of MYBPC1 mutations, along with other clinical findings, such as positive deep tendon reflexes and normal peripheral nerve conduction velocity testing, which could help in considering other diseases as differential diagnoses.


Subject(s)
Muscular Atrophy, Spinal , Tremor , Child, Preschool , Humans , Mutation/genetics , Tongue/metabolism
3.
Brain Dev ; 44(5): 353-356, 2022 May.
Article in English | MEDLINE | ID: mdl-35058083

ABSTRACT

INTRODUCTION: Early disease control with disease-modifying drugs is important for improving the prognosis of multiple sclerosis (MS) in children. Dimethyl fumarate (DMF) is an oral disease-modifying drug for MS in adults with relatively stable disease; however, its use in young children has not been heavily documented in the current literature. We report the case of a pediatric patient with relapsing-remitting MS who was treated with DMF. CASE REPORT: A 3-year-old boy with a history of common cold symptoms developed unsteadiness and somnolence. Magnetic resonance imaging revealed multiple white matter lesions. Symptoms were recurrent, and DMF was prescribed at 6 years of age due to a relapse episode with oculomotor disability and facial paralysis. However, disease progression continued, and new lesions were noted at age 7; thus, the dose of DMF was increased to 240 mg/day. No relapse has been observed for over three years; sequelae or severe side effects were absent. CONCLUSIONS: DMF may be a useful oral disease-modifying drug for preventing recurrence in young children with MS.


Subject(s)
Multiple Sclerosis, Relapsing-Remitting , Multiple Sclerosis , Adult , Child , Child, Preschool , Dimethyl Fumarate/therapeutic use , Humans , Immunosuppressive Agents/therapeutic use , Magnetic Resonance Imaging , Male , Multiple Sclerosis/chemically induced , Multiple Sclerosis/drug therapy , Multiple Sclerosis, Relapsing-Remitting/chemically induced , Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Multiple Sclerosis, Relapsing-Remitting/drug therapy
4.
Brain Dev ; 44(2): 148-152, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34579981

ABSTRACT

INTRODUCTION: Pitt-Hopkins syndrome (PTHS) is a neurodevelopmental disorder caused by mutations in TCF4. Seizures have been found to vary among patients with PTHS. We report the case of a PTHS patient with a novel missense mutation in the gene TCF4, presenting with two types of early epileptic encephalopathy. CASE REPORT: The patient was a Japanese boy. His first seizure was reported at 17 days of age, with twitching of the left eyelid and tonic-clonic seizures on either side of his body. An ictal electroencephalogram (EEG) showed epileptic discharges arising independently from both hemispheres, occasionally resembling migrating partial seizures of infancy (MPSI) that migrated from one side to the other. Brain magnetic resonance imaging revealed agenesis of the corpus callosum. His facial characteristics included a distinctive upper lip and thickened helices. His seizures were refractory, and psychomotor development was severely delayed. At the age of 10 months, he developed West syndrome with spasms and hypsarrhythmia. After being prescribed topiramate (TPM), his seizures and EEG abnormalities dramatically improved. Also, psychomotor development progressed. Whole-exome sequencing revealed a novel de novo missense mutation in exon 18 (NM_001083962.2:c.1718A > T, p.(Asn573Ile)), corresponding to the basic region of the basic helix-loop-helix domain, which may be a causative gene for epileptic encephalopathy. CONCLUSIONS: To our knowledge, this is the first report of a patient with PTHS treated with TPM, who presented with both MPSI as well as West syndrome. This may help provide new insights regarding the phenotypes caused by mutations in TCF4.


Subject(s)
Facies , Hyperventilation , Intellectual Disability , Spasms, Infantile , Transcription Factor 4/genetics , Anticonvulsants/pharmacology , Humans , Hyperventilation/diagnosis , Hyperventilation/drug therapy , Hyperventilation/genetics , Hyperventilation/physiopathology , Infant , Intellectual Disability/diagnosis , Intellectual Disability/drug therapy , Intellectual Disability/genetics , Intellectual Disability/physiopathology , Male , Mutation, Missense , Spasms, Infantile/diagnosis , Spasms, Infantile/drug therapy , Spasms, Infantile/genetics , Spasms, Infantile/physiopathology , Topiramate/pharmacology
5.
Am J Med Genet A ; 188(4): 1293-1298, 2022 04.
Article in English | MEDLINE | ID: mdl-34971077

ABSTRACT

Congenital myasthenic syndromes (CMS) is a group of diseases that causes abnormalities at the neuromuscular junction owing to genetic anomalies. The pathogenic variant in ALG14 results in a severe pathological form of CMS causing end-plate acetylcholine receptor deficiency. Here, we report the cases of two siblings with CMS associated with a novel variant in ALG14. Immediately after birth, they showed hypotonia and multiple joint contractures with low Apgar scores. Ptosis, low-set ears, and high-arched palate were noted. Deep tendon reflexes were symmetrical. They showed worsening swallowing and respiratory problems; hence, nasal feeding and tracheotomy were performed. Cranial magnetic resonance imaging scans revealed delayed myelination and cerebral atrophy. Exome sequencing indicated that the siblings had novel compound heterozygous missense variants, c.590T>G (p.Val197Gly) and c.433G>A (p.Gly145Arg), in exon 4 of ALG14. Repetitive nerve stimulation test showed an abnormal decrease in compound muscle action potential. After treatment with pyridostigmine, the time off the respirator increased. Their epileptic seizures were well controlled by anti-epileptic drugs. Their clinical course is stable even now at the ages of 5 and 2 years, making them the longest reported survivors of a severe form of CMS with the ALG14 variant thus far.


Subject(s)
Myasthenic Syndromes, Congenital , Exons , Humans , Mutation , Myasthenic Syndromes, Congenital/complications , Myasthenic Syndromes, Congenital/diagnosis , Myasthenic Syndromes, Congenital/genetics , Siblings , Survivors , Exome Sequencing
6.
Physiol Rep ; 9(24): e15092, 2021 12.
Article in English | MEDLINE | ID: mdl-34921520

ABSTRACT

Sodium-dependent glucose cotransporters (SGLTs) have attracted considerable attention as new targets for type 2 diabetes mellitus. In the kidney, SGLT2 is the major glucose uptake transporter in the proximal tubules, and inhibition of SGLT2 in the proximal tubules shows renoprotective effects. On the other hand, SGLT1 plays a role in glucose absorption from the gastrointestinal tract, and the relationship between SGLT1 inhibition in the gut and renal function remains unclear. Here, we examined the effect of SGL5213, a novel and potent intestinal SGLT1 inhibitor, in a renal failure (RF) model. SGL5213 improved renal function and reduced gut-derived uremic toxins (phenyl sulfate and trimethylamine-N-oxide) in an adenine-induced RF model. Histological analysis revealed that SGL5213 ameliorated renal fibrosis and inflammation. SGL5213 also reduced gut inflammation and fibrosis in the ileum, which is a primary target of SGL5213. Examination of the gut microbiota community revealed that the Firmicutes/Bacteroidetes ratio, which suggests gut dysbiosis, was increased in RF and SGL5213 rebalanced the ratio by increasing Bacteroidetes and reducing Firmicutes. At the genus level, Allobaculum (a major component of Erysipelotrichaceae) was significantly increased in the RF group, and this increase was canceled by SGL5213. We also measured the effect of SGL5213 on bacterial phenol-producing enzymes that catalyze tyrosine into phenol, following the reduction of phenyl sulfate, which is a novel marker and a therapeutic target for diabetic kidney disease DKD. We found that the enzyme inhibition was less potent, suggesting that the change in the microbial community and the reduction of uremic toxins may be related to the renoprotective effect of SGL5213. Because SGL5213 is a low-absorbable SGLT1 inhibitor, these data suggest that the gastrointestinal inhibition of SGLT1 is also a target for chronic kidney diseases.


Subject(s)
Adenine/toxicity , Gastrointestinal Microbiome/drug effects , Renal Insufficiency/chemically induced , Renal Insufficiency/drug therapy , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sorbitol/analogs & derivatives , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Gastrointestinal Microbiome/physiology , Mice , Mice, Inbred C57BL , Renal Insufficiency/metabolism , Sorbitol/pharmacology , Sorbitol/therapeutic use
7.
Mol Genet Metab Rep ; 29: 100800, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34522618

ABSTRACT

Biallelic 4-hydroxyphenylpyruvate dioxygenase-like protein (HPDL) variants were recently reported as a cause of progressive and incurable neurodegenerative diseases ranging from neonatal-onset leukoencephalopathy with severe neurodevelopmental delay to spastic paraplegia. Although the physiological function of HPDL remains unknown, its subcellular localization in the mitochondria has been reported. Here, we report a case of HPDL-related neurological disease that was clinically and neuroimaging compatible with Leigh syndrome, previously unreported, and was treated with a ketogenic diet.

8.
PLoS One ; 15(12): e0231064, 2020.
Article in English | MEDLINE | ID: mdl-33264289

ABSTRACT

Sporadic inclusion body myositis (sIBM) is the most common idiopathic inflammatory myopathy, and several reports have suggested that mitochondrial abnormalities are involved in its etiology. We recruited 9 sIBM patients and found significant histological changes and an elevation of growth differential factor 15 (GDF15), a marker of mitochondrial disease, strongly suggesting the involvement of mitochondrial dysfunction. Bioenergetic analysis of sIBM patient myoblasts revealed impaired mitochondrial function. Decreased ATP production, reduced mitochondrial size and reduced mitochondrial dynamics were also observed in sIBM myoblasts. Cell vulnerability to oxidative stress also suggested the existence of mitochondrial dysfunction. Mitochonic acid-5 (MA-5) increased the cellular ATP level, reduced mitochondrial ROS, and provided protection against sIBM myoblast death. MA-5 also improved the survival of sIBM skin fibroblasts as well as mitochondrial morphology and dynamics in these cells. The reduction in the gene expression levels of Opa1 and Drp1 was also reversed by MA-5, suggesting the modification of the fusion/fission process. These data suggest that MA-5 may provide an alternative therapeutic strategy for treating not only mitochondrial diseases but also sIBM.


Subject(s)
Indoleacetic Acids/therapeutic use , Mitochondria, Muscle/metabolism , Myositis, Inclusion Body/drug therapy , Phenylbutyrates/therapeutic use , Adenosine Triphosphate/biosynthesis , Aged , Aged, 80 and over , Buthionine Sulfoximine/pharmacology , Cell Survival/drug effects , Cells, Cultured , DNA, Mitochondrial/genetics , Drug Evaluation, Preclinical , Dynamins/biosynthesis , Dynamins/genetics , Female , Fibroblast Growth Factors/blood , Fibroblasts/drug effects , GTP Phosphohydrolases/biosynthesis , GTP Phosphohydrolases/genetics , Growth Differentiation Factor 15/biosynthesis , Growth Differentiation Factor 15/blood , Growth Differentiation Factor 15/genetics , Humans , Indoleacetic Acids/pharmacology , Male , Middle Aged , Mitochondria, Muscle/pathology , Myoblasts/drug effects , Myoblasts/metabolism , Myoblasts/ultrastructure , Myositis, Inclusion Body/metabolism , Myositis, Inclusion Body/pathology , Oxygen Consumption , Phenylbutyrates/pharmacology , Reactive Oxygen Species/metabolism , Retrospective Studies
9.
CEN Case Rep ; 9(3): 210-214, 2020 08.
Article in English | MEDLINE | ID: mdl-32128695

ABSTRACT

Unlike complete deficiency of hypoxanthine phosphoribosyltransferase (HPRT) (i.e., Lesch-Nyhan syndrome), partial HPRT deficiency causes HPRT-related hyperuricemia without neurological symptoms. Herein, we describe a 22-year-old man without neurological symptoms that presented gout, hyperuricemia (serum urate level, 12.2 mg/dL), multiple renal microcalculi, and a family history of juvenile gout that was exhibited by his brother and grandfather. Genetic testing revealed a novel missense mutation, c.103G>A (p.V35M), in the HPRT1 gene, and biochemical testing (conducted using the patient's erythrocytes) showed that the patient retained only 12.4% HPRT enzymatic activity compared to that exhibited by a healthy control subject. We thus diagnosed the patient with HPRT-related hyperuricemia caused by partial HPRT deficiency. After his serum urate level was controlled via treatment with febuxostat, his gout did not recur. Thus, this study emphasizes that HPRT deficiency should be considered as a potential cause of familial juvenile gout, even in the absence of neurological symptoms.


Subject(s)
Gout/genetics , Hyperuricemia/genetics , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Kidney Diseases/genetics , Febuxostat/administration & dosage , Febuxostat/therapeutic use , Gout/complications , Gout/diagnosis , Gout/drug therapy , Gout Suppressants/administration & dosage , Gout Suppressants/therapeutic use , Humans , Hyperuricemia/blood , Hyperuricemia/diagnosis , Hyperuricemia/drug therapy , Hyperuricemia/etiology , Kidney Calculi/diagnosis , Kidney Calculi/etiology , Kidney Calculi/pathology , Kidney Diseases/diagnosis , Kidney Diseases/drug therapy , Male , Mutation, Missense/genetics , Treatment Outcome , Young Adult
10.
Nephrol Dial Transplant ; 35(2): 250-264, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31411705

ABSTRACT

BACKGROUND: Cardiorenal syndrome is a major cause of mortality in patients with chronic kidney disease (CKD). However, the involvement of detrimental humoral mediators in the pathogenesis of cardiorenal syndrome is still controversial. Trimethylamine-N-oxide (TMAO), a hepatic metabolic product of trimethylamine generated from dietary phosphatidylcholine or carnitine derived by the gut microbiota, has been linked directly with progression of cardiovascular disease and renal dysfunction. Thus, targeting TMAO may be a novel strategy for the prevention of cardiovascular disease and chronic kidney disease. METHODS: Linaclotide, a guanylate cyclase C agonist, was administered to adenine-induced renal failure (RF) mice and changes in renal function and levels of gut-derived uremic toxins, as well as the gut microbiota community, were analyzed using metabolomic and metagenomic methods to reveal its cardiorenal effect. RESULTS: Linaclotide decreased the plasma levels of TMAO at a clinically used low dose of 10 µg/kg in the adenine-induced RF mouse model. At a high concentration of 100 µg/kg, linaclotide clearly improved renal function and reduced the levels of various uremic toxins. A reduction in TMAO levels following linaclotide treatment was also observed in a choline-fed pro-atherosclerotic model. Linaclotide ameliorated renal inflammation and fibrosis and cardiac fibrosis, as well as decreased the expression of collagen I, transforming growth factor-ß, galectin-3 (Gal-3) and ST2 genes. Plasma levels of Gal-3 and ST2 were also reduced. Because exposure of cardiomyocytes to TMAO increased fibronectin expression, these data suggest that linaclotide reduced the levels of TMAO and various uremic toxins and may result in not only renal, but also cardiac, fibrosis. F4/80-positive macrophages were abundant in small intestinal crypts in RF mice, and this increased expression was decreased by linaclotide. Reduced colonic claudin-1 levels were also restored by linaclotide, suggesting that linaclotide ameliorated the 'leaky gut' in RF mice. Metagenomic analysis revealed that the microbial order Clostridiales could be responsible for the change in TMAO levels. CONCLUSION: Linaclotide reduced TMAO and uremic toxin levels and could be a powerful tool for the prevention and control of the cardiorenal syndrome by modification of the gut-cardio-renal axis.


Subject(s)
Adenine/toxicity , Cardio-Renal Syndrome/drug therapy , Gastrointestinal Microbiome/drug effects , Guanylate Cyclase/chemistry , Guanylyl Cyclase C Agonists/pharmacology , Peptides/pharmacology , Renal Insufficiency, Chronic/drug therapy , Animals , Cardio-Renal Syndrome/chemically induced , Cardio-Renal Syndrome/metabolism , Cardio-Renal Syndrome/pathology , Disease Models, Animal , Disease Progression , Fibrosis/chemically induced , Fibrosis/drug therapy , Fibrosis/metabolism , Fibrosis/pathology , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology
11.
J Am Soc Nephrol ; 31(2): 280-296, 2020 02.
Article in English | MEDLINE | ID: mdl-31767624

ABSTRACT

BACKGROUND: Ferroptosis, nonapoptotic cell death mediated by free radical reactions and driven by the oxidative degradation of lipids, is a therapeutic target because of its role in organ damage, including AKI. Ferroptosis-causing radicals that are targeted by ferroptosis suppressors have not been unequivocally identified. Because certain cytochrome P450 substrate drugs can prevent lipid peroxidation via obscure mechanisms, we evaluated their antiferroptotic potential and used them to identify ferroptosis-causing radicals. METHODS: Using a cell-based assay, we screened cytochrome P450 substrate compounds to identify drugs with antiferroptotic activity and investigated the underlying mechanism. To evaluate radical-scavenging activity, we used electron paramagnetic resonance-spin trapping methods and a fluorescence probe for lipid radicals, NBD-Pen, that we had developed. We then assessed the therapeutic potency of these drugs in mouse models of cisplatin-induced AKI and LPS/galactosamine-induced liver injury. RESULTS: We identified various US Food and Drug Administration-approved drugs and hormones that have antiferroptotic properties, including rifampicin, promethazine, omeprazole, indole-3-carbinol, carvedilol, propranolol, estradiol, and thyroid hormones. The antiferroptotic drug effects were closely associated with the scavenging of lipid peroxyl radicals but not significantly related to interactions with other radicals. The elevated lipid peroxyl radical levels were associated with ferroptosis onset, and known ferroptosis suppressors, such as ferrostatin-1, also functioned as lipid peroxyl radical scavengers. The drugs exerted antiferroptotic activities in various cell types, including tubules, podocytes, and renal fibroblasts. Moreover, in mice, the drugs ameliorated AKI and liver injury, with suppression of tissue lipid peroxidation and decreased cell death. CONCLUSIONS: Although elevated lipid peroxyl radical levels can trigger ferroptosis onset, some drugs that scavenge lipid peroxyl radicals can help control ferroptosis-related disorders, including AKI.


Subject(s)
Acute Kidney Injury/prevention & control , Drug Repositioning , Ferroptosis/drug effects , Free Radical Scavengers/pharmacology , Lipid Peroxidation/drug effects , Animals , Cells, Cultured , Humans , Male , Mice , Mice, Inbred C57BL , Rats
12.
Nat Commun ; 10(1): 1835, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015435

ABSTRACT

Diabetic kidney disease is a major cause of renal failure that urgently necessitates a breakthrough in disease management. Here we show using untargeted metabolomics that levels of phenyl sulfate, a gut microbiota-derived metabolite, increase with the progression of diabetes in rats overexpressing human uremic toxin transporter SLCO4C1 in the kidney, and are decreased in rats with limited proteinuria. In experimental models of diabetes, phenyl sulfate administration induces albuminuria and podocyte damage. In a diabetic patient cohort, phenyl sulfate levels significantly correlate with basal and predicted 2-year progression of albuminuria in patients with microalbuminuria. Inhibition of tyrosine phenol-lyase, a bacterial enzyme responsible for the synthesis of phenol from dietary tyrosine before it is metabolized into phenyl sulfate in the liver, reduces albuminuria in diabetic mice. Together, our results suggest that phenyl sulfate contributes to albuminuria and could be used as a disease marker and future therapeutic target in diabetic kidney disease.


Subject(s)
Albuminuria/etiology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/blood , Gastrointestinal Microbiome/physiology , Sulfuric Acid Esters/metabolism , Adult , Aged , Aged, 80 and over , Albuminuria/blood , Albuminuria/drug therapy , Albuminuria/pathology , Animals , Animals, Genetically Modified , Cohort Studies , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/urine , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 2/blood , Diabetic Nephropathies/etiology , Diabetic Nephropathies/pathology , Dogs , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Humans , Madin Darby Canine Kidney Cells , Male , Metabolomics/methods , Mice , Mice, Inbred C57BL , Middle Aged , Organic Anion Transporters/genetics , Podocytes/metabolism , Podocytes/pathology , Rats , Streptozocin/toxicity , Sulfuric Acid Esters/blood , Tyrosine Phenol-Lyase/antagonists & inhibitors , Tyrosine Phenol-Lyase/metabolism , Young Adult
13.
EBioMedicine ; 20: 27-38, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28579242

ABSTRACT

Mitochondrial dysfunction increases oxidative stress and depletes ATP in a variety of disorders. Several antioxidant therapies and drugs affecting mitochondrial biogenesis are undergoing investigation, although not all of them have demonstrated favorable effects in the clinic. We recently reported a therapeutic mitochondrial drug mitochonic acid MA-5 (Tohoku J. Exp. Med., 2015). MA-5 increased ATP, rescued mitochondrial disease fibroblasts and prolonged the life span of the disease model "Mitomouse" (JASN, 2016). To investigate the potential of MA-5 on various mitochondrial diseases, we collected 25 cases of fibroblasts from various genetic mutations and cell protective effect of MA-5 and the ATP producing mechanism was examined. 24 out of the 25 patient fibroblasts (96%) were responded to MA-5. Under oxidative stress condition, the GDF-15 was increased and this increase was significantly abrogated by MA-5. The serum GDF-15 elevated in Mitomouse was likewise reduced by MA-5. MA-5 facilitates mitochondrial ATP production and reduces ROS independent of ETC by facilitating ATP synthase oligomerization and supercomplex formation with mitofilin/Mic60. MA-5 reduced mitochondria fragmentation, restores crista shape and dynamics. MA-5 has potential as a drug for the treatment of various mitochondrial diseases. The diagnostic use of GDF-15 will be also useful in a forthcoming MA-5 clinical trial.


Subject(s)
Indoleacetic Acids/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Phenylbutyrates/pharmacology , Protein Multimerization/drug effects , Adenosine Triphosphate/metabolism , Animals , Biomarkers , Cell Line , Cell Survival/drug effects , DNA, Mitochondrial , Disease Models, Animal , Fibroblast Growth Factors/metabolism , Fibroblasts/metabolism , Growth Differentiation Factor 15/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/genetics , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/genetics , Mitochondrial Dynamics/drug effects , Mitochondrial Proton-Translocating ATPases/chemistry , Multiprotein Complexes/metabolism , Mutation , Organelle Biogenesis , Prognosis , Protective Agents , Protein Binding
14.
Sci Rep ; 7(1): 1884, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28507324

ABSTRACT

Renal fibrosis is closely related to chronic inflammation and is under the control of epigenetic regulations. Because the signaling of transforming growth factor-ß1 (TGF-ß1) and tumor necrosis factor-α (TNF-α) play key roles in progression of renal fibrosis, dual blockade of TGF-ß1 and TNF-α is desired as its therapeutic approach. Here we screened small molecules showing anti-TNF-α activity in the compound library of indole derivatives. 11 out of 41 indole derivatives inhibited the TNF-α effect. Among them, Mitochonic Acid 35 (MA-35), 5-(3, 5-dimethoxybenzyloxy)-3-indoleacetic acid, showed the potent effect. The anti-TNF-α activity was mediated by inhibiting IκB kinase phosphorylation, which attenuated the LPS/GaIN-induced hepatic inflammation in the mice. Additionally, MA-35 concurrently showed an anti-TGF-ß1 effect by inhibiting Smad3 phosphorylation, resulting in the downregulation of TGF-ß1-induced fibrotic gene expression. In unilateral ureter obstructed mouse kidney, which is a renal fibrosis model, MA-35 attenuated renal inflammation and fibrosis with the downregulation of inflammatory cytokines and fibrotic gene expressions. Furthermore, MA-35 inhibited TGF-ß1-induced H3K4me1 histone modification of the fibrotic gene promoter, leading to a decrease in the fibrotic gene expression. MA-35 affects multiple signaling pathways involved in the fibrosis and may recover epigenetic modification; therefore, it could possibly be a novel therapeutic drug for fibrosis.


Subject(s)
Indoles/pharmacology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line , Disease Models, Animal , Extracellular Matrix/metabolism , Fibrosis , Hepatitis/drug therapy , Hepatitis/etiology , Hepatitis/metabolism , Hepatitis/pathology , Histones/metabolism , Humans , I-kappa B Kinase/metabolism , Kidney Diseases/drug therapy , Kidney Diseases/etiology , Lipopolysaccharides/adverse effects , Male , Methylation , Mice , Models, Biological , Phosphorylation/drug effects , Smad3 Protein/metabolism
15.
Brain Dev ; 39(6): 532-535, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28143689

ABSTRACT

Glutaric aciduria type 1 (GA1) is a rare metabolic disorder caused by a deficiency of glutaryl-CoA dehydrogenase. The typical clinical onset features an acute encephalopathic crisis developed in early childhood, causing irreversible striatal injury. Recently, tandem mass spectrometry of spots of dried blood has allowed pre-symptomatic detection of GA1 in newborns. Early treatment can prevent irreversible neurological injury. We report the case of a girl with GA1 who exhibited a characteristic reversible change upon brain magnetic resonance imaging (MRI). She was diagnosed with GA1 as a newborn. She commenced dietary carnitine and her intake of lysine and tryptophan were reduced at the age of 4weeks. After treatment commenced, her mean glutarylcarnitine level was lower than that in the previous reports. The plasma lysine and tryptophan levels were maintained below the normal ranges. At 4months, brain MRI revealed a widened operculum with dilatation of the subarachnoid spaces surrounding the atrophic bilateral frontotemporal lobes; this is typical of GA1 patients. However, at 17months, MRI revealed that the atrophic lesion had disappeared and she subsequently underwent normal maturation. She has never suffered a metabolic decompensation episode. At 26months, her development and brain MRI were normal. The present reversible brain atrophy in a patient with GA1 indicates that early dietary modifications with a lower level of glutarylcarnitine and administration of carnitine can lead to normal development.


Subject(s)
Amino Acid Metabolism, Inborn Errors/complications , Amino Acid Metabolism, Inborn Errors/pathology , Atrophy/etiology , Brain Diseases, Metabolic/complications , Brain Diseases, Metabolic/pathology , Brain/pathology , Glutaryl-CoA Dehydrogenase/deficiency , Amino Acid Metabolism, Inborn Errors/diagnostic imaging , Amino Acids/blood , Brain/diagnostic imaging , Brain Diseases, Metabolic/diagnostic imaging , Child, Preschool , Female , Humans , Magnetic Resonance Imaging
16.
J Clin Neurosci ; 39: 108-110, 2017 May.
Article in English | MEDLINE | ID: mdl-28209305

ABSTRACT

We describe the case of a 10-year-old boy who developed reversible cerebral vasoconstriction syndrome (RCVS) after cerebellitis. He received intravenous immunoglobulin and methylprednisolone to treat the cerebellitis. However, he then presented with a sudden severe headache, vomiting, and generalized tonic-clonic seizure. Brain magnetic resonance angiography (MRA) initially revealed diffuse cerebral vasodilatations, and diffuse multifocal segmental vasoconstrictions developed several days later. His clinical symptoms gradually resolved after several days, in the absence of any specific therapy. MRA performed 46days after symptom onset showed that the multifocal segmental vasoconstrictions had resolved, suggesting a diagnosis of RCVS. The imaging features of RCVS include multifocal segmental vasoconstriction. However, our case suggests that diffuse cerebral vasodilatation may in fact be evident during the early stage of disease.


Subject(s)
Brain Diseases/diagnostic imaging , Brain Diseases/physiopathology , Vasoconstriction , Vasodilation , Child , Humans , Magnetic Resonance Angiography , Male , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...