Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 301
Filter
1.
Nature ; 626(8001): 1042-1048, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38418917

ABSTRACT

The loss of the tail is among the most notable anatomical changes to have occurred along the evolutionary lineage leading to humans and to the 'anthropomorphous apes'1-3, with a proposed role in contributing to human bipedalism4-6. Yet, the genetic mechanism that facilitated tail-loss evolution in hominoids remains unknown. Here we present evidence that an individual insertion of an Alu element in the genome of the hominoid ancestor may have contributed to tail-loss evolution. We demonstrate that this Alu element-inserted into an intron of the TBXT gene7-9-pairs with a neighbouring ancestral Alu element encoded in the reverse genomic orientation and leads to a hominoid-specific alternative splicing event. To study the effect of this splicing event, we generated multiple mouse models that express both full-length and exon-skipped isoforms of Tbxt, mimicking the expression pattern of its hominoid orthologue TBXT. Mice expressing both Tbxt isoforms exhibit a complete absence of the tail or a shortened tail depending on the relative abundance of Tbxt isoforms expressed at the embryonic tail bud. These results support the notion that the exon-skipped transcript is sufficient to induce a tail-loss phenotype. Moreover, mice expressing the exon-skipped Tbxt isoform develop neural tube defects, a condition that affects approximately 1 in 1,000 neonates in humans10. Thus, tail-loss evolution may have been associated with an adaptive cost of the potential for neural tube defects, which continue to affect human health today.


Subject(s)
Alternative Splicing , Evolution, Molecular , Hominidae , T-Box Domain Proteins , Tail , Animals , Humans , Mice , Alternative Splicing/genetics , Alu Elements/genetics , Disease Models, Animal , Genome/genetics , Hominidae/anatomy & histology , Hominidae/genetics , Introns/genetics , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Phenotype , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Tail/anatomy & histology , Tail/embryology , Exons/genetics
2.
Int J Mol Sci ; 22(20)2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34681657

ABSTRACT

BACKGROUND: The transient receptor potential ankyrin 1 (TRPA1) cation channels function as broadly-tuned sensors of noxious chemicals in many species. Recent studies identified four functional TRPA1 isoforms in Drosophila melanogaster (dTRPA1(A) to (D)), but their responses to non-electrophilic chemicals are yet to be fully characterized. METHODS: We determined the behavioral responses of adult flies to the mammalian TRPA1 non-electrophilic activators citronellal and menthol, and characterized the effects of these compounds on all four dTRPA1 channel isoforms using intracellular Ca2+ imaging and whole-cell patch-clamp recordings. RESULTS: Wild type flies avoided citronellal and menthol in an olfactory test and this behavior was reduced in dTrpA1 mutant flies. Both compounds activate all dTRPA1 isoforms in the heterologous expression system HEK293T, with the following sensitivity series: dTRPA1(C) = dTRPA1(D) > dTRPA1(A) ≫ dTRPA1(B) for citronellal and dTRPA1(A) > dTRPA1(D) > dTRPA1(C) > dTRPA1(B) for menthol. CONCLUSIONS: dTrpA1 was required for the normal avoidance of Drosophila melanogaster towards citronellal and menthol. All dTRPA1 isoforms are activated by both compounds, but the dTRPA1(B) is consistently the least sensitive. We discuss how these findings may guide further studies on the physiological roles and the structural bases of chemical sensitivity of TRPA1 channels.


Subject(s)
Acyclic Monoterpenes/pharmacology , Aldehydes/pharmacology , Drosophila Proteins/metabolism , Drosophila melanogaster/drug effects , Menthol/pharmacology , TRPA1 Cation Channel/metabolism , Animals , Animals, Genetically Modified/metabolism , Calcium/metabolism , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Female , HEK293 Cells , Humans , Insect Repellents/pharmacology , Male , Patch-Clamp Techniques , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , TRPA1 Cation Channel/deficiency , TRPA1 Cation Channel/genetics
3.
Am J Physiol Cell Physiol ; 321(3): C607-C614, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34378992

ABSTRACT

Bovine milk exosomes (BMEs) are being explored in drug delivery despite their rapid elimination by macrophages. We aimed at identifying the BME transporter in murine bone marrow-derived macrophages (BMDMs). Fluorophore-labeled BMEs were used in transport studies in BMDMs from C57BL/6J and class A scavenger receptor type 1/2 (CASR-1/2) knockout mice and tissue accumulation in macrophage-depleted C57BL/6J mice. Parametric and nonparametric statistics tests for pairwise and multiple comparisons were used. Chemical inhibitors of phagocytosis by cytochalasin D led to a 69 ± 18% decrease in BME uptake compared with controls (P < 0.05), whereas inhibitors of endocytic pathways other than phagocytosis had a modest effect on uptake (P > 0.05). Inhibitors of class A scavenger receptors (CASRs) including CASR-1/2 caused a 70% decrease in BME uptake (P < 0.05). The uptake of BMEs by BMDMs from CASR-1/2 knockout mice was smaller by 58 ± 23% compared with wild-type controls (P < 0.05). Macrophage depletion by clodronate caused a more than 44% decrease in BME uptake in the spleen and lungs (P < 0.05), whereas the decrease observed in liver was not statistically significant. In conclusion, CASR-1/2 facilitates the uptake of BMEs in BMDMs and C57BL/6J mice.


Subject(s)
Exosomes/metabolism , Macrophages/metabolism , Milk/chemistry , Scavenger Receptors, Class A/genetics , Animals , Cattle , Clodronic Acid/pharmacology , Cytochalasin D/pharmacology , Endocytosis/drug effects , Exosomes/chemistry , Female , Fluorescent Dyes/chemistry , Gene Expression , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Macrophages/cytology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/drug effects , Protein Isoforms/deficiency , Protein Isoforms/genetics , Scavenger Receptors, Class A/deficiency , Spleen/drug effects , Spleen/metabolism , Staining and Labeling/methods
5.
Int J Mol Sci ; 22(11)2021 May 30.
Article in English | MEDLINE | ID: mdl-34070744

ABSTRACT

The ClC-2 channel plays a critical role in maintaining ion homeostasis in the brain and the testis. Loss-of-function mutations in the ClC-2-encoding human CLCN2 gene are linked to the white matter disease leukodystrophy. Clcn2-deficient mice display neuronal myelin vacuolation and testicular degeneration. Leukodystrophy-causing ClC-2 mutant channels are associated with anomalous proteostasis manifesting enhanced endoplasmic reticulum (ER)-associated degradation. The molecular nature of the ER quality control system for ClC-2 protein remains elusive. In mouse testicular tissues and Leydig cells, we demonstrated that endogenous ClC-2 co-existed in the same protein complex with the molecular chaperones heat shock protein 90ß (Hsp90ß) and heat shock cognate protein (Hsc70), as well as the associated co-chaperones Hsp70/Hsp90 organizing protein (HOP), activator of Hsp90 ATPase homolog 1 (Aha1), and FK506-binding protein 8 (FKBP8). Further biochemical analyses revealed that the Hsp90ß-Hsc70 chaperone/co-chaperone system promoted mouse and human ClC-2 protein biogenesis. FKBP8 additionally facilitated membrane trafficking of ClC-2 channels. Interestingly, treatment with the Hsp90-targeting small molecule 17-allylamino-17-demethoxygeldanamycin (17-AAG) substantially boosted ClC-2 protein expression. Also, 17-AAG effectively increased both total and cell surface protein levels of leukodystrophy-causing loss-of-function ClC-2 mutant channels. Our findings highlight the therapeutic potential of 17-AAG in correcting anomalous ClC-2 proteostasis associated with leukodystrophy.


Subject(s)
Brain/metabolism , Chloride Channels/genetics , Leydig Cells/metabolism , Neurons/metabolism , Pelizaeus-Merzbacher Disease/genetics , Proteostasis/genetics , Animals , Benzoquinones/pharmacology , Brain/drug effects , Brain/pathology , CHO Cells , CLC-2 Chloride Channels , Chloride Channels/deficiency , Cricetulus , Disease Models, Animal , Endoplasmic Reticulum-Associated Degradation/drug effects , Gene Expression Regulation , HEK293 Cells , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Lactams, Macrocyclic/pharmacology , Leydig Cells/drug effects , Leydig Cells/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Neurons/drug effects , Neurons/pathology , Pelizaeus-Merzbacher Disease/drug therapy , Pelizaeus-Merzbacher Disease/metabolism , Pelizaeus-Merzbacher Disease/pathology , Protein Isoforms/deficiency , Protein Isoforms/genetics , Signal Transduction , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism
6.
PLoS Genet ; 17(6): e1009603, 2021 06.
Article in English | MEDLINE | ID: mdl-34143769

ABSTRACT

The inability to maintain a strictly regulated endo(lyso)somal acidic pH through the proton-pumping action of the vacuolar-ATPases (v-ATPases) has been associated with various human diseases including heritable connective tissue disorders. Autosomal recessive (AR) cutis laxa (CL) type 2C syndrome is associated with genetic defects in the ATP6V1E1 gene and is characterized by skin wrinkles or loose redundant skin folds with pleiotropic systemic manifestations. The underlying pathological mechanisms leading to the clinical presentations remain largely unknown. Here, we show that loss of atp6v1e1b in zebrafish leads to early mortality, associated with craniofacial dysmorphisms, vascular anomalies, cardiac dysfunction, N-glycosylation defects, hypotonia, and epidermal structural defects. These features are reminiscent of the phenotypic manifestations in ARCL type 2C patients. Our data demonstrates that loss of atp6v1e1b alters endo(lyso)somal protein levels, and interferes with non-canonical v-ATPase pathways in vivo. In order to gain further insights into the processes affected by loss of atp6v1e1b, we performed an untargeted analysis of the transcriptome, metabolome, and lipidome in early atp6v1e1b-deficient larvae. We report multiple affected pathways including but not limited to oxidative phosphorylation, sphingolipid, fatty acid, and energy metabolism together with profound defects on mitochondrial respiration. Taken together, our results identify complex pathobiological effects due to loss of atp6v1e1b in vivo.


Subject(s)
Abnormalities, Multiple/genetics , Cutis Laxa/genetics , Epithelial Cells/metabolism , Skin/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Zebrafish Proteins/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Animals , Cutis Laxa/metabolism , Cutis Laxa/pathology , Disease Models, Animal , Endosomes/metabolism , Endosomes/pathology , Epithelial Cells/pathology , Gene Expression Regulation , Humans , Larva/genetics , Larva/growth & development , Larva/metabolism , Lipidomics , Longevity/genetics , Lysosomes/metabolism , Lysosomes/pathology , Metabolome/genetics , Mitochondria/metabolism , Mitochondria/pathology , Oxidative Phosphorylation , Protein Isoforms/deficiency , Protein Isoforms/genetics , Skin/pathology , Syndrome , Transcriptome , Vacuolar Proton-Translocating ATPases/deficiency , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/deficiency
7.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Article in English | MEDLINE | ID: mdl-33972431

ABSTRACT

Febrile seizures (FSs) are the most common convulsion in infancy and childhood. Considering the limitations of current treatments, it is important to examine the mechanistic cause of FSs. Prompted by a genome-wide association study identifying TMEM16C (also known as ANO3) as a risk factor of FSs, we showed previously that loss of TMEM16C function causes hippocampal neuronal hyperexcitability [Feenstra et al., Nat. Genet. 46, 1274-1282 (2014)]. Our previous study further revealed a reduction in the number of warm-sensitive neurons that increase their action potential firing rate with rising temperature of the brain region harboring these hypothalamic neurons. Whereas central neuronal hyperexcitability has been implicated in FSs, it is unclear whether the maximal temperature reached during fever or the rate of body temperature rise affects FSs. Here we report that mutant rodent pups with TMEM16C eliminated from all or a subset of their central neurons serve as FS models with deficient thermoregulation. Tmem16c knockout (KO) rat pups at postnatal day 10 (P10) are more susceptible to hyperthermia-induced seizures. Moreover, they display a more rapid rise of body temperature upon heat exposure. In addition, conditional knockout (cKO) mouse pups (P11) with TMEM16C deletion from the brain display greater susceptibility of hyperthermia-induced seizures as well as deficiency in thermoregulation. We also found similar phenotypes in P11 cKO mouse pups with TMEM16C deletion from Ptgds-expressing cells, including temperature-sensitive neurons in the preoptic area (POA) of the anterior hypothalamus, the brain region that controls body temperature. These findings suggest that homeostatic thermoregulation plays an important role in FSs.


Subject(s)
Body Temperature Regulation/genetics , Chloride Channels/genetics , Fever/genetics , Hyperthermia/genetics , Preoptic Area/metabolism , Seizures, Febrile/genetics , Action Potentials/physiology , Animals , Animals, Newborn , Body Temperature/drug effects , Body Temperature/physiology , Chloride Channels/deficiency , Female , Fever/chemically induced , Fever/metabolism , Fever/physiopathology , Gene Expression , Hippocampus/metabolism , Hippocampus/physiopathology , Hyperthermia/metabolism , Hyperthermia/physiopathology , Kainic Acid/administration & dosage , Male , Mice , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Preoptic Area/physiopathology , Protein Isoforms/deficiency , Protein Isoforms/genetics , Rats , Seizures, Febrile/chemically induced , Seizures, Febrile/metabolism , Seizures, Febrile/physiopathology
8.
Cell Rep ; 35(2): 108981, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852861

ABSTRACT

Despite the central role of chromosomal context in gene transcription, human noncoding DNA variants are generally studied outside of their genomic location. This limits our understanding of disease-causing regulatory variants. INS promoter mutations cause recessive neonatal diabetes. We show that all INS promoter point mutations in 60 patients disrupt a CC dinucleotide, whereas none affect other elements important for episomal promoter function. To model CC mutations, we humanized an ∼3.1-kb region of the mouse Ins2 gene. This recapitulated developmental chromatin states and cell-specific transcription. A CC mutant allele, however, abrogated active chromatin formation during pancreas development. A search for transcription factors acting through this element revealed that another neonatal diabetes gene product, GLIS3, has a pioneer-like ability to derepress INS chromatin, which is hampered by the CC mutation. Our in vivo analysis, therefore, connects two human genetic defects in an essential mechanism for developmental activation of the INS gene.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/genetics , Diabetes Mellitus/genetics , Insulin/genetics , Pancreas/metabolism , Point Mutation , Repressor Proteins/genetics , Trans-Activators/genetics , Alleles , Animals , Chromatin/chemistry , Chromatin/pathology , DNA-Binding Proteins/deficiency , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Embryo, Mammalian , Gene Expression Regulation, Developmental , Humans , Infant, Newborn , Infant, Newborn, Diseases , Insulin/deficiency , Mice , Mice, Transgenic , Pancreas/growth & development , Pancreas/pathology , Promoter Regions, Genetic , Protein Isoforms/deficiency , Protein Isoforms/genetics , Repressor Proteins/deficiency , Trans-Activators/deficiency , Transcription, Genetic
9.
Cell Rep ; 35(3): 109015, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33882302

ABSTRACT

Osteoblasts orchestrate bone formation through the secretion of type I collagen and other constituents of the matrix on which hydroxyapatite crystals mineralize. Here, we show that TENT5A, whose mutations were found in congenital bone disease osteogenesis imperfecta patients, is a cytoplasmic poly(A) polymerase playing a crucial role in regulating bone mineralization. Direct RNA sequencing revealed that TENT5A is induced during osteoblast differentiation and polyadenylates mRNAs encoding Col1α1, Col1α2, and other secreted proteins involved in osteogenesis, increasing their expression. We postulate that TENT5A, possibly together with its paralog TENT5C, is responsible for the wave of cytoplasmic polyadenylation of mRNAs encoding secreted proteins occurring during bone mineralization. Importantly, the Tent5a knockout (KO) mouse line displays bone fragility and skeletal hypomineralization phenotype resulting from quantitative and qualitative collagen defects. Thus, we report a biologically relevant posttranscriptional regulator of collagen production and, more generally, bone formation.


Subject(s)
Calcification, Physiologic/genetics , Osteoblasts/metabolism , Osteogenesis Imperfecta/genetics , Osteogenesis/genetics , Polynucleotide Adenylyltransferase/genetics , RNA, Messenger/genetics , Animals , Cell Differentiation , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain/genetics , Collagen Type I, alpha 1 Chain/metabolism , Disease Models, Animal , Eye Proteins/genetics , Eye Proteins/metabolism , Female , Gene Expression Regulation, Developmental , Humans , Male , Mice , Mice, Knockout , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Osteoblasts/pathology , Osteogenesis Imperfecta/metabolism , Osteogenesis Imperfecta/pathology , Osteonectin/genetics , Osteonectin/metabolism , Polyadenylation , Polynucleotide Adenylyltransferase/metabolism , Protein Isoforms/deficiency , Protein Isoforms/genetics , RNA, Messenger/metabolism , Sequence Analysis, RNA , Serpins/genetics , Serpins/metabolism , Signal Transduction
10.
Cells ; 10(3)2021 03 03.
Article in English | MEDLINE | ID: mdl-33802526

ABSTRACT

Vertebrate postembryonic development is regulated by thyroid hormone (T3). Of particular interest is anuran metamorphosis, which offers several unique advantages for studying the role of T3 and its two nuclear receptor genes, TRα and TRß, during postembryonic development. We have recently generated TR double knockout (TRDKO) Xenopus tropicalis animals and reported that TR is essential for the completion of metamorphosis. Furthermore, TRDKO tadpoles are stalled at the climax of metamorphosis before eventual death. Here we show that TRDKO intestine lacked larval epithelial cell death and adult stem cell formation/proliferation during natural metamorphosis. Interestingly, TRDKO tadpole intestine had premature formation of adult-like epithelial folds and muscle development. In addition, T3 treatment of premetamorphic TRDKO tadpoles failed to induce any metamorphic changes in the intestine. Furthermore, RNA-seq analysis revealed that TRDKO altered the expression of many genes in biological pathways such as Wnt signaling and the cell cycle that likely underlay the inhibition of larval epithelial cell death and adult stem cell development caused by removing both TR genes. Our data suggest that liganded TR is required for larval epithelial cell degeneration and adult stem cell formation, whereas unliganded TR prevents precocious adult tissue morphogenesis such as smooth-muscle development and epithelial folding.


Subject(s)
Adult Stem Cells/metabolism , Amphibian Proteins/genetics , Epithelial Cells/metabolism , Intestines/cytology , Larva/genetics , Receptors, Thyroid Hormone/genetics , Thyroid Hormones/genetics , Xenopus/genetics , Adult Stem Cells/cytology , Adult Stem Cells/drug effects , Amphibian Proteins/classification , Amphibian Proteins/metabolism , Animals , Animals, Genetically Modified , Apoptosis/genetics , Cell Cycle/genetics , Cell Differentiation/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Gene Ontology , Gene Regulatory Networks , Intestines/drug effects , Intestines/growth & development , Larva/cytology , Larva/drug effects , Larva/growth & development , Metabolic Networks and Pathways/genetics , Metamorphosis, Biological , Molecular Sequence Annotation , Protein Isoforms/deficiency , Protein Isoforms/genetics , Receptors, Thyroid Hormone/deficiency , Thyroid Hormones/metabolism , Thyroid Hormones/pharmacology , Wnt Signaling Pathway/genetics , Xenopus/growth & development , Xenopus/metabolism
11.
Nat Commun ; 12(1): 833, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33547280

ABSTRACT

The structure of proline prevents it from adopting an optimal position for rapid protein synthesis. Poly-proline-tract (PPT) associated ribosomal stalling is resolved by highly conserved eIF5A, the only protein to contain the amino acid hypusine. We show that de novo heterozygous EIF5A variants cause a disorder characterized by variable combinations of developmental delay, microcephaly, micrognathia and dysmorphism. Yeast growth assays, polysome profiling, total/hypusinated eIF5A levels and PPT-reporters studies reveal that the variants impair eIF5A function, reduce eIF5A-ribosome interactions and impair the synthesis of PPT-containing proteins. Supplementation with 1 mM spermidine partially corrects the yeast growth defects, improves the polysome profiles and restores expression of PPT reporters. In zebrafish, knockdown eif5a partly recapitulates the human phenotype that can be rescued with 1 µM spermidine supplementation. In summary, we uncover the role of eIF5A in human development and disease, demonstrate the mechanistic complexity of EIF5A-related disorder and raise possibilities for its treatment.


Subject(s)
Developmental Disabilities/genetics , Gene Expression Regulation, Developmental , Microcephaly/genetics , Micrognathism/genetics , Peptide Initiation Factors/genetics , RNA-Binding Proteins/genetics , Adolescent , Amino Acid Sequence , Animals , Child , Developmental Disabilities/metabolism , Developmental Disabilities/pathology , Embryo, Nonmammalian , Female , Humans , Lysine/analogs & derivatives , Lysine/genetics , Lysine/metabolism , Male , Microcephaly/metabolism , Microcephaly/pathology , Micrognathism/metabolism , Micrognathism/pathology , Peptide Initiation Factors/deficiency , Peptides/genetics , Peptides/metabolism , Protein Biosynthesis , Protein Conformation , Protein Isoforms/deficiency , Protein Isoforms/genetics , Ribosomes/genetics , Ribosomes/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Spermidine/pharmacology , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Eukaryotic Translation Initiation Factor 5A
12.
FASEB J ; 35(2): e21224, 2021 02.
Article in English | MEDLINE | ID: mdl-33236473

ABSTRACT

GABA is synthesized by glutamate decarboxylase (GAD), which has two isoforms, namely, GAD65 and GAD67, encoded by the Gad2 and Gad1 genes, respectively. GAD65-deficient (Gad2-/- ) mice exhibit a reduction in brain GABA content after 1 month of age and show spontaneous seizures in adulthood. Approximately 25% of Gad2-/- mice died by 6 months of age. Our Western blot analysis demonstrated that the protein expression ratio of GAD65 to GAD67 in the brain was greater in rats than in mice during postnatal development, suggesting that the contribution of each GAD isoform to GABA functions differs between these two species. To evaluate whether GAD65 deficiency causes different phenotypes between rats and mice, we generated Gad2-/- rats using TALEN genome editing technology. Western blot and immunohistochemical analyses with new antibodies demonstrated that the GAD65 protein was undetectable in the Gad2-/- rat brain. Gad2-/- pups exhibited spontaneous seizures and paroxysmal discharge in EEG at postnatal weeks 3-4. More than 80% of the Gad2-/- rats died at postnatal days (PNDs) 17-23. GABA content in Gad2-/- brains was significantly lower than those in Gad2+/- and Gad2+/+ brains at PND17-19. These results suggest that the low levels of brain GABA content in Gad2-/- rats may lead to epilepsy followed by premature death, and that Gad2-/- rats are more severely affected than Gad2-/- mice. Considering that the GAD65/GAD67 ratio in human brains is more similar to that in rat brains than in mouse brains, Gad2-/- rats would be useful for further investigating the roles of GAD65 in vivo.


Subject(s)
Epilepsy/genetics , Glutamate Decarboxylase/genetics , Animals , Brain/metabolism , Brain/physiopathology , Epilepsy/metabolism , Glutamate Decarboxylase/deficiency , Glutamate Decarboxylase/metabolism , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Rats, Long-Evans , Receptors, GABA/metabolism , Synaptic Potentials , gamma-Aminobutyric Acid/metabolism
13.
Cell ; 182(2): 404-416.e14, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32610081

ABSTRACT

Problems arising during translation of mRNAs lead to ribosome stalling and collisions that trigger a series of quality control events. However, the global cellular response to ribosome collisions has not been explored. Here, we uncover a function for ribosome collisions in signal transduction. Using translation elongation inhibitors and general cellular stress conditions, including amino acid starvation and UV irradiation, we show that ribosome collisions activate the stress-activated protein kinase (SAPK) and GCN2-mediated stress response pathways. We show that the MAPKKK ZAK functions as the sentinel for ribosome collisions and is required for immediate early activation of both SAPK (p38/JNK) and GCN2 signaling pathways. Selective ribosome profiling and biochemistry demonstrate that although ZAK generally associates with elongating ribosomes on polysomal mRNAs, it specifically auto-phosphorylates on the minimal unit of colliding ribosomes, the disome. Together, these results provide molecular insights into how perturbation of translational homeostasis regulates cell fate.


Subject(s)
Ribosomes/metabolism , Stress, Physiological , ATP-Binding Cassette Transporters/metabolism , Anisomycin/pharmacology , Apoptosis/drug effects , DNA Damage/radiation effects , Enzyme Activation , Humans , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/metabolism , Phosphorylation , Polyribosomes/metabolism , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Signal Transduction , Ultraviolet Rays , eIF-2 Kinase/metabolism
14.
Infect Immun ; 88(9)2020 08 19.
Article in English | MEDLINE | ID: mdl-32631916

ABSTRACT

Enteric pathogens exploit chemical and nutrient signaling to gauge their location within a host and control expression of traits important for infection. Ethanolamine-containing molecules are essential in host physiology and play important roles in intestinal processes. The transcription factor EutR is conserved in the Enterobacteriaceae and is required for ethanolamine sensing and metabolism. In enterohemorrhagic Escherichia coli (EHEC) O157:H7, EutR responds to ethanolamine to activate expression of traits required for host colonization and disease; however, the importance of EutR to EHEC intestinal infection has not been examined. Because EHEC does not naturally colonize or cause disease in mice, we employed the natural murine pathogen Citrobacter rodentium as a model of EHEC virulence to investigate the importance of EutR in vivo EHEC and C. rodentium possess the locus of enterocyte effacement (LEE), which is the canonical virulence trait of attaching and effacing pathogens. Our findings demonstrate that ethanolamine sensing and EutR-dependent regulation of the LEE are conserved in C. rodentium Moreover, during infection, EutR is required for maximal LEE expression, colonization, and transmission efficiency. These findings reveal that EutR not only is important for persistence during the primary host infection cycle but also is required for maintenance in a host population.


Subject(s)
Citrobacter rodentium/genetics , Enterobacteriaceae Infections/microbiology , Enterohemorrhagic Escherichia coli/genetics , Escherichia coli Proteins/genetics , Ethanolamine/metabolism , Gene Expression Regulation, Bacterial , Phosphoproteins/genetics , Transcription Factors/genetics , Animals , Citrobacter rodentium/pathogenicity , Colony Count, Microbial , Conserved Sequence , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/pathology , Enterobacteriaceae Infections/transmission , Enterocytes/microbiology , Enterocytes/pathology , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Proteins/metabolism , Female , Host Microbial Interactions/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/metabolism , Protein Isoforms/deficiency , Protein Isoforms/genetics , Signal Transduction , Transcription Factors/deficiency , Virulence
15.
Mol Metab ; 34: 72-84, 2020 04.
Article in English | MEDLINE | ID: mdl-32180561

ABSTRACT

OBJECTIVE: The liver is regularly exposed to changing metabolic and inflammatory environments. It must sense and adapt to metabolic need while balancing resources required to protect itself from insult. Peroxisome proliferator activated receptor gamma coactivator-1 alpha (PGC-1α) is a transcriptional coactivator expressed as multiple, alternatively spliced variants transcribed from different promoters that coordinate metabolic adaptation and protect against inflammation. It is not known how PGC-1α integrates extracellular signals to balance metabolic and anti-inflammatory outcomes. METHODS: Primary mouse hepatocytes were used to evaluate the role(s) of different PGC-1α proteins in regulating hepatic metabolism and inflammatory signaling downstream of tumor necrosis factor alpha (TNFα). Gene expression and signaling analysis were combined with biochemical measurement of apoptosis using gain- and loss-of-function in vitro and in vivo. RESULTS: Hepatocytes expressed multiple isoforms of PGC-1α, including PGC-1α4, which microarray analysis showed had common and isoform-specific functions linked to metabolism and inflammation compared with canonical PGC-1α1. Whereas PGC-1α1 primarily impacted gene programs of nutrient metabolism and mitochondrial biology, TNFα signaling showed several pathways related to innate immunity and cell death downstream of PGC-1α4. Gain- and loss-of-function models illustrated that PGC-1α4 uniquely enhanced expression of anti-apoptotic gene programs and attenuated hepatocyte apoptosis in response to TNFα or lipopolysaccharide (LPS). This was in contrast to PGC-1α1, which decreased the expression of a wide inflammatory gene network but did not prevent hepatocyte death in response to cytokines. CONCLUSIONS: PGC-1α variants have distinct, yet complementary roles in hepatic responses to metabolism and inflammation, and we identify PGC-1α4 as an important mitigator of apoptosis.


Subject(s)
Apoptosis , Hepatocytes/metabolism , Inflammation/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Animals , Cell Line , Female , Hepatocytes/pathology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/deficiency , Protein Isoforms/deficiency , Protein Isoforms/metabolism
16.
Mol Brain ; 13(1): 7, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31959219

ABSTRACT

Clustered protocadherins (Pcdhs), a large group of adhesion molecules, are important for axonal projections and dendritic spread, but little is known about how they influence neuronal activity. The Pcdhß cluster is strongly expressed in the hippocampus, and in vivo Ca2+ imaging in Pcdhß-deficient mice revealed altered activity of neuronal ensembles but not of individual cells in this region in freely moving animals. Specifically, Pcdhß deficiency increased the number of large-size neuronal ensembles and the proportion of cells shared between ensembles. Furthermore, Pcdhß-deficient mice exhibited reduced repetitive neuronal population activity during exploration of a novel context and were less able to discriminate contexts in a contextual fear conditioning paradigm. These results suggest that one function of Pcdhßs is to modulate neural ensemble activity in the hippocampus to promote context discrimination.


Subject(s)
CA1 Region, Hippocampal/physiology , Cadherins/physiology , Conditioning, Classical/physiology , Discrimination Learning/physiology , Fear/physiology , Animals , Cadherins/deficiency , Calcium/analysis , Electroshock , Exploratory Behavior , Genes, Reporter , Genetic Vectors , Male , Mice , Mice, Knockout , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods , Neurons/chemistry , Neurons/ultrastructure , Open Field Test , Protein Isoforms/deficiency , Protein Isoforms/physiology
17.
J Leukoc Biol ; 107(6): 907-915, 2020 06.
Article in English | MEDLINE | ID: mdl-31749173

ABSTRACT

The TNFR superfamily of receptors, the major focus of the recent TNFR Superfamily Conference held in June 2019, employ the TNFR-associated factor (TRAF) family of adaptor proteins in key aspects of their signaling pathways. Although many early studies investigated TRAF functions via exogenous overexpression in nonhematopoietic cell lines, it has subsequently become clear that whereas TRAFs share some overlap in function, each also plays unique biologic roles, that can be highly context dependent. This brief review summarizes the current state of knowledge of functions of each of the TRAF molecules that mediate important functions in T lymphocytes: TRAFs 1, 2, 3, 5, and 6. Due to our current appreciation of the contextual nature of TRAF function, our focus is upon findings made specifically in T lymphocytes. Key T cell functions for each TRAF are detailed, as well as future knowledge gaps of interest and importance.


Subject(s)
Immunologic Deficiency Syndromes/genetics , Receptors, Tumor Necrosis Factor/genetics , Signal Transduction/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Disease Models, Animal , Gene Expression Regulation , Humans , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/pathology , Immunologic Memory , Mice , Mice, Knockout , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/immunology , Receptors, Tumor Necrosis Factor/immunology , Signal Transduction/immunology , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/deficiency , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/immunology , Tumor Necrosis Factor-alpha/immunology
18.
Elife ; 82019 10 02.
Article in English | MEDLINE | ID: mdl-31577230

ABSTRACT

Toxoplasma gondii contains a limited subset of actin binding proteins. Here we show that the putative actin regulator cyclase-associated protein (CAP) is present in two different isoforms and its deletion leads to significant defects in some but not all actin dependent processes. We observe defects in cell-cell communication, daughter cell orientation and the juxtanuclear accumulation of actin, but only modest defects in synchronicity of division and no defect in the replication of the apicoplast. 3D electron microscopy reveals that loss of CAP results in a defect in formation of a normal central residual body, but parasites remain connected within the vacuole. This dissociates synchronicity of division and parasite rosetting and reveals that establishment and maintenance of the residual body may be more complex than previously thought. These results highlight the different spatial requirements for F-actin regulation in Toxoplasma which appear to be achieved by partially overlapping functions of actin regulators.


Subject(s)
Actins/metabolism , Protozoan Proteins/metabolism , Toxoplasma/metabolism , Cell Communication , Cell Division , Gene Deletion , Protein Isoforms/deficiency , Protein Isoforms/metabolism , Protozoan Proteins/genetics
19.
Elife ; 82019 10 11.
Article in English | MEDLINE | ID: mdl-31603426

ABSTRACT

Dynamin-related protein 1 (Drp1) divides mitochondria as a mechano-chemical GTPase. However, the function of Drp1 beyond mitochondrial division is largely unknown. Multiple Drp1 isoforms are produced through mRNA splicing. One such isoform, Drp1ABCD, contains all four alternative exons and is specifically expressed in the brain. Here, we studied the function of Drp1ABCD in mouse neurons in both culture and animal systems using isoform-specific knockdown by shRNA and isoform-specific knockout by CRISPR/Cas9. We found that the expression of Drp1ABCD is induced during postnatal brain development. Drp1ABCD is enriched in dendritic spines and regulates postsynaptic clathrin-mediated endocytosis by positioning the endocytic zone at the postsynaptic density, independently of mitochondrial division. Drp1ABCD loss promotes the formation of ectopic dendrites in neurons and enhanced sensorimotor gating behavior in mice. These data reveal that Drp1ABCD controls postsynaptic endocytosis, neuronal morphology and brain function.


Subject(s)
Brain/metabolism , Dendrites/metabolism , Dynamins/metabolism , Endocytosis , Mitochondrial Dynamics , Synapses/metabolism , Animals , Dynamins/deficiency , Mice , Mice, Knockout , Protein Isoforms/deficiency , Protein Isoforms/metabolism
20.
J Immunol ; 203(8): 2141-2149, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31541023

ABSTRACT

Foxo3 acts as an important central regulator that integrates signaling pathways and coordinates cellular responses to environmental changes. Recent studies show the involvement of Foxo3 in osteoclastogenesis and rheumatoid arthritis, which prompted us to further investigate the FOXO3 locus. Several databases document FOXO3 isoform2, an N-terminal truncated mutation of the full-length FOXO3 However, the biological function of FOXO3 isoform2 is unclear. In this study, we established a conditional allele of Foxo3 in mice that deletes the full-length Foxo3 except isoform2, a close ortholog of the human FOXO3 isoform2. Expression of Foxo3 isoform2 specifically in macrophage/osteoclast lineage suppresses osteoclastogenesis and leads to the osteopetrotic phenotype in mice. Mechanistically, Foxo3 isoform2 enhances the expression of type I IFN response genes to RANKL stimulation and thus inhibits osteoclastogenesis via endogenous IFN-ß-mediated feedback inhibition. Our findings identify, to our knowledge, the first known biological function of Foxo3 isoform2 that acts as a novel osteoclastic inhibitor in bone remodeling.


Subject(s)
Forkhead Box Protein O3/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Animals , Cell Differentiation/drug effects , Cells, Cultured , Forkhead Box Protein O3/deficiency , HEK293 Cells , Humans , Interferon Type I/pharmacology , Mice , Mice, Inbred C57BL , Osteoclasts/drug effects , Osteogenesis/drug effects , Phenotype , Protein Isoforms/analysis , Protein Isoforms/deficiency , Protein Isoforms/metabolism , RAW 264.7 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...