Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 265
Filter
1.
Nat Commun ; 15(1): 4866, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849373

ABSTRACT

Dense and aligned Collagen I fibers are associated with collective cancer invasion led by protrusive tumor cells, leader cells. In some breast tumors, a population of cancer cells (basal-like cells) maintain several epithelial characteristics and express the myoepithelial/basal cell marker Keratin 14 (K14). Emergence of leader cells and K14 expression are regarded as interconnected events triggered by Collagen I, however the underlying mechanisms remain unknown. Using breast carcinoma organoids, we show that Collagen I drives a force-dependent loop, specifically in basal-like cancer cells. The feed-forward loop is centered around the mechanotransducer Yap and independent of K14 expression. Yap promotes a transcriptional program that enhances Collagen I alignment and tension, which further activates Yap. Active Yap is detected in invading breast cancer cells in patients and required for collective invasion in 3D Collagen I and in the mammary fat pad of mice. Our work uncovers an essential function for Yap in leader cell selection during collective cancer invasion.


Subject(s)
Adaptor Proteins, Signal Transducing , Breast Neoplasms , Collagen Type I , Mechanotransduction, Cellular , Neoplasm Invasiveness , Transcription Factors , YAP-Signaling Proteins , Animals , Female , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Cell Line, Tumor , Collagen Type I/metabolism , Gene Expression Regulation, Neoplastic , Organoids/metabolism , Organoids/pathology , Transcription Factors/metabolism , Transcription Factors/genetics , YAP-Signaling Proteins/metabolism
2.
Metabolites ; 14(5)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38786724

ABSTRACT

Direct infusion-high-resolution mass spectrometry (DI-HRMS) allows for rapid profiling of complex mixtures of metabolites in blood, cerebrospinal fluid, tissue samples and cultured cells. Here, we present a DI-HRMS method suitable for the rapid determination of metabolic fluxes of isotopically labeled substrates in cultured cells and organoids. We adapted an automated annotation pipeline by selecting labeled adducts that best represent the majority of 13C and/or 15N-labeled glycolytic and tricarboxylic acid cycle intermediates as well as a number of their derivatives. Furthermore, valine, leucine and several of their degradation products were included. We show that DI-HRMS can determine anticipated and unanticipated alterations in metabolic fluxes along these pathways that result from the genetic alteration of single metabolic enzymes, including pyruvate dehydrogenase (PDHA1) and glutaminase (GLS). In addition, it can precisely pinpoint metabolic adaptations to the loss of methylmalonyl-CoA mutase in patient-derived liver organoids. Our results highlight the power of DI-HRMS in combination with stable isotopically labeled compounds as an efficient screening method for fluxomics.

3.
Metabolites ; 13(12)2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38132878

ABSTRACT

NAD synthetase 1 (encoded by the gene NADSYN1) is a cytosolic enzyme that catalyzes the final step in the biosynthesis of nicotinamide adenine dinucleotide (NAD+) from tryptophan and nicotinic acid. NADSYN1 deficiency has recently been added to the spectrum of congenital NAD+ deficiency disorders. To gain insight into the metabolic consequences of NADSYN1 deficiency, the encoding gene was disrupted in A549 and HEK293T cells, and the metabolome was profiled in the presence of different NAD+ precursors, including tryptophan, nicotinamide and nicotinic acid. We demonstrate that when precursors of the NAD+ salvage pathway in the form of nicotinamide become limiting, NADSYN1 deficiency results in a decline in intracellular NAD+ levels even in the presence of other potential NAD+ sources such as tryptophan and nicotinic acid. As a consequence, alterations in 122 and 69 metabolites are observed in NADSYN1-deficient A549 and HEK293T cells compared to the wild-type cell line (FC > 2 and p < 0.05). We thus show that NADSYN1 deficiency results in a metabolic phenotype characterized by alterations in glycolysis, the TCA cycle, the pentose phosphate pathway, and the polyol pathway.

4.
Cell Rep ; 42(9): 113043, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37647199

ABSTRACT

The malate-aspartate shuttle (MAS) is a redox shuttle that transports reducing equivalents across the inner mitochondrial membrane while recycling cytosolic NADH to NAD+. We genetically disrupted each MAS component to generate a panel of MAS-deficient HEK293 cell lines in which we performed [U-13C]-glucose tracing. MAS-deficient cells have reduced serine biosynthesis, which strongly correlates with the lactate M+3/pyruvate M+3 ratio (reflective of the cytosolic NAD+/NADH ratio), consistent with the NAD+ dependency of phosphoglycerate dehydrogenase in the serine synthesis pathway. Among the MAS-deficient cells, those lacking malate dehydrogenase 1 (MDH1) show the most severe metabolic disruptions, whereas oxoglutarate-malate carrier (OGC)- and MDH2-deficient cells are less affected. Increasing the NAD+-regenerating capacity using pyruvate supplementation resolves most of the metabolic disturbances. Overall, we show that the MAS is important for de novo serine biosynthesis, implying that serine supplementation could be used as a therapeutic strategy for MAS defects and possibly other redox disorders.


Subject(s)
Aspartic Acid , Malates , Humans , Aspartic Acid/metabolism , Malates/metabolism , NAD/metabolism , HEK293 Cells , Oxidation-Reduction , Pyruvates
5.
J Biol Chem ; 299(9): 105047, 2023 09.
Article in English | MEDLINE | ID: mdl-37451483

ABSTRACT

Recently, biallelic variants in PLPBP coding for pyridoxal 5'-phosphate homeostasis protein (PLPHP) were identified as a novel cause of early-onset vitamin B6-dependent epilepsy. The molecular function and precise role of PLPHP in vitamin B6 metabolism are not well understood. To address these questions, we used PLPHP-deficient patient skin fibroblasts and HEK293 cells and YBL036C (PLPHP ortholog)-deficient yeast. We showed that independent of extracellular B6 vitamer type (pyridoxine, pyridoxamine, or pyridoxal), intracellular pyridoxal 5'-phosphate (PLP) was lower in PLPHP-deficient fibroblasts and HEK293 cells than controls. Culturing cells with pyridoxine or pyridoxamine led to the concentration-dependent accumulation of pyridoxine 5'-phosphate and pyridoxamine 5'-phosphate (PMP), respectively, suggesting insufficient pyridox(am)ine 5'-phosphate oxidase activity. Experiments utilizing 13C4-pyridoxine confirmed lower pyridox(am)ine 5'-phosphate oxidase activity and revealed increased fractional turnovers of PLP and pyridoxal, indicating increased PLP hydrolysis to pyridoxal in PLPHP-deficient cells. This effect could be partly counteracted by inactivation of pyridoxal phosphatase. PLPHP deficiency had a distinct effect on mitochondrial PLP and PMP, suggesting impaired activity of mitochondrial transaminases. Moreover, in YBL036C-deficient yeast, PLP was depleted and PMP accumulated only with carbon sources requiring mitochondrial metabolism. Lactate and pyruvate accumulation along with the decrease of tricarboxylic acid cycle intermediates downstream of α-ketoglutarate suggested impaired mitochondrial oxidative metabolism in PLPHP-deficient HEK293 cells. We hypothesize that impaired activity of mitochondrial transaminases may contribute to this depletion. Taken together, our study provides new insights into the pathomechanisms of PLPBP deficiency and reinforces the link between PLPHP function, vitamin B6 metabolism, and mitochondrial oxidative metabolism.


Subject(s)
Mitochondria , Vitamin B 6 , Humans , HEK293 Cells , Proteins/genetics , Proteins/metabolism , Pyridoxal Phosphate/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transaminases/metabolism , Vitamin B 6/metabolism , Fibroblasts , Cells, Cultured , Pyridoxaminephosphate Oxidase/metabolism , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/metabolism , Oxidation-Reduction , Amino Acids/metabolism
6.
Orphanet J Rare Dis ; 17(1): 252, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35804402

ABSTRACT

BACKGROUND: MTOR inhibition is an effective treatment for many manifestations of tuberous sclerosis complex. Because mTOR inhibition is a disease modifying therapy, lifelong use will most likely be necessary. This study addresses the long-term effects of mTOR inhibitors on lipid and glucose metabolism and aims to provide better insight in the incidence and time course of these metabolic adverse effects in treated TSC patients. METHODS: All patients who gave informed consent for the nationwide TSC Registry and were ever treated with mTOR inhibitors (sirolimus and/or everolimus) were included. Lipid profiles, HbA1c and medication were analysed in all patients before and during mTOR inhibitor treatment. RESULTS: We included 141 patients, the median age was 36 years, median use of mTOR inhibitors 5.1 years (aimed serum levels 3.0-5.0 µg/l). Total cholesterol, LDL- and HDL-cholesterol levels at baseline were similar to healthy reference data. After start of mTOR inhibition therapy, total cholesterol, LDL-cholesterol and triglycerides increased significantly and were higher compared to healthy reference population. Mean total cholesterol levels increased by 1.0 mmol/L after 3-6 months of mTOR inhibition therapy but did not increase further during follow-up. In this study, 2.5% (3/118) of patients developed diabetes (defined as an HbA1c ≥ 48 mmol/mol) during a median follow-up of 5 years. CONCLUSIONS: Hypercholesterolemia is a frequent side effect of mTOR inhibition in TSC patients, and predominantly occurs within the first year of treatment. Although hyperglycemia is a frequent side effect in other indications for mTOR inhibition, incidence of diabetes mellitus in TSC patients was only 2.5%. This may reflect the difference of mTOR inhibition in patients with normal mTOR complex pathway function versus patients with overactive mTOR complex signaling due to a genetic defect (TSC patients).


Subject(s)
Tuberous Sclerosis , Adult , Humans , Cholesterol, LDL , Glucose/therapeutic use , Glycated Hemoglobin/therapeutic use , Registries , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/metabolism
7.
Mol Cell Proteomics ; 21(9): 100263, 2022 09.
Article in English | MEDLINE | ID: mdl-35863698

ABSTRACT

In Birt-Hogg-Dubé (BHD) syndrome, germline loss-of-function mutations in the Folliculin (FLCN) gene lead to an increased risk of renal cancer. To address how FLCN inactivation affects cellular kinase signaling pathways, we analyzed comprehensive phosphoproteomic profiles of FLCNPOS and FLCNNEG human renal tubular epithelial cells (RPTEC/TERT1). In total, 15,744 phosphorylated peptides were identified from 4329 phosphorylated proteins. INKA analysis revealed that FLCN loss alters the activity of numerous kinases, including tyrosine kinases EGFR, MET, and the Ephrin receptor subfamily (EPHA2 and EPHB1), as well their downstream targets MAPK1/3. Validation experiments in the BHD renal tumor cell line UOK257 confirmed that FLCN loss contributes to enhanced MAPK1/3 and downstream RPS6K1/3 signaling. The clinically available MAPK inhibitor Ulixertinib showed enhanced toxicity in FLCNNEG cells. Interestingly, FLCN inactivation induced the phosphorylation of PIK3CD (Tyr524) without altering the phosphorylation of canonical Akt1/Akt2/mTOR/EIF4EBP1 phosphosites. Also, we identified that FLCN inactivation resulted in dephosphorylation of TFEB Ser109, Ser114, and Ser122, which may be linked to increased oxidative stress levels in FLCNNEG cells. Together, our study highlights differential phosphorylation of specific kinases and substrates in FLCNNEG renal cells. This provides insight into BHD-associated renal tumorigenesis and may point to several novel candidates for targeted therapies.


Subject(s)
Birt-Hogg-Dube Syndrome , Kidney Neoplasms , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Birt-Hogg-Dube Syndrome/genetics , Birt-Hogg-Dube Syndrome/metabolism , Birt-Hogg-Dube Syndrome/pathology , Ephrins , ErbB Receptors , Humans , Kidney Neoplasms/genetics , Phosphoserine , Proto-Oncogene Proteins , TOR Serine-Threonine Kinases , Tumor Suppressor Proteins , Tyrosine
8.
J Am Soc Nephrol ; 32(11): 2885-2899, 2021 11.
Article in English | MEDLINE | ID: mdl-34607910

ABSTRACT

BACKGROUND: Over the last decade, advances in genetic techniques have resulted in the identification of rare hereditary disorders of renal magnesium and salt handling. Nevertheless, approximately 20% of all patients with tubulopathy lack a genetic diagnosis. METHODS: We performed whole-exome and -genome sequencing of a patient cohort with a novel, inherited, salt-losing tubulopathy; hypomagnesemia; and dilated cardiomyopathy. We also conducted subsequent in vitro functional analyses of identified variants of RRAGD, a gene that encodes a small Rag guanosine triphosphatase (GTPase). RESULTS: In eight children from unrelated families with a tubulopathy characterized by hypomagnesemia, hypokalemia, salt wasting, and nephrocalcinosis, we identified heterozygous missense variants in RRAGD that mostly occurred de novo. Six of these patients also had dilated cardiomyopathy and three underwent heart transplantation. We identified a heterozygous variant in RRAGD that segregated with the phenotype in eight members of a large family with similar kidney manifestations. The GTPase RagD, encoded by RRAGD, plays a role in mediating amino acid signaling to the mechanistic target of rapamycin complex 1 (mTORC1). RagD expression along the mammalian nephron included the thick ascending limb and the distal convoluted tubule. The identified RRAGD variants were shown to induce a constitutive activation of mTOR signaling in vitro. CONCLUSIONS: Our findings establish a novel disease, which we call autosomal dominant kidney hypomagnesemia (ADKH-RRAGD), that combines an electrolyte-losing tubulopathy and dilated cardiomyopathy. The condition is caused by variants in the RRAGD gene, which encodes Rag GTPase D; these variants lead to an activation of mTOR signaling, suggesting a critical role of Rag GTPase D for renal electrolyte handling and cardiac function.


Subject(s)
Cardiomyopathy, Dilated/genetics , Hypercalciuria/genetics , Kidney Diseases/genetics , Monomeric GTP-Binding Proteins/genetics , Mutation, Missense , Nephrocalcinosis/genetics , Renal Tubular Transport, Inborn Errors/genetics , TOR Serine-Threonine Kinases/metabolism , Cardiomyopathy, Dilated/metabolism , Female , HEK293 Cells , Humans , Hypercalciuria/metabolism , Kidney Diseases/metabolism , Kidney Tubules, Distal/metabolism , Male , Models, Molecular , Natriuresis/genetics , Nephrocalcinosis/metabolism , Pedigree , Protein Conformation , Renal Tubular Transport, Inborn Errors/metabolism , Seizures/genetics , Seizures/metabolism , Signal Transduction , Exome Sequencing , Whole Genome Sequencing
9.
EMBO Mol Med ; 13(5): e13258, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33851776

ABSTRACT

Vacuolar protein sorting 41 (VPS41) is as part of the Homotypic fusion and Protein Sorting (HOPS) complex required for lysosomal fusion events and, independent of HOPS, for regulated secretion. Here, we report three patients with compound heterozygous mutations in VPS41 (VPS41S285P and VPS41R662* ; VPS41c.1423-2A>G and VPS41R662* ) displaying neurodegeneration with ataxia and dystonia. Cellular consequences were investigated in patient fibroblasts and VPS41-depleted HeLa cells. All mutants prevented formation of a functional HOPS complex, causing delayed lysosomal delivery of endocytic and autophagic cargo. By contrast, VPS41S285P enabled regulated secretion. Strikingly, loss of VPS41 function caused a cytosolic redistribution of mTORC1, continuous nuclear localization of Transcription Factor E3 (TFE3), enhanced levels of LC3II, and a reduced autophagic response to nutrient starvation. Phosphorylation of mTORC1 substrates S6K1 and 4EBP1 was not affected. In a C. elegans model of Parkinson's disease, co-expression of VPS41S285P /VPS41R662* abolished the neuroprotective function of VPS41 against α-synuclein aggregates. We conclude that the VPS41 variants specifically abrogate HOPS function, which interferes with the TFEB/TFE3 axis of mTORC1 signaling, and cause a neurodegenerative disease.


Subject(s)
Neurodegenerative Diseases , Animals , Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Caenorhabditis elegans/genetics , HeLa Cells , Humans , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Neurodegenerative Diseases/genetics , Protein Transport , Vesicular Transport Proteins/metabolism
10.
Brain Pathol ; 31(5): e12949, 2021 09.
Article in English | MEDLINE | ID: mdl-33786950

ABSTRACT

Tuberous sclerosis complex (TSC) is a congenital disorder characterized by cortical malformations and concomitant epilepsy caused by loss-of-function mutations in the mTOR suppressors TSC1 or TSC2. While the underlying molecular changes caused by mTOR activation in TSC have previously been investigated, the drivers of these transcriptional change have not been fully elucidated. A better understanding of the perturbed transcriptional regulation could lead to the identification of novel pathways for therapeutic intervention not only in TSC, but other genetic epilepsies in which mTOR activation plays a key role, such as focal cortical dysplasia 2b (FCD). Here, we analyzed RNA sequencing data from cortical tubers and a tsc2-/- zebrafish. We identified differential expression of the transcription factors (TFs) SPI1/PU.1, IRF8, GBX2, and IKZF1 of which SPI1/PU.1 and IRF8 targets were enriched among the differentially expressed genes. Furthermore, for SPI1/PU.1 these findings were conserved in TSC zebrafish model. Next, we confirmed overexpression of SPI1/PU.1 on the RNA and protein level in a separate cohort of surgically resected TSC tubers and FCD tissue, in fetal TSC tissue, and a Tsc1GFAP-/- mouse model of TSC. Subsequently, we validated the expression of SPI1/PU.1 in dysmorphic cells with mTOR activation in TSC tubers. In fetal TSC, we detected SPI1/PU.1 expression prenatally and elevated RNA Spi1 expression in Tsc1GFAP-/- mice before the development of seizures. Finally, in vitro, we identified that in astrocytes and neurons SPI1 transcription was driven by H2 O2 -induced oxidative stress, independent of mTOR. We identified SPI1/PU.1 as a novel TF involved in the pro-inflammatory gene expression of malformed cells in TSC and FCD 2b. This transcriptional program is activated in response to oxidative stress and already present prenatally. Importantly, SPI1/PU.1 protein appears to be strictly limited to malformed cells, as we did not find SPI1/PU.1 protein expression in mice nor in our in vitro models.


Subject(s)
Oxidative Stress/physiology , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Tuberous Sclerosis Complex 2 Protein/metabolism , Tuberous Sclerosis/pathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain/metabolism , Brain/pathology , Humans , Malformations of Cortical Development/metabolism , Malformations of Cortical Development/pathology , Mice, Transgenic , Neurons/pathology , Tuberous Sclerosis/genetics , Tuberous Sclerosis/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics , Up-Regulation
11.
Rhinology ; 59(2): 114-132, 2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33760909

ABSTRACT

BACKGROUND: Viral respiratory infections are a leading cause of worldwide mortality and exert the potential to cause global socioeconomic crises. However, inexpensive, efficacious, and rapidly deployable strategies to reduce viral transmission are increasingly important in the setting of an ongoing pandemic, though not entirely understood. This article provides a comprehensive review of commonly employed nonpharmacological interventions to interrupt viral spread and provides evidence-based recommendations for their use. METHODOLOGY: A systematic review of three databases was performed. Studies with defined endpoints of subjects receiving one of five interventions (nasal washing, gargling, personal protective equipment (PPE), social distancing, and hand hygiene) were included. An evidence-based review of the highest level of evidence, with recommendations, was created in accordance with a previously described, rigorous, iterative process. RESULTS: Fifty-four primary studies were included. The most commonly studied intervention was hand hygiene, followed by PPE, gargling, saline nasal washing, and social distancing. CONCLUSIONS: Mask use and hand hygiene are strong recommendations for prevention of viral transmission. Donning gloves, gowns, and eye protection are a recommendation in healthcare settings. Saline nasal washing and gargling are options in selected populations. Although an aggregate level of evidence is not provided, the authors recommend social distancing.


Subject(s)
Personal Protective Equipment , Virus Diseases , Humans , Pandemics , Protective Clothing
12.
Biochim Biophys Acta Mol Basis Dis ; 1866(3): 165607, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31759955

ABSTRACT

Pyridox(am)ine 5'-phosphate oxidase (PNPO) catalyzes oxidation of pyridoxine 5'-phosphate (PNP) and pyridoxamine 5'-phosphate (PMP) to pyridoxal 5'-phosphate (PLP), the active form of vitamin B6. PNPO deficiency results in neonatal/infantile seizures and neurodevelopmental delay. To gain insight into this disorder we generated Pnpo deficient (pnpo-/-) zebrafish (CRISPR/Cas9 gene editing). Locomotion analysis showed that pnpo-/- zebrafish develop seizures resulting in only 38% of pnpo-/- zebrafish surviving beyond 20 days post fertilization (dpf). The age of seizure onset varied and survival after the onset was brief. Biochemical profiling at 20 dpf revealed a reduction of PLP and pyridoxal (PL) and accumulation of PMP and pyridoxamine (PM). Amino acids involved in neurotransmission including glutamate, γ-aminobutyric acid (GABA) and glycine were decreased. Concentrations of several, mostly essential, amino acids were increased in pnpo-/- zebrafish suggesting impaired activity of PLP-dependent transaminases involved in their degradation. PLP treatment increased survival at 20 dpf and led to complete normalization of PLP, PL, glutamate, GABA and glycine. However, amino acid profiles only partially normalized and accumulation of PMP and PM persisted. Taken together, our data indicate that not only decreased PLP but also accumulation of PMP may play a role in the clinical phenotype of PNPO deficiency.


Subject(s)
Brain Diseases, Metabolic/metabolism , Hypoxia-Ischemia, Brain/metabolism , Metabolic Diseases/etiology , Metabolic Diseases/metabolism , Pyridoxaminephosphate Oxidase/deficiency , Seizures/etiology , Seizures/metabolism , Zebrafish/metabolism , Amino Acids/metabolism , Animals , Brain Diseases, Metabolic/etiology , Oxidoreductases/metabolism , Pyridoxal Phosphate/analogs & derivatives , Pyridoxal Phosphate/metabolism , Pyridoxamine/metabolism , Pyridoxaminephosphate Oxidase/metabolism , Synaptic Transmission/physiology
13.
Hum Mol Genet ; 28(1): 96-104, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30239721

ABSTRACT

Loss-of-function mutations in glutaminase (GLS), the enzyme converting glutamine into glutamate, and the counteracting enzyme glutamine synthetase (GS) cause disturbed glutamate homeostasis and severe neonatal encephalopathy. We report a de novo Ser482Cys gain-of-function variant in GLS encoding GLS associated with profound developmental delay and infantile cataract. Functional analysis demonstrated that this variant causes hyperactivity and compensatory downregulation of GLS expression combined with upregulation of the counteracting enzyme GS, supporting pathogenicity. Ser482Cys-GLS likely improves the electrostatic environment of the GLS catalytic site, thereby intrinsically inducing hyperactivity. Alignment of +/-12.000 GLS protein sequences from >1000 genera revealed extreme conservation of Ser482 to the same degree as catalytic residues. Together with the hyperactivity, this indicates that Ser482 is evolutionarily preserved to achieve optimal-but submaximal-GLS activity. In line with GLS hyperactivity, increased glutamate and decreased glutamine concentrations were measured in urine and fibroblasts. In the brain (both grey and white matter), glutamate was also extremely high and glutamine was almost undetectable, demonstrated with magnetic resonance spectroscopic imaging at clinical field strength and subsequently supported at ultra-high field strength. Considering the neurotoxicity of glutamate when present in excess, the strikingly high glutamate concentrations measured in the brain provide an explanation for the developmental delay. Cataract, a known consequence of oxidative stress, was evoked in zebrafish expressing the hypermorphic Ser482Cys-GLS and could be alleviated by inhibition of GLS. The capacity to detoxify reactive oxygen species was reduced upon Ser482Cys-GLS expression, providing an explanation for cataract formation. In conclusion, we describe an inborn error of glutamate metabolism caused by a GLS hyperactivity variant, illustrating the importance of balanced GLS activity.


Subject(s)
Glutaminase/genetics , Glutaminase/physiology , Adolescent , Animals , Brain/metabolism , Cataract/genetics , Child, Preschool , Developmental Disabilities/genetics , Disease Models, Animal , Female , Fibroblasts , Gain of Function Mutation/genetics , Glutamate-Ammonia Ligase/genetics , Glutamate-Ammonia Ligase/physiology , Glutamic Acid/genetics , Glutamic Acid/metabolism , Glutamine/metabolism , HEK293 Cells , Humans , Male , Oxidative Stress , Reactive Oxygen Species/metabolism , Zebrafish
14.
JAMA Neurol ; 76(3): 342-350, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30575854

ABSTRACT

Importance: The identification and understanding of the monogenic causes of neurodevelopmental disorders are of high importance for personalized treatment and genetic counseling. Objective: To identify and characterize novel genes for a specific neurodevelopmental disorder characterized by refractory seizures, respiratory failure, brain abnormalities, and death in the neonatal period; describe the outcome of glutaminase deficiency in humans; and understand the underlying pathological mechanisms. Design, Setting, and Participants: We performed exome sequencing of cases of neurodevelopmental disorders without a clear genetic diagnosis, followed by genetic and bioinformatic evaluation of candidate variants and genes. Establishing pathogenicity of the variants was achieved by measuring metabolites in dried blood spots by a hydrophilic interaction liquid chromatography method coupled with tandem mass spectrometry. The participants are 2 families with a total of 4 children who each had lethal, therapy-refractory early neonatal seizures with status epilepticus and suppression bursts, respiratory insufficiency, simplified gyral structures, diffuse volume loss of the brain, and cerebral edema. Data analysis occurred from October 2017 to June 2018. Main Outcomes and Measures: Early neonatal epileptic encephalopathy with glutaminase deficiency and lethal outcome. Results: A total of 4 infants from 2 unrelated families, each of whom died less than 40 days after birth, were included. We identified a homozygous frameshift variant p.(Asp232Glufs*2) in GLS in the first family, as well as compound heterozygous variants p.(Gln81*) and p.(Arg272Lys) in GLS in the second family. The GLS gene encodes glutaminase (Enzyme Commission 3.5.1.2), which plays a major role in the conversion of glutamine into glutamate, the main excitatory neurotransmitter of the central nervous system. All 3 variants probably lead to a loss of function and thus glutaminase deficiency. Indeed, glutamine was increased in affected children (available z scores, 3.2 and 11.7). We theorize that the potential reduction of glutamate and the excess of glutamine were a probable cause of the described physiological and structural abnormalities of the central nervous system. Conclusions and Relevance: We identified a novel autosomal recessive neurometabolic disorder of loss of function of glutaminase that leads to lethal early neonatal encephalopathy. This inborn error of metabolism underlines the importance of GLS for appropriate glutamine homeostasis and respiratory regulation, signal transduction, and survival.


Subject(s)
Brain Diseases/genetics , Epilepsy/genetics , Glutaminase/deficiency , Mutation/genetics , Brain/metabolism , Brain Diseases/diagnosis , Epilepsy/diagnosis , Female , Glutamine/blood , Humans , Infant , Infant, Newborn , Male , Seizures/diagnosis , Seizures/genetics
15.
PLoS One ; 13(11): e0207159, 2018.
Article in English | MEDLINE | ID: mdl-30408122

ABSTRACT

Apico-basal polarity establishment is a seminal process in tissue morphogenesis. To function properly it is often imperative that epithelial cells limit apical membrane formation to a single domain. We previously demonstrated that signaling by the small GTPase Cdc42, together with its guanine nucleotide exchange factor (GEF) Tuba, is required to prevent the formation of multiple apical domains in polarized Ls174T:W4 cells, a single cell model for enterocyte polarization. To further chart the molecular signaling mechanisms that safeguard singularity during enterocyte polarization we generated knockout cells for the Cdc42 effector protein Par6A. Par6A loss results in the formation of multiple apical domains, similar to loss of Cdc42. In Par6A knockout cells, we find that active Cdc42 is more mobile at the apical membrane compared to control cells and that wild type Cdc42 is more diffusely localized throughout the cell, indicating that Par6A is required to restrict Cdc42 signaling. Par6A, Cdc42 and its GEF Tuba bind in a co-immunoprecipitation experiment and they partially colocalize at the apical membrane in polarized Ls174T:W4 cells, suggesting the formation of a trimeric complex. Indeed, in a rescue experiment using Par6A mutants, we show that the ability to establish this trimeric complex correlates with the ability to restore singularity in Par6A knockout cells. Together, these experiments therefore indicate that a Tuba/Cdc42/Par6A complex is required to ensure the formation of a single apical domain during enterocyte polarization.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Polarity/physiology , Cytoskeletal Proteins/metabolism , Enterocytes/cytology , Enterocytes/metabolism , Guanine Nucleotide Exchange Factors/metabolism , cdc42 GTP-Binding Protein/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Cell Line , Cell Polarity/genetics , Cytoskeletal Proteins/chemistry , Gene Knockout Techniques , Guanine Nucleotide Exchange Factors/chemistry , Humans , Microvilli/metabolism , Microvilli/ultrastructure , Protein Structure, Quaternary , Signal Transduction , cdc42 GTP-Binding Protein/chemistry
16.
Chem Commun (Camb) ; 54(37): 4689-4691, 2018 May 07.
Article in English | MEDLINE | ID: mdl-29676428

ABSTRACT

Four variants of a synthetic chloride anion channel composed of dialkyamines linked to a heptapeptide have been studied in different lipid and polymer membranes using electrophysiological techniques. For the first time, the values of conductance states, voltage gating properties, and qualitative state transition frequency were measured in different lipid systems.

17.
Mol Cell Biol ; 38(12)2018 06 15.
Article in English | MEDLINE | ID: mdl-29581186

ABSTRACT

PTEN is a tumor suppressor that is frequently lost in epithelial malignancies. A part of the tumor-suppressive properties of PTEN is attributed to its function in cell polarization and consequently its role in maintaining epithelial tissue integrity. However, surprisingly little is known about the function and regulation of PTEN during epithelial cell polarization. We used clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9-mediated gene disruption to delete PTEN in intestinal epithelial Ls174T:W4 cells, which upon differentiation form a microvillus-covered apical membrane (brush border) on a part of the cell cortex, independent of cell-cell junctions. We show that loss of PTEN results in the formation of a larger brush border that, in a fraction of the cells, even spans the entire plasma membrane, revealing that PTEN functions in the regulation of apical membrane size. Depletion of the phosphatase PTPL1 resulted in a similar defect. PTPL1 interacts with PTEN, and this interaction is necessary for apical membrane enrichment of PTEN. Importantly, phosphatase activity of PTPL1 is not required, indicating that PTPL1 functions as an anchor protein in this process. Our work thus demonstrates a novel function for PTEN during cell polarization in controlling apical membrane size and identifies PTPL1 as a critical apical membrane anchor for PTEN in this process.


Subject(s)
Cell Membrane/metabolism , Cell Polarity/physiology , Microvilli/metabolism , PTEN Phosphohydrolase/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 13/genetics , Animals , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Epithelial Cells/physiology , Gene Knockout Techniques , HEK293 Cells , Humans , Male , Mice , Microvilli/genetics , Neoplasms/pathology , PTEN Phosphohydrolase/genetics
18.
Mol Cell Biol ; 37(7)2017 04 01.
Article in English | MEDLINE | ID: mdl-28069739

ABSTRACT

Signaling by the small GTPase Cdc42 governs a diverse set of cellular processes that contribute to tissue morphogenesis. Since these processes often require highly localized signaling, Cdc42 activity must be clustered in order to prevent ectopic signaling. During cell polarization, apical Cdc42 signaling directs the positioning of the nascent apical membrane. However, the molecular mechanisms that drive Cdc42 clustering during polarity establishment are largely unknown. Here, we demonstrate that during cell polarization localized Cdc42 signaling is enabled via activity-dependent control of Cdc42 mobility. By performing photoconversion experiments, we show that inactive Cdc42-GDP is 30-fold more mobile than active Cdc42-GTP. This switch in apical mobility originates from a dual mechanism involving RhoGDI-mediated membrane dissociation of Cdc42-GDP and Tuba-mediated immobilization of Cdc42-GTP. Interference with either mechanism affects Cdc42 clustering and as a consequence impairs Cdc42-mediated apical membrane clustering. We therefore identify a molecular network, comprised of Cdc42, the guanine nucleotide exchange factor (GEF) Tuba, and RhoGDI, that enables differential diffusion of inactive and active Cdc42 and is required to establish localized Cdc42 signaling during enterocyte polarization.


Subject(s)
Cell Polarity , Enterocytes/cytology , Enterocytes/metabolism , Signal Transduction , cdc42 GTP-Binding Protein/metabolism , Cell Line , Cell Membrane/metabolism , HEK293 Cells , Humans , Microvilli/metabolism , Protein Binding , Tubulin/metabolism , rho-Specific Guanine Nucleotide Dissociation Inhibitors
19.
Langmuir ; 33(5): 1171-1179, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28040902

ABSTRACT

This work explores methods for forming and characterizing biomimetic planar membranes composed of amphiphilic block copolymers. The membranes are diblocks and triblocks with hydrophilic blocks of poly(2-methyl-2-oxazoline) (PMOXA) and hydrophobic blocks of poly(dimethylsiloxane) (PDMS). Experiments with the lipid diphytanoyl phosphocholine (DPhPC) serve as a basis for comparison with the polymeric membranes. Phase-contrast microscopy is used to study how membranes evolve over time after their formation. Capacitance measurements as a function of the thinned membrane area (prepared from two separate solvent systems) are performed to clarify the importance of the Plateau-Gibbs border in electrical measurements. Finally, functional reconstitution of the two ion channels, alamethicin and gramicidin, is investigated. Imaging in transmitted phase-contrast mode provides visualization of thinned regions that contain monolayers or bilayers (in the case of diblock copolymer). The specific capacitance measurements yield 0.28 µF/cm2 with a corresponding thickness of 8.5 nm for PMOXA6-PDMS35-PMOXA6 (blocks of 6 PMOXA and 35 PDMS repeat units) formed from a solution of ethanol-decane, 0.55 µF/cm2 and 4.4 nm in chloroform-toluene, and 0.46 µF/cm2 and 5.4 nm for the diblock PMOXA6-PDMS17 in ethanol-decane. Alamethicin reconstitution in the block copolymers shows slower channel-forming kinetics with somewhat higher conductance values than found in DPhPC. Gramicidin in the block copolymer shows a slightly voltage-dependent conductance as a function of time, with little stochastic conductance state switching, in contrast to reconstitution in DPhPC where gramicidin switches states at ∼3 Hz.

20.
J Biomol Struct Dyn ; 34(3): 529-39, 2016.
Article in English | MEDLINE | ID: mdl-26046587

ABSTRACT

All-atom molecular dynamics is used to investigate the transport of Na(+) across a 1,2-dioleoyl-sn-glycero-3-phosphocholine lipid bilayer facilitated by a diazacrown hydraphile. Specifically, the free energy of Na(+) passing through the bilayer is calculated using the adaptive biasing force method to study the free energy associated with the increase in Na(+) transport in the presence of the hydraphile molecule. The results show that water interaction greatly influences Na(+) transport through the lipid bilayer as water is pulled through the bilayer with Na(+) forming a water channel. The hydraphile causes a reduction in the free energy barrier for the transport of Na(+) through the head group part of the lipid bilayer since it complexes the Na(+) reducing the necessity for water to be complexed and, therefore, dragged through with Na(+), an energetically unfavorable process. The free energy associated with Na(+) being desolvated within the bilayer is significantly decreased in the presence of the hydraphile molecule; the hydraphile increases the number of solvation states of Na(+) that can be adopted, and this increase in the number of available configurations provides an entropic explanation for the success of the hydraphile.


Subject(s)
Lipid Bilayers/chemistry , Molecular Dynamics Simulation , Sodium Channels/chemistry , Hydrophobic and Hydrophilic Interactions , Ions/chemistry , Molecular Conformation , Quantum Theory , Sodium/chemistry , Solvents/chemistry , Water/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...