794 results on '"neurodegeneration with brain iron accumulation"'
Search Results
102. Novel biallelic FA2H mutations in a Japanese boy with fatty acid hydroxylase-associated neurodegeneration
- Author
-
Akio Kihara, Tadashi Kaname, Masahiro Kawaguchi, Yusuke Okuno, Kohji Kato, Hideki Muramatsu, Tomohiko Nakata, Hiroyuki Yamamoto, Hiroyuki Kidokoro, Jun Natsume, and Takayuki Sassa
- Subjects
medicine.medical_specialty ,Spastic gait ,Neurodegeneration with brain iron accumulation ,Hereditary spastic paraplegia ,Neurodegeneration ,Leukodystrophy ,General Medicine ,Biology ,medicine.disease ,Compound heterozygosity ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Developmental Neuroscience ,Internal medicine ,Pediatrics, Perinatology and Child Health ,medicine ,Missense mutation ,Neurology (clinical) ,030217 neurology & neurosurgery ,Exome sequencing - Abstract
FA2H encodes fatty acid 2-hydroxylase, which plays a significant role in maintaining the neuronal myelin sheath. Previous reports have revealed that a FA2H mutation leads to spastic paraplegia, leukodystrophy, and neurodegeneration with brain iron accumulation, collectively referred to as fatty acid hydroxylase-associated neurodegeneration (FAHN). The disease severity of FAHN varies among individual patients and may be explained by the enzyme activity of FA2H mutant proteins. Here we report a 10-year-old Japanese boy with FAHN having novel heterozygous mutations in FA2H. The patient presented with a spastic gait since the age of 5 years and was unable to walk without a cane by the time he was 8 years old. Brain MRI demonstrated a partial thinning of the corpus callosum, slight reduction of cerebellar volume, and posterior dominant periventricular leukodystrophy. Whole exome sequencing revealed two novel missense mutations in FA2H with compound heterozygous inheritance (NM_024306, p.Val149Leu, and p.His260Gln mutations). The enzyme activities of the p.Val149Leu and p.His260Gln variants were 60%–80% and almost 0%, respectively. Our cell-based enzyme assay demonstrated partial functionality for one of the variants, indicating a milder phenotype. However, considered along with previous reports, there was no definite relationship between the disease severity and residual enzyme activity measured using a similar method. Further research is needed to precisely predict the phenotypic severity of this disorder.
- Published
- 2020
- Full Text
- View/download PDF
103. A case of novel WDR45 mutation with beta-propeller protein-associated neurodegeneration (BPAN) presenting asymmetrical extrapyramidal signs
- Author
-
Michiaki Koga, Naomichi Matsumoto, Takashi Kanda, Tsuyoshi Mizuguchi, Kazuhiro Iwama, and Ryota Sato
- Subjects
Pathology ,medicine.medical_specialty ,Neurodegeneration with brain iron accumulation ,business.industry ,Parkinsonism ,Neurodegeneration ,Substantia nigra ,medicine.disease ,Hyperintensity ,nervous system diseases ,03 medical and health sciences ,0302 clinical medicine ,WDR45 ,Globus pallidus ,nervous system ,medicine ,Neurology (clinical) ,Global developmental delay ,business ,030217 neurology & neurosurgery - Abstract
Beta-propeller protein-associated neurodegeneration (BPAN) is categorized in Neurodegeneration with brain iron accumulation. The clinical feature of BPAN is global developmental delay in early childhood, followed rapid progression of cognitive disfunction and parkinsonism in adulthood. This case was pointed out intellectual disability at the age of 9, followed left dominant progressive parkinsonism from the age of 31. Brain MRI showed the T1-weighted signal hyperintensity of the substantia nigra with a central band of hypointensity and the T2 star weighted image hypointensity of substantia nigra and globus pallidus presenting dominant at right side. DAT SPECT also showed specific binding ratio decreased dominant in right side. She was diagnosed BPAN based on her genetic test revealing a novel mutation (c.411dupT) in WDR45. No studies reported detailed parkinsonism like laterality in BPAN. This case indicates the left dominant parkinsonism was caused by right dominant iron deposition to substantia nigra and globus pallidus in view of MRI findings and DAT SPECT.
- Published
- 2020
- Full Text
- View/download PDF
104. Spectrum of Truncal Dystonia and Response to Treatment: A Retrospective Analysis
- Author
-
Sahil Mehta, Sucharita Ray, Kamalesh Chakravarty, and Vivek Lal
- Subjects
medicine.medical_specialty ,Neurology ,Parkinson's disease ,parkinson’s disease ,Neurodegeneration with brain iron accumulation ,camptocormia ,lcsh:RC346-429 ,03 medical and health sciences ,Camptocormia ,0302 clinical medicine ,truncal dystonia ,Internal medicine ,medicine ,030212 general & internal medicine ,botulinum toxin ,Adverse effect ,lcsh:Neurology. Diseases of the nervous system ,Dystonia ,business.industry ,medicine.disease ,Botulinum toxin ,Etiology ,Original Article ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Background: Presence of truncal dystonia usually points to a secondary cause of dystonia like exposure to dopamine receptor blockers or neurodegenerative illness. Rarely, it can occur as an idiopathic focal or segmental dystonia. Methods: Retrospective review of medical records and videos of patients of truncal dystonia presenting in the Botulinum Toxin Clinic of Department of Neurology at Post Graduate Institute of Medical Education and Research, Chandigarh between May 2016 and February 2019. Results: A total of 16 patients with predominant truncal dystonia were recruited. There were ten males and six females with mean age of 49.1 ± 15.1 years (range 22–70). Extensor truncal dystonia was the most common (12/16) followed by camptocormia (4/16). Various etiologies included Idiopathic Parkinson’s disease (4/16), Tardive dystonia (5/16), Neurodegeneration with brain iron accumulation (genetically confirmed) (2/16) and idiopathic (5/16). All patients were refractory to a combination of oral medications tried over a period of 1.82 ± 1.93 years. All patients received electromyographic-guided botulinum toxin in paraspinals or rectus abdominis muscles depending upon the type of dystonia. The mean dose of abobotulinum toxin used was 286.7 ± 108.6 units (range 200–500 units) for paraspinals and 297.5 ± 68.5 (range 200–350) for rectus abdominis muscles per session. Average subjective response after botulinum toxin injection session was 31.2 ± 21.5% (range 0–70). No adverse effects were reported. Conclusion: Botulinum toxin is an acceptable alternative to patients presenting with medically refractory truncal dystonia and may offer modest benefit.
- Published
- 2020
105. Novel PANK2 mutation identified in patient with pantothenate kinase-associated neurodegeneration
- Author
-
Nikola Kresojević, Vladimir S. Kostic, Svetlana Tomić, Ivana Novakovic, and Marina Svetel
- Subjects
Pathology ,medicine.medical_specialty ,Neurodegeneration with brain iron accumulation ,Hypomimia ,lcsh:Medicine ,Neurological examination ,Pantothenate kinase-associated neurodegeneration ,03 medical and health sciences ,Exon ,0302 clinical medicine ,pantothenate kinase-associated neurodegeneration ,medicine ,neurodegeneration with brain iron accumulation ,PANK2 ,030212 general & internal medicine ,pank2 ,Dystonia ,medicine.diagnostic_test ,business.industry ,Putamen ,lcsh:R ,General Medicine ,medicine.disease ,3. Good health ,medicine.symptom ,business - Abstract
Introduction. Pantothenate kinase-associated neurodegeneration (PKAN) is a rare, recessively inherited disorder caused by mutations in the pantothenate kinase 2 (PANK2) gene on chromosome 20p13. The objective of this report is to present a patient with atypical PKAN with the novel heterozygous PANK2 mutation. Case outline. We present a 32-year-old female who had disease onset at the age 20 (depression, speech, chewing problems and backward falls) with progressive course. Neurological examination revealed hypomimia, risus sardonicus, dysphagia, tachylalia and severe dystonic dysarthria, moderate arms, legs, and jaw-opening dystonia, postural instability, urge incontinence, and decreased visual acuity. Brain magnetic resonance imaging revealed iron accumulation in the bilateral globus pallidus and putamen (?eye-of-the-tiger?), a radiological finding pathognomonic for PKAN. Genetic analysis revealed known mutation p.T528M (c.1583C>T) in exon 6, and novel p.Y405D (c.1213T>G) in exon 3 of the PANK2 gene. In silico analyses strongly suggested this mutation to be pathogenic. Conclusion. We report a patient with PKAN, and novel substitution p.Y405D (c.1213T>G) in PANK2 that has not been previously described in PKAN patients.
- Published
- 2020
106. Different cortical excitability profiles in hereditary brain iron and copper accumulation
- Author
-
Fiore Manganelli, Rosa Iodice, Lorenzo Ugga, Marcello Esposito, Lucia Ruggiero, Raffaele Iorio, Sabina Pappatà, Alberto Piperno, Marcello De Angelis, Lucio Santoro, Paola Tarantino, Raffaele Dubbioso, Francesco Aruta, Dubbioso, Raffaele, Ruggiero, L, Esposito, M, Tarantino, Paola, DE ANGELIS, Marcello, Aruta, Francesco, Pappatà, Sabina, Ugga, Lorenzo, Piperno, Aldo, Iorio, Raffaele, Santoro, Lucio, Iodice, Rosa, Manganelli, Fiore, Dubbioso, R, Tarantino, P, De Angelis, M, Aruta, F, Pappatà, S, Ugga, L, Piperno, A, Iorio, R, Santoro, L, Iodice, R, and Manganelli, F
- Subjects
Adult ,Male ,medicine.medical_specialty ,Neurology ,Neurodegeneration with brain iron accumulation ,Wilson’s disease ,medicine.medical_treatment ,Neuroaxonal Dystrophies ,Dermatology ,Young Adult ,GABA ,03 medical and health sciences ,0302 clinical medicine ,Hepatolenticular Degeneration ,Parkinsonian Disorders ,Humans ,Medicine ,030212 general & internal medicine ,Neurodegeneration ,Aceruloplasminemia ,Neurodegeneration, Metals, Wilson’s disease, NBIA, TMS, GABA ,NBIA ,Metal ,business.industry ,Parkinsonism ,Ceruloplasmin ,Neurodegenerative Diseases ,General Medicine ,Middle Aged ,medicine.disease ,Iron Metabolism Disorders ,Transcranial Magnetic Stimulation ,Transcranial magnetic stimulation ,Psychiatry and Mental health ,medicine.anatomical_structure ,TMS ,Cortical Excitability ,Female ,Neurology (clinical) ,Alzheimer's disease ,business ,Neuroscience ,030217 neurology & neurosurgery ,Motor cortex - Abstract
Background and aim: Neurodegeneration with brain iron accumulation (NBIA) and Wilson’s disease (WD) is considered the prototype of neurodegenerative disorders characterised by the overloading of iron and copper in the central nervous system. Growing evidence has unveiled the involvement of these metals in brain cortical neurotransmission. Aim of this study was to assess cortical excitability profile due to copper and iron overload. Methods: Three patients affected by NBIA, namely two patients with a recessive hereditary parkinsonism (PARK9) and one patient with aceruloplasminemia and 7 patients with neurological WD underwent transcranial magnetic stimulation (TMS) protocols to assess cortical excitability. Specifically, we evaluated the motor thresholds that reflect membrane excitability related to the voltage-gated sodium channels in the neurons of the motor system and the ease of activation of motor cortex via glutamatergic networks, and ad hoc TMS protocols to probe inhibitory-GABAergic (short interval intracortical inhibition, SICI; short-latency afferent inhibition, SAI; cortical silent period, CSP) and excitatory intracortical circuitry (intracortical facilitation, ICF). Results: Patients with NBIA exhibited an abnormal prolongation of CSP respect to HC and WD patients. On the contrary, neurological WD displayed higher motor thresholds and reduced CSP and SICI. Conclusion: Hereditary conditions due to overload of copper and iron exhibited peculiar cortical excitability profiles that can help during differential diagnosis between these conditions. Moreover, such results can give us more clues about the role of metals in acquired neurodegenerative disorders, such as Parkinson disease, Alzheimer disease, and multiple sclerosis.
- Published
- 2019
- Full Text
- View/download PDF
107. Cortical pencil lining on SWI MRI in NBIA and healthy aging
- Author
-
Roald A. Lambrechts, Marlous C M van der Weijden, Peter Jan van Laar, Dineke S. Verbeek, Marina A. J. Tijssen, Molecular Neuroscience and Ageing Research (MOLAR), and Movement Disorder (MD)
- Subjects
Adult ,Male ,Pathology ,medicine.medical_specialty ,Neurodegeneration with brain iron accumulation ,Red nucleus ,Iron ,Caudate nucleus ,lcsh:RC346-429 ,030218 nuclear medicine & medical imaging ,Young Adult ,03 medical and health sciences ,Iron accumulation ,0302 clinical medicine ,Cortical pencil lining ,medicine ,Humans ,Child ,lcsh:Neurology. Diseases of the nervous system ,Aged ,NBIA ,business.industry ,Putamen ,Brain ,Neurodegenerative Diseases ,General Medicine ,Susceptibility-weighted imaging ,Middle Aged ,Magnetic Resonance Imaging ,Dentate nucleus ,Globus pallidus ,medicine.anatomical_structure ,Healthy aging ,Susceptibility weighted imaging ,Female ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Research Article ,Motor cortex - Abstract
BackgroundNeurodegeneration with brain iron accumulation (NBIA) is characterized by pathological iron accumulation in the subcortical nuclei and the cortex. As age-related iron accumulation studies in these structures are lacking in healthy aging, we aimed to characterize the dynamics of age-dependent iron accumulation in subcortical nuclei in healthy aging and selected NBIA cases. This is fundamental to understand the natural age-related iron deposition in the healthy brain prior to using this marker as a potential prognostic or diagnostic tool in neurodegenerative disorders.MethodsSusceptibility-weighted imaging (SWI) scans from 81 healthy volunteers (0-79 years) and four genetically confirmed patients suffering from NBIA (2-14 years) were obtained. We scored the presence or absence of pencil lining of the motor cortex and putamen and analyzed the normalized SWI signal intensity ratio (NSIR) in five subcortical nuclei.ResultsIn healthy subjects, an age-dependent increase of pencil lining occurred starting from the second decade of life and was present in all cases at the age of 50. In their first decade, NBIA patients showed no cortical pencil lining, but we did observe putaminal pencil lining at this stage. In healthy subjects, age and NSIR of all nuclei correlated positively and was particularly dynamic in early childhood until young adulthood in the globus pallidus, dentate nucleus and red nucleus, but not in the caudate nucleus and putamen. NBIA patients showed an increased NSIR in the globus pallidus only and not in the other subcortical nuclei compared to age-matched healthy subjects.ConclusionsCortical pencil lining is part of healthy aging. This should be considered when assessing this as a potential marker in NBIA diagnosis and prognosis. Putaminal pencil lining has the potential to become a specific marker for some subtypes of NBIA in the first decade of life, as it was only observed in NBIA and not in age-matched healthy subjects. NSIR in the subcortical nuclei during healthy aging was shown to be dynamic, accentuating the importance of having an age-dependent baseline.
- Published
- 2019
- Full Text
- View/download PDF
108. Brain iron transport
- Author
-
Ya Ke and Zhong-Ming Qian
- Subjects
0106 biological sciences ,Neurodegeneration with brain iron accumulation ,Iron ,Regulator ,010603 evolutionary biology ,01 natural sciences ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Receptors, Transferrin ,medicine ,Animals ,Humans ,Cerebrospinal Fluid ,030304 developmental biology ,Neurons ,0303 health sciences ,Microglia ,Chemistry ,Transferrin ,Brain ,Metabolism ,Oligodendrocyte ,Oligodendroglia ,medicine.anatomical_structure ,Blood-Brain Barrier ,Astrocytes ,Endothelium, Vascular ,Neuron ,General Agricultural and Biological Sciences ,Neuroscience ,Homeostasis ,Astrocyte - Abstract
Brain iron is a crucial participant and regulator of normal physiological activity. However, excess iron is involved in the formation of free radicals, and has been associated with oxidative damage to neuronal and other brain cells. Abnormally high brain iron levels have been observed in various neurodegenerative diseases, including neurodegeneration with brain iron accumulation, Alzheimer's disease, Parkinson's disease and Huntington's disease. However, the key question of why iron levels increase in the relevant regions of the brain remains to be answered. A full understanding of the homeostatic mechanisms involved in brain iron transport and metabolism is therefore critical not only for elucidating the pathophysiological mechanisms responsible for excess iron accumulation in the brain but also for developing pharmacological interventions to disrupt the chain of pathological events occurring in these neurodegenerative diseases. Numerous studies have been conducted, but to date no effort to synthesize these studies and ideas into a systematic and coherent summary has been made, especially concerning iron transport across the luminal (apical) membrane of the capillary endothelium and the membranes of different brain cell types. Herein, we review key findings on brain iron transport, highlighting the mechanisms involved in iron transport across the luminal (apical) as well as the abluminal (basal) membrane of the blood-brain barrier, the blood-cerebrospinal fluid barrier, and iron uptake and release in neurons, oligodendrocytes, astrocytes and microglia within the brain. We offer suggestions for addressing the many important gaps in our understanding of this important topic, and provide new insights into the potential causes of abnormally increased iron levels in regions of the brain in neurodegenerative disorders.
- Published
- 2019
- Full Text
- View/download PDF
109. Impaired iPLA2β activity affects iron uptake and storage without iron accumulation: An in vitro study excluding decreased iPLA2β activity as the cause of iron deposition in PLAN
- Author
-
Xinxiang Yan, Qiying Sun, Yu-pei Guo, Jifeng Guo, Qian Xu, Bei-sha Tang, Hong-li Liu, and Juan-juan Huang
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Neurodegeneration with brain iron accumulation ,Phospholipase ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Molecular Biology ,Mutation ,biology ,Chemistry ,General Neuroscience ,Neurodegeneration ,Antagonist ,DMT1 ,medicine.disease ,Ferritin ,030104 developmental biology ,Endocrinology ,Apoptosis ,biology.protein ,Neurology (clinical) ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
PLA2G6-associated neurodegeneration (PLAN, NBIA2) is the second most common type of neurodegeneration with brain iron accumulation (NBIA), caused by recessive mutations of PLA2G6 gene, which encodes Ca2+-independent phospholipase A2β (iPLA2β). In most PLAN cases, decreased iPLA2β activity and iron deposition was observed meanwhile, and researchers also identified a PLA2G6 mutation family without iron deposition shown by MRI images. This brought us the question of whether decreased iPLA2β activity was the cause of iron deposition in PLAN. In this study, we used S-BEL as the antagonist of iPLA2β to block its activity and used SH-SY5Y cells as the expression system. We incubated SH-SY5Y cells with different concentrations of S-BEL. The results showed that decreased iPLA2β activity led no obvious iron accumulation, while changes of cells state and activation of apoptosis were observed. To further investigate the cause of unchanged iron level, we examined the cellular iron regulatory proteins involved in iron uptake, storage and export. The results were as follows: TfR1 (iron uptake protein) expression was decreased, the expression of ferritin heavy chain and light chain (iron storage protein) was increased. There was no alteration of the expression of DMT1 (iron uptake protein) and FPN1 (iron export protein). Under the condition of decreased iPLA2β activity, there was no obvious iron accumulation but iron uptake activity decreased and iron storage activity increased. Therefore, we speculate that the decreased iPLA2β activity may not be the main reason for iron deposition in PLAN.
- Published
- 2019
- Full Text
- View/download PDF
110. Challenges in the approach and reporting of atypical manifestations of membrane protein-associated neurodegeneration (MPAN): An editorial
- Author
-
Philippe A. Salles
- Subjects
Pathology ,medicine.medical_specialty ,Atypical manifestations ,business.industry ,Neurodegeneration with brain iron accumulation ,Neurodegeneration ,Spastic paraparesis ,Membrane Proteins ,Neurodegenerative Diseases ,medicine.disease ,Mitochondrial Proteins ,Neurology ,Membrane protein ,Medicine ,Humans ,Neurology (clinical) ,Geriatrics and Gerontology ,business - Published
- 2021
111. BPAN manifesting with febrile seizures and language delay:a case report from Brazil
- Author
-
Thayane Rosas Batista Rezende, Zumira Aparecida Carneiro, Charles Marques Lourenço, and Maria Cecília de Mattos Alves Silva
- Subjects
Language delay ,Neurodegeneration with brain iron accumulation ,bpan ,Febrile seizures ,wdr45 mutation ,Neurodevelopmental diseases ,Bioinformatics ,medicine.disease_cause ,nbia ,WDR45 ,Basal ganglia ,medicine ,febrile seizures ,WDR45 mutation ,Exome sequencing ,NBIA disorders ,Mutation ,NBIA ,business.industry ,Inborn Errors of Metabolism ,Neurodegeneration ,General Medicine ,medicine.disease ,Dentate nucleus ,BPAN ,Medicine ,business - Abstract
Neurodegeneration with brain iron accumulation (NBIA) is a complex group of hereditary progressive neurodegenerative diseases characterized by deposition of iron in the basal ganglia. Twelve genetic forms of this disorder have been identified in previous studies. Though they have different inheritance mechanisms all are usually associated with abnormal brain MRI findings. One of NBIA types is an X-linked disorder known as Beta-propeller Protein Associated Neurodegeneration (BPAN). Herein we describe the case of a 4-year-old girl with 2 episodes of febrile seizures, a brain MRI showing nonspecific hyperintense signal in the dentate nucleus area, and delays in language and communication development. Her diagnosis was made based on a genetic evaluation where exome sequencing revealed a mutation in the position chrX:48.933.022 region of the WDR45 gene. The literature describes different clinical presentations for BPAN, each with a different prognosis, suggesting a wide range of possible symptoms of BPAN, including mild cognitive delay and even epileptic encephalopathy (EE).
- Published
- 2021
112. Loss of PLA2G6 leads to elevated mitochondrial lipid peroxidation and mitochondrial dysfunction.
- Author
-
Kinghorn, Kerri J., Castillo-Quan, Jorge Iván, Bartolome, Fernando, Angelova, Plamena R., Li Li, Pope, Simon, Cochemé, Helena M., Khan, Shabana, Asghari, Shabnam, Bhatia, Kailash P., Hardy, John, Abramov, Andrey Y., Partridge, Linda, and Li, Li
- Subjects
- *
MITOCHONDRIAL pathology , *PROTEIN metabolism , *ANIMALS , *BIOLOGICAL transport , *CELL lines , *ESTERASES , *FIBROBLASTS , *GENETIC techniques , *INSECTS , *MASS spectrometry , *LIPID peroxidation (Biology) , *MICROSCOPY , *MITOCHONDRIA , *NEURODEGENERATION , *POLYMERASE chain reaction , *PROTEINS , *OXIDATIVE stress , *REVERSE transcriptase polymerase chain reaction - Abstract
The PLA2G6 gene encodes a group VIA calcium-independent phospholipase A2 beta enzyme that selectively hydrolyses glycerophospholipids to release free fatty acids. Mutations in PLA2G6 have been associated with disorders such as infantile neuroaxonal dystrophy, neurodegeneration with brain iron accumulation type II and Karak syndrome. More recently, PLA2G6 was identified as the causative gene in a subgroup of patients with autosomal recessive early-onset dystonia-parkinsonism. Neuropathological examination revealed widespread Lewy body pathology and the accumulation of hyperphosphorylated tau, supporting a link between PLA2G6 mutations and parkinsonian disorders. Here we show that knockout of the Drosophila homologue of the PLA2G6 gene, iPLA2-VIA, results in reduced survival, locomotor deficits and organismal hypersensitivity to oxidative stress. Furthermore, we demonstrate that loss of iPLA2-VIA function leads to a number of mitochondrial abnormalities, including mitochondrial respiratory chain dysfunction, reduced ATP synthesis and abnormal mitochondrial morphology. Moreover, we show that loss of iPLA2-VIA is strongly associated with increased lipid peroxidation levels. We confirmed our findings using cultured fibroblasts taken from two patients with mutations in the PLA2G6 gene. Similar abnormalities were seen including elevated mitochondrial lipid peroxidation and mitochondrial membrane defects, as well as raised levels of cytoplasmic and mitochondrial reactive oxygen species. Finally, we demonstrated that deuterated polyunsaturated fatty acids, which inhibit lipid peroxidation, were able to partially rescue the locomotor abnormalities seen in aged flies lacking iPLA2-VIA gene function, and restore mitochondrial membrane potential in fibroblasts from patients with PLA2G6 mutations. Taken together, our findings demonstrate that loss of normal PLA2G6 gene activity leads to lipid peroxidation, mitochondrial dysfunction and subsequent mitochondrial membrane abnormalities. Furthermore we show that the iPLA2-VIA knockout fly model provides a useful platform for the further study of PLA2G6-associated neurodegeneration. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
113. Mutations of C19orf12, coding for a transmembrane glycine zipper containing mitochondrial protein, cause mis-localization of the protein, inability to respond to oxidative stress and increased mitochondrial Ca2+.
- Author
-
Venco, Paola, Bonora, Massimo, Giorgi, Carlotta, Papaleo, Elena, Iuso, Arcangela, Prokisch, Holger, Pinton, Paolo, and Tiranti, Valeria
- Subjects
NEURODEGENERATION ,BASAL ganglia ,MITOCHONDRIA ,ENDOPLASMIC reticulum ,PROTEIN-protein interactions - Abstract
Mutations in C19orf12 have been identified in patients affected by Neurodegeneration with Brain Iron Accumulation (NBIA), a clinical entity characterized by iron accumulation in the basal ganglia. By using western blot analysis with specific antibody and confocal studies, we showed that wild-type C19orf12 protein was not exclusively present in mitochondria, but also in the Endoplasmic Reticulum (ER) and MAM (Mitochondria Associated Membrane), while mutant C19orf12 variants presented a different localization. Moreover, after induction of oxidative stress, a GFP-tagged C19orf12 wild-type protein was able to relocate to the cytosol. On the contrary, mutant isoforms were not able to respond to oxidative stress. High mitochondrial calcium concentration and increased H
2 O2 induced apoptosis were found in fibroblasts derived from one patient as compared to controls. C19orf12 protein is a 17 kDa mitochondrial membrane-associated protein whose function is still unknown. Our in silico investigation suggests that, the glycine zipper motifs of C19orf12 form helical regions spanning the membrane. The N- and C-terminal regions with respect to the transmembrane portion, on the contrary, are predicted to rearrange in a structural domain, which is homologs to the N-terminal regulatory domain of the magnesium transporter MgtE, suggesting that C19orf12 may act as a regulatory protein for human MgtE transporters. The mutations here described affect respectively one glycine residue of the glycine zipper motifs, which are involved in dimerization of transmembrane helices and predicted to impair the correct localization of the protein into the membranes, and one residue present in the regulatory domain, which is important for protein-protein interaction. [ABSTRACT FROM AUTHOR]- Published
- 2015
- Full Text
- View/download PDF
114. Parkinsonism and ataxia
- Author
-
Giulia Lazzeri, Giulia Franco, and Alessio Di Fonzo
- Subjects
Ataxia ,Movement disorders ,Parkinson's disease ,Cerebellar ataxia ,Cerebellar Ataxia ,Neurodegeneration with brain iron accumulation ,business.industry ,Parkinsonism ,Neurodegeneration ,Parkinson Disease ,Disease ,Multiple System Atrophy ,medicine.disease ,Neurology ,Parkinsonian Disorders ,medicine ,Humans ,Neurology (clinical) ,medicine.symptom ,business ,Neuroscience - Abstract
Ataxia is not a common feature in Parkinson's disease. Nevertheless, some rare forms of parkinsonism have ataxia as one of the main features in their clinical picture, especially those with juvenile or early-onset. On the other side, in cerebellar degenerative diseases, parkinsonism might accompany the typical symptoms and even become predominant in some cases. Many disorders involving different neurological systems present with a movement phenomenology reflecting the underlying pattern of pathological involvement, such as neurodegeneration with brain iron accumulation, neurodegeneration associated with calcium deposition, and metabolic and mitochondrial disorders. The prototype of sporadic disorders that present with a constellation of symptoms due to the involvement of multiple Central Nervous System regions is multiple system atrophy, whose motor symptoms at onset can be cerebellar ataxia or parkinsonism. Clinical syndromes encompassing both parkinsonian and cerebellar features might represent a diagnostic challenge for neurologists. Recognizing acquired and potentially treatable causes responsible for complex movement disorders is of paramount importance, since an early diagnosis is essential to prevent permanent consequences. The present review aims to provide a pragmatic overview of the most common diseases characterized by the coexistence of cerebellar and parkinsonism features and suggests a possible diagnostic approach for both inherited and sporadic disorders. This article is part of the Special Issue "Parkinsonism across the spectrum of movement disorders and beyond" edited by Joseph Jankovic, Daniel D. Truong and Matteo Bologna.
- Published
- 2021
115. Neurodegeneration with brain iron accumulation: Characterization of clinical, radiological, and genetic features of pediatric patients from Southern India
- Author
-
Anita Mahadevan, Rashmi Santhoshkumar, Naveen Kumar Bhardwaj, Ashwin Vivek Sardesai, Jitendra Kumar Saini, and Vykuntaraju K Gowda
- Subjects
Male ,Pediatrics ,medicine.medical_specialty ,Movement disorders ,Neurodegeneration with brain iron accumulation ,Neuroaxonal Dystrophies ,India ,Epilepsy ,Developmental Neuroscience ,Neuroimaging ,Medicine ,Humans ,Child ,Pantothenate Kinase-Associated Neurodegeneration ,Retrospective Studies ,business.industry ,Neurodegeneration ,General Medicine ,medicine.disease ,Iron Metabolism Disorders ,Kufor Rakeb syndrome ,Pediatrics, Perinatology and Child Health ,Cohort ,Cerebellar atrophy ,Female ,Neurology (clinical) ,medicine.symptom ,business - Abstract
Background Neurodegeneration with brain iron accumulation (NBIA) is a group of rare inherited neurodegenerative disorders. Ten types of NBIA are known. Studies reporting various NBIA subtypes together are few. This study was aimed at describing clinical features, neuroimaging findings, and genetic mutations of different NBIA group disorders. Methods Clinical, radiological, and genetic data of patients diagnosed with NBIA in a tertiary care centre in Southern India from 2014 to 2020 was retrospectively collected and analysed. Results In our cohort of 27 cases, PLA2G6-associated neurodegeneration (PLAN) was most common (n = 13) followed by Pantothenate kinase-associated neurodegeneration (PKAN) (n = 9). We had 2 cases each of Mitochondrial membrane-associated neurodegeneration (MPAN) and Beta-propeller protein- associated neurodegeneration (BPAN) and 1 case of Kufor-Rakeb Syndrome (KRS). Walking difficulty was the presenting complaint in all PKAN cases, whereas the presentation in PLAN was that of development regression with onset at a mean age of 2 years. Overall, 50% patients of them presented with development regression and one-third had epilepsy. Presence of pyramidal signs was most common examination feature (89%) followed by one or more eye findings (81%) and movement disorders (50%). Neuroimaging was abnormal in 24/27 cases and cerebellar atrophy was the commonest finding (52%) followed by globus pallidus hypointensities (44%). Conclusions One should have a high index of clinical suspicion for the diagnosis of NBIA in children presenting with neuroregression and vision abnormalities in presence of pyramidal signs or movement disorders. Neuroimaging and ophthalmological evaluation provide important clues to diagnosis in NBIA syndromes.
- Published
- 2021
116. Down regulation of the expression of mitochondrial phosphopantetheinyl-proteins in pantothenate kinase-associated neurodegeneration: pathophysiological consequences and therapeutic perspectives
- Author
-
Alejandra Suárez-Carrillo, Suleva Povea-Cabello, Marta Talaverón-Rey, Juan M. Suárez-Rivero, José Antonio Sánchez-Alcázar, Irene Villalón-García, Joaquín J. Salas, Manuel Munuera-Cabeza, Mónica Álvarez-Córdoba, Instituto de Salud Carlos III, European Commission, Junta de Andalucía, Asociación de Enfermos de Neurodegeneración con Acumulación Cerebral de Hierro (España), Asociación de Enfermos de Patologías Mitocondriales (España), Federación Española de Enfermedades Raras, and Fundación Merck Salud
- Subjects
0301 basic medicine ,Neurodegeneration with brain iron accumulation ,Coenzyme A ,Down-Regulation ,Mitochondrion ,Pantothenate kinase-associated neurodegeneration ,Mitochondrial Proteins ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Pantothenate ,Humans ,Pharmacology (medical) ,Acyl carrier protein ,Genetics (clinical) ,biology ,4′-phosphopantetheinylation ,Chemistry ,Pantothenate kinase ,Research ,Induced neurons ,Neurodegeneration ,General Medicine ,PANK2 ,medicine.disease ,Mitochondria ,Fatty acid synthase ,Phosphotransferases (Alcohol Group Acceptor) ,030104 developmental biology ,Biochemistry ,biology.protein ,Medicine ,030217 neurology & neurosurgery - Abstract
16 Páginas.-- 7 Figuras, Background: Neurodegeneration with brain iron accumulation (NBIA) is a group of genetic neurological disorders frequently associated with iron accumulation in the basal nuclei of the brain characterized by progressive spasticity, dystonia, muscle rigidity, neuropsychiatric symptoms, and retinal degeneration or optic nerve atrophy. Pantothenate kinase-associated neurodegeneration (PKAN) is the most widespread NBIA disorder. It is caused by mutations in the gene of pantothenate kinase 2 (PANK2) which catalyzes the first reaction of coenzyme A (CoA) biosynthesis. Thus, altered PANK2 activity is expected to induce CoA deficiency as well as low levels of essential metabolic intermediates such as 4'-phosphopantetheine which is a necessary cofactor for critical proteins involved in cytosolic and mitochondrial pathways such as fatty acid biosynthesis, mitochondrial respiratory complex I assembly and lysine and tetrahydrofolate metabolism, among other metabolic processes. Methods: In this manuscript, we examined the effect of PANK2 mutations on the expression levels of proteins with phosphopantetheine cofactors in fibroblast derived from PKAN patients. These proteins include cytosolic acyl carrier protein (ACP), which is integrated within the multifunctional polypeptide chain of the fatty acid synthase involved in cytosolic fatty acid biosynthesis type I (FASI); mitochondrial ACP (mtACP) associated with mitocondrial fatty acid biosynthesis type II (FASII); mitochondrial alpha-aminoadipic semialdehyde synthase (AASS); and 10-formyltetrahydrofolate dehydrogenases (cytosolic, ALD1L1, and mitochondrial, ALD1L2). Results: In PKAN fibroblasts the expression levels of cytosolic FAS and ALD1L1 were not affected while the expression levels of mtACP, AASS and ALD1L2 were markedly reduced, suggesting that 4'-phosphopantetheinylation of mitochondrial but no cytosolic proteins were markedly affected in PKAN patients. Furthermore, the correction of PANK2 expression levels by treatment with pantothenate in selected mutations with residual enzyme content was able to correct the expression levels of mitochondrial phosphopantetheinyl-proteins and restore the affected pathways. The positive effects of pantothenate in particular mutations were also corroborated in induced neurons obtained by direct reprograming of mutant PANK2 fibroblasts. Conclusions: Our results suggest that the expression levels of mitochondrial phosphopantetheinyl-proteins are severely reduced in PKAN cells and that in selected mutations pantothenate increases the expression levels of both PANK2 and mitochondrial phosphopantetheinyl-proteins associated with remarkable improvement of cell pathophysiology., This work was supported by FIS PI16/00786 and PI19/00377 grants, Instituto de Salud Carlos III, Spain and Fondo Europeo de Desarrollo Regional (FEDER-Unión Europea), Proyectos de Investigación de Excelencia de la Junta de Andalucía CTS-5725 and PY18-850 and by ENACH (Asociación de Enfermos de Neurodegeneración con Acumulación Cerebral de Hierro), AEPMI (Asociación de Enfermos de Patología Mitocondrial), FEDER (Federación Española de Enfermedades Raras) and Fundación MERK Salud. S. Povea-Cabello is a recipient of a PhD fellowship from the Ministerio de Economía y Competitividad (MINECO).
- Published
- 2021
117. Generation of a human induced pluripotent stem cell (iPSC) line (IBMS-iPSC-070-02) from a patient with neurodegeneration with brain iron accumulation (NBIA) having compound heterozygous mutations in PANK2 gene
- Author
-
Chin-Hsien Lin, Cheng-Yen Huang, Chih-Hsin Ou-Yang, Chun-Hwei Tai, and Han-Yi Lin
- Subjects
0301 basic medicine ,Neurodegeneration with brain iron accumulation ,Induced Pluripotent Stem Cells ,Mutant ,Neuroaxonal Dystrophies ,Biology ,Mitochondrion ,Compound heterozygosity ,Peripheral blood mononuclear cell ,03 medical and health sciences ,0302 clinical medicine ,Humans ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Pantothenate Kinase-Associated Neurodegeneration ,Brain ,Cell Biology ,General Medicine ,PANK2 ,Iron Metabolism Disorders ,Phosphotransferases (Alcohol Group Acceptor) ,030104 developmental biology ,lcsh:Biology (General) ,Mutation ,Leukocytes, Mononuclear ,Cancer research ,Female ,Cellular model ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Neurodegeneration with brain iron accumulation (NBIA) is a genetically and phenotypically heterogeneous group of inherited neurodegenerative disorder characterized by basal ganglia iron deposition. Mutations in Pantothenate Kinase 2 (PANK2) are major genetic causes for patients with NBIA. The location of PANK2 in the mitochondria suggests mutant PANK2 causing mitochondrial dysfunction in the pathogenesis of NBIA. Here, we used the Sendai virus delivery system to generate induced pluripotent stem cells (iPSCs) from peripheral blood mononuclear cells of a female patient having compound heterozygous mutations in PANK2. This cellular model could provide a platform for pathophysiological studies of NBIA in the future.
- Published
- 2021
118. Towards Precision Therapies for Inherited Disorders of Neurodegeneration with Brain Iron Accumulation
- Author
-
Manju A. Kurian, Robert V.V. Spaull, Audrey Ker Shin Soo, Penelope Hogarth, and Susan J. Hayflick
- Subjects
NBIA ,business.industry ,Neurodegeneration with brain iron accumulation ,Iron ,PLAN ,Clinical study design ,PKAN ,Brain ,Neurodegenerative Diseases ,Review ,Disease ,Bioinformatics ,medicine.disease ,gene therapy ,Pantothenate kinase-associated neurodegeneration ,MPAN ,Clinical trial ,Genome editing ,pantothenate kinase-associated neurodegeneration ,Increased iron ,Humans ,Medicine ,Personalized medicine ,business - Abstract
Background: Neurodegeneration with brain iron accumulation (NBIA) disorders comprise a group of rare but devastating inherited neurological diseases with unifying features of progressive cognitive and motor decline, and increased iron deposition in the basal ganglia. Although at present there are no proven disease-modifying treatments, the severe nature of these monogenic disorders lends to consideration of personalized medicine strategies, including targeted gene therapy. In this review we summarize the progress and future direction towards precision therapies for NBIA disorders. Methods: This review considered all relevant publications up to April 2021 using a systematic search strategy of PubMed and clinical trials databases. Results: We review what is currently known about the underlying pathophysiology of NBIA disorders, common NBIA disease pathways, and how this knowledge has influenced current management strategies and clinical trial design. The safety profile, efficacy and clinical outcome of clinical studies are reviewed. Furthermore, the potential for future therapeutic approaches is also discussed. Discussion: Therapeutic options in NBIAs remain very limited, with no proven disease-modifying treatments at present. However, a number of different approaches are currently under development with increasing focus on targeted precision therapies. Recent advances in the field give hope that novel strategies, such as gene therapy, gene editing and substrate replacement therapies are both scientifically and financially feasible for these conditions. Highlights This article provides an up-to-date review of the current literature about Neurodegeneration with Brain Iron Accumulation (NBIA), with a focus on disease pathophysiology, current and previously trialed therapies, and future treatments in development, including consideration of potential genetic therapy approaches.
- Published
- 2021
- Full Text
- View/download PDF
119. A comprehensive phenotypic characterization of a whole-body Wdr45 knock-out mouse
- Author
-
Svenja-Viola Hensler, Martin Hrabě de Angelis, Wolfgang Wurst, Antonio Aguilar-Pimentel, Thomas Meitinger, Patricia da Silva-Buttkus, Jan Rozman, Julia Calzada-Wack, Oana V. Amarie, Enrica Zanuttigh, Thomas Klopstock, Silvia Vidali, Irina Treise, Dirk Janik, Tanja Klein-Rodewald, Arcangela Iuso, Michael Faerberboeck, Lillian Garrett, Lore Becker, Helmut Fuchs, Caroline Biagosch, Johannes A. Mayr, Valerie Gailus-Durner, Holger Prokisch, and Birgit Rathkolb
- Subjects
Male ,medicine.medical_specialty ,Neurodegeneration with brain iron accumulation ,Neuropathology ,Biology ,genetics [Carrier Proteins] ,Article ,Exon ,Mice ,WDR45 ,Internal medicine ,ddc:570 ,Genetics ,medicine ,Animals ,Gene ,Mice, Knockout ,Neurodegeneration ,medicine.disease ,Phenotype ,ddc ,Endocrinology ,Knockout mouse ,Female ,Carrier Proteins - Abstract
Pathogenic variants in the WDR45 (OMIM: 300,526) gene on chromosome Xp11 are the genetic cause of a rare neurological disorder characterized by increased iron deposition in the basal ganglia. As WDR45 encodes a beta-propeller scaffold protein with a putative role in autophagy, the disease has been named Beta-Propeller Protein-Associated Neurodegeneration (BPAN). BPAN represents one of the four most common forms of Neurodegeneration with Brain Iron Accumulation (NBIA). In the current study, we generated and characterized a whole-body Wdr45 knock-out (KO) mouse model. The model, developed using TALENs, presents a 20-bp deletion in exon 2 of Wdr45. Homozygous females and hemizygous males are viable, proving that systemic depletion of Wdr45 does not impair viability and male fertility in mice. The in-depth phenotypic characterization of the mouse model revealed neuropathology signs at four months of age, neurodegeneration progressing with ageing, hearing and visual impairment, specific haematological alterations, but no brain iron accumulation. Biochemically, Wdr45 KO mice presented with decreased complex I (CI) activity in the brain, suggesting that mitochondrial dysfunction accompanies Wdr45 deficiency. Overall, the systemic Wdr45 KO described here complements the two mouse models previously reported in the literature (PMIDs: 26,000,824, 31,204,559) and represents an additional robust model to investigate the pathophysiology of BPAN and to test therapeutic strategies for the disease.
- Published
- 2021
- Full Text
- View/download PDF
120. Iron Metabolism and Related Disorders
- Author
-
Karin E. Finberg and Yelena Ginzburg
- Subjects
Genetics ,Sideroblastic anemia ,biology ,Iron-deficiency anemia ,Hepcidin ,Neurodegeneration with brain iron accumulation ,Microcytic anemia ,Ferroportin ,medicine ,biology.protein ,medicine.disease ,Aceruloplasminemia ,Hemochromatosis - Abstract
Iron is an essential element for numerous fundamental biological processes, including erythropoiesis; however, in large quantities and when unbound, iron can be highly toxic to cells. Much of iron metabolism involves maintaining a delicate balance between sequestered and available iron by regulating iron movement between compartments where iron can be absorbed, recycled, and stored. A wide spectrum of clinical disorders result from mutations in gene products with key roles in cellular and systemic iron regulation. Mutations in the gene products that encode the master iron regulatory hormone hepcidin or its receptor ferroportin lead to several hereditary forms of hemochromatosis with juvenile or adult onset. Genetic defects that restrict iron availability to erythroid precursors underlie several clinically-distinct forms of microcytic anemia that are also characterized by dysregulated systemic iron balance, including iron-refractory iron deficiency anemia, DMT1 mutation-related hypochromic microcytic anemia, and congenital atransferrinemia. A variety of other inherited conditions, including hereditary hyperferritinemia-cataract syndrome, neuroferritinopathy, aceruloplasminemia, Friedreich ataxia, and X-linked sideroblastic anemia with ataxia, also result from genetic defects that disrupt iron homeostasis at the local or systemic level. Furthermore, several genetic forms of neurodegeneration with brain iron accumulation have been described for which the relationship of the underlying mutated gene product to iron metabolism remains to be elucidated. In this chapter, we review the clinical features, molecular genetics, pathophysiology, diagnosis, and management of the major genetic disorders of iron metabolism.
- Published
- 2021
- Full Text
- View/download PDF
121. Novel Mutations In Atp13A2 Associated With Mixed Neurological Presentations And Iron Toxicity Due To Nonsense-Mediated Decay
- Author
-
Koray Kırımtay, Arzu Karabay, Zuhal Yapici, Murat Gultekin, and Benan Temizci
- Subjects
Adult ,Male ,0301 basic medicine ,genetic structures ,Neurodegeneration with brain iron accumulation ,Iron ,Primary Cell Culture ,Nonsense-mediated decay ,Mutant ,Biology ,medicine.disease_cause ,Frameshift mutation ,03 medical and health sciences ,0302 clinical medicine ,Parkinsonian Disorders ,Western blot ,Exome Sequencing ,mental disorders ,medicine ,Humans ,Molecular Biology ,Mutation ,Messenger RNA ,medicine.diagnostic_test ,Siblings ,General Neuroscience ,Parkinsonism ,Brain ,medicine.disease ,Magnetic Resonance Imaging ,Molecular biology ,eye diseases ,Nonsense Mediated mRNA Decay ,nervous system diseases ,Proton-Translocating ATPases ,030104 developmental biology ,Female ,Neurology (clinical) ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Background Kufor-Rakeb Syndrome (KRS) is an autosomal recessive disease characterized by Parkinsonism, pyramidal signs, dementia, and supranuclear gaze palsy. KRS is caused by mutations in ATP13A2 producing a transmembrane protein responsible for the regulation of intracellular inorganic cations. Objective Two siblings born to a Turkish family of consanguineous marriage had mixed neurological presentations with the presence of hypointense images on T2-weighted MRI and were pre-diagnosed as having autosomal recessive spastic paraparesis or ataxia. We aimed to identify the disease-causing mutation by whole-exome sequencing and elucidate the underlying molecular mechanism of the causative mutation. Methods Prussian blue staining was conducted for the detection of cellular iron accumulation. Disease-causing mutation in ATP13A2 was detected by whole-exome sequencing. Expression levels of ATP13A2 mRNA and protein were assessed by qRT-PCR and Western Blot. Results Iron deposits in the patients’ fibroblasts were detected by Prussian blue staining. Novel homozygous mutation c.1422_1423del:p.P474fs was detected in the ATP13A2. As this mutation caused a premature termination codon (PTC), the expression of mutant ATP13A2 mRNA through qRT-PCR analysis was found to be degraded by nonsense-mediated decay and this prevented the expression of ATP13A2 protein in the patients’ fibroblasts. Conclusions Novel frameshift mutation causing a PTC in ATP13A2 lead to degradation of ATP13A2 mRNA by NMD. Iron accumulation due to the absence of ATP13A2 protein in the patient’s fibroblasts and hypointense areas on T2-weighted images may expand the spectrum of KRS to consider it as neurodegeneration with brain iron accumulation disorders.
- Published
- 2021
122. Pantothenate kinase-associated neurodegeneration: Clinical aspects, diagnosis and treatments.
- Author
-
Razmeh, Saeed, Habibi, Amir Hassan, Orooji, Maryam, Alizadeh, Elham, Moradiankokhdan, Karim, and Razmeh, Behroz
- Subjects
- *
NEURODEGENERATION , *TREATMENT of neurodegeneration , *MAGNETIC resonance imaging , *DIAGNOSIS - Abstract
Pantothenate Kinase-Associated Neurodegeneration (PKAN) is an autosomal recessive disorder characterized by a mutation in the PANK2 gene. The clinical presentation may range from only speech disorder to severe generalized dystonia, spasticity, Visual loss, dysphagia and dementia. The hallmark of this disease is eyes of the tiger sign in the medial aspect of bilateral globus pallidus on T2-weighted MRI that is a hyperintense lesion surrounded by hypointensity. Common treatments for PKAN disease include anticholinergics, botulinum toxin, Oral and Intrathecal baclofen, Iron chelation drugs and surgical procedures such as ablative pallidotomy or thalamotomy, Deep brain stimulation. There are many controversies about the pathogenesis and treatment of this disease, and in recent years interesting studies have been done on PKAN disease and other similar diseases. This review summarizes the clinical presentation, etiology, imaging modalities and treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
123. A pilot trial of deferiprone in pantothenate kinase-associated neurodegeneration patients.
- Author
-
Rohani, Mohammad, Razmeh, Saeed, Ali Shahidi, Gholam, Alizadeh, Elham, and Orooji, Maryam
- Subjects
NEURODEGENERATION ,CHROMOSOMES ,MAGNETIC resonance imaging - Abstract
Pantothenate kinase-associated neurodegeneration (PKAN) is the most common form of neurodegeneration with brain iron accumulation, it is an autosomal recessive disease due to mutation in PANK 2 on chromosome 20, which causes the accumulation of iron in basal ganglia and production of free radicals that cause degeneration of the cells. Deferiprone is an iron chelator that was used in treatment of thalassemia patients, it can cross the blood-brain barrier and reverse the iron deposition in the brain. Five patients with genetically confirmed PKAN received 15 mg/kg deferiprone twice daily. All patients were examined at baseline, 12 and 18 months and magnetic resonance imaging (MRI) was done at the baseline and after 18 months. In our study qualitative evaluation of MRI showed that deferiprone was able to reduce the iron load in globus pallidus of all the patients and the results of clinical rating scales show that in four patients, there is an improvement in the first 12 months. The results of our paper show that deferiprone can prevent the progression of the disease. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
124. Tremor-Dominant Pantothenate Kinase-associated Neurodegeneration.
- Author
-
Rohani, Mohammad, Shahidi, Gholamali, Alavi, Afagh, Lang, Anthony E., Yousefi, Niloufar, Razme, Said, and Fasano, Alfonso
- Subjects
- *
NEUROLOGICAL disorders , *PARKINSON'S disease , *NEURODEGENERATION , *MOVEMENT disorders , *GENETIC testing - Abstract
Neurodegeneration with brain iron accumulation ( NBIA) includes a rare and heterogeneous group of disorders characterized by iron deposition in the basal ganglia. Pantothenate kinase-associated neurodegeneration ( PKAN) is the most common NBIA and has 2 main presentations: typical and atypical, the latter rarely presents with tremor. Our reported patients underwent full neurologic examination, standard brain magnetic imaging, and genetic testing for PKAN. Three patients who had 'tremor-dominant' PKAN with a relatively benign course were reported, including 1 with dystonic tremor and 2 with parkinsonian tremor. All 3 patients had homozygous mutations in the PANK2 gene and typical eye of the tiger sign on brain imaging. PKAN (and NBIA in general) may be a potential cause of tremor, thus emphasizing the need to consider this diagnosis even in patients with a clinical diagnosis of essential, dystonic, or parkinsonian tremor. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
125. Seizure in Neurodegeneration with Brain Iron Accumulation: A Systematic Review.
- Author
-
Emamikhah M, Saiyarsarai P, Schneider SA, Fasano A, Mohammadzadeh N, and Rohani M
- Subjects
- Female, Humans, Child, Preschool, Child, Adolescent, Young Adult, Adult, Male, Seizures, Brain, Iron, Pantothenate Kinase-Associated Neurodegeneration, Epilepsy
- Abstract
Background: Neurodegeneration with brain iron accumulation (NBIA) is a rare genetic disorder. Its clinical manifestations comprise a wide spectrum mainly movement disorders. Seizure as a clinical manifestation is known to occur in some NBIAs, but the exact prevalence of epilepsy in each individual disorder is not well elucidated. The aim of this review was to investigate the frequency of seizures in NBIA disorders as well as to determine the associated features of patients with seizures., Method: The electronic bibliographic databases PubMed, Scopus, Embase, and Google Scholar were systematically searched for all cases in any type of article from inception to December 16, 2019. All the reported cases of NBIA (with or without genetic confirmation) were identified. Case reports with an explicit diagnosis of any types of NBIA, which have reported occurrence (or absence) of any type of seizure or epilepsy, in the English language, were included. Seizure incidence rate, type, and age of onset were reported as frequencies and percentages., Result: 1698 articles were identified and 51 were included in this review. Of 305 reported cases, 150 (49.2%) had seizures (phospholipase A2-associated neurodegeneration (PLAN) = 64 (50.8%), beta-propeller protein-associated neurodegeneration (BPAN) = 57 (72.1%), pantothenate kinase-associated neurodegeneration (PKAN) = 11 (23.4%), and others = 18 (very variable proportions)). The most frequent seizure type in NBIA patients was generalized tonic-clonic seizure with the mean age of seizure onset between 2 and 36 years. However, most of these papers had been published before the new classification of epilepsy became accessible. Affected patients were more likely to be females., Conclusion: Seizures are common in NBIA, particularly in PLAN and BPAN. In PKAN, the most common type of NBIA, around 10% of patients are affected by seizures. BPAN is the most possible NBIA accompanying seizure. Most of the findings regarding the seizure characteristics in the NBIAs are biased due to the huge missing data. Therefore, any conclusions should be made with caution and need further investigations.
- Published
- 2023
- Full Text
- View/download PDF
126. Paramagnetic Signals in the Globus Pallidus as Late Radiographic Sign of Juvenile-Onset GM1 Gangliosidosis.
- Author
-
Takenouchi, Toshiki, Kosaki, Rika, Nakabayashi, Kazuhiko, Hata, Kenichiro, Takahashi, Takao, and Kosaki, Kenjiro
- Subjects
- *
PARAMAGNETIC materials , *CELLULAR signal transduction , *GLOBUS pallidus , *BRAIN , *RADIOGRAPHY , *GANGLIOSIDOSES , *LYSOSOMAL storage diseases , *PHYSICIANS , *DIAGNOSIS - Abstract
Background The juvenile form of GM1 gangliosidosis lacks specific physical findings and thus is often a diagnostic challenge for clinicians. T 2 hypodensity in the globus pallidus is a characteristic radiographic sign of neurodegeneration with iron accumulation in the brain that is observed in GM1 gangliosidosis, but the exact timing when this radiographic sign becomes apparent remains to be elucidated. Patients Two male siblings had normal development until 2 years of age and then developed psychomotor regression with dystonia. Their neuroimaging studies indicated progressive global cerebral atrophy. Exome sequencing identified compound heterozygous missense mutations in GLB1 , leading to a diagnosis of GM1 gangliosidosis. Results A retrospective review of neuroimaging studies revealed that the two patients had strikingly similar clinical courses and radiographic progressions with cortical atrophy that preceded the T 2 hypointensity in the globus pallidus. Conclusions Paramagnetic signals in the globus pallidus become apparent relatively late during the disease course, once cerebral atrophy has already become prominent. A comprehensive diagnostic approach involving clinical, radiographic, and genetic testing is necessary for the early identification of affected individuals. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
127. Neurodegeneration with Brain Iron Accumulation: An Overview.
- Author
-
TONEKABONI, Seyed Hassan and MOLLAMOHAMMADI, Mohsen
- Subjects
BRAIN anatomy ,DIAGNOSIS of brain diseases ,SPASTICITY ,ENZYMES ,DIAGNOSTIC imaging ,IRON ,NEURODEGENERATION ,NEUROLOGY ,SERIAL publications ,PANTOTHENIC acid ,DIAGNOSIS ,PHYSIOLOGY ,THERAPEUTICS - Abstract
Objective Neurodegeneration with brain iron accumulation (NBIA) is a group of neurodegenerative disorder with deposition of iron in the brain (mainly Basal Ganglia) leading to a progressive Parkinsonism, spasticity, dystonia, retinal degeneration, optic atrophy often accompanied by psychiatric manifestations and cognitive decline. 8 of the 10 genetically defined NBIA types are inherited as autosomal recessive and the remaining two by autosomal dominant and X-linked dominant manner. Brain MRI findings are almost specific and show abnormal brain iron deposition in basal ganglia some other related anatomical locations. In some types of NBIA cerebellar atrophy is the major finding in MRI. [ABSTRACT FROM AUTHOR]
- Published
- 2014
128. Late diagnosis of fucosidosis in a child with progressive fixed dystonia, bilateral pallidal lesions and red spots on the skin.
- Author
-
Gautschi, Matthias, Merlini, Laura, Calza, Anne-Marie, Hayflick, Susan, Nuoffer, Jean-Marc, and Fluss, Joel
- Abstract
Abstract: Fucosidosis is a rare lysosomal storage disease. A 14-year-old girl is presented, with recurrent infections, progressive dystonic movement disorder and mental retardation with onset in early childhood. The clinical picture was also marked by mild morphologic features, but absent dysostosis multiplex and organomegaly. MRI images at 6.5 years of age were reminiscent of pallidal iron deposition (“eye-of-the-tiger” sign) seen in neurodegeneration with brain iron accumulation (NBIA) disorders. Progressively spreading angiokeratoma corporis diffusum led to the correct diagnosis. This case extends the scope of clinical and neuroradiological manifestations of fucosidosis. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
129. Molecular Defects in Friedreich’s Ataxia: Convergence of Oxidative Stress and Cytoskeletal Abnormalities
- Author
-
Frances M. Smith and Daniel J. Kosman
- Subjects
0301 basic medicine ,Ataxia ,Neurodegeneration with brain iron accumulation ,Friedreich’s ataxia ,Context (language use) ,Review ,medicine.disease_cause ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Biochemistry ,Nrf2 ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Molecular Biosciences ,glutathione ,Cytoskeleton ,Molecular Biology ,lcsh:QH301-705.5 ,Actin ,frataxin ,biology ,Neurodegeneration ,cytoskeleton ,medicine.disease ,Cell biology ,030104 developmental biology ,PIP5K1β ,lcsh:Biology (General) ,030220 oncology & carcinogenesis ,Frataxin ,biology.protein ,medicine.symptom ,Oxidative stress - Abstract
Friedreich's ataxia (FRDA) is a multi-faceted disease characterized by progressive sensory-motor loss, neurodegeneration, brain iron accumulation, and eventual death by hypertrophic cardiomyopathy. FRDA follows loss of frataxin (FXN), a mitochondrial chaperone protein required for incorporation of iron into iron-sulfur cluster and heme precursors. After the discovery of the molecular basis of FRDA in 1996, over two decades of research have been dedicated to understanding the temporal manifestations of disease both at the whole body and molecular level. Early research indicated strong cellular iron dysregulation in both human and yeast models followed by onset of oxidative stress. Since then, the pathophysiology due to dysregulation of intracellular iron chaperoning has become central in FRDA relative to antioxidant defense and run-down in energy metabolism. At the same time, limited consideration has been given to changes in cytoskeletal organization, which was one of the first molecular defects noted. These alterations include both post-translational oxidative glutathionylation of actin monomers and differential DNA processing of a cytoskeletal regulator PIP5K1β. Currently unknown in respect to FRDA but well understood in the context of FXN-deficient cell physiology is the resulting impact on the cytoskeleton; this disassembly of actin filaments has a particularly profound effect on cell-cell junctions characteristic of barrier cells. With respect to a neurodegenerative disorder such as FRDA, this cytoskeletal and tight junction breakdown in the brain microvascular endothelial cells of the blood-brain barrier is likely a component of disease etiology. This review serves to outline a brief history of this research and hones in on pathway dysregulation downstream of iron-related pathology in FRDA related to actin dynamics. The review presented here was not written with the intent of being exhaustive, but to instead urge the reader to consider the essentiality of the cytoskeleton and appreciate the limited knowledge on FRDA-related cytoskeletal dysfunction as a result of oxidative stress. The review examines previous hypotheses of neurodegeneration with brain iron accumulation (NBIA) in FRDA with a specific biochemical focus.
- Published
- 2020
- Full Text
- View/download PDF
130. PLA2G6-associated neurodegeneration (PLAN): Further expansion of the clinical, radiological and mutation spectrum associated with infantile and atypical childhood-onset disease.
- Author
-
Illingworth, M.A., Meyer, E., Chong, W.K., Manzur, A.Y., Carr, L.J., Younis, R., Hardy, C., McDonald, F., Childs, A.M., Stewart, B., Warren, D., Kneen, R., King, M.D., Hayflick, S.J., and Kurian, M.A.
- Subjects
- *
NEURODEGENERATION , *GENETIC mutation , *PHOSPHOLIPASE A2 , *PHENOTYPES , *BRAIN imaging , *JUVENILE diseases - Abstract
Abstract: Phospholipase A2 associated neurodegeneration (PLAN) is a major phenotype of autosomal recessive Neurodegeneration with Brain Iron Accumulation (NBIA). We describe the clinical phenotypes, neuroimaging features and PLA2G6 mutations in 5 children, of whom 4 presented with infantile neuroaxonal dystrophy (INAD). One other patient was diagnosed with the onset of PLAN in childhood, and our report highlights the diagnostic challenges associated with this atypical PLAN subtype. In this series, the neuroradiological relevance of classical PLAN features as well as apparent claval hypertrophy’ is explored. Novel PLA2G6 mutations were identified in all patients. PLAN should be considered not only in patients presenting with a classic INAD phenotype but also in older patients presenting later in childhood with non-specific progressive neurological features including social communication difficulties, gait disturbance, dyspraxia, neuropsychiatric symptoms and extrapyramidal motor features. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
131. Novel homozygous PANK2 mutation causing atypical pantothenate kinase-associated neurodegeneration (PKAN) in a Cypriot family.
- Author
-
Tanteles, George A., Spanou-Aristidou, Elena, Antoniou, Chloe, Christophidou-Anastasiadou, Violetta, and Kleopa, Kleopas A.
- Subjects
- *
PROTEIN kinases , *GENETIC mutation , *NEURODEGENERATION , *HUMAN chromosomes , *MISSENSE mutation , *HYPOKINESIA - Abstract
Abstract: Pantothenate kinase-associated neurodegeneration (PKAN) is the commonest, recessively inherited form of neurodegeneration with brain iron accumulation (NBIA) resulting from mutations in the pantothenate kinase 2 (PANK2) gene on chromosome 20. PKAN is usually rapidly progressive, presenting in the vast majority in the first decade of life (classic form). A rarer, later onset and slowly progressive (atypical) PKAN form also exists. We present two siblings of Cypriot descent, a 27-year-old man and his clinically asymptomatic younger sister, both of whom were found to be homozygous for a novel c.695A>G (p.Asp232Gly) missense mutation in exon 2 of the PANK2 gene. The index patient presented with a 5-year history of slowly progressive gait disturbance, dysarthria, mild axial rigidity and bradykinesia. His brain MRI scan revealed the characteristic “eye-of-the-tiger” sign. Atypical genetically confirmed PKAN cases are sparsely reported and should be considered in the differential diagnosis of patients presenting with a progressive extrapyramidal syndrome particularly if the radiographic findings are suggestive of iron accumulation. Effective treatment strategies for PKAN are not currently available and symptomatic therapy is often unsatisfactory. However, early diagnosis including the presymptomatic stage is important for genetic counseling and will be crucial for testing novel therapeutics in the future. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
132. Mitochondrial Dysfunction, Oxidative Stress and Neuroinflammation in Neurodegeneration with Brain Iron Accumulation (NBIA)
- Author
-
Candela Machuca, Isabel Hinarejos, Carmen Espinós, and Paula Sancho
- Subjects
0301 basic medicine ,autophagy ,brain iron accumulation ,Physiology ,Neurodegeneration with brain iron accumulation ,Clinical Biochemistry ,Choreoathetosis ,rare disease ,Review ,medicine.disease_cause ,Biochemistry ,neuroinflammation ,03 medical and health sciences ,0302 clinical medicine ,WDR45 ,lipid metabolism ,mitochondrial dysfunction ,Medicine ,oxidative stress ,iron metabolism ,Molecular Biology ,Neuroinflammation ,Dystonia ,business.industry ,Parkinsonism ,lcsh:RM1-950 ,Cell Biology ,medicine.disease ,PANK2 ,030104 developmental biology ,lcsh:Therapeutics. Pharmacology ,membrane remodelling ,medicine.symptom ,business ,neurodegenerative disorder ,Neuroscience ,030217 neurology & neurosurgery ,Oxidative stress - Abstract
The syndromes of neurodegeneration with brain iron accumulation (NBIA) encompass a group of invalidating and progressive rare diseases that share the abnormal accumulation of iron in the basal ganglia. The onset of NBIA disorders ranges from infancy to adulthood. Main clinical signs are related to extrapyramidal features (dystonia, parkinsonism and choreoathetosis), and neuropsychiatric abnormalities. Ten NBIA forms are widely accepted to be caused by mutations in the genes PANK2, PLA2G6, WDR45, C19ORF12, FA2H, ATP13A2, COASY, FTL1, CP, and DCAF17. Nonetheless, many patients remain without a conclusive genetic diagnosis, which shows that there must be additional as yet undiscovered NBIA genes. In line with this, isolated cases of known monogenic disorders, and also, new genetic diseases, which present with abnormal brain iron phenotypes compatible with NBIA, have been described. Several pathways are involved in NBIA syndromes: iron and lipid metabolism, mitochondrial dynamics, and autophagy. However, many neurodegenerative conditions share features such as mitochondrial dysfunction and oxidative stress, given the bioenergetics requirements of neurons. This review aims to describe the existing link between the classical ten NBIA forms by examining their connection with mitochondrial impairment as well as oxidative stress and neuroinflammation.
- Published
- 2020
133. Impaired proteasome activity and neurodegeneration with brain iron accumulation in FBXO7 defect
- Author
-
Correa-Vela, Marta, Lupo, Vincenzo, Montpeyó Garcia-Moreno, Marta, Sancho, Paula, Marcé-Grau, Anna, Hernández-Vara, Jorge, Darling, Alejandra, Jenkins, Alison, Fernández-Rodríguez, Sandra, Tello, Cristina, Ramírez-Jiménez, Laura, Pérez, Belén, Sánchez-Montáñez, Ángel, Macaya Ruiz, Alfons, Sobrido, María J., Martinez-Vicente, Marta, Pérez-Dueñas, Belén, Espinós, Carmen, Universitat Autònoma de Barcelona, Fundació La Marató de TV3, Instituto de Salud Carlos III, Generalitat Valenciana, Fundació per Amor a L'Art, and Ministerio de Educación, Cultura y Deporte (España)
- Subjects
0301 basic medicine ,Adult ,medicine.medical_specialty ,Levodopa ,Proteasome Endopeptidase Complex ,Neurodegeneration with brain iron accumulation ,Neuroaxonal Dystrophies ,Neurosciences. Biological psychiatry. Neuropsychiatry ,medicine.disease_cause ,Brief Communication ,03 medical and health sciences ,Epilepsy ,Consanguinity ,Young Adult ,0302 clinical medicine ,Parkinsonian Disorders ,Internal medicine ,Mitophagy ,medicine ,Cerebellar Degeneration ,Humans ,RC346-429 ,health care economics and organizations ,Spinocerebellar Degenerations ,Paraplegia ,Mutation ,business.industry ,General Neuroscience ,Parkinsonism ,F-Box Proteins ,Syndrome ,medicine.disease ,Phenotype ,Iron Metabolism Disorders ,nervous system diseases ,030104 developmental biology ,Endocrinology ,Female ,Neurology (clinical) ,Neurology. Diseases of the nervous system ,business ,030217 neurology & neurosurgery ,medicine.drug ,RC321-571 - Abstract
FBXO7 is implicated in the ubiquitin–proteasome system and parkin-mediated mitophagy. FBXO7defects cause a levodopa-responsive parkinsonian-pyramidal syndrome(PPS). Methods: We investigated the disease molecular bases in a child with PPS and brain iron accumulation. Results: A novel homozygous c.368C>G (p.S123*) FBXO7 mutation was identified in a child with spastic paraplegia, epilepsy, cerebellar degeneration, levodopa nonresponsive parkinsonism, and brain iron deposition. Patient’s fibroblasts assays demonstrated an absence of FBXO7 RNA expression leading to impaired proteasome degradation and accumulation of poly-ubiquitinated proteins. Conclusion: This novel FBXO7 phenotype associated with impaired proteasome activity overlaps with neurodegeneration with brain iron accumulation disorders., Fundacio La Marato de TV3 [Grants 20143130 to BPD, and 20143131 to CE], by the Instituto de Salud Carlos III (ISCIII) - Subdireccion General de Evaluacion y Fomento de la Investigacion within the framework of the National R + D+I Plan cofunded with ERDF funds [Grants PI18/01319 to BPD and PI18/00147 to CE], and by the Generalitat Valenciana [Grant PROMETEO/2018/ 135 to CE]. Part of the equipment employed in this work has been funded by Generalitat Valenciana and cofinanced with ERDF funds (OP ERDF of Comunitat Valenciana 2014-2020). SFR had a contract funded by the Spanish Foundation Per Amor a l’Art (FPAA). PS had a FPU-PhD fellowship funded by the Spanish Ministry of Education, Culture and Sport Inmunoterapia
- Published
- 2020
134. Iron deposition in Parkinsonisms: A Quantitative Susceptibility Mapping study in the deep grey matter
- Author
-
Fabio Triulzi, Giorgio Sacilotto, Silvia Siggillino, Gianni Pezzoli, Elisa Scola, Luca Melazzini, Nina Samoylova, Valeria Elisa Contarino, Sonia F. Calloni, Maria Paola Fedeli, and Giorgio Conte
- Subjects
Pathology ,medicine.medical_specialty ,Parkinson's disease ,Neurodegeneration with brain iron accumulation ,Iron ,030218 nuclear medicine & medical imaging ,Progressive supranuclear palsy ,03 medical and health sciences ,0302 clinical medicine ,Atrophy ,Parkinsonian Disorders ,Medicine ,Corticobasal degeneration ,Humans ,Radiology, Nuclear Medicine and imaging ,Gray Matter ,business.industry ,Putamen ,Parkinsonism ,Quantitative susceptibility mapping ,General Medicine ,Multiple System Atrophy ,medicine.disease ,Magnetic Resonance Imaging ,nervous system diseases ,nervous system ,030220 oncology & carcinogenesis ,Supranuclear Palsy, Progressive ,business - Abstract
Purpose The aim of the study is to quantify the susceptibility in deep grey nuclei that are affected by pathological processes related to iron accumulation in patients with Parkinson's disease and primary atypical parkinsonisms such as Progressive Supranuclear Palsy, Multiple System Atrophy and Cortico-Basal Degeneration, in order to assist the differential diagnosis among parkinsonian syndromes. Methods We enrolled 49 patients with Parkinson’s disease and 26 patients with primary atypical parkinsonisms. Automatic segmentation of putamen, globus pallidus, caudate nucleus and thalamus and manual segmentation of red nuclei and substantia nigra were performed, and region of interest-based Quantitative Susceptibility Mapping analysis were performed. Statistical comparisons of the mean susceptibility values in the segmented brain regions were performed among primary atypical parkinsonisms and Parkinson's disease. Results Susceptibility values in red nuclei were increased in Progressive Supranuclear Palsy patients compared to parkinsonian phenotype Multiple System Atrophy (p = 0.004), and Parkinson's disease patients (p = 0.006). Susceptibility in thalamus was decreased in Cortico-Basal Degeneration patients compared to Parkinson's disease (p = 0.006), Multiple System Atrophy with cerebellar phenotype (p = 0.031) and parkinsonian phenotype (p = 0.001) patients, and in Progressive Supranuclear Palsy patients compared to Multiple System Atrophy with parkinsonian phenotype patients (p = 0.012). Conclusions Quantitative Susceptibility Mapping allows the depiction and quantification of different patterns of iron deposition in the deep gray nuclei occurring in primary atypical parkinsonisms and Parkinson's disease and it may help as a non-invasive tool in the differential diagnosis between parkinsonian syndromes.
- Published
- 2020
135. Phenotypic and imaging spectrum associated with WDR45
- Author
-
Dorothée Ville, Amy Goldstein, Holly Dubbs, Adeline Vanderver, Anne-Lise Poulat, Tanner Hagelstrom, Amy Pizzino, Christian P. Schaaf, Denise L. Perry, Alka Malhotra, Arastoo Vossough, Charles Perrine, Gaetan Lesca, Laura Adang, Matthieu Milh, Ingo Helbig, Anna-Kaisa Anttonen, Catherine Williams, Omar Sherbini, Chloé Laurencin, Tarja Linnankivi, Ryan J. Taft, Medicum, HUSLAB, Department of Medical and Clinical Genetics, Neuroscience Center, University of Helsinki, Helsinki University Hospital Area, and HUS Children and Adolescents
- Subjects
Male ,Pediatrics ,Neurodegeneration with brain iron accumulation ,Developmental delay ,Developmental Disabilities ,CHILDHOOD ,3124 Neurology and psychiatry ,Cohort Studies ,Epilepsy ,0302 clinical medicine ,3123 Gynaecology and paediatrics ,Global developmental delay ,Child ,EXOME SEQUENCING REVEALS ,Dystonia ,Epileptic encephalopathy ,Parkinsonism ,WDR45 ,Middle Aged ,FEMALE ,Phenotype ,Neurology ,Child, Preschool ,Cohort ,Hypomyelination ,Adult ,medicine.medical_specialty ,Adolescent ,Neuroaxonal Dystrophies ,Article ,03 medical and health sciences ,Young Adult ,PROTEIN-ASSOCIATED NEURODEGENERATION ,Developmental Neuroscience ,030225 pediatrics ,Exome Sequencing ,medicine ,Dementia ,Humans ,business.industry ,MUTATIONS ,Infant ,medicine.disease ,Iron Metabolism Disorders ,Pediatrics, Perinatology and Child Health ,Neurology (clinical) ,3111 Biomedicine ,business ,Carrier Proteins ,030217 neurology & neurosurgery ,Demyelinating Diseases - Abstract
Background Mutations in the X-linked gene WDR45 cause neurodegeneration with brain iron accumulation type 5. Global developmental delay occurs at an early age with slow progression to dystonia, parkinsonism, and dementia due to progressive iron accumulation in the brain. Methods We present 17 new cases and reviewed 106 reported cases of neurodegeneration with brain iron accumulation type 5. Detailed information related to developmental history and key time to event measures was collected. Results Within this cohort, there were 19 males. Most individuals were molecularly diagnosed by whole-exome testing. Overall 10 novel variants were identified across 11 subjects. All individuals were affected by developmental delay, most prominently in verbal skills. Most individuals experienced a decline in motor and cognitive skills. Although most individuals were affected by seizures, the spectrum ranged from provoked seizures to intractable epilepsy. The imaging findings varied as well, often evolving over time. The classic iron accumulation in the globus pallidus and substantia nigra was noted in half of our cohort and was associated with older age of image acquisition, whereas myelination abnormalities were associated with younger age. Conclusions WDR45 mutations lead to a progressive and evolving disorder whose diagnosis is often delayed. Developmental delay and seizures predominate in early childhood, followed by a progressive decline of neurological function. There is variable expressivity in the clinical phenotypes of individuals with WDR45 mutations, suggesting that this gene should be considered in the diagnostic evaluation of children with myelination abnormalities, iron deposition, developmental delay, and epilepsy depending on the age at evaluation.
- Published
- 2020
136. WDR45 Contributes to Iron Accumulation Through Dysregulation of Neuronal Iron Homeostasis
- Author
-
Eun-kyeong Choi, Young Ah Seo, and Luisa Aring
- Subjects
chemistry.chemical_classification ,Reactive oxygen species ,Nutrition and Dietetics ,biology ,Neurodegeneration with brain iron accumulation ,Chemistry ,Catabolism ,Autophagy ,Medicine (miscellaneous) ,Mitochondrion ,Cell biology ,Ferritin ,WDR45 ,Transferrin ,Neuroscience, Nutrition and the Brain ,biology.protein ,Food Science - Abstract
OBJECTIVES: Neurodegeneration with brain iron accumulation (NBIA) is a clinically and genetically heterogeneous group of neurodegenerative diseases characterized by an abnormal accumulation of brain iron and progressive degeneration of the nervous system. β-propeller protein-associated neurodegeneration (BPAN) (OMIM #300,894) is a recently identified subtype of NBIA. BPAN is caused by de novo mutations in the WD repeat domain 45 (WDR45) gene. WDR45 deficiency in BPAN patients and animal models has shown defects in autophagic flux, suggesting a role for WDR45 in autophagy. How WDR45 deficiency leads to brain iron overload remains unclear. The goal of the present study is to identify the pathogenic mechanisms of WDR45 deficiency that cause iron overload and neurodegeneration. METHODS: To elucidate the role of WDR45 in dopaminergic neuronal cells, we generated a WDR45-knockout (KO) SH-SY5Y cell line by CRISPR/Cas9-mediated genome editing. To identify mechanisms underlying iron homeostasis and transport, we examined two cellular iron acquisition pathways in these cells using radioactive isotope (59)Fe: 1) the canonical transferrin-bound iron (TBI) uptake pathway and 2) the nontransferrin-bound iron (NTBI) pathway. RESULTS: Loss of WDR45 increased total iron levels with a concomitant increase in the iron storage protein ferritin in neuronal cells. Specifically, WDR45-KO cells preferentially took up NTBI compared to wild-type cells. Concordant with these functional data, the level of divalent metal transporter-1 (DMT1) expression was upregulated in WDR45-KO cells, providing a causal link to iron overload in WDR45 deficiency. In addition, loss of WDR45 led to defects in autophagic flux and impaired ferritinophagy, a lysosomal process that promotes ferritin degradation, suggesting that iron overload is driven by impaired ferritinophagy. Interestingly, WDR45 deficiency increased iron accumulation in the mitochondria, impaired mitochondrial function, and in turn, elevated reactive oxygen species generation. CONCLUSIONS: Our study provides the first evidence that WDR45 deficiency alters cellular iron acquisition pathways thereby leading to iron accumulation in neuronal cells. These findings will serve as a basis for developing therapeutic strategies for patients with NBIA. FUNDING SOURCES: NIH, NBIA Disorder Association.
- Published
- 2020
137. Brain MRI Pattern Recognition in Neurodegeneration With Brain Iron Accumulation
- Author
-
Ji Young Yun, Penelope Hogarth, Susan J. Hayflick, Allison Gregory, and Jae-Hyeok Lee
- Subjects
0301 basic medicine ,Neurodegeneration with brain iron accumulation ,Disease ,pattern ,lcsh:RC346-429 ,03 medical and health sciences ,0302 clinical medicine ,iron ,medicine ,Brain mri ,magnetic resonance imaging ,lcsh:Neurology. Diseases of the nervous system ,Genetic testing ,Natural course ,medicine.diagnostic_test ,NBIA ,business.industry ,Neurodegeneration ,neurodegeneration ,Magnetic resonance imaging ,medicine.disease ,030104 developmental biology ,Neurology ,Pattern recognition (psychology) ,Neurology (clinical) ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Most neurodegeneration with brain iron accumulation (NBIA) disorders can be distinguished by identifying characteristic changes on magnetic resonance imaging (MRI) in combination with clinical findings. However, a significant number of patients with an NBIA disorder confirmed by genetic testing have MRI features that are atypical for their specific disease. The appearance of specific MRI patterns depends on the stage of the disease and the patient's age at evaluation. MRI interpretation can be challenging because of heterogeneously acquired MRI datasets, individual interpreter bias, and lack of quantitative data. Therefore, optimal acquisition and interpretation of MRI data are needed to better define MRI phenotypes in NBIA disorders. The stepwise approach outlined here may help to identify NBIA disorders and delineate the natural course of MRI-identified changes.
- Published
- 2020
138. Neurodegeneration with Brain Iron Accumulation
- Author
-
Chandana Gaddipati and Amit Batla
- Subjects
Pathology ,medicine.medical_specialty ,Neurodegeneration with brain iron accumulation ,global developmental delay ,AIAN Review ,lcsh:RC346-429 ,03 medical and health sciences ,0302 clinical medicine ,Basal ganglia ,medicine ,030212 general & internal medicine ,Global developmental delay ,lcsh:Neurology. Diseases of the nervous system ,parkinsonism ,Genetic testing ,dystonia parkinsonism ,medicine.diagnostic_test ,NBIA ,business.industry ,Parkinsonism ,medicine.disease ,Phenotype ,Globus pallidus ,Neurology (clinical) ,Differential diagnosis ,business ,030217 neurology & neurosurgery - Abstract
The term NBIA encompasses a heterogeneous group of inherited disorders characterized clinically by progressive extra pyramidal syndrome and pathologically by excessive iron deposition in brain, primarily affecting the basal ganglia (globus pallidus mainly). The hallmark of this syndrome is the age specific phenotypic presentation and intraphenotypic heterogeneity. NBIAs at present include ten subtypes with genes identified in nine subtypes. They form an important differential diagnosis for the phenotype of global developmental delay in infancy/childhood to dystonia-parkinsonism or isolated parkinsonism at all ages and also for the isolated craniocervical dystonia of adult onset. There needs to be a high index of clinical suspicion for this syndrome and the evaluation includes MRI brain T2* weighted imaging which reveal symmetrical iron deposition in bilateral globus pallidi and other basal ganglia. The T2 * imaging pattern of iron deposition varies amongst the different subtypes and the combination of clinical phenotype and MRI signature makes it easier to confidently make a diagnosis of NBIA and to recommend genetic testing. The treatment to date is mostly symptomatic with targeted therapies on the horizon.
- Published
- 2019
139. Is there heart disease in cases of neurodegeneration associated with mutations in C19orf12?
- Author
-
Anna Członkowska, Tomasz Litwin, Małgorzata Buksińska-Lisik, Tomasz Kmieć, Iwona Kurkowska-Jastrzębska, and Marta Skowrońska
- Subjects
0301 basic medicine ,Tachycardia ,Adult ,Male ,medicine.medical_specialty ,Mitochondrial Diseases ,Heart disease ,Adolescent ,Heart Diseases ,Neurodegeneration with brain iron accumulation ,Cardiomyopathy ,Neuroaxonal Dystrophies ,Mitochondrial Proteins ,03 medical and health sciences ,Electrocardiography ,Young Adult ,0302 clinical medicine ,Internal medicine ,Heart rate ,medicine ,Dementia ,Heart rate variability ,Humans ,business.industry ,Neurodegenerative Diseases ,medicine.disease ,Iron Metabolism Disorders ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Ventricle ,Cardiology ,Female ,Neurology (clinical) ,Geriatrics and Gerontology ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Introduction In mitochondrial membrane protein-associated neurodegeneration (MPAN), a subtype of neurodegeneration with brain iron accumulation (NBIA), patients suffer from optic nerve atrophy and dementia, which are also typical for another group of diseases, the mitochondrial diseases (MD). Around 30% of patients with MD have heart disease, commonly cardiomyopathy and arrhythmias, and 10% experience a major adverse cardiovascular event. The aim of this study was to assess cardiac involvement in MPAN. Methods Thirteen patients with MPAN were evaluated after written informed consent. All patients had echocardiography and 12 patients had 24-h Holter electrocardiogram (ECG) monitoring using 3-channel digital recorders. Results Echocardiography revealed normal values for the dimensions of all heart chambers. The systolic function of the left ventricle was normal in all cases. Right ventricle systolic impairment was found in three patients. 24-hour Holter ECG revealed predominant resting tachycardia during daytime with no physiological slowing of heart rate during sleep in seven cases. No significant arrhythmias were found. In nine patients, selected heart rate variability (HRV) parameters were lower than reference values. Conclusion Cardiomyopathy, typical of MD, was not found in patients with MPAN. There were no significant arrhythmias, but disturbances in the circadian rhythm of the heart rate were observed in most cases. The decrease in HRV may reflect an early sign of autonomic dysfunction. A standard cardiac work-up is recommended for patients with MPAN to assess if additional treatment is needed.
- Published
- 2020
140. Oxidative Stress, a Crossroad Between Rare Diseases and Neurodegeneration
- Author
-
Federico V. Pallardó, Pascual Sanz, Teresa Sevilla, J.M. Millán, Marta Seco-Cervera, Carmen Espinós, Andrea Tapia, Regina Rodrigo, Máximo Ibo Galindo, Dolores Martínez-Rubio, José Santiago Ibáñez-Cabellos, and Maria Adelaida Garcia-Gimeno
- Subjects
0301 basic medicine ,Ataxia ,Unverricht–Lundborg disease (ULD) ,Physiology ,Neurodegeneration with brain iron accumulation ,Clinical Biochemistry ,Friedreich’s ataxia ,Review ,medicine.disease_cause ,Bioinformatics ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Lafora disease (LD) ,0302 clinical medicine ,Medicine ,progressive myoclonus epilepsy (PME) ,Molecular Biology ,Neuroinflammation ,Reactive nitrogen species ,neurodegenerative disorders with brain iron accumulation (NBIA) ,business.industry ,Neurodegeneration ,lcsh:RM1-950 ,Neurotoxicity ,Cell Biology ,medicine.disease ,Dravet syndrome ,Charcot-Marie-Tooth disease (CMT) ,030104 developmental biology ,lcsh:Therapeutics. Pharmacology ,chemistry ,medicine.symptom ,business ,Myoclonus ,inherited retinal dystrophy (IRD) ,030217 neurology & neurosurgery ,Oxidative stress - Abstract
Oxidative stress is an imbalance between production and accumulation of oxygen reactive species and/or reactive nitrogen species in cells and tissues, and the capacity of detoxifying these products, using enzymatic and non-enzymatic components, such as glutathione. Oxidative stress plays roles in several pathological processes in the nervous system, such as neurotoxicity, neuroinflammation, ischemic stroke, and neurodegeneration. The concepts of oxidative stress and rare diseases were formulated in the eighties, and since then, the link between them has not stopped growing. The present review aims to expand knowledge in the pathological processes associated with oxidative stress underlying some groups of rare diseases: Friedreich’s ataxia, diseases with neurodegeneration with brain iron accumulation, Charcot-Marie-Tooth as an example of rare neuromuscular disorders, inherited retinal dystrophies, progressive myoclonus epilepsies, and pediatric drug-resistant epilepsies. Despite the discrimination between cause and effect may not be easy on many occasions, all these conditions are Mendelian rare diseases that share oxidative stress as a common factor, and this may represent a potential target for therapies., Generalitat Valenciana [Grant PROMETEO/2018/135].
- Published
- 2020
141. Human patient SFPQ homozygous mutation is found deleterious for brain and motor development in a zebrafish model
- Author
-
Henry Houlden, Stephanie Efthymiou, Corinne Houart, Eugenia Borgione, Vincenzo Salpietro, Triona Fielding, Carmela Scuderi, and Patricia M. Gordon
- Subjects
Neurodegeneration with brain iron accumulation ,Parkinsonism ,Central nervous system ,Substantia nigra ,Biology ,medicine.disease ,biology.organism_classification ,medicine.anatomical_structure ,Globus pallidus ,Genetic model ,medicine ,Neuroscience ,Zebrafish ,Exome sequencing - Abstract
SFPQ (Splicing factor proline- and glutamine-rich) is a DNA and RNA binding protein involved in transcription, pre-mRNA splicing, and DNA damage repair and it has been previously implicated in neurodegenerative disorders. A homozygous p.Ser660Asn variant in SFPQ was identified through whole exome sequencing (WES) in an Italian woman presented a complex neurological phenotype with intellectual disability, peripheral neuropathy, bradykinesia, extrapyramidal rigidity and rest (heads) tremor and neuroradiological anomalies including thin dysplastic corpus callosum, hypomyelination and hypointensity of the globus pallidus and of the mesencephalic substantia nigra (resembling neurodegeneration with brain iron accumulation; NBIA). Using a zebrafish SFPQ genetic model we have showed that a rescue with this SFPQS660N mutant revealed robust defects in the developing central nervous system (CNS) of the embryos, including abnormal branching of the motor axons innervating body muscles and misfolding of the posterior brain neuroepithelium. The defects hereby identified in the model organism indicate a potential contribution of the homozygous SFPQ p.Ser660Asn variant in some of the patient's neurodegenerative features, including the clinical parkinsonism and the NBIA-like pattern on brain imaging.
- Published
- 2020
- Full Text
- View/download PDF
142. Mitochondrial membrane protein–associated neurodegeneration: A case series of six children
- Author
-
Ozlem M Herguner, Faruk Incecik, and Atil Bisgin
- Subjects
Mutation ,Neurodegeneration with brain iron accumulation ,business.industry ,Neurodegeneration ,Substantia nigra ,Case Reports ,medicine.disease_cause ,Bioinformatics ,medicine.disease ,mitochondrial membrane protein–associated neurodegeneration ,lcsh:RC346-429 ,Globus pallidus ,children ,Mitochondrial Membrane Protein ,medicine ,Neurology (clinical) ,Spasticity ,medicine.symptom ,Inner mitochondrial membrane ,business ,c19orf12 mutations ,lcsh:Neurology. Diseases of the nervous system - Abstract
Neurodegeneration with brain iron accumulation (NBIA) is a group of genetic disorders with a progressive extrapyramidal syndrome and excessive iron deposition in the brain, particularly in the globus pallidus and substantia nigra. Mitochondrial membrane protein–associated neurodegeneration (MPAN), a subtype of NBIA, is caused by mutation in the orphan gene C19orf12. A slowly progressive gait disorder from generalized dystonia and spasticity and cognitive impairment constitute the main features of MPAN. The C19orf12 p.Thr11Met mutation is frequent among Turkish patients with MPAN. Here, we report the clinical manifestations and genetic study results of six Turkish patients with MPAN due to different mutations from previous.
- Published
- 2020
143. Pantothenate Kinase-Associated Neurodegeneration Causing Paradoxical Vocal Fold Motion
- Author
-
Rafizadeh, Sassan and Long, Jennifer L
- Subjects
Allied Health and Rehabilitation Science ,Music ,Health Sciences ,Creative Arts and Writing ,Neurosciences ,Neurodegenerative ,Lung ,Clinical Research ,Aetiology ,2.1 Biological and endogenous factors ,Neurological ,Adult ,Female ,Humans ,Neurodegenerative Diseases ,Phosphotransferases (Alcohol Group Acceptor) ,Vocal Cord Dysfunction ,Paradoxical vocal fold motion ,Pantothenate kinase-associated neurodegeneration ,Neurodegeneration with brain iron accumulation ,Clinical Sciences ,Performing Arts and Creative Writing ,Speech-Language Pathology & Audiology ,Allied health and rehabilitation science - Abstract
We report a patient with paradoxical vocal fold motion (PVFM) due to pantothenate kinase-associated neurodegeneration, a rare neurodegenerative disease and a subclass of neurodegeneration with brain iron accumulation disorders. PVFM is marked by normal vocal fold anatomy and physiology with intermittent adduction during the respiratory cycle. Many etiologies have been reported and include laryngeal hypersensitivity such as asthma and gastroesophageal reflux, functional disorders, and neurologic disorders such as focal respiratory dystonia. This case highlights the occasional association of PVFM with underlying neurologic disorders, especially those that disrupt autonomic functioning.
- Published
- 2013
144. Iron overload is accompanied by mitochondrial and lysosomal dysfunction in WDR45 mutant cells
- Author
-
Christine Klein, Lena F. Burbulla, Franziska Rudolph, Philip Seibler, Dimitri Krainc, Ana Westenberger, Thomas G. P. M. Schmidt, Simone Zittel, Alexander Münchau, Johanne Heine, Marija Dulovic, and Aleksandar Rakovic
- Subjects
0301 basic medicine ,Iron Overload ,Neurodegeneration with brain iron accumulation ,Iron ,Induced Pluripotent Stem Cells ,Mutant ,Mitochondrion ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Autophagy ,medicine ,Humans ,Induced pluripotent stem cell ,Cells, Cultured ,Mutation ,Chemistry ,Dopaminergic Neurons ,Neurodegeneration ,Neurodegenerative Diseases ,Fibroblasts ,medicine.disease ,Corrigenda ,Mitochondria ,Cell biology ,030104 developmental biology ,Nerve Degeneration ,Female ,Neurology (clinical) ,Carrier Proteins ,Lysosomes ,030217 neurology & neurosurgery ,Oxidative stress - Abstract
Beta-propeller protein-associated neurodegeneration is a subtype of monogenic neurodegeneration with brain iron accumulation caused by de novo mutations in WDR45. The WDR45 protein functions as a beta-propeller scaffold and plays a putative role in autophagy through its interaction with phospholipids and autophagy-related proteins. Loss of WDR45 function due to disease-causing mutations has been linked to defects in autophagic flux in patient and animal cells. However, the role of WDR45 in iron homeostasis remains elusive. Here we studied patient-specific WDR45 mutant fibroblasts and induced pluripotent stem cell-derived midbrain neurons. Our data demonstrated that loss of WDR45 increased cellular iron levels and oxidative stress, accompanied by mitochondrial abnormalities, autophagic defects, and diminished lysosomal function. Restoring WDR45 levels partially rescued oxidative stress and the susceptibility to iron treatment, and activation of autophagy reduced the observed iron overload in WDR45 mutant cells. Our data suggest that iron-containing macromolecules and organelles cannot effectively be degraded through the lysosomal pathway due to loss of WDR45 function.
- Published
- 2018
- Full Text
- View/download PDF
145. Mutations in PPCS, Encoding Phosphopantothenoylcysteine Synthetase, Cause Autosomal-Recessive Dilated Cardiomyopathy
- Author
-
Arcangela Iuso, Nicola A. Grzeschik, Holger Prokisch, Iris Barshack, Muhamad Kumbar, Bart Kanon, Thomas Schwarzmayr, Gal Dubnov-Raz, Dorothea Haas, Riccardo Berutti, Bader Alhaddad, Marit Wiersma, Zeev Perles, Ben Pode-Shakked, Georg F. Hoffmann, Mathias Grigat, Tal Tirosh, Caterina Terrile, Elisa Mastantuono, Tim M. Strom, Jürgen G. Okun, Marina Rubinshtein, Matthias C. Braunisch, Yair Anikster, Shachar Abudi, Camilla Avivi, Ana C. Messias, Amir Vardi, Brundel Bianca Johanna Josephina Maria, Ody C. M. Sibon, Eran Eyal, Dina Marek-Yagel, Tobias B. Haack, Yishay Salem, Thomas Meitinger, A Volkov, Ortal Barel, Hans Joachim Schüller, Molecular Neuroscience and Ageing Research (MOLAR), Movement Disorder (MD), Physiology, and ACS - Heart failure & arrhythmias
- Subjects
Male ,0301 basic medicine ,Neurodegeneration with brain iron accumulation ,COENZYME-A SYNTHESIS ,chemistry.chemical_compound ,0302 clinical medicine ,Enzyme Stability ,PANTETHINE RESCUES ,Phosphopantothenoylcysteine synthetase ,Peptide Synthases ,Genetics (clinical) ,Exome sequencing ,2. Zero hunger ,chemistry.chemical_classification ,biology ,Coenzyme A ,Dilated Cardiomyopathy ,Pantethine Treatment ,Pentothenate ,Phospohopantothenoylcysteine Synthetase ,Ppcs ,Pantethine ,Homozygote ,Neurodegeneration ,NEURODEGENERATION ,High-Throughput Nucleotide Sequencing ,Heart ,Magnetic Resonance Imaging ,Pedigree ,Biochemistry ,ESCHERICHIA-COLI ,Child, Preschool ,Pantetheine ,Drosophila ,Female ,PROTEIN-STRUCTURE ,Cardiomyopathy, Dilated ,COA ,Genes, Recessive ,Saccharomyces cerevisiae ,Article ,Cofactor ,03 medical and health sciences ,BRAIN IRON ACCUMULATION ,Carnitine ,Genetics ,medicine ,Animals ,Humans ,Amino Acid Sequence ,MACROMOLECULES ,Demography ,Infant, Newborn ,Infant ,Reproducibility of Results ,Fibroblasts ,medicine.disease ,Biosynthetic Pathways ,SWISS-MODEL ,030104 developmental biology ,Enzyme ,chemistry ,Mutation ,biology.protein ,SYSTEM ,030217 neurology & neurosurgery - Abstract
Coenzyme A (CoA) is an essential metabolic cofactor used by around 4% of cellular enzymes. Its role is to carry and transfer acetyl and acyl groups to other molecules. Cells can synthesize CoA de novo from vitamin B5 (pantothenate) through five consecutive enzymatic steps. Phosphopantothenoylcysteine synthetase (PPCS) catalyzes the second step of the pathway during which phosphopantothenate reacts with ATP and cysteine to form phosphopantothenoylcysteine. Inborn errors of CoA biosynthesis have been implicated in neurodegeneration with brain iron accumulation (NBIA), a group of rare neurological disorders characterized by accumulation of iron in the basal ganglia and progressive neurodegeneration. Exome sequencing in five individuals from two unrelated families presenting with dilated cardiomyopathy revealed biallelic mutations in PPCS, linking CoA synthesis with a cardiac phenotype. Studies in yeast and fruit flies confirmed the pathogenicity of identified mutations. Biochemical analysis revealed a decrease in CoA levels in fibroblasts of all affected individuals. CoA biosynthesis can occur with pantethine as a source independent from PPCS, suggesting pantethine as targeted treatment for the affected individuals still alive.
- Published
- 2018
- Full Text
- View/download PDF
146. A Patient with Beta-Propeller Protein-Associated Neurodegeneration: Treatment with Iron Chelation Therapy
- Author
-
Mohamad Imran Idris, Ai Huey Tan, Ping Chong Bee, Norlisah Ramli, Azlina Ahmad-Annuar, Susanne A. Schneider, Jia Lun Lim, and Shen-Yang Lim
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Neurology ,stereotypies ,Neurodegeneration with brain iron accumulation ,Case Report ,Bioinformatics ,lcsh:RC346-429 ,lcsh:RC321-571 ,Beta-propeller ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,WDR45 ,medicine ,deferiprone ,Beta-propeller protein-associated neurodegeneration ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,lcsh:Neurology. Diseases of the nervous system ,neurodegeneration with brain iron accumulation ,business.industry ,Neurodegeneration ,Cognition ,Iron chelation therapy ,medicine.disease ,030104 developmental biology ,chemistry ,Neurology (clinical) ,Deferiprone ,business ,030217 neurology & neurosurgery - Abstract
We present a case of beta-propeller protein-associated neurodegeneration, a form of neurodegeneration with brain iron accumulation. The patient harbored a novel mutation in the WDR45 gene. A detailed video and description of her clinical condition are provided. Her movement disorder phenomenology was characterized primarily by limb stereotypies and gait dyspraxia. The patient's disability was advanced by the time iron-chelating therapy with deferiprone was initiated, and no clinical response in terms of cognitive function, behavior, speech, or movements were observed after one year of treatment.
- Published
- 2018
147. Classification and molecular pathogenesis of NBIA syndromes
- Author
-
Valeria Tiranti and Ivano Di Meo
- Subjects
0301 basic medicine ,Neurodegeneration with brain iron accumulation ,Neuroaxonal Dystrophies ,Biology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Gene ,Exome sequencing ,Dystonia ,Genetics ,Genetic heterogeneity ,Parkinsonism ,Autophagy ,Lipid metabolism ,Syndrome ,General Medicine ,medicine.disease ,Iron Metabolism Disorders ,030104 developmental biology ,Mutation ,Pediatrics, Perinatology and Child Health ,Neurology (clinical) ,030217 neurology & neurosurgery - Abstract
Brain iron accumulation is the hallmark of a group of seriously invalidating and progressive rare diseases collectively denominated Neurodegeneration with Brain Iron Accumulation (NBIA), characterized by movement disorder, painful dystonia, parkinsonism, mental disability and early death. Currently there is no established therapy available to slow down or reverse the progression of these conditions. Several genes have been identified as responsible for NBIA but only two encode for proteins playing a direct role in iron metabolism. The other genes encode for proteins either with various functions in lipid metabolism, lysosomal activity and autophagic processes or with still unknown roles. The different NBIA subtypes have been classified and denominated on the basis of the mutated genes and, despite genetic heterogeneity, some of them code for proteins, which share or converge on common metabolic pathways. In the last ten years, the implementation of genetic screening based on Whole Exome Sequencing has greatly accelerated gene discovery, nevertheless our knowledge of the pathogenic mechanisms underlying the NBIA syndromes is still largely incomplete.
- Published
- 2018
- Full Text
- View/download PDF
148. Challenges in the approach and reporting of atypical manifestations of membrane protein-associated neurodegeneration (MPAN): An editorial.
- Author
-
Salles, Philippe A.
- Subjects
- *
SYMPTOMS , *NEURODEGENERATION , *MITOCHONDRIAL membranes , *PROTEINS , *MEMBRANE proteins - Published
- 2021
- Full Text
- View/download PDF
149. Precision Medicine in Rare Diseases
- Author
-
Instituto de Salud Carlos III, European Commission, Ministerio de Educación, Cultura y Deporte (España), Villalón-García, Irene, Álvarez-Córdoba, Mónica, Suarez-Rivero, Juan M., Povea-Cabello, Suleva, Talaverón-Rey, Marta, Suárez-Carrillo, Alejandra, Munuera, Manuel, Sánchez-Alcázar, José Antonio, Instituto de Salud Carlos III, European Commission, Ministerio de Educación, Cultura y Deporte (España), Villalón-García, Irene, Álvarez-Córdoba, Mónica, Suarez-Rivero, Juan M., Povea-Cabello, Suleva, Talaverón-Rey, Marta, Suárez-Carrillo, Alejandra, Munuera, Manuel, and Sánchez-Alcázar, José Antonio
- Abstract
Rare diseases are those that have a low prevalence in the population (less than 5 individuals per 10,000 inhabitants). However, infrequent pathologies affect a large number of people, since according to the World Health Organization (WHO), there are about 7000 rare diseases that affect 7% of the world’s population. Many patients with rare diseases have suffered the consequences of what is called the diagnostic odyssey, that is, extensive and prolonged serial tests and clinical visits, sometimes for many years, all with the hope of identifying the etiology of their disease. For patients with rare diseases, obtaining the genetic diagnosis can mean the end of the diagnostic odyssey, and the beginning of another, the therapeutic odyssey. This scenario is especially challenging for the scientific community, since more than 90% of rare diseases do not currently have an effective treatment. This therapeutic failure in rare diseases means that new approaches are necessary. Our research group proposes that the use of precision or personalized medicine techniques can be an alternative to find potential therapies in these diseases. To this end, we propose that patients’ own cells can be used to carry out personalized pharmacological screening for the identification of potential treatments.
- Published
- 2020
150. Movement disorders: Indian scenario: A clinico-genetic review.
- Author
-
Das, Shyamal Kumar, Ghosh, Bhaskar, Das, Gautami, Biswas, Arindam, and Ray, Jharna
- Subjects
- *
MOVEMENT disorders , *NEUROLOGY , *DISEASE prevalence , *DIAGNOSIS , *THERAPEUTICS - Abstract
Movement disorder (MD) is an important branch of neurology and has great potentiality in management because of improved diagnosis and therapeutic strategies. Over the last three decades, emphasis has been laid on the evaluation of various MDs in India by a limited number of interested neurologists and basic scientists. In this review, we want to highlight common problems of MDs in India with regard to epidemiology, clinical features and genetics. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.