Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Microbiol Spectr ; 12(3): e0375022, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38299851

ABSTRACT

Chronic wasting disease (CWD) is a naturally occurring prion disease in cervids that has been rapidly proliferating in the United States. Here, we investigated a potential link between CWD infection and gut microbiome by analyzing 50 fecal samples obtained from CWD-positive animals of different sexes from various regions in the USA compared to 50 CWD-negative controls using high throughput sequencing of the 16S ribosomal RNA and targeted metabolomics. Our analysis reveals promising trends in the gut microbiota that could potentially be CWD-dependent, including several bacterial taxa at each rank level, as well as taxa pairs, that can differentiate between CWD-negative and CWD-positive deer. Through machine-learning, these taxa and taxa pairs at each rank level could facilitate identification of around 70% of both the CWD-negative and the CWD-positive samples. Our results provide a potential tool for diagnostics and surveillance of CWD in the wild, as well as conceptual advances in our understanding of the disease.IMPORTANCEThis is a comprehensive study that tests the connection between the composition of the gut microbiome in deer in response to chronic wasting disease (CWD). We analyzed 50 fecal samples obtained from CWD-positive animals compared to 50 CWD-negative controls to identify CWD-dependent changes in the gut microbiome, matched with the analysis of fecal metabolites. Our results show promising trends suggesting that fecal microbial composition can directly correspond to CWD disease status. These results point to the microbial composition of the feces as a potential tool for diagnostics and surveillance of CWD in the wild, including non-invasive CWD detection in asymptomatic deer and deer habitats, and enable conceptual advances in our understanding of the disease.


Subject(s)
Deer , Wasting Disease, Chronic , Animals , Wasting Disease, Chronic/diagnosis , Wasting Disease, Chronic/genetics , Wasting Disease, Chronic/metabolism , Prospective Studies , Feces , Biomarkers/metabolism
2.
bioRxiv ; 2023 Mar 27.
Article in English | MEDLINE | ID: mdl-37034790

ABSTRACT

Lack of non-muscle ß -actin gene (Actb) leads to early embryonic lethality in mice, however mice with ß - to γ -actin replacement develop normally and show no detectable phenotypes at young age. Here we investigated the effect of this replacement in the retina. During aging, these mice have accelerated de-generation of retinal structure and function, including elongated microvilli and defective mitochondria of retinal pigment epithelium (RPE), abnormally bulging photoreceptor outer segments (OS) accompanied by reduced transducin concentration and light sensitivity, and accumulation of autofluorescent microglia cells in the subretinal space between RPE and OS. These defects are accompanied by changes in the F-actin binding of several key actin interacting partners, including ezrin, myosin, talin, and vinculin known to play central roles in modulating actin cytoskeleton and cell adhesion and mediating the phagocytosis of OS. Our data show that ß -actin protein is essential for maintaining normal retinal structure and function.

3.
iScience ; 25(10): 105186, 2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36248738

ABSTRACT

Cytoplasmic beta- and gamma-actin are ubiquitously expressed in every eukaryotic cell. They are encoded by different genes, but their amino acid sequences differ only by four conservative substitutions at the N-termini, making it difficult to dissect their individual regulation. Here, we analyzed actin from cultured cells and tissues by mass spectrometry and found that beta, unlike gamma actin, undergoes sequential removal of N-terminal Asp residues, leading to truncated actin species found in both F- and G-actin preparations. This processing affects up to ∼3% of beta actin in different cell types. We used CRISPR/Cas-9 in cultured cells to delete two candidate enzymes capable of mediating this type of processing. This deletion abolishes most of the beta actin N-terminal processing and results in changes in F-actin levels, cell spreading, filopodia formation, and cell migration. Our results demonstrate previously unknown isoform-specific actin regulation that can potentially affect actin functions in cells.

4.
Transl Neurodegener ; 11(1): 20, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35395956

ABSTRACT

BACKGROUND: Alpha-synuclein (α-syn) exhibits pathological misfolding in many human neurodegenerative disorders. We previously showed that α-syn is arginylated in the mouse brain and that lack of arginylation leads to neurodegeneration in mice. METHODS: Here, we tested α-syn arginylation in human brain pathology using newly derived antibodies in combination with Western blotting, biochemical assays, and experiments in live neurons. RESULTS: We found that α-syn was arginylated in the human brain on E46 and E83, two sites previously implicated in α-syn pathology and familial cases of Parkinson's disease. The levels of arginylation in different brain samples ranged between ~ 3% and ~ 50% of the total α-syn pool, and this arginylation nearly exclusively concentrated in the subcellular α-syn fraction that sedimented at low centrifugation speeds and appeared to be simultaneously targeted by multiple posttranslational modifications. Arginylated α-syn was less susceptible to S129 phosphorylation and pathological aggregation in neurons. The arginylation level inversely correlated with the overall α-syn levels and with patient age, suggesting a possible causal relationship between arginylation decline and α-syn-dependent neuropathology. CONCLUSION: We propose that α-syn arginylation constitutes a potential neuroprotective mechanism that prevents its abnormal accumulation during neurodegeneration and aging in the human brain.


Subject(s)
Neurodegenerative Diseases , Parkinson Disease , Synucleinopathies , Animals , Brain/metabolism , Humans , Mice , Parkinson Disease/pathology , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
5.
Elife ; 102021 06 24.
Article in English | MEDLINE | ID: mdl-34165080

ABSTRACT

ß- and γ-cytoplasmic actins are ubiquitously expressed in every cell type and are nearly identical at the amino acid level but play vastly different roles in vivo. Their essential roles in embryogenesis and mesenchymal cell migration critically depend on the nucleotide sequences of their genes, rather than their amino acid sequences; however, it is unclear which gene elements underlie this effect. Here we address the specific role of the coding sequence in ß- and γ-cytoplasmic actins' intracellular functions, using stable polyclonal populations of immortalized mouse embryonic fibroblasts with exogenously expressed actin isoforms and their 'codon-switched' variants. When targeted to the cell periphery using ß-actin 3'UTR; ß-actin and γ-actin have differential effects on cell migration. These effects directly depend on the coding sequence. Single-molecule measurements of actin isoform translation, combined with fluorescence recovery after photobleaching, demonstrate a pronounced difference in ß- and γ-actins' translation elongation rates in cells, leading to changes in their dynamics at focal adhesions, impairments in actin bundle formation, and reduced cell anchoring to the substrate during migration. Our results demonstrate that coding sequence-mediated differences in actin translation play a key role in cell migration.


Most mammalian cells make both ß- and γ-actin, two proteins which shape the cell's internal skeleton and its ability to migrate. The molecules share over 99% of their sequence, yet they play distinct roles. In fact, deleting the ß-actin gene in mice causes death in the womb, while the animals can survive with comparatively milder issues without their γ-actin gene. How two similar proteins can have such different biological roles is a long-standing mystery. A closer look could hold some clues: ß- and γ-actin may contain the same blocks (or amino acids), but the genetic sequences that encode these proteins differ by about 13%. This is because different units of genetic information ­ known as synonymous codons ­ can encode the same amino acid. These 'silent substitutions' have no effect on the sequence of the proteins, yet a cell reads synonymous codons (and therefore produces proteins) at different speeds. To find out the impact of silent substitutions, Vedula et al. swapped the codons for the two proteins, forcing mouse cells to produce ß-actin using γ-actin codons, and vice versa. Cells with non-manipulated γ-actin and those with ß-actin made using γ-actin codons could move much faster than cells with ß-actin. This suggested that silent substitutions were indeed affecting the role of the protein. Vedula et al. found that cells read γ-codons ­ and therefore made γ-actin ­ much more slowly than ß-codons: this also affected how quickly the protein could be dispatched where it was needed in the cell. Slower production meant that bundles of γ-actin were shorter, which allowed cells to move faster by providing a weaker anchoring system. Overall, this work provides new links between silent substitutions and protein behavior, a relatively new research area which is likely to shed light on other protein families.


Subject(s)
Actins/metabolism , Cell Movement/physiology , Protein Biosynthesis/physiology , Actins/genetics , Amino Acid Substitution , Animals , Base Sequence , Focal Adhesions , Gene Expression Regulation , Mice , Protein Isoforms , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Sci Rep ; 11(1): 9376, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33931669

ABSTRACT

Regulator of G-protein signaling 7 (RGS7) is predominately present in the nervous system and is essential for neuronal signaling involving G-proteins. Prior studies in cultured cells showed that RGS7 is regulated via proteasomal degradation, however no protein is known to facilitate proteasomal degradation of RGS7 and it has not been shown whether this regulation affects G-protein signaling in neurons. Here we used a knockout mouse model with conditional deletion of arginyltransferase (Ate1) in the nervous system and found that in retinal ON bipolar cells, where RGS7 modulates a G-protein to signal light increments, deletion of Ate1 raised the level of RGS7. Electroretinographs revealed that lack of Ate1 leads to increased light-evoked response sensitivities of ON-bipolar cells, as well as their downstream neurons. In cultured mouse embryonic fibroblasts (MEF), RGS7 was rapidly degraded via proteasome pathway and this degradation was abolished in Ate1 knockout MEF. Our results indicate that Ate1 regulates RGS7 protein level by facilitating proteasomal degradation of RGS7 and thus affects G-protein signaling in neurons.


Subject(s)
Aminoacyltransferases/physiology , Fibroblasts/metabolism , Light , Nervous System/metabolism , RGS Proteins/metabolism , Retinal Bipolar Cells/metabolism , Animals , Female , Fibroblasts/pathology , Fibroblasts/radiation effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System/pathology , Nervous System/radiation effects , RGS Proteins/genetics , Retinal Bipolar Cells/pathology , Retinal Bipolar Cells/radiation effects , Signal Transduction
7.
Front Cell Dev Biol ; 9: 807345, 2021.
Article in English | MEDLINE | ID: mdl-35127722

ABSTRACT

Arginylation is a post-translational modification mediated by the arginyltransferase (Ate1). We recently showed that conditional deletion of Ate1 in the nervous system leads to increased light-evoked response sensitivities of ON-bipolar cells in the retina, indicating that arginylation regulates the G-protein signaling complexes of those neurons and/or photoreceptors. However, none of the key players in the signaling pathway were previously shown to be arginylated. Here we show that Gαt1, Gß1, RGS6, and RGS7 are arginylated in the retina and RGS6 and RGS7 protein levels are elevated in Ate1 knockout, suggesting that arginylation plays a direct role in regulating their protein level and the G-protein-mediated responses in the retina.

8.
Cell Chem Biol ; 27(7): 839-849.e4, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32553119

ABSTRACT

Arginyltransferase ATE1 mediates posttranslational arginylation and plays key roles in multiple physiological processes. ATE1 utilizes arginyl (Arg)-tRNAArg as the donor of Arg, putting this reaction into a direct competition with the protein synthesis machinery. Here, we address the question of ATE1- Arg-tRNAArg specificity as a potential mechanism enabling this competition in vivo. Using in vitro arginylation assays and Ate1 knockout models, we find that, in addition to full-length tRNA, ATE1 is also able to utilize short tRNAArg fragments that bear structural resemblance to tRNA-derived fragments (tRF), a recently discovered class of small regulatory non-coding RNAs with global emerging biological role. Ate1 knockout cells show a decrease in tRFArg generation and a significant increase in the ratio of tRNAArg:tRFArg compared with wild type, suggesting a functional link between tRFArg and arginylation. We propose that generation of physiologically important tRFs can serve as a switch between translation and protein arginylation.


Subject(s)
Aminoacyltransferases/metabolism , Arginine/metabolism , RNA, Transfer, Arg/metabolism , Aminoacyltransferases/genetics , Angiotensin II/metabolism , Animals , Cell Line , Humans , Mice , Protein Binding , Protein Processing, Post-Translational , Substrate Specificity
9.
Elife ; 62017 12 15.
Article in English | MEDLINE | ID: mdl-29244021

ABSTRACT

ß- and γ-cytoplasmic actin are nearly indistinguishable in their amino acid sequence, but are encoded by different genes that play non-redundant biological roles. The key determinants that drive their functional distinction are unknown. Here, we tested the hypothesis that ß- and γ-actin functions are defined by their nucleotide, rather than their amino acid sequence, using targeted editing of the mouse genome. Although previous studies have shown that disruption of ß-actin gene critically impacts cell migration and mouse embryogenesis, we demonstrate here that generation of a mouse lacking ß-actin protein by editing ß-actin gene to encode γ-actin protein, and vice versa, does not affect cell migration and/or organism survival. Our data suggest that the essential in vivo function of ß-actin is provided by the gene sequence independent of the encoded protein isoform. We propose that this regulation constitutes a global 'silent code' mechanism that controls the functional diversity of protein isoforms.


Subject(s)
Actins/genetics , Actins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Amino Acid Sequence , Animals , Base Sequence , Gene Editing , Mice
10.
Sci Rep ; 7(1): 11323, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28900170

ABSTRACT

Alpha synuclein (α-syn) is a central player in neurodegeneration, but the mechanisms triggering its pathology are not fully understood. Here we found that α-syn is a highly efficient substrate for arginyltransferase ATE1 and is arginylated in vivo by a novel mid-chain mechanism that targets the acidic side chains of E46 and E83. Lack of arginylation leads to increased α-syn aggregation and causes the formation of larger pathological aggregates in neurons, accompanied by impairments in its ability to be cleared via normal degradation pathways. In the mouse brain, lack of arginylation leads to an increase in α-syn's insoluble fraction, accompanied by behavioral changes characteristic for neurodegenerative pathology. Our data show that lack of arginylation in the brain leads to neurodegeneration, and suggests that α-syn arginylation can be a previously unknown factor that facilitates normal α-syn folding and function in vivo.


Subject(s)
Arginine/metabolism , Brain/physiology , Neurodegenerative Diseases/metabolism , alpha-Synuclein/metabolism , Amino Acid Sequence , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Humans , Mass Spectrometry , Mice , Mice, Knockout , Models, Biological , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/prevention & control , Neurons/metabolism , Neurons/pathology , Peptides/chemistry , Peptides/metabolism , Protein Aggregates , Protein Aggregation, Pathological/metabolism , Protein Processing, Post-Translational , Proteolysis , Recombinant Proteins , Substrate Specificity , alpha-Synuclein/chemistry
11.
Dev Biol ; 430(1): 41-51, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28844905

ABSTRACT

Arginylation is an emerging protein modification mediated by arginyltransferase ATE1, shown to regulate embryogenesis and actin cytoskeleton, however its functions in different physiological systems are not well understood. Here we analyzed the role of ATE1 in brain development and neuronal growth by producing a conditional mouse knockout with Ate1 deletion in the nervous system driven by Nestin promoter (Nes-Ate1 mice). These mice were weaker than wild type, resulting in low postnatal survival rates, and had abnormalities in the brain that suggested defects in neuronal migration. Cultured Ate1 knockout neurons showed a reduction in the neurite outgrowth and the levels of doublecortin and F-actin in the growth cones. In wild type, ATE1 prominently localized to the growth cones, in addition to the cell bodies. Examination of the Ate1 mRNA sequence reveals the existence of putative zipcode-binding sequences involved in mRNA targeting to the cell periphery and local translation at the growth cones. Fluorescence in situ hybridization showed that Ate1 mRNA localized to the tips of the growth cones, likely due to zipcode-mediated targeting, and this localization coincided with spots of localization of arginylated ß-actin, which disappeared in the presence of protein synthesis inhibitors. We propose that zipcode-mediated co-targeting of Ate1 and ß-actin mRNA leads to localized co-translational arginylation of ß-actin that drives the growth cone migration and neurite outgrowth.


Subject(s)
Aminoacyltransferases/metabolism , Brain/growth & development , Brain/metabolism , Growth Cones/enzymology , Neurites/enzymology , Neuronal Outgrowth , Actins/metabolism , Animals , Arginine/metabolism , Brain/abnormalities , Brain/pathology , Cell Movement , Doublecortin Domain Proteins , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Models, Biological , Neuropeptides/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Genom Data ; 6: 99-107, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26697345

ABSTRACT

Mitochondrial metabolic dysfunction is often seen in cancers. This paper shows that the defect in a mitochondrial electron transport component, the cytochrome c oxidase (CcO), leads to increased glycolysis and carcinogenesis. Using whole genome microarray expression analysis we show that genetic silencing of the CcO subunit Cox4i1 in mouse C2C12 myoblasts resulted in metabolic shift to glycolysis, activated a retrograde stress signaling, and induced carcinogenesis. In the knockdown cells, the expression of Cox4i1 was less than 5% of the control and the expression of the irreversible glycolytic enzymes (Hk1, Pfkm and Pkm) increased two folds, facilitating metabolic shift to glycolysis. The expression of Ca (2+) sensitive Calcineurin (Ppp3ca) and the expression of PI3-kinase (Pik3r4 and Pik3cb) increased by two folds. This Ca (2+)/Calcineurin/PI3K retrograde stress signaling induced the up-regulation of many nuclear genes involved in tumor progression. Overall, we found 1047 genes with 2-folds expression change (with p-value less than 0.01) between the knockdown and the control, among which were 35 up-regulated genes in pathways in cancer (enrichment p-value less than 10(- 5)). Functional analysis revealed that the up-regulated genes in pathways in cancer were dominated by genes in signal transduction, regulation of transcription and PI3K signaling pathway. These results suggest that a defect in CcO complex initiates a retrograde signaling which can induce tumor progression. Physiological studies of these cells and esophageal tumors from human patients support these results. GEO accession number = GSE68525.

13.
BMC Genomics ; 15: 677, 2014 Aug 13.
Article in English | MEDLINE | ID: mdl-25124333

ABSTRACT

BACKGROUND: Mitochondrial DNA (mtDNA) deletions cause disease and accumulate during aging, yet our understanding of the molecular mechanisms underlying their formation remains rudimentary. Guanine-quadruplex (GQ) DNA structures are associated with nuclear DNA instability in cancer; recent evidence indicates they can also form in mitochondrial nucleic acids, suggesting that these non-B DNA structures could be associated with mtDNA deletions. Currently, the multiple types of GQ sequences and their association with human mtDNA stability are unknown. RESULTS: Here, we show an association between human mtDNA deletion breakpoint locations (sites where DNA ends rejoin after deletion of a section) and sequences with G-quadruplex forming potential (QFP), and establish the ability of selected sequences to form GQ in vitro. QFP contain four runs of either two or three consecutive guanines (2G and 3G, respectively), and we identified four types of QFP for subsequent analysis: intrastrand 2G, intrastrand 3G, duplex derived interstrand (ddi) 2G, and ddi 3G QFP sequences. We analyzed the position of each motif set relative to either 5' or 3' unique mtDNA deletion breakpoints, and found that intrastrand QFP sequences, but not ddi QFP sequences, showed significant association with mtDNA deletion breakpoint locations. Moreover, a large proportion of these QFP sequences occur at smaller distances to breakpoints relative to distribution-matched controls. The positive association of 2G QFP sequences persisted when breakpoints were divided into clinical subgroups. We tested in vitro GQ formation of representative mtDNA sequences containing these 2G QFP sequences and detected robust GQ structures by UV-VIS and CD spectroscopy. Notably, the most frequent deletion breakpoints, including those of the "common deletion", are bounded by 2G QFP sequence motifs. CONCLUSIONS: The potential for GQ to influence mitochondrial genome stability supports a high-priority investigation of these structures and their regulation in normal and pathological mitochondrial biology. These findings emphasize the potential importance of helicases that subsequently resolve GQ to maintain the stability of the mitochondrial genome.


Subject(s)
DNA, Mitochondrial/genetics , Chromosome Breakage , G-Quadruplexes , Gene Deletion , Genome, Mitochondrial , Genomic Instability , Humans , Inverted Repeat Sequences
14.
Development ; 135(23): 3881-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18948421

ABSTRACT

Post-translational arginylation mediated by arginyltransferase (Ate1) is essential for cardiovascular development and angiogenesis in mammals and directly affects myocardium structure in the developing heart. We recently showed that arginylation exerts a number of intracellular effects by modifying proteins involved in the functioning of the actin cytoskeleton and in cell motility. Here, we investigated the role of arginylation in the development and function of cardiac myocytes and their actin-containing structures during embryogenesis. Biochemical and mass spectrometry analyses showed that alpha cardiac actin undergoes arginylation at four sites during development. Ultrastructural analysis of the myofibrils in wild-type and Ate1 knockout mouse hearts showed that the absence of arginylation results in defects in myofibril structure that delay their development and affect the continuity of myofibrils throughout the heart, predicting defects in cardiac contractility. Comparison of cardiac myocytes derived from wild-type and Ate1 knockout mouse embryos revealed that the absence of arginylation results in abnormal beating patterns. Our results demonstrate cell-autonomous cardiac myocyte defects in arginylation knockout mice that lead to severe congenital abnormalities similar to those observed in human disease, and outline a new function of arginylation in the regulation of the actin cytoskeleton in cardiac myocytes.


Subject(s)
Actins/metabolism , Aminoacyltransferases/metabolism , Heart/embryology , Myocardial Contraction , Myofibrils/enzymology , Actins/chemistry , Animals , Arginine/metabolism , Cells, Cultured , Mice , Mice, Knockout , Models, Cardiovascular , Myocardium/enzymology , Myocardium/pathology , Myocardium/ultrastructure , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Myocytes, Cardiac/ultrastructure , Myofibrils/pathology , Myofibrils/ultrastructure , Protein Structure, Secondary , Sarcomeres/pathology , Sarcomeres/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...