Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Brain ; 147(4): 1197-1205, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38141063

ABSTRACT

Dysfunctional RNA processing caused by genetic defects in RNA processing enzymes has a profound impact on the nervous system, resulting in neurodevelopmental conditions. We characterized a recessive neurological disorder in 18 children and young adults from 10 independent families typified by intellectual disability, motor developmental delay and gait disturbance. In some patients peripheral neuropathy, corpus callosum abnormalities and progressive basal ganglia deposits were present. The disorder is associated with rare variants in NUDT2, a mRNA decapping and Ap4A hydrolysing enzyme, including novel missense and in-frame deletion variants. We show that these NUDT2 variants lead to a marked loss of enzymatic activity, strongly implicating loss of NUDT2 function as the cause of the disorder. NUDT2-deficient patient fibroblasts exhibit a markedly altered transcriptome, accompanied by changes in mRNA half-life and stability. Amongst the most up-regulated mRNAs in NUDT2-deficient cells, we identified host response and interferon-responsive genes. Importantly, add-back experiments using an Ap4A hydrolase defective in mRNA decapping highlighted loss of NUDT2 decapping as the activity implicated in altered mRNA homeostasis. Our results confirm that reduction or loss of NUDT2 hydrolase activity is associated with a neurological disease, highlighting the importance of a physiologically balanced mRNA processing machinery for neuronal development and homeostasis.


Subject(s)
Intellectual Disability , Neurodevelopmental Disorders , Child , Young Adult , Humans , RNA, Messenger/genetics , Phosphoric Monoester Hydrolases/genetics , Neurodevelopmental Disorders/genetics , Intellectual Disability/genetics , Nudix Hydrolases
2.
J Clin Invest ; 131(9)2021 05 03.
Article in English | MEDLINE | ID: mdl-33755596

ABSTRACT

GDP-mannose-pyrophosphorylase-B (GMPPB) facilitates the generation of GDP-mannose, a sugar donor required for glycosylation. GMPPB defects cause muscle disease due to hypoglycosylation of α-dystroglycan (α-DG). Alpha-DG is part of a protein complex, which links the extracellular matrix with the cytoskeleton, thus stabilizing myofibers. Mutations of the catalytically inactive homolog GMPPA cause alacrima, achalasia, and mental retardation syndrome (AAMR syndrome), which also involves muscle weakness. Here, we showed that Gmppa-KO mice recapitulated cognitive and motor deficits. As structural correlates, we found cortical layering defects, progressive neuron loss, and myopathic alterations. Increased GDP-mannose levels in skeletal muscle and in vitro assays identified GMPPA as an allosteric feedback inhibitor of GMPPB. Thus, its disruption enhanced mannose incorporation into glycoproteins, including α-DG in mice and humans. This increased α-DG turnover and thereby lowered α-DG abundance. In mice, dietary mannose restriction beginning after weaning corrected α-DG hyperglycosylation and abundance, normalized skeletal muscle morphology, and prevented neuron degeneration and the development of motor deficits. Cortical layering and cognitive performance, however, were not improved. We thus identified GMPPA defects as the first congenital disorder of glycosylation characterized by α-DG hyperglycosylation, to our knowledge, and we have unraveled underlying disease mechanisms and identified potential dietary treatment options.


Subject(s)
Dystroglycans , Guanosine Diphosphate Mannose , Muscle, Skeletal/metabolism , Neuromuscular Diseases , Nucleotidyltransferases/deficiency , Animals , Dystroglycans/genetics , Dystroglycans/metabolism , Glycosylation , Guanosine Diphosphate Mannose/genetics , Guanosine Diphosphate Mannose/metabolism , Humans , Mice , Mice, Knockout , Neuromuscular Diseases/diet therapy , Neuromuscular Diseases/genetics , Neuromuscular Diseases/metabolism , Nucleotidyltransferases/metabolism
3.
Am J Hum Genet ; 107(2): 364-373, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32707086

ABSTRACT

We report bi-allelic pathogenic HPDL variants as a cause of a progressive, pediatric-onset spastic movement disorder with variable clinical presentation. The single-exon gene HPDL encodes a protein of unknown function with sequence similarity to 4-hydroxyphenylpyruvate dioxygenase. Exome sequencing studies in 13 families revealed bi-allelic HPDL variants in each of the 17 individuals affected with this clinically heterogeneous autosomal-recessive neurological disorder. HPDL levels were significantly reduced in fibroblast cell lines derived from more severely affected individuals, indicating the identified HPDL variants resulted in the loss of HPDL protein. Clinical presentation ranged from severe, neonatal-onset neurodevelopmental delay with neuroimaging findings resembling mitochondrial encephalopathy to milder manifestation of adolescent-onset, isolated hereditary spastic paraplegia. All affected individuals developed spasticity predominantly of the lower limbs over the course of the disease. We demonstrated through bioinformatic and cellular studies that HPDL has a mitochondrial localization signal and consequently localizes to mitochondria suggesting a putative role in mitochondrial metabolism. Taken together, these genetic, bioinformatic, and functional studies demonstrate HPDL is a mitochondrial protein, the loss of which causes a clinically variable form of pediatric-onset spastic movement disorder.


Subject(s)
Brain Diseases/genetics , Mitochondrial Proteins/genetics , Neurodegenerative Diseases/genetics , Spastic Paraplegia, Hereditary/genetics , Adolescent , Adult , Alleles , Amino Acid Sequence , Child , Female , Humans , Male , Mitochondria/genetics , Pedigree , Phenotype , Young Adult
4.
Cereb Cortex ; 29(10): 4263-4276, 2019 09 13.
Article in English | MEDLINE | ID: mdl-30541023

ABSTRACT

Brain functions are extremely sensitive to pH changes because of the pH-dependence of proteins involved in neuronal excitability and synaptic transmission. Here, we show that the Na+/H+ exchanger Nhe1, which uses the Na+ gradient to extrude H+, is expressed at both inhibitory and excitatory presynapses. We disrupted Nhe1 specifically in mice either in Emx1-positive glutamatergic neurons or in parvalbumin-positive cells, mainly GABAergic interneurons. While Nhe1 disruption in excitatory neurons had no effect on overall network excitability, mice with disruption of Nhe1 in parvalbumin-positive neurons displayed epileptic activity. From our electrophysiological analyses in the CA1 of the hippocampus, we conclude that the disruption in parvalbumin-positive neurons impairs the release of GABA-loaded vesicles, but increases the size of GABA quanta. The latter is most likely an indirect pH-dependent effect, as Nhe1 was not expressed in purified synaptic vesicles itself. Conclusively, our data provide first evidence that Nhe1 affects network excitability via modulation of inhibitory interneurons.


Subject(s)
CA1 Region, Hippocampal/physiology , Membrane Potentials , Presynaptic Terminals/physiology , Sodium-Hydrogen Exchanger 1/physiology , gamma-Aminobutyric Acid/physiology , Animals , Epilepsy/physiopathology , Female , GABAergic Neurons/physiology , Glutamic Acid/metabolism , Interneurons/physiology , Male , Mice, Inbred C57BL , Mice, Transgenic , Presynaptic Terminals/metabolism , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , gamma-Aminobutyric Acid/metabolism
5.
J Am Soc Nephrol ; 28(1): 209-217, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27335120

ABSTRACT

Chloride transport by the renal tubule is critical for blood pressure (BP), acid-base, and potassium homeostasis. Chloride uptake from the urinary fluid is mediated by various apical transporters, whereas basolateral chloride exit is thought to be mediated by ClC-Ka/K1 and ClC-Kb/K2, two chloride channels from the ClC family, or by KCl cotransporters from the SLC12 gene family. Nevertheless, the localization and role of ClC-K channels is not fully resolved. Because inactivating mutations in ClC-Kb/K2 cause Bartter syndrome, a disease that mimics the effects of the loop diuretic furosemide, ClC-Kb/K2 is assumed to have a critical role in salt handling by the thick ascending limb. To dissect the role of this channel in detail, we generated a mouse model with a targeted disruption of the murine ortholog ClC-K2. Mutant mice developed a Bartter syndrome phenotype, characterized by renal salt loss, marked hypokalemia, and metabolic alkalosis. Patch-clamp analysis of tubules isolated from knockout (KO) mice suggested that ClC-K2 is the main basolateral chloride channel in the thick ascending limb and in the aldosterone-sensitive distal nephron. Accordingly, ClC-K2 KO mice did not exhibit the natriuretic response to furosemide and exhibited a severely blunted response to thiazide. We conclude that ClC-Kb/K2 is critical for salt absorption not only by the thick ascending limb, but also by the distal convoluted tubule.


Subject(s)
Anion Transport Proteins/physiology , Chloride Channels/physiology , Nephrons/metabolism , Sodium Chloride/metabolism , Animals , Diuretics/pharmacology , Furosemide/pharmacology , Mice , Mice, Knockout , Nephrons/drug effects , Sodium Chloride Symporter Inhibitors/pharmacology
6.
J Am Soc Nephrol ; 28(5): 1507-1520, 2017 May.
Article in English | MEDLINE | ID: mdl-27932475

ABSTRACT

Distal nephron acid secretion is mediated by highly specialized type A intercalated cells (A-ICs), which contain vacuolar H+-ATPase (V-type ATPase)-rich vesicles that fuse with the apical plasma membrane on demand. Intracellular bicarbonate generated by luminal H+ secretion is removed by the basolateral anion-exchanger AE1. Chronically reduced renal acid excretion in distal renal tubular acidosis (dRTA) may lead to nephrocalcinosis and renal failure. Studies in MDCK monolayers led to the proposal of a dominant-negative trafficking mechanism to explain AE1-associated dominant dRTA. To test this hypothesis in vivo, we generated an Ae1 R607H knockin mouse, which corresponds to the most common dominant dRTA mutation in human AE1, R589H. Compared with wild-type mice, heterozygous and homozygous R607H knockin mice displayed incomplete dRTA characterized by compensatory upregulation of the Na+/HCO3- cotransporter NBCn1. Red blood cell Ae1-mediated anion-exchange activity and surface polypeptide expression did not change. Mutant mice expressed far less Ae1 in A-ICs, but basolateral targeting of the mutant protein was preserved. Notably, mutant mice also exhibited reduced expression of V-type ATPase and compromised targeting of this proton pump to the plasma membrane upon acid challenge. Accumulation of p62- and ubiquitin-positive material in A-ICs of knockin mice suggested a defect in the degradative pathway, which may explain the observed loss of A-ICs. R607H knockin did not affect type B intercalated cells. We propose that reduced basolateral anion-exchange activity in A-ICs inhibits trafficking and regulation of V-type ATPase, compromising luminal H+ secretion and possibly lysosomal acidification.


Subject(s)
Acidosis, Renal Tubular/enzymology , Anion Exchange Protein 1, Erythrocyte/physiology , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/enzymology , Vacuolar Proton-Translocating ATPases/physiology , Animals , Anion Exchange Protein 1, Erythrocyte/genetics , Male , Mice , Models, Biological
7.
PLoS Genet ; 11(8): e1005454, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26284655

ABSTRACT

Hereditary spastic paraplegia (HSP) is characterized by a dying back degeneration of corticospinal axons which leads to progressive weakness and spasticity of the legs. SPG11 is the most common autosomal-recessive form of HSPs and is caused by mutations in SPG11. A recent in vitro study suggested that Spatacsin, the respective gene product, is needed for the recycling of lysosomes from autolysosomes, a process known as autophagic lysosome reformation. The relevance of this observation for hereditary spastic paraplegia, however, has remained unclear. Here, we report that disruption of Spatacsin in mice indeed causes hereditary spastic paraplegia-like phenotypes with loss of cortical neurons and Purkinje cells. Degenerating neurons accumulate autofluorescent material, which stains for the lysosomal protein Lamp1 and for p62, a marker of substrate destined to be degraded by autophagy, and hence appears to be related to autolysosomes. Supporting a more generalized defect of autophagy, levels of lipidated LC3 are increased in Spatacsin knockout mouse embryonic fibrobasts (MEFs). Though distinct parameters of lysosomal function like processing of cathepsin D and lysosomal pH are preserved, lysosome numbers are reduced in knockout MEFs and the recovery of lysosomes during sustained starvation impaired consistent with a defect of autophagic lysosome reformation. Because lysosomes are reduced in cortical neurons and Purkinje cells in vivo, we propose that the decreased number of lysosomes available for fusion with autophagosomes impairs autolysosomal clearance, results in the accumulation of undegraded material and finally causes death of particularly sensitive neurons like cortical motoneurons and Purkinje cells in knockout mice.


Subject(s)
Autophagy , Lysosomes/physiology , Proteins/genetics , Spastic Paraplegia, Hereditary/pathology , Animals , Cells, Cultured , Cerebellum/pathology , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Motor Cortex/pathology , Purkinje Cells/pathology , Spastic Paraplegia, Hereditary/genetics
8.
Anal Biochem ; 474: 35-7, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25615417

ABSTRACT

Following locus-specific genome editing of mouse embryonic stem cells (ESCs), the identification of correctly targeted clones remains a challenge. We applied multiplex ligation-dependent probe amplification (MLPA) to screen for homologous recombination-based genomic integration of a knockout construct in which part of a gene is deleted. All candidate ESCs thereby identified were subsequently validated by conventional methods. Thus, MLPA represents a highly reliable as well as cost- and time-efficient alternative to currently applied methods such as Southern blotting and polymerase chain reaction (PCR)-based approaches. It is also applicable to knockin recombination strategies and compatible with the CRISPR/Cas9 system and other genome editing strategies.


Subject(s)
Embryonic Stem Cells/cytology , Multiplex Polymerase Chain Reaction/methods , Animals , Clone Cells , Electroporation , Embryonic Stem Cells/metabolism , Homologous Recombination/genetics , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice, Knockout
9.
Cell Physiol Biochem ; 34(5): 1566-77, 2014.
Article in English | MEDLINE | ID: mdl-25402438

ABSTRACT

BACKGROUND: The Slc4 family of bicarbonate transporters consists of several members, many of which are highly expressed in the kidney and play an important role in acid-base homeostasis. Among them are Ae1 (Slc4a1) and Ae2 (Slc4a2). Another member, Ae3 (Slc4a3), is suggested to be expressed in the kidney, however, its localization and impact on renal function is still unknown. Ae3 has also been implicated in the central control of breathing. AIMS: Here, we analyzed the expression of Ae3 transcripts in isolated nephron segments and investigated systemic and renal acid-base homeostasis and renal electrolyte handling in the absence of Ae3, using a knock out mouse model. METHODS: qPCR was used to localize Ae3 transcripts in the murine nephron, metabolic studies and whole body plethysmography to assess the role of Ae3 in renal functions. RESULTS: Two Ae3 transcripts, the brain variant bAe3 and the cardiac variant cAe3, are expressed at low levels in the murine kidney. Although differentially distributed, they localize mostly to the distal nephron and renal collecting duct system. At baseline and after an acid challenge, mice deficient for Ae3 did not show major disturbances in renal acid-base excretion. Respiratory responses in whole body plethysmography to acid loading and CO2 and O2 challenges were normal. No major differences in renal electrolyte handling were discovered except for small changes in magnesium, potassium and sodium excretion after 7 days of acid loading. We therefore challenged mice with diets with high and low magnesium diets and found no differences in renal magnesium excretion but elevated expression of the Trpm6 magnesium channel in Ae3 KO mice. In conclusion, Ae3 is expressed in murine kidney at very low levels. CONCLUSIONS: Ae3 plays no role in systemic acid-base homeostasis but may modify renal magnesium handling inducing a higher expression of Trpm6.


Subject(s)
Acid-Base Equilibrium/physiology , Antiporters/metabolism , Chloride-Bicarbonate Antiporters/metabolism , Kidney/metabolism , Magnesium/metabolism , Animals , Carbon Dioxide/metabolism , Electrolytes/metabolism , Homeostasis/physiology , Kidney/physiology , Male , Mice , Mice, Knockout , Oxygen/metabolism , Potassium/metabolism , Sodium/metabolism
10.
PLoS Genet ; 9(12): e1003988, 2013.
Article in English | MEDLINE | ID: mdl-24367272

ABSTRACT

Hereditary spastic paraplegias (HSPs) are characterized by progressive weakness and spasticity of the legs because of the degeneration of cortical motoneuron axons. SPG15 is a recessively inherited HSP variant caused by mutations in the ZFYVE26 gene and is additionally characterized by cerebellar ataxia, mental decline, and progressive thinning of the corpus callosum. ZFYVE26 encodes the FYVE domain-containing protein ZFYVE26/SPASTIZIN, which has been suggested to be associated with the newly discovered adaptor protein 5 (AP5) complex. We show that Zfyve26 is broadly expressed in neurons, associates with intracellular vesicles immunopositive for the early endosomal marker EEA1, and co-fractionates with a component of the AP5 complex. As the function of ZFYVE26 in neurons was largely unknown, we disrupted Zfyve26 in mice. Zfyve26 knockout mice do not show developmental defects but develop late-onset spastic paraplegia with cerebellar ataxia confirming that SPG15 is caused by ZFYVE26 deficiency. The morphological analysis reveals axon degeneration and progressive loss of both cortical motoneurons and Purkinje cells in the cerebellum. Importantly, neuron loss is preceded by accumulation of large intraneuronal deposits of membrane-surrounded material, which co-stains with the lysosomal marker Lamp1. A density gradient analysis of brain lysates shows an increase of Lamp1-positive membrane compartments with higher densities in Zfyve26 knockout mice. Increased levels of lysosomal enzymes in brains of aged knockout mice further support an alteration of the lysosomal compartment upon disruption of Zfyve26. We propose that SPG15 is caused by an endolysosomal membrane trafficking defect, which results in endolysosomal dysfunction. This appears to be particularly relevant in neurons with highly specialized neurites such as cortical motoneurons and Purkinje cells.


Subject(s)
Carrier Proteins/genetics , Endosomes/metabolism , Lysosomes/metabolism , Retinal Degeneration/genetics , Spastic Paraplegia, Hereditary/genetics , Animals , Brain/metabolism , Brain/pathology , Carrier Proteins/metabolism , Corpus Callosum/metabolism , Corpus Callosum/pathology , Disease Models, Animal , Endosomes/pathology , Humans , Lysosomes/genetics , Mice , Mice, Knockout , Motor Neurons/metabolism , Mutation , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Spastic Paraplegia, Hereditary/metabolism , Spastic Paraplegia, Hereditary/pathology
11.
Nat Genet ; 45(11): 1399-404, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24036948

ABSTRACT

The sensation of pain protects the body from serious injury. Using exome sequencing, we identified a specific de novo missense mutation in SCN11A in individuals with the congenital inability to experience pain who suffer from recurrent tissue damage and severe mutilations. Heterozygous knock-in mice carrying the orthologous mutation showed reduced sensitivity to pain and self-inflicted tissue lesions, recapitulating aspects of the human phenotype. SCN11A encodes Nav1.9, a voltage-gated sodium ion channel that is primarily expressed in nociceptors, which function as key relay stations for the electrical transmission of pain signals from the periphery to the central nervous system. Mutant Nav1.9 channels displayed excessive activity at resting voltages, causing sustained depolarization of nociceptors, impaired generation of action potentials and aberrant synaptic transmission. The gain-of-function mechanism that underlies this channelopathy suggests an alternative way to modulate pain perception.


Subject(s)
Pain Perception/physiology , Pain/genetics , Action Potentials/genetics , Animals , Channelopathies/genetics , Gene Knock-In Techniques , Humans , Mice , Mice, Inbred C57BL , NAV1.9 Voltage-Gated Sodium Channel/genetics , Nociceptors/physiology
12.
Am J Hum Genet ; 93(4): 727-34, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-24035193

ABSTRACT

In guanosine diphosphate (GDP)-mannose pyrophosphorylase A (GMPPA), we identified a homozygous nonsense mutation that segregated with achalasia and alacrima, delayed developmental milestones, and gait abnormalities in a consanguineous Pakistani pedigree. Mutations in GMPPA were subsequently found in ten additional individuals from eight independent families affected by the combination of achalasia, alacrima, and neurological deficits. This autosomal-recessive disorder shows many similarities with triple A syndrome, which is characterized by achalasia, alacrima, and variable neurological deficits in combination with adrenal insufficiency. GMPPA is a largely uncharacterized homolog of GMPPB. GMPPB catalyzes the formation of GDP-mannose, which is an essential precursor of glycan moieties of glycoproteins and glycolipids and is associated with congenital and limb-girdle muscular dystrophies with hypoglycosylation of α-dystroglycan. Surprisingly, GDP-mannose pyrophosphorylase activity was unchanged and GDP-mannose levels were strongly increased in lymphoblasts of individuals with GMPPA mutations. This suggests that GMPPA might serve as a GMPPB regulatory subunit mediating feedback inhibition of GMPPB instead of displaying catalytic enzyme activity itself. Thus, a triple-A-like syndrome can be added to the growing list of congenital disorders of glycosylation, in which dysregulation rather than mere enzyme deficiency is the basal pathophysiological mechanism.


Subject(s)
Codon, Nonsense , Genes, Recessive/genetics , Guanosine Diphosphate Mannose/genetics , Intellectual Disability/genetics , Nucleotidyltransferases/genetics , Adolescent , Adrenal Insufficiency/genetics , Adult , Child , Consanguinity , Esophageal Achalasia/genetics , Eye Diseases, Hereditary/genetics , Glycosylation , Guanosine Diphosphate Mannose/metabolism , Homozygote , Humans , Intellectual Disability/enzymology , Lacrimal Apparatus Diseases/genetics , Nervous System Diseases/genetics , Nucleotidyltransferases/metabolism , Pedigree , Young Adult
13.
EMBO J ; 32(16): 2275-86, 2013 Aug 14.
Article in English | MEDLINE | ID: mdl-23881097

ABSTRACT

Brain carbonic anhydrases (CAs) are known to modulate neuronal signalling. Using a novel CA VII (Car7) knockout (KO) mouse as well as a CA II (Car2) KO and a CA II/VII double KO, we show that mature hippocampal pyramidal neurons are endowed with two cytosolic isoforms. CA VII is predominantly expressed by neurons starting around postnatal day 10 (P10). The ubiquitous isoform II is expressed in neurons at P20. Both isoforms enhance bicarbonate-driven GABAergic excitation during intense GABAA-receptor activation. P13-14 CA VII KO mice show behavioural manifestations atypical of experimental febrile seizures (eFS) and a complete absence of electrographic seizures. A low dose of diazepam promotes eFS in P13-P14 rat pups, whereas seizures are blocked at higher concentrations that suppress breathing. Thus, the respiratory alkalosis-dependent eFS are exacerbated by GABAergic excitation. We found that CA VII mRNA is expressed in the human cerebral cortex before the age when febrile seizures (FS) occur in children. Our data indicate that CA VII is a key molecule in age-dependent neuronal pH regulation with consequent effects on generation of FS.


Subject(s)
Carbonic Anhydrase II/metabolism , Carbonic Anhydrases/metabolism , Cerebral Cortex/cytology , GABAergic Neurons/metabolism , Seizures, Febrile/enzymology , Age Factors , Analysis of Variance , Animals , Blotting, Northern , Blotting, Western , Carbonic Anhydrase II/genetics , Carbonic Anhydrases/genetics , Cerebral Cortex/metabolism , Diazepam/toxicity , Electroencephalography , Fluorescence , Humans , Hydrogen-Ion Concentration , Mice , Mice, Knockout , Rats , Seizures, Febrile/chemically induced , Seizures, Febrile/metabolism
14.
EMBO Mol Med ; 4(10): 1057-71, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22933323

ABSTRACT

The V-ATPase is a multisubunit complex that transports protons across membranes. Mutations of its B1 or a4 subunit are associated with distal renal tubular acidosis and deafness. In the kidney, the a4 subunit is expressed in intercalated cells of the distal nephron, where the V-ATPase controls acid/base secretion, and in proximal tubule cells, where its role is less clear. Here, we report that a4 KO mice suffer not only from severe acidosis but also from proximal tubule dysfunction with defective endocytic trafficking, proteinuria, phosphaturia and accumulation of lysosomal material and we provide evidence that these findings may be also relevant in patients. In the inner ear, the a4 subunit co-localized with pendrin at the apical side of epithelial cells lining the endolymphatic sac. As a4 KO mice were profoundly deaf and displayed enlarged endolymphatic fluid compartments mirroring the alterations in pendrin KO mice, we propose that pendrin and the proton pump co-operate in endolymph homeostasis. Thus, our mouse model gives new insights into the divergent functions of the V-ATPase and the pathophysiology of a4-related symptoms.


Subject(s)
Acidosis, Renal Tubular/physiopathology , Kidney Tubules, Proximal/enzymology , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Child , Child, Preschool , Disease Models, Animal , Humans , Infant , Mice , Mice, Knockout , Protein Subunits/deficiency , Protein Subunits/metabolism , Vacuolar Proton-Translocating ATPases/deficiency
15.
Nat Genet ; 41(11): 1179-81, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19838196

ABSTRACT

Hereditary sensory and autonomic neuropathy type II (HSAN II) leads to severe mutilations because of impaired nociception and autonomic dysfunction. Here we show that loss-of-function mutations in FAM134B, encoding a newly identified cis-Golgi protein, cause HSAN II. Fam134b knockdown results in structural alterations of the cis-Golgi compartment and induces apoptosis in some primary dorsal root ganglion neurons. This implicates FAM134B as critical in long-term survival of nociceptive and autonomic ganglion neurons.


Subject(s)
Golgi Apparatus/metabolism , Hereditary Sensory and Autonomic Neuropathies/genetics , Membrane Proteins/genetics , Mutation , Neoplasm Proteins/genetics , Adult , Animals , Female , Hereditary Sensory and Autonomic Neuropathies/metabolism , Hereditary Sensory and Autonomic Neuropathies/pathology , Humans , Intracellular Signaling Peptides and Proteins , Male , Membrane Proteins/metabolism , Mice , Pedigree , RNA Interference
16.
Free Radic Biol Med ; 43(8): 1189-96, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17854714

ABSTRACT

Activation of leukocytes and in particular polymorphonuclear neutrophils (PMN) has emerged as a critical confounder in the pathophysiology of cardiovascular disease: Myeloperoxidase (MPO), one of the principal proteins hosted in and secreted by activated PMN, has been mechanistically linked to endothelial and left ventricular (LV) dysfunction in rodent models of sepsis and ischemic cardiomyopathy. Whether PMN activation is also overt in patients with LV dysfunction of ischemic and nonischemic origin, however, remains elusive. Prospectively, 447 consecutive, stable outpatients were included in this single-center study. In 113 patients with impaired left ventricular function (ejection fraction <50%; nonischemic cardiomyopathy, n=52; ischemic cardiomyopathy, n=61), MPO plasma levels were elevated (24.5 [IR:15.8-54.0] vs 15.5 [IR:8.9-39.2] ng/ml in controls, P<0.01) as was elastase (111.5 [IR:63.8-233.3] vs 70.5 [IR:45.0-129.0] ng/ml, P<0.01) and NT-proBNP plasma levels (747.4 [IR:216.3-1958.3] vs 264.1 [IR:82.5-671.8] ng/L, P<0.01). Elevation of circulating MPO was irrespective of the etiology of heart failure and independent of traditional confounding variables. No association was observed between MPO -463 promoter polymorphism genotype and LV dysfunction. MPO plasma levels correlated with ejection fraction (P<0.01) and left ventricular end-diastolic diameter (P<0.01), respectively. Myeloperoxidase mRNA expression levels obtained from circulating leukocytes were significantly increased in patients with LV dysfunction. Systemic leukocyte activation with increased transcription of MPO mRNA and augmented release of MPO appears to represent a so far underrecognized characteristic in LV dysfunction, which was revealed to be irrespective of the underlying pathology. Given its potent proinflammatory properties, MPO may represent an important mechanistic link to LV dysfunction and deserves to be evaluated as both marker and therapeutic target in this disease.


Subject(s)
Neutrophils/physiology , Ventricular Dysfunction, Left/physiopathology , Aged , Cardiomyopathies/blood , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/physiopathology , Female , Humans , Logistic Models , Male , Middle Aged , Myocardial Ischemia/blood , Myocardial Ischemia/diagnostic imaging , Myocardial Ischemia/physiopathology , Natriuretic Peptide, Brain/blood , Pancreatic Elastase/blood , Peroxidase/blood , Peroxidase/genetics , Polymorphism, Genetic , Predictive Value of Tests , Prospective Studies , RNA, Messenger/metabolism , Ultrasonography , Ventricular Dysfunction, Left/blood , Ventricular Dysfunction, Left/diagnostic imaging
17.
Pflugers Arch ; 454(2): 195-207, 2007 May.
Article in English | MEDLINE | ID: mdl-17242957

ABSTRACT

Recent studies showed that the Ca(2+)-independent transient outward current (I (to)) is very small or even not detectable in the porcine left ventricle. We investigated whether an altered molecular expression or function of voltage-dependent potassium channels belonging to the Kv4 sub-family and their ancillary Ca(2+)-binding beta sub-unit KChIP2, which contribute to the major fraction of I (to )in other species, may underlie this lack of a significant I (to) in the porcine left ventricle. RT-PCR analysis with degenerate primers showed that both Kv4 mRNA and KChIP2 mRNA are expressed in porcine left ventricular tissue and in isolated ventricular myocytes. PCR cloning and sequence analysis predicted proteins with >98% identity to rat and human Kv4.2 and >99% identity to rat and human KChIP2. Heterologous expression of porcine Kv4.2 in Xenopus laevis oocytes gave rise to currents with characteristic properties of rat and human Kv4.2, and co-expression with its KChIP2 sub-unit increased current density (tenfold), slowed inactivation (twofold) and accelerated recovery from inactivation (tenfold). Kv4.2 immunohistochemistry in porcine left ventricular tissue revealed a predominant membrane-bound signal. Relative quantification of gene expression indicated that Kv4.2 and KChIP2 mRNA and protein are expressed at comparable ratios in porcine and rat left ventricular tissues, which displays a large I (to). Collectively, these data demonstrate that the lack of a significant I (to) in the porcine left ventricle does not result from dysfunctional or insufficiently expressed Kv4.2 and KChIP2 sub-units.


Subject(s)
Heart/physiology , Kv Channel-Interacting Proteins/physiology , Shal Potassium Channels/physiology , 4-Aminopyridine/pharmacology , Amino Acid Sequence , Animals , Barium/pharmacology , Cadmium/pharmacology , Calcium/metabolism , Cloning, Molecular , Electric Stimulation , Electrophysiology , Female , Gene Expression , Kv Channel-Interacting Proteins/genetics , Male , Membrane Potentials/drug effects , Molecular Sequence Data , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Oocytes/drug effects , Oocytes/metabolism , Oocytes/physiology , Rats , Rats, Sprague-Dawley , Sequence Homology, Amino Acid , Shal Potassium Channels/genetics , Sus scrofa , Ventricular Function , Xenopus laevis
18.
Development ; 132(13): 3139-49, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15944193

ABSTRACT

In diabetic individuals, the imbalance in glucose homeostasis is caused by loss or dysfunction of insulin-secreting beta-cells of the pancreatic islets. As successful generation of insulin-producing cells in vitro could constitute a cure for diabetes, recent studies have explored the molecular program that underlies beta-cell formation. From these studies, the homeodomain transcription factor NKX6.1 has proven to be a key player. In Nkx6.1 mutants, beta-cell numbers are selectively reduced, while other islet cell types develop normally. However, the molecular events downstream of NKX6.1, as well as the molecular pathways that ensure residual beta-cell formation in the absence of NKX6.1 are largely unknown. Here, we show that the Nkx6.1 paralog, Nkx6.2, is expressed during pancreas development and partially compensates for NKX6.1 function. Surprisingly, our analysis of Nkx6 compound mutant mice revealed a previously unrecognized requirement for NKX6 activity in alpha-cell formation. This finding suggests a more general role for NKX6 factors in endocrine cell differentiation than formerly suggested. Similar to NKX6 factors, the transcription factor MYT1 has recently been shown to regulate alpha- as well as beta-cell development. We demonstrate that expression of Myt1 depends on overall Nkx6 gene dose, and therefore identify Myt1 as a possible downstream target of Nkx6 genes in the endocrine differentiation pathway.


Subject(s)
Cell Differentiation/physiology , Homeodomain Proteins/metabolism , Islets of Langerhans/physiology , Pancreas/embryology , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/genetics , Islets of Langerhans/cytology , Mice , Mutation , Nerve Tissue Proteins/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...