Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 198
1.
Int J Mol Sci ; 23(21)2022 Oct 26.
Article En | MEDLINE | ID: mdl-36361705

Coenzyme A (CoA) is an essential cofactor in all living organisms, being involved in a large number of chemical reactions. Sequence variations in pantothenate kinase 2 (PANK2), the first enzyme of CoA biosynthesis, are found in patients affected by Pantothenate Kinase Associated Neurodegeneration (PKAN), one of the most common forms of neurodegeneration, with brain iron accumulation. Knowledge about the biochemical and molecular features of this disorder has increased a lot in recent years. Nonetheless, the main culprit of the pathology is not well defined, and no treatment option is available yet. In order to contribute to the understanding of this disease and facilitate the search for therapies, we explored the potential of the zebrafish animal model and generated lines carrying biallelic mutations in the pank2 gene. The phenotypic characterization of pank2-mutant embryos revealed anomalies in the development of venous vascular structures and germ cells. Adult fish showed testicular atrophy and altered behavioral response in an anxiety test but no evident signs of neurodegeneration. The study suggests that selected cell and tissue types show a higher vulnerability to pank2 deficiency in zebrafish. Deciphering the biological basis of this phenomenon could provide relevant clues for better understanding and treating PKAN.


Pantothenate Kinase-Associated Neurodegeneration , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Pantothenate Kinase-Associated Neurodegeneration/genetics , Pantothenate Kinase-Associated Neurodegeneration/pathology , Mutation , Coenzyme A/genetics , Atrophy
2.
Cell Death Dis ; 13(2): 185, 2022 02 25.
Article En | MEDLINE | ID: mdl-35217637

Neurodegeneration associated with defective pantothenate kinase-2 (PKAN) is an early-onset monogenic autosomal-recessive disorder. The hallmark of the disease is the massive accumulation of iron in the globus pallidus brain region of patients. PKAN is caused by mutations in the PANK2 gene encoding the mitochondrial enzyme pantothenate kinase-2, whose function is to catalyze the first reaction of the CoA biosynthetic pathway. To date, the way in which this alteration leads to brain iron accumulation has not been elucidated. Starting from previously obtained hiPS clones, we set up a differentiation protocol able to generate inhibitory neurons. We obtained striatal-like medium spiny neurons composed of approximately 70-80% GABAergic neurons and 10-20% glial cells. Within this mixed population, we detected iron deposition in both PKAN cell types, however, the viability of PKAN GABAergic neurons was strongly affected. CoA treatment was able to reduce cell death and, notably, iron overload. Further differentiation of hiPS clones in a pure population of astrocytes showed particularly evident iron accumulation, with approximately 50% of cells positive for Perls staining. The analysis of these PKAN astrocytes indicated alterations in iron metabolism, mitochondrial morphology, respiratory activity, and oxidative status. Moreover, PKAN astrocytes showed signs of ferroptosis and were prone to developing a stellate phenotype, thus gaining neurotoxic features. This characteristic was confirmed in iPS-derived astrocyte and glutamatergic neuron cocultures, in which PKAN glutamatergic neurons were less viable in the presence of PKAN astrocytes. This newly generated astrocyte model is the first in vitro disease model recapitulating the human phenotype and can be exploited to deeply clarify the pathogenetic mechanisms underlying the disease.


Astrocytes , Pantothenate Kinase-Associated Neurodegeneration , Astrocytes/metabolism , Coenzyme A/genetics , Coenzyme A/metabolism , Humans , Iron/metabolism , Neurons/metabolism , Pantothenate Kinase-Associated Neurodegeneration/genetics , Pantothenate Kinase-Associated Neurodegeneration/metabolism , Pantothenate Kinase-Associated Neurodegeneration/pathology , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/metabolism
3.
J Transl Med ; 20(1): 103, 2022 02 23.
Article En | MEDLINE | ID: mdl-35197056

BACKGROUND: Pantothenate kinase (PANK) is the first and rate-controlling enzymatic step in the only pathway for cellular coenzyme A (CoA) biosynthesis. PANK-associated neurodegeneration (PKAN), formerly known as Hallervorden-Spatz disease, is a rare, life-threatening neurologic disorder that affects the CNS and arises from mutations in the human PANK2 gene. Pantazines, a class of small molecules containing the pantazine moiety, yield promising therapeutic effects in an animal model of brain CoA deficiency. A reliable technique to identify the neurometabolic effects of PANK dysfunction and to monitor therapeutic responses is needed. METHODS: We applied 1H magnetic resonance spectroscopy as a noninvasive technique to evaluate the therapeutic effects of the newly developed Pantazine BBP-671. RESULTS: 1H MRS reliably quantified changes in cerebral metabolites, including glutamate/glutamine, lactate, and N-acetyl aspartate in a neuronal Pank1 and Pank2 double-knockout (SynCre+ Pank1,2 dKO) mouse model of brain CoA deficiency. The neuronal SynCre+ Pank1,2 dKO mice had distinct decreases in Glx/tCr, NAA/tCr, and lactate/tCr ratios compared to the wildtype matched control mice that increased in response to BBP-671 treatment. CONCLUSIONS: BBP-671 treatment completely restored glutamate/glutamine levels in the brains of the mouse model, suggesting that these metabolites are promising clinically translatable biomarkers for future therapeutic trials.


Coenzyme A , Pantothenate Kinase-Associated Neurodegeneration , Animals , Brain/pathology , Coenzyme A/metabolism , Disease Models, Animal , Mice , Pantothenate Kinase-Associated Neurodegeneration/genetics , Pantothenate Kinase-Associated Neurodegeneration/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proton Magnetic Resonance Spectroscopy
4.
Biomolecules ; 12(2)2022 02 18.
Article En | MEDLINE | ID: mdl-35204826

Pantothenate kinase-associated neurodegeneration (PKAN) is a progressive neurodegenerative disease caused by mutations in the pantothenate kinase 2 (PANK2) gene and associated with iron deposition in basal ganglia. Pantothenate kinase isoforms catalyze the first step in coenzyme A (CoA) biosynthesis. Since PANK2 is the only isoform in erythrocytes, these cells are an excellent ex vivo model to study the effect of PANK2 point mutations on expression/stability and activity of the protein as well as on the downstream molecular consequences. PKAN erythrocytes containing the T528M PANK2 mutant had residual enzyme activities but variable PANK2 abundances indicating an impaired regulation of the protein. Patients with G521R/G521R, G521R/G262R, and R264N/L275fs PANK2 mutants had no residual enzyme activity and strongly reduced PANK2 abundance. G521R inactivates the catalytic activity of the enzyme, whereas G262R and the R264N point mutations impair the switch from the inactive to the active conformation of the PANK2 dimer. Metabolites in cytosolic extracts were analyzed by gas chromatography-mass spectrometry and multivariate analytic methods revealing changes in the carboxylate metabolism of erythrocytes from PKAN patients as compared to that of the carrier and healthy control. Assuming low/absent CoA levels in PKAN erythrocytes, changes are consistent with a model of altered citrate channeling where citrate is preferentially converted to α-ketoglutarate and α-hydroxyglutarate instead of being used for de novo acetyl-CoA generation. This finding hints at the importance of carboxylate metabolism in PKAN pathology with potential links to reduced cytoplasmic acetyl-CoA levels in neurons and to aberrant brain iron regulation.


Neurodegenerative Diseases , Pantothenate Kinase-Associated Neurodegeneration , Acetyl Coenzyme A , Citrates , Citric Acid , Erythrocytes/metabolism , Humans , Iron/metabolism , Mutation , Pantothenate Kinase-Associated Neurodegeneration/genetics , Pantothenate Kinase-Associated Neurodegeneration/pathology , Phosphotransferases (Alcohol Group Acceptor) , Protein Isoforms/genetics
5.
J Biol Chem ; 298(3): 101577, 2022 03.
Article En | MEDLINE | ID: mdl-35041826

Pantothenate kinase-associated neurodegeneration (PKAN) is an incurable rare genetic disorder of children and young adults caused by mutations in the PANK2 gene, which encodes an enzyme critical for the biosynthesis of coenzyme A. Although PKAN affects only a small number of patients, it shares several hallmarks of more common neurodegenerative diseases of older adults such as Alzheimer's disease and Parkinson's disease. Advances in etiological understanding and treatment of PKAN could therefore have implications for our understanding of more common diseases and may shed new lights on the physiological importance of coenzyme A, a cofactor critical for the operation of various cellular metabolic processes. The large body of knowledge that accumulated over the years around PKAN pathology, including but not limited to studies of various PKAN models and therapies, has contributed not only to progress in our understanding of the disease but also, importantly, to the crystallization of key questions that guide future investigations of the disease. In this review, we will summarize this knowledge and demonstrate how it forms the backdrop to new avenues of research.


Neurodegenerative Diseases , Pantothenate Kinase-Associated Neurodegeneration , Animals , Coenzyme A/genetics , Coenzyme A/metabolism , Humans , Mutation , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/therapy , Pantothenate Kinase-Associated Neurodegeneration/genetics , Pantothenate Kinase-Associated Neurodegeneration/metabolism , Pantothenate Kinase-Associated Neurodegeneration/pathology , Pantothenate Kinase-Associated Neurodegeneration/therapy , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism
6.
Int J Neurosci ; 132(6): 582-588, 2022 Jun.
Article En | MEDLINE | ID: mdl-33043782

AIM: Pantothenate kinase associated neurodegeneration (PKAN) is a severe autosomal recessive rare disease and characterized by iron accumulation in the basal ganglia. To investigate the pathogenesis of this disease in two sibling patients with PANK in a Chinese family, whole-exome variant detection and functional analysis were performed. MATERIALS AND METHODS: Clinical and radiographic investigations were performed in the two brother patients. Whole exome sequencing (WES) was used in mutation detection, and the mutations were confirmed by Sanger sequencing. A longevity cohort genetic database was applied as Chinese urban controls. Bioinformatic analysis was performed to predict the pathogenicity. RESULTS: Compound heterozygous mutations of PANK2 were detected in two sibling brothers with PKAN in a Chinese family: c.510_522del (p.A170fs) and c.1319G > C (p.R440P) in the transcript NM_153638. PANK2: c.510_522del (p.A170fs) was absent in public data and the Chinese urban controls. Bioinformatics analysis showed that the above two variants were pathogenicity. CONCLUSIONS: We identified a rare compound heterozygous combination of PANK2 mutations found in a Chinese family in which two sibling brothers suffered from PKAN. PANK2 c.510_522del (p.A170fs) was the first reported to be a PKAN pathogenic variant.


Pantothenate Kinase-Associated Neurodegeneration , Phosphotransferases (Alcohol Group Acceptor) , Asian People/genetics , China , Humans , Male , Mutation , Pantothenate Kinase-Associated Neurodegeneration/genetics , Pantothenate Kinase-Associated Neurodegeneration/pathology , Phosphotransferases (Alcohol Group Acceptor)/genetics
7.
J Med Chem ; 63(24): 15785-15801, 2020 12 24.
Article En | MEDLINE | ID: mdl-33320012

Mutations in the human PANK2 gene are implicated in neurodegenerative diseases such as pantothenate kinase-associated neurodegeneration (PKAN) and result in low levels of coenzyme-A (CoA) in the CNS due to impaired production of phosphopantothenic acid (PPA) from vitamin B5. Restoration of central PPA levels by delivery of exogenous PPA is a recent strategy to reactivate CoA biosynthesis in PKAN patients. Fosmetpantotenate is an oral PPA prodrug. We report here the development of a new PANk2-/- knockout model that allows CoA regeneration in brain cells to be evaluated and describe two new series of cyclic phosphate prodrugs of PPA capable of regenerating excellent levels of CoA in this system. A proof-of-concept study in mouse demonstrates the potential of this new class of prodrugs to deliver PPA to the brain following oral administration and confirms incorporation of the prodrug-derived PPA into CoA.


Pantothenic Acid/analogs & derivatives , Prodrugs/chemistry , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Coenzyme A/metabolism , Cyclization , Disease Models, Animal , Half-Life , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Lipid Droplets/chemistry , Lipid Droplets/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pantothenate Kinase-Associated Neurodegeneration/drug therapy , Pantothenate Kinase-Associated Neurodegeneration/pathology , Pantothenic Acid/chemistry , Pantothenic Acid/metabolism , Pantothenic Acid/therapeutic use , Prodrugs/metabolism , Prodrugs/therapeutic use , Structure-Activity Relationship
8.
Bull Exp Biol Med ; 170(1): 58-63, 2020 Nov.
Article En | MEDLINE | ID: mdl-33237527

Mutations in pank2 gene encoding pantothenate kinase 2 determine a pantothenate kinase-associated neurodegeneration, a rare disorder characterized by iron deposition in the globus pallidus. To extend our previous work, we performed microinjections of a new pank2-specific morpholino to zebrafish embryos and thoroughly analyzed vasculature development. Vessels development was severely perturbed in the head, trunk, and tail, where blood accumulation was remarkable and associated with dilation of the posterior cardinal vein. This phenotype was specific as confirmed by p53 expression analysis and injection of the same morpholino in pank2-mutant embryos. We can conclude that pank2 gene is involved in vasculature development in zebrafish embryos. The comprehension of the underlining mechanisms could be of relevance for understanding of pantothenate kinase-associated neurodegeneration.


Blood Vessels/metabolism , Coenzyme A/pharmacology , Globus Pallidus/metabolism , Pantothenate Kinase-Associated Neurodegeneration/prevention & control , Phosphotransferases (Alcohol Group Acceptor)/genetics , Animals , Blood Vessels/growth & development , Blood Vessels/pathology , Disease Models, Animal , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Globus Pallidus/blood supply , Globus Pallidus/drug effects , Globus Pallidus/pathology , Head/blood supply , Head/growth & development , Humans , Morpholinos/administration & dosage , Morpholinos/genetics , Morpholinos/metabolism , Pantothenate Kinase-Associated Neurodegeneration/genetics , Pantothenate Kinase-Associated Neurodegeneration/metabolism , Pantothenate Kinase-Associated Neurodegeneration/pathology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Tail/blood supply , Tail/growth & development , Tail/metabolism , Torso/blood supply , Torso/growth & development , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Zebrafish
9.
Ann Clin Transl Neurol ; 7(8): 1340-1351, 2020 08.
Article En | MEDLINE | ID: mdl-32705819

OBJECTIVE: Pantothenate kinase 2-associated neurodegeneration (PKAN) is a rare neurodegenerative disease caused by mutations in the pantothenate kinase 2 (PANK2) gene. PKAN is associated with iron deposition in the basal ganglia and, occasionally, with the occurrence of misshaped erythrocytes (acanthocytes). The aim of this study was to assess residual activity of PANK2 in erythrocytes of PKAN patients and to correlate these data with the type of PANK2 mutations and the progression of neurodegeneration. METHODS: Residual PANK2 activities in erythrocytes of 14 PKAN patients and 14 related carriers were assessed by a radiometric assay. Clinical data on neurodegeneration included the Barry-Albright Dystonia Scale (BAD-Scale) besides further general patient features. A molecular visualization and analysis program was used to rationalize the influence of the PKAN causing mutations on a molecular level. RESULTS: Erythrocytes of PKAN patients had markedly reduced or no PANK2 activity. However, patients with at least one allele of the c.1583C > T (T528M) or the c.833G > T (R278L) variant exhibited 12-56% of residual PANK2 activity. In line, molecular modeling indicated only minor effects on enzyme structure for these point mutations. On average, these patients with c.1583C > T or c.833G > T variant had lower BAD scores corresponding to lower symptom severity than patients with other PANK2 point mutations. INTERPRETATION: Residual erythrocyte PANK2 activity could be a predictor for the progression of neurodegeneration in PKAN patients. Erythrocytes are an interesting patient-derived cell system with still underestimated diagnostic potential.


Disease Progression , Erythrocytes/metabolism , Pantothenate Kinase-Associated Neurodegeneration/blood , Pantothenate Kinase-Associated Neurodegeneration/diagnosis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Registries , Adolescent , Adult , Biological Specimen Banks , Female , Humans , Male , Pantothenate Kinase-Associated Neurodegeneration/genetics , Pantothenate Kinase-Associated Neurodegeneration/pathology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Prognosis , Young Adult
10.
Int J Mol Sci ; 21(10)2020 May 22.
Article En | MEDLINE | ID: mdl-32456086

Pantothenate Kinase-associated Neurodegeneration (PKAN) belongs to a wide spectrum of diseases characterized by brain iron accumulation and extrapyramidal motor signs. PKAN is caused by mutations in PANK2, encoding the mitochondrial pantothenate kinase 2, which is the first enzyme of the biosynthesis of Coenzyme A. We established and characterized glutamatergic neurons starting from previously developed PKAN Induced Pluripotent Stem Cells (iPSCs). Results obtained by inductively coupled plasma mass spectrometry indicated a higher amount of total cellular iron in PKAN glutamatergic neurons with respect to controls. PKAN glutamatergic neurons, analyzed by electron microscopy, exhibited electron dense aggregates in mitochondria that were identified as granules containing calcium phosphate. Calcium homeostasis resulted compromised in neurons, as verified by monitoring the activity of calcium-dependent enzyme calpain1, calcium imaging and voltage dependent calcium currents. Notably, the presence of calcification in the internal globus pallidus was confirmed in seven out of 15 genetically defined PKAN patients for whom brain CT scan was available. Moreover, we observed a higher prevalence of brain calcification in females. Our data prove that high amount of iron coexists with an impairment of cytosolic calcium in PKAN glutamatergic neurons, indicating both, iron and calcium dys-homeostasis, as actors in pathogenesis of the disease.


Calcium/metabolism , Iron/metabolism , Mitochondria/metabolism , Neurons/metabolism , Pantothenate Kinase-Associated Neurodegeneration/metabolism , Adolescent , Brain/diagnostic imaging , Brain/pathology , Calcium/adverse effects , Calpain/metabolism , Child , Child, Preschool , Cohort Studies , Cytoplasm/physiology , Female , Homeostasis , Humans , Induced Pluripotent Stem Cells , Infant , Iron/adverse effects , Magnetic Resonance Imaging , Male , Mass Spectrometry , Microscopy, Electron , Mitochondria/enzymology , Mitochondria/ultrastructure , Neurons/physiology , Neurons/ultrastructure , Pantothenate Kinase-Associated Neurodegeneration/pathology , Phosphotransferases (Alcohol Group Acceptor) , Tomography, X-Ray Computed , Young Adult
11.
Neurocase ; 26(3): 175-182, 2020 06.
Article En | MEDLINE | ID: mdl-32310012

Panthothenate kinase-associated neurodegeneration (PKAN) is arare neurodegeneration caused by mutations in the pantothenate kinase (PANK2) gene, which is located on chromosome 20p13. These mutations result in iron accumulation in the brain basal ganglia leading to parkinsonism, dysarthria, spasticity, cognitive impairment, and retinopathy. Herein, we report acase of adult-onset PKAN who presented with young-onset action tremor, bradykinesia, dysarthria, and bilateral interossei atrophy.  Neuroimaging demonstrated "eye-of-the-tiger signs". Through analyzing PANK2 gene, PANK2 NM_153638:c.1133A>G (p.Asp378 Gly) and PANK2 NM_153638:c.1502 T > A (p.lle501Asn), were detected. In addition, we reviewed the clinical and genetic features and therapeutic strategies for patients with PKAN.


Pantothenate Kinase-Associated Neurodegeneration , Phosphotransferases (Alcohol Group Acceptor)/genetics , Adult , Humans , Male , Pantothenate Kinase-Associated Neurodegeneration/genetics , Pantothenate Kinase-Associated Neurodegeneration/pathology , Pantothenate Kinase-Associated Neurodegeneration/physiopathology , Young Adult
12.
Biochim Biophys Acta Mol Basis Dis ; 1866(5): 165663, 2020 05 01.
Article En | MEDLINE | ID: mdl-31918006

Pantothenate kinase (PanK) is the first enzyme in the coenzyme A (CoA) biosynthetic pathway. The differential expression of the four-active mammalian PanK isoforms regulates CoA levels in different tissues and PANK2 mutations lead to Pantothenate Kinase Associated Neurodegeneration (PKAN). The molecular mechanisms that potentially underlie PKAN pathophysiology are investigated in a mouse model of CoA deficiency in the central nervous system (CNS). Both PanK1 and PanK2 contribute to brain CoA levels in mice and so a mouse model with a systemic deletion of Pank1 together with neuronal deletion of Pank2 was generated. Neuronal Pank2 expression in double knockout mice decreased starting at P9-11 triggering a significant brain CoA deficiency. The depressed brain CoA in the mice correlates with abnormal forelimb flexing and weakness that, in turn, contributes to reduced locomotion and abnormal gait. Biochemical analysis reveals a reduction in short-chain acyl-CoAs, including acetyl-CoA and succinyl-CoA. Comparative gene expression analysis reveals that the CoA deficiency in brain is associated with a large elevation of Hif3a transcript expression and significant reduction of gene transcripts in heme and hemoglobin synthesis. Reduction of brain heme levels is associated with the CoA deficiency. The data suggest a response to oxygen/glucose deprivation and indicate a disruption of oxidative metabolism arising from a CoA deficiency in the CNS.


Brain Chemistry/genetics , Brain/pathology , Coenzyme A/deficiency , Pantothenate Kinase-Associated Neurodegeneration/pathology , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Animals , Apoptosis Regulatory Proteins/metabolism , Brain/cytology , Coenzyme A/analysis , Coenzyme A/biosynthesis , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation/genetics , Heme/analysis , Heme/metabolism , Hemoglobins/analysis , Hemoglobins/metabolism , Humans , Male , Mice , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Oxidation-Reduction , Pantothenate Kinase-Associated Neurodegeneration/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Repressor Proteins/metabolism
13.
Pediatr Neurol ; 103: 76-78, 2020 02.
Article En | MEDLINE | ID: mdl-31371123

BACKGROUND: Tongue protrusion dystonia is an uncommon focal dystonia involving the lingual muscles. Causes of tongue protrusion dystonia include tardive dystonia, posthypoxic dystonia, neuroacanthocytosis, pantothenate kinase-associated neurodegeneration, and Lesch-Nyhan syndrome. METHOD: We summarize three children with pantothenate kinase-associated neurodegeneration and tongue protrusion dystonia. All three patients underwent careful neurological examination, brain magnetic resonance imaging, and genetic testing. RESULTS: Tongue protrusion dystonia was a prominent and disabling symptom in all three patients. Brain magnetic resonance imaging revealed a typical eye of the tiger sign in all patients. Two patients had the same genetic mutation (c.1168 A>T mutation, p.I390F). CONCLUSIONS: Tongue protrusion dystonia may be a clue to the underlying etiology of dystonia, including hereditary forms of dystonia. Among them, pantothenate kinase-associated neurodegeneration is an important cause, especially in children.


Dystonic Disorders , Pantothenate Kinase-Associated Neurodegeneration , Tongue/physiopathology , Adolescent , Child , Dystonic Disorders/diagnosis , Dystonic Disorders/etiology , Dystonic Disorders/genetics , Dystonic Disorders/pathology , Female , Humans , Magnetic Resonance Imaging , Male , Pantothenate Kinase-Associated Neurodegeneration/complications , Pantothenate Kinase-Associated Neurodegeneration/diagnosis , Pantothenate Kinase-Associated Neurodegeneration/genetics , Pantothenate Kinase-Associated Neurodegeneration/pathology
14.
Mol Neurobiol ; 56(5): 3638-3656, 2019 May.
Article En | MEDLINE | ID: mdl-30173408

Neurodegeneration with brain iron accumulation (NBIA) is a group of inherited neurologic disorders in which iron accumulates in the basal ganglia resulting in progressive dystonia, spasticity, parkinsonism, neuropsychiatric abnormalities, and optic atrophy or retinal degeneration. The most prevalent form of NBIA is pantothenate kinase-associated neurodegeneration (PKAN) associated with mutations in the gene of pantothenate kinase 2 (PANK2), which is essential for coenzyme A (CoA) synthesis. There is no cure for NBIA nor is there a standard course of treatment. In the current work, we describe that fibroblasts derived from patients harbouring PANK2 mutations can reproduce many of the cellular pathological alterations found in the disease, such as intracellular iron and lipofuscin accumulation, increased oxidative stress, and mitochondrial dysfunction. Furthermore, mutant fibroblasts showed a characteristic senescent morphology. Treatment with pantothenate, the PANK2 enzyme substrate, was able to correct all pathological alterations in responder mutant fibroblasts with residual PANK2 enzyme expression. However, pantothenate had no effect on mutant fibroblasts with truncated/incomplete protein expression. The positive effect of pantothenate in particular mutations was also confirmed in induced neurons obtained by direct reprograming of mutant fibroblasts. Our results suggest that pantothenate treatment can stabilize the expression levels of PANK2 in selected mutations. These results encourage us to propose our screening model as a quick and easy way to detect pantothenate-responder patients with PANK2 mutations. The existence of residual enzyme expression in some affected individuals raises the possibility of treatment using high dose of pantothenate.


Iron/metabolism , Mutation/genetics , Pantothenate Kinase-Associated Neurodegeneration/drug therapy , Pantothenate Kinase-Associated Neurodegeneration/genetics , Pantothenic Acid/therapeutic use , Cell Death/drug effects , Cell Shape/drug effects , Coenzyme A/metabolism , Energy Metabolism/drug effects , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , Fibroblasts/ultrastructure , Gene Expression Regulation, Enzymologic/drug effects , Humans , Lipid Peroxidation/drug effects , Lipofuscin/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Pantothenate Kinase-Associated Neurodegeneration/pathology , Pantothenic Acid/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Carbonylation/drug effects
15.
J Clin Neurosci ; 59: 20-28, 2019 Jan.
Article En | MEDLINE | ID: mdl-30392836

Pantothenate kinase-associated neurodegeneration (PKAN) is extremely rare. In this study, we aimed to evaluate the magnetic resonance imaging (MRI) findings of PKAN patients. Conventional MRI and susceptibility weighted imaging (SWI) sequences and quantitative susceptibility mapping (QSM) maps of six patients from three PKAN families and eight healthy male volunteers were retrospectively analyzed. Iron content was represented by QSM values. The typical eye-of-the-tiger sign (n = 4) and hyperintensity (n = 2) of the bilateral globus pallidus (GP) were observed on T2WI sequences. The SWI signal was low (n = 5), and the QSM values were obviously higher (n = 2), which manifested as a reversed eye-of-the-tiger sign (n = 4) and hyperintensity (n = 2) on the QSM map. The QSM values were higher in the bilateral central GP, bilateral peripheral GP, and bilateral substantia nigra (SN) and lower in the left red nucleus (RN) compared with the healthy controls. No significant differences were observed in the right RN, bilateral thalamus and bilateral occipital regions. Low signals on SWI sequences and high QSM values with a reversed eye-of-the-tiger sign on QSM maps are important for the diagnosis of PKAN, especially in patients who do not show the eye-of-the-tiger sign in early stages. The eye-of-the-tiger sign observed on T2WI is helpful in diagnosing PKAN when the disease has already progressed to an advanced stage.


Magnetic Resonance Imaging/methods , Pantothenate Kinase-Associated Neurodegeneration/diagnostic imaging , Adolescent , Adult , Brain Mapping/methods , Child , Humans , Male , Pantothenate Kinase-Associated Neurodegeneration/pathology
16.
Nat Commun ; 9(1): 4399, 2018 10 23.
Article En | MEDLINE | ID: mdl-30352999

Pantothenate kinase (PANK) is a metabolic enzyme that regulates cellular coenzyme A (CoA) levels. There are three human PANK genes, and inactivating mutations in PANK2 lead to pantothenate kinase associated neurodegeneration (PKAN). Here we performed a library screen followed by chemical optimization to produce PZ-2891, an allosteric PANK activator that crosses the blood brain barrier. PZ-2891 occupies the pantothenate pocket and engages the dimer interface to form a PANK•ATP•Mg2+•PZ-2891 complex. The binding of PZ-2891 to one protomer locks the opposite protomer in a catalytically active conformation that is refractory to acetyl-CoA inhibition. Oral administration of PZ-2891 increases CoA levels in mouse liver and brain. A knockout mouse model of brain CoA deficiency exhibited weight loss, severe locomotor impairment and early death. Knockout mice on PZ-2891 therapy gain weight, and have improved locomotor activity and life span establishing pantazines as novel therapeutics for the treatment of PKAN.


Pantothenate Kinase-Associated Neurodegeneration/therapy , Phosphotransferases (Alcohol Group Acceptor)/genetics , Adenosine Triphosphate/metabolism , Allosteric Regulation , Animals , Cells, Cultured , Coenzyme A/deficiency , Coenzyme A/metabolism , Disease Models, Animal , Enzyme Stability , Female , Ligands , Magnesium/metabolism , Male , Mice, Knockout , Neurons/metabolism , Organ Specificity , Pantothenate Kinase-Associated Neurodegeneration/pathology , Protein Conformation , Protein Multimerization
17.
Mol Med Rep ; 18(5): 4739-4746, 2018 Nov.
Article En | MEDLINE | ID: mdl-30221726

Coenzyme A (CoA) is an essential cofactor of cellular metabolism that is involved in ~4% of cellular reactions. Its de novo production relies on five subsequent enzymatic steps, starting with the phosphorylation of vitamin B5. Pantothenate kinase 2 (PANK2) and coenzyme A synthase (COASY) catalyze the first and last steps of this pathway. Mutations in these genes lead to severe and progressive movement disorders, with neurodegeneration and iron accumulation in the basal ganglia, known as PANK2­ and COASY protein­associated neurodegeneration, respectively. Given the ubiquitous role of CoA in cellular metabolism, it is still not clear why patients carrying PANK2 and COASY mutations develop almost exclusively neurological symptoms. Important clues are the energetic profile of neural cells as well as the high levels of PANK2 expression in the brain; however, other features may contribute to this selective tissue vulnerability. Notably, when pank2 or coasy expression was suppressed in zebrafish evident perturbation of neuronal development was observed, as well as severe defects in vasculature formation. Supplementation of CoA to fish water prevented the appearance of the phenotype, thereby confirming the specific connection with the availability of the metabolic cofactor. The present study investigated the associations between PANK2 defects and angiogenesis in a mammalian setting, and revealed that PANK2 expression was required for normal angiogenetic properties of human umbilical vein endothelial cells.


Morphogenesis/genetics , Neovascularization, Physiologic/genetics , Pantothenate Kinase-Associated Neurodegeneration/genetics , Transferases/genetics , Zebrafish Proteins/genetics , Animals , Brain/growth & development , Brain/metabolism , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Iron/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mutation , Pantothenate Kinase-Associated Neurodegeneration/pathology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Zebrafish/genetics , Zebrafish/growth & development
18.
Int J Neurosci ; 128(12): 1109-1113, 2018 Dec.
Article En | MEDLINE | ID: mdl-29962256

AIM: Pantothenate-kinase-associated neurodegeneration (PKAN), which is characterised by iron accumulation in the basal ganglia, is a rare autosomal recessive neurodegenerative disorder caused by pantothenate kinase 2 (PANK2) gene mutations. The PANK2 gene is located on chromosome 20p13 and encodes pantothenate kinase. Herein, we identified one patient with PKAN who had mutations in the PANK2 gene. MATERIALS AND METHODS: We performed clinical and radiographic investigations, and diagnosed this disease at the clinical and genetic levels. RESULTS: It is worth mentioning that the patient displayed an eye-of-the-tiger sign. Through scanning the exons and flanking intronic sequences of PANK2 in patient and control subjects, we report a compound heterozygote c. 260A > G (NM_001324191) and c.405dupC (NM_153638) for PANK2 mutations in a Chinese patient with clinical manifestation of progressive prosopospasm, dysarthria and gait disturbance. Bioinformatics analysis showed that two variants exhibited highly conserved residues across species. CONCLUSION: we reported a patient presenting with atypical PKAN, and identified novel compound heterozygous PANK2 gene mutations..


Pantothenate Kinase-Associated Neurodegeneration/genetics , Pantothenate Kinase-Associated Neurodegeneration/pathology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Adult , Asian People/genetics , China , Female , Heterozygote , Humans , Male , Mutation , Pedigree
20.
J Assist Reprod Genet ; 34(1): 109-116, 2017 Jan.
Article En | MEDLINE | ID: mdl-27815806

PURPOSE: We aim to present a case of a healthy infant born after intracytoplasmic sperm injection-in vitro fertilization (ICSI-IVF) with a preimplantation genetic diagnosis (PGD) for pantothenate kinase-associated neurodegeneration (PKAN) due to PANK2 mutation. METHODS: ICSI-IVF was performed on a Thai couple, 34-year-old female and 33-year-old male, with a family history of PKAN in their first child. Following fertilization, each of the embryos were biopsied in the cleavage stage and subsequently processed for whole-genome amplification. Genetic status of the embryos was diagnosed by linkage analysis and direct mutation testing using primer extension-based mini-sequencing. Comprehensive chromosomal aneuploidy screening was performed using a next-generation sequencing-based strategy. RESULTS: Only a single cycle of ICSI-IVF was processed. There were seven embryos from this couple-two were likely affected, three were likely carriers, one was likely unaffected, and one failed in target genome amplification. Aneuploidy screening was performed before making a decision on embryo transfer, and only one unaffected embryo passed the screening. That embryo was transferred in a frozen thawed cycle, and the pregnancy was successful. The diagnosis was confirmed by amniocentesis, which presented with a result consistent with PGD. At 38 weeks of gestational age, a healthy male baby was born. Postnatal genetic confirmation was also consistent with PGD and the prenatal results. At the age of 24 months, the baby presented with normal growth and development lacking any neurological symptoms. CONCLUSIONS: We report the first successful trial of PGD for PKAN in a developing country using linkage analysis and mini-sequencing in cleavage stage embryos.


Embryo Transfer/methods , Fertilization in Vitro/methods , Pantothenate Kinase-Associated Neurodegeneration/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Preimplantation Diagnosis , Adult , Aneuploidy , Female , Humans , Infant, Newborn , Male , Mutation , Pantothenate Kinase-Associated Neurodegeneration/diagnosis , Pantothenate Kinase-Associated Neurodegeneration/pathology , Pregnancy , Sperm Injections, Intracytoplasmic
...