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










Publication year range
1.
Mov Disord ; 39(2): 339-349, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38014556

ABSTRACT

BACKGROUND: Recent studies have advanced our understanding of the genetic drivers of Parkinson's disease (PD). Rare variants in more than 20 genes are considered causal for PD, and the latest PD genome-wide association study (GWAS) identified 90 independent risk loci. However, there remains a gap in our understanding of PD genetics outside of the European populations in which the vast majority of these studies were focused. OBJECTIVE: The aim was to identify genetic risk factors for PD in a South Asian population. METHODS: A total of 674 PD subjects predominantly with age of onset (AoO) ≤50 years (encompassing juvenile, young, or early-onset PD) were recruited from 10 specialty movement disorder centers across India over a 2-year period; 1376 control subjects were selected from the reference population GenomeAsia, Phase 2. We performed various case-only and case-control genetic analyses for PD diagnosis and AoO. RESULTS: A genome-wide significant signal for PD diagnosis was identified in the SNCA region, strongly colocalizing with SNCA region signal from European PD GWAS. PD cases with pathogenic mutations in PD genes exhibited, on average, lower PD polygenic risk scores than PD cases lacking any PD gene mutations. Gene burden studies of rare, predicted deleterious variants identified BSN, encoding the presynaptic protein Bassoon that has been previously associated with neurodegenerative disease. CONCLUSIONS: This study constitutes the largest genetic investigation of PD in a South Asian population to date. Future work should seek to expand sample numbers in this population to enable improved statistical power to detect PD genes in this understudied group. © 2023 Denali Therapeutics and The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Neurodegenerative Diseases , Parkinson Disease , Humans , Middle Aged , Parkinson Disease/epidemiology , Parkinson Disease/genetics , Parkinson Disease/diagnosis , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Mutation
2.
Elife ; 122023 Oct 24.
Article in English | MEDLINE | ID: mdl-37874617

ABSTRACT

Leucine-rich repeat kinase 2 (LRRK2) variants associated with Parkinson's disease (PD) and Crohn's disease lead to increased phosphorylation of its Rab substrates. While it has been recently shown that perturbations in cellular homeostasis including lysosomal damage can increase LRRK2 activity and localization to lysosomes, the molecular mechanisms by which LRRK2 activity is regulated have remained poorly defined. We performed a targeted siRNA screen to identify regulators of LRRK2 activity and identified Rab12 as a novel modulator of LRRK2-dependent phosphorylation of one of its substrates, Rab10. Using a combination of imaging and immunopurification methods to isolate lysosomes, we demonstrated that Rab12 is actively recruited to damaged lysosomes and leads to a local and LRRK2-dependent increase in Rab10 phosphorylation. PD-linked variants, including LRRK2 R1441G and VPS35 D620N, lead to increased recruitment of LRRK2 to the lysosome and a local elevation in lysosomal levels of pT73 Rab10. Together, these data suggest a conserved mechanism by which Rab12, in response to damage or expression of PD-associated variants, facilitates the recruitment of LRRK2 and phosphorylation of its Rab substrate(s) at the lysosome.


Lysosomes are cellular compartments tasked with breaking down large molecules such as lipids or proteins. They perform an essential role in helping cells dispose of obsolete or harmful components; in fact, defects in lysosome function are associated with a range of health conditions. For instance, many genes associated with an increased risk of developing Parkinson's disease code for proteins required for lysosomes to work properly, such as the kinase LRRK2. Previous work has shown that this enzyme gets recruited to the surface of damaged lysosomes, where it can modulate the function of another set of molecular actors by modifying them through a chemical process known as phosphorylation. Such activity is increased in harmful versions of LRRK2 linked to Parkinson's disease. However, the molecular mechanisms which control LRRK2 activity or its recruitment to lysosomes remain unclear. To examine this question, Wang, Bondar et al. first performed a targeted screen to identify proteins that can regulate LRRK2 activity. This revealed that Rab12, one of molecular actors that LRRK2 phosphorylates, can in turn modulate the activity of the enzyme. Further imaging and biochemical experiments then showed that Rab12 is recruited to damaged lysosomes and that this step was in fact necessary for LRRK2 to also relocate to these compartments. The data suggest that this Rab12-driven recruitment process increases the local concentration of LRRK2 near its Rab targets on the membrane of damaged lysosomes, and therefore leads to enhanced LRRK2 activity. Crucially, Wang, Bondar et al. showed that Rab12 also plays a role in the increased LRRK2 activity observed with two Parkinson's disease-linked mutations (one in LRRK2 itself and one in another lysosomal regulator, VPS35), suggesting that increased LRRK2 concentration on lysosomes may be a conserved mechanism that leads to increased LRRK2 activity in disease. Overall, these results highlight a new, Rab12-dependent mechanism that results in enhanced activity at the lysosomal membrane with variants associated with Parkinson's disease, and for LRRK2 in general when lysosomes are damaged. This knowledge will be helpful to develop therapeutic strategies that target LRRK2, and to better understand how increased LRRK2 activity and lysosomal injury may be linked to Parkinson's disease.


Subject(s)
Biological Phenomena , Lysosomes , rab GTP-Binding Proteins , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Lysosomes/metabolism , Mutation , Phosphorylation , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , Humans
3.
Dev Cell ; 58(18): 1782-1800.e10, 2023 09 25.
Article in English | MEDLINE | ID: mdl-37494933

ABSTRACT

Despite the key roles of perilipin-2 (PLIN2) in governing lipid droplet (LD) metabolism, the mechanisms that regulate PLIN2 levels remain incompletely understood. Here, we leverage a set of genome-edited human PLIN2 reporter cell lines in a series of CRISPR-Cas9 loss-of-function screens, identifying genetic modifiers that influence PLIN2 expression and post-translational stability under different metabolic conditions and in different cell types. These regulators include canonical genes that control lipid metabolism as well as genes involved in ubiquitination, transcription, and mitochondrial function. We further demonstrate a role for the E3 ligase MARCH6 in regulating triacylglycerol biosynthesis, thereby influencing LD abundance and PLIN2 stability. Finally, our CRISPR screens and several published screens provide the foundation for CRISPRlipid (http://crisprlipid.org), an online data commons for lipid-related functional genomics data. Our study identifies mechanisms of PLIN2 and LD regulation and provides an extensive resource for the exploration of LD biology and lipid metabolism.


Subject(s)
CRISPR-Cas Systems , Lipid Droplets , Humans , Perilipin-2/genetics , Perilipin-2/metabolism , Lipid Droplets/metabolism , CRISPR-Cas Systems/genetics , Lipid Metabolism/genetics , Cell Line
4.
Methods Enzymol ; 619: 97-120, 2019.
Article in English | MEDLINE | ID: mdl-30910031

ABSTRACT

Identification and degradation of misfolded proteins by the ubiquitin-proteasome system (UPS) is crucial for maintaining proteostasis, but only a handful of UPS components have been linked to the recognition of specific substrates. Studies in Saccharomyces cerevisiae using systematic perturbation of nonessential genes have uncovered UPS components that recognize and ubiquitylate model substrates of the UPS; however, similar analyses in metazoans have been limited. In this chapter, we describe methods for using CRISPR/Cas9 technology combined with genome-wide high complexity single guide (sgRNA) libraries and a transcriptional shutoff strategy for phenotypic selection based on kinetic measurements of protein turnover to identify the genes required to degrade model clients of the mammalian ER-associated degradation system. We also discuss considerations for screen design, execution, and interpretation.


Subject(s)
CRISPR-Cas Systems , Endoplasmic Reticulum-Associated Degradation , Animals , Cell Line , Gene Editing/methods , Humans , RNA, Guide, Kinetoplastida/genetics , Transduction, Genetic
5.
Proc Natl Acad Sci U S A ; 116(4): 1299-1308, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30626644

ABSTRACT

Ubiquitin fold modifier 1 (UFM1) is a small, metazoan-specific, ubiquitin-like protein modifier that is essential for embryonic development. Although loss-of-function mutations in UFM1 conjugation are linked to endoplasmic reticulum (ER) stress, neither the biological function nor the relevant cellular targets of this protein modifier are known. Here, we show that a largely uncharacterized ribosomal protein, RPL26, is the principal target of UFM1 conjugation. RPL26 UFMylation and de-UFMylation is catalyzed by enzyme complexes tethered to the cytoplasmic surface of the ER and UFMylated RPL26 is highly enriched on ER membrane-bound ribosomes and polysomes. Biochemical analysis and structural modeling establish that UFMylated RPL26 and the UFMylation machinery are in close proximity to the SEC61 translocon, suggesting that this modification plays a direct role in cotranslational protein translocation into the ER. These data suggest that UFMylation is a ribosomal modification specialized to facilitate metazoan-specific protein biogenesis at the ER.


Subject(s)
Ribosomal Proteins/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Carrier Proteins/metabolism , Cell Line , Cell Line, Tumor , Cytoplasm/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/physiology , HEK293 Cells , Humans , K562 Cells , Polyribosomes/metabolism , Protein Binding/physiology , Protein Transport/physiology , Ribosomes/metabolism
6.
Mol Cell ; 73(2): 377-389.e11, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30581143

ABSTRACT

The ubiquitin proteasome system (UPS) maintains the integrity of the proteome by selectively degrading misfolded or mis-assembled proteins, but the rules that govern how conformationally defective proteins in the secretory pathway are selected from the structurally and topologically diverse constellation of correctly folded membrane and secretory proteins for efficient degradation by cytosolic proteasomes is not well understood. Here, we combine parallel pooled genome-wide CRISPR-Cas9 forward genetic screening with a highly quantitative and sensitive protein turnover assay to discover a previously undescribed collaboration between membrane-embedded cytoplasmic ubiquitin E3 ligases to conjugate heterotypic branched or mixed ubiquitin (Ub) chains on substrates of endoplasmic-reticulum-associated degradation (ERAD). These findings demonstrate that parallel CRISPR analysis can be used to deconvolve highly complex cell biological processes and identify new biochemical pathways in protein quality control.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Endoplasmic Reticulum-Associated Degradation , Genome-Wide Association Study/methods , Proteasome Endopeptidase Complex/metabolism , Proteostasis , CRISPR-Associated Protein 9/metabolism , Endoplasmic Reticulum-Associated Degradation/drug effects , Endoplasmic Reticulum-Associated Degradation/genetics , HEK293 Cells , Humans , K562 Cells , Kinetics , Proteasome Endopeptidase Complex/genetics , Protein Folding , Proteolysis , Proteostasis/drug effects , Proteostasis/genetics , Ricin/pharmacology , Substrate Specificity , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
7.
Mol Cell ; 70(3): 516-530.e6, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29706535

ABSTRACT

Glycoproteins engaged in unproductive folding in the ER are marked for degradation by a signal generated by progressive demannosylation of substrate N-glycans that is decoded by ER lectins, but how the two lectins, OS9 and XTP3B, contribute to non-glycosylated protein triage is unknown. We generated cell lines with homozygous deletions of both lectins individually and in combination. We found that OS9 and XTP3B redundantly promote glycoprotein degradation and stabilize the SEL1L/HRD1 dislocon complex, that XTP3B profoundly inhibits the degradation of non-glycosylated proteins, and that OS9 antagonizes this inhibition. The relative expression of OS9 and XTP3B and the distribution of glycan and non-glycan degrons within the same protein contribute to the fidelity and processivity of glycoprotein triage and, therefore, determine the fates of newly synthesized proteins in the early secretory pathway.


Subject(s)
Endoplasmic Reticulum-Associated Degradation/physiology , Endoplasmic Reticulum/metabolism , Lectins/metabolism , Neoplasm Proteins/metabolism , Polysaccharides/metabolism , Cell Line , Cell Line, Tumor , Glycoproteins/metabolism , Glycosylation , HEK293 Cells , Humans , K562 Cells , Protein Folding , Protein Translocation Systems/metabolism
8.
Mol Biol Cell ; 25(19): 3059-69, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25103239

ABSTRACT

Insulin-stimulated glucose uptake in fat and muscle is mediated by the major facilitative glucose transporter Glut4. Insulin controls the trafficking of Glut4 to the plasma membrane via regulation of a series of small G proteins, including RalA and Rab10. We demonstrate here that Rab10 is a bona fide target of the GTPase-activating protein AS160, which is inhibited after phosphorylation by the protein kinase Akt. Once activated, Rab10 can increase the GTP binding of RalA by recruiting the Ral guanyl nucleotide exchange factor, Rlf/Rgl2. Rab10 and RalA reside in the same pool of Glut4-storage vesicles in untreated cells, and, together with Rlf, they ensure maximal glucose transport. Overexpression of membrane-tethered Rlf compensates for the loss of Rab10 in Glut4 translocation, suggesting that Rab10 recruits Rlf to membrane compartments for RalA activation and that RalA is downstream of Rab10. Together these studies identify a new G protein cascade in the regulation of insulin-stimulated Glut4 trafficking and glucose uptake.


Subject(s)
Glucose Transporter Type 4/metabolism , Insulin/metabolism , Transcription Factors/metabolism , rab GTP-Binding Proteins/metabolism , ral GTP-Binding Proteins/metabolism , 3T3 Cells , Adipocytes/metabolism , Animals , Biological Transport , COS Cells , Cell Line , Chlorocebus aethiops , Enzyme Activation , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Glucose/metabolism , Guanine Nucleotide Exchange Factors , Guanosine Triphosphate/metabolism , HEK293 Cells , Humans , Mice , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering , Transcription Factors/biosynthesis , Transcription Factors/genetics , rab GTP-Binding Proteins/agonists
9.
Elife ; 2: e01119, 2013 Dec 24.
Article in English | MEDLINE | ID: mdl-24368730

ABSTRACT

Obesity produces a chronic inflammatory state involving the NFκB pathway, resulting in persistent elevation of the noncanonical IκB kinases IKKε and TBK1. In this study, we report that these kinases attenuate ß-adrenergic signaling in white adipose tissue. Treatment of 3T3-L1 adipocytes with specific inhibitors of these kinases restored ß-adrenergic signaling and lipolysis attenuated by TNFα and Poly (I:C). Conversely, overexpression of the kinases reduced induction of Ucp1, lipolysis, cAMP levels, and phosphorylation of hormone sensitive lipase in response to isoproterenol or forskolin. Noncanonical IKKs reduce catecholamine sensitivity by phosphorylating and activating the major adipocyte phosphodiesterase PDE3B. In vivo inhibition of these kinases by treatment of obese mice with the drug amlexanox reversed obesity-induced catecholamine resistance, and restored PKA signaling in response to injection of a ß-3 adrenergic agonist. These studies suggest that by reducing production of cAMP in adipocytes, IKKε and TBK1 may contribute to the repression of energy expenditure during obesity. DOI: http://dx.doi.org/10.7554/eLife.01119.001.


Subject(s)
Adipocytes/enzymology , Adipose Tissue, White/enzymology , Catecholamines/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , I-kappa B Kinase/metabolism , Inflammation/enzymology , Obesity/enzymology , Protein Serine-Threonine Kinases/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipose Tissue, White/drug effects , Adrenergic beta-3 Receptor Agonists/pharmacology , Aminopyridines/pharmacology , Animals , COS Cells , Chlorocebus aethiops , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Dioxoles/pharmacology , Disease Models, Animal , Energy Metabolism , Enzyme Activation , HEK293 Cells , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/genetics , Inflammation/genetics , Ion Channels/metabolism , Lipolysis , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Obesity/genetics , Phosphorylation , Poly I-C/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Receptors, Adrenergic, beta/metabolism , Signal Transduction , Sterol Esterase/metabolism , Time Factors , Transfection , Tumor Necrosis Factor-alpha/pharmacology , Uncoupling Protein 1
10.
J Biol Chem ; 288(13): 9272-83, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23386617

ABSTRACT

RGC1 and RGC2 comprise a functional RalGAP complex (RGC) that suppresses RalA activity. The PI3-kinase/Akt signaling pathway activates RalA through phosphorylation-mediated inhibition of the RGC. Here we identify a novel phosphorylation-dependent interaction between 14-3-3 and the RGC. 14-3-3 binds to the complex through an Akt-phosphorylated residue, threonine 715, on RGC2. Interaction with 14-3-3 does not alter in vitro activity of the GTPase-activating protein complex. However, blocking the interaction between 14-3-3 and RGC2 in cells increases suppression of RalA activity by the RGC, suggesting that 14-3-3 inhibits the complex through a non-catalytic mechanism. Together, these data show that 14-3-3 negatively regulates the RGC downstream of the PI3-kinase/Akt signaling pathway.


Subject(s)
14-3-3 Proteins/metabolism , GTPase-Activating Proteins/metabolism , Gene Expression Regulation , Nerve Tissue Proteins/metabolism , ral GTP-Binding Proteins/metabolism , 3T3 Cells , Adipocytes/cytology , Amino Acid Motifs , Androstadienes/pharmacology , Animals , DNA/metabolism , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Protein Binding , Signal Transduction , Wortmannin
11.
Nat Rev Mol Cell Biol ; 13(6): 383-96, 2012 May 23.
Article in English | MEDLINE | ID: mdl-22617471

ABSTRACT

Despite daily fasting and feeding, plasma glucose levels are normally maintained within a narrow range owing to the hormones insulin and glucagon. Insulin increases glucose uptake into fat and muscle cells through the regulated trafficking of vesicles that contain glucose transporter type 4 (GLUT4). New insights into insulin signalling reveal that phosphorylation events initiated by the insulin receptor regulate key GLUT4 trafficking proteins, including small GTPases, tethering complexes and the vesicle fusion machinery. These proteins, in turn, control GLUT4 movement through the endosomal system, formation and retention of specialized GLUT4 storage vesicles and targeted exocytosis of these vesicles. Understanding these processes may help to explain the development of insulin resistance in type 2 diabetes and provide new potential therapeutic targets.


Subject(s)
Glucose Transporter Type 4/metabolism , Glucose/metabolism , Insulin/metabolism , Animals , Biological Transport , Endocytosis , Humans , Signal Transduction
12.
Nat Cell Biol ; 13(5): 580-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21516108

ABSTRACT

The exocyst complex tethers vesicles at sites of fusion through interactions with small GTPases. The G protein RalA resides on Glut4 vesicles, and binds to the exocyst after activation by insulin, but must then disengage to ensure continuous exocytosis. Here we report that, after recognition of the exocyst by activated RalA, disengagement occurs through phosphorylation of its effector Sec5, rather than RalA inactivation. Sec5 undergoes phosphorylation in the G-protein binding domain, allosterically reducing RalA interaction. The phosphorylation event is catalysed by protein kinase C and is reversed by an exocyst-associated phosphatase. Introduction of Sec5 bearing mutations of the phosphorylation site to either alanine or aspartate disrupts insulin-stimulated Glut4 exocytosis, as well as other trafficking processes in polarized epithelial cells and during development of zebrafish embryos. The exocyst thus serves as a 'gatekeeper' for exocytic vesicles through a circuit of engagement, disengagement and re-engagement with G proteins.


Subject(s)
Exocytosis , Allosteric Regulation , Animals , Biocatalysis , GTP Phosphohydrolases/metabolism , Phosphorylation , Protein Kinase C/metabolism , Zebrafish
13.
Mol Biol Cell ; 22(1): 141-52, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21148297

ABSTRACT

Insulin stimulates glucose transport in muscle and adipose tissue by translocation of glucose transporter 4 (GLUT4) to the plasma membrane. We previously reported that activation of the small GTPase RalA downstream of PI 3-kinase plays a critical role in this process by mobilizing the exocyst complex for GLUT4 vesicle targeting in adipocytes. Here we report the identification and characterization of a Ral GAP complex (RGC) that mediates the activation of RalA downstream of the PI 3-kinase/Akt pathway. The complex is composed of an RGC1 regulatory subunit and an RGC2 catalytic subunit (previously identified as AS250) that directly stimulates the guanosine triphosphate hydrolysis of RalA. Knockdown of RGC proteins leads to increased RalA activity and glucose uptake in adipocytes. Insulin inhibits the GAP complex through Akt2-catalyzed phosphorylation of RGC2 in vitro and in vivo, while activated Akt relieves the inhibitory effect of RGC proteins on RalA activity. The RGC complex thus connects PI 3-kinase/Akt activity to the transport machineries responsible for GLUT4 translocation.


Subject(s)
GTPase-Activating Proteins/metabolism , Insulin/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , ral GTP-Binding Proteins/metabolism , Adipocytes/metabolism , Animals , Blotting, Western , COS Cells , Catalytic Domain , Chlorocebus aethiops , Gene Knockdown Techniques , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Humans , Mice , NIH 3T3 Cells , Phosphorylation , ral GTP-Binding Proteins/genetics
14.
Dev Cell ; 13(3): 391-404, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17765682

ABSTRACT

Insulin stimulates glucose transport in muscle and adipose tissue by producing translocation of the glucose transporter Glut4. The exocyst, an evolutionarily conserved vesicle tethering complex, is crucial for targeting Glut4 to the plasma membrane. Here we report that insulin regulates this process via the G protein RalA, which is present in Glut4 vesicles and interacts with the exocyst in adipocytes. Insulin stimulates the activity of RalA in a PI 3-kinase-dependent manner. Disruption of RalA function by dominant-negative mutants or siRNA-mediated knockdown attenuates insulin-stimulated glucose transport. RalA also interacts with Myo1c, a molecular motor implicated in Glut4 trafficking. This interaction is modulated by Calmodulin, which functions as the light chain for Myo1c during insulin-stimulated glucose uptake. Thus, RalA serves two functions in insulin action: as a cargo receptor for the Myo1c motor, and as a signal for the unification of the exocyst to target Glut4 vesicles to the plasma membrane.


Subject(s)
Cell Membrane/drug effects , Exocytosis/drug effects , GTPase-Activating Proteins/metabolism , Glucose Transporter Type 4/metabolism , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , CHO Cells , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Cricetinae , Cricetulus , Exocytosis/physiology , Lentivirus/genetics , Mice , Models, Biological , Myosin Type I , Myosins/metabolism , Protein Transport/drug effects , RNA, Small Interfering/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...