498 results on '"rab GTP-Binding Proteins physiology"'
Search Results
2. Mevalonate pathway orchestrates insulin signaling via RAB14 geranylgeranylation-mediated phosphorylation of AKT to regulate hepatic glucose metabolism.
- Author
-
Wang L, Zhu L, Zheng Z, Meng L, Liu H, Wang K, Chen J, Li P, and Yang H
- Subjects
- Animals, Diterpenes metabolism, Hep G2 Cells, Humans, Insulin Resistance, Male, Mechanistic Target of Rapamycin Complex 2 physiology, Mice, Mice, Inbred C57BL, Phosphorylation, Signal Transduction, Simvastatin pharmacology, Transferases antagonists & inhibitors, Glucose metabolism, Insulin pharmacology, Liver metabolism, Mevalonic Acid metabolism, Proto-Oncogene Proteins c-akt metabolism, rab GTP-Binding Proteins physiology
- Abstract
Statin use accompanies with increased risk of new onset of type 2 diabetes, however, the underlying mechanisms remain not be fully understood and effective prevention strategies are still lacking. Herein, we find that both pharmacological and genetic inhibition of GGTase II mimic the disruption of simvastatin on hepatic insulin signaling and glucose metabolism in vitro. AAV8-mediated knockdown of liver RABGGTA, the specific subunit of GGTase II, triggers systemic glucose metabolism disorders in vivo. By adopting a small-scale siRNA screening, we identify RAB14 as a regulator of hepatic insulin signaling and glucose metabolism. Geranylgeranylation deficiency of RAB14 inhibits the phosphorylation of AKT (Ser473) and disrupts hepatic insulin signaling and glucose metabolism possibly via impeding mTORC2 complex assembly. Finally, geranylgeranyl pyrophosphate (GGPP) supplementation is sufficient to prevent simvastatin-caused disruption of hepatic insulin signaling and glucose metabolism in vitro. Geranylgeraniol (GGOH), a precursor of GGPP, is able to ameliorate simvastatin-induced systemic glucose metabolism disorders in vivo. In conclusion, our data indicate that statins-targeted mevalonate pathway regulates hepatic insulin signaling and glucose metabolism via geranylgeranylation of RAB14. GGPP/GGOH supplementation might be an effective strategy for the prevention of the diabetic effects of statins., Competing Interests: Declaration of competing interest The authors declare that they have no conflict of interests., (Copyright © 2021. Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
3. Rab35 regulates insulin secretion via phogrin in pancreatic β cells.
- Author
-
Lu C, Zhao Q, Wang D, Feng Y, Feng L, Li Z, and Shi Q
- Subjects
- HEK293 Cells, Humans, Insulin-Secreting Cells physiology, Receptor-Like Protein Tyrosine Phosphatases, Class 8 metabolism, rab GTP-Binding Proteins metabolism, Insulin metabolism, Insulin-Secreting Cells metabolism, Receptor-Like Protein Tyrosine Phosphatases, Class 8 physiology, rab GTP-Binding Proteins physiology
- Abstract
Dysfunction of pancreatic β cell insulin secretion is related to the pathogenesis of type 2 diabetes (T2D). Rab proteins have been shown to be key players in insulin secretion by pancreatic β cells, and phogrin is a marker for the processes of exocytosis and insulin secretion. The purposes of this study were to clarify the regulatory role of Rab35 in insulin secretion and analyse the Rab35/phogrin interaction mechanism in β-TC-6 cells. We studied the effects of Rab35 gene overexpression and interference on insulin secretion and phogrin expression and levels in β-TC-6 cells. The Rab35/phogrin interaction was verified by GST pulldown, co-IP and co-localisation experiments. Here, we report that Rab35 is mainly distributed in the β-TC-6-cell plasma membrane and cytoplasm. Rab35 overexpression promotes insulin secretion and decreases phogrin expression in β-TC-6 cells, whereas its silencing significantly inhibits insulin secretion, promotes phogrin expression (p < 0.05) and causes phogrin redistribution. Furthermore, Rab35 silencing suppresses exocytosis of insulin. Rab35 interacts with phogrin, and both proteins co-localise in the plasma membranes and cytoplasm of β-TC-6 cells. Our study presents novel evidence that Rab35 regulates insulin secretion by inhibiting phogrin expression and causing intracellular phogrin redistribution in pancreatic β cells., (© 2021 John Wiley & Sons Australia, Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
4. Extracellular CIRP-Impaired Rab26 Restrains EPOR-Mediated Macrophage Polarization in Acute Lung Injury.
- Author
-
Zhang W, Wang Y, Li C, Xu Y, Wang X, Wu D, Gao Z, Qian H, You Z, Zhang Z, He B, and Wang G
- Subjects
- Animals, Cell Polarity, Cells, Cultured, Inflammation etiology, Mice, Mice, Inbred C57BL, PPAR gamma physiology, Acute Lung Injury immunology, Macrophages physiology, RNA-Binding Proteins physiology, Receptors, Erythropoietin physiology, rab GTP-Binding Proteins physiology
- Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a condition with an imbalanced inflammatory response and delayed resolution of inflammation. Macrophage polarization plays an important role in inflammation and resolution. However, the mechanism of macrophage polarization in ALI/ARDS is not fully understood. We found that mice with lipopolysaccharide administration developed lung injury with the accumulation of extracellular cold-inducible RNA-binding protein (eCIRP) in the lungs. eCIRP, as a damage-associated molecular pattern (DAMP), inhibited M2 macrophage polarization, thereby tipping the balance toward inflammation rather than resolution. Anti-CIRP antibodies reversed such phenotypes. The levels of macrophage erythropoietin (EPO) receptor (EPOR) were reduced after eCIRP treatment. Myeloid-specific EPOR-deficient mice displayed restrained M2 macrophage polarization and impaired inflammation resolution. Mechanistically, eCIRP impaired Rab26, a member of Ras superfamilies of small G proteins, and reduced the transportation of surface EPOR, which resulted in macrophage polarization toward the M1 phenotype. Moreover, EPO treatment hardly promotes M2 polarization in Rab26 knockout (KO) macrophages through EPOR. Collectively, macrophage EPOR signaling is impaired by eCIRP through Rab26 during ALI/ARDS, leading to the restrained M2 macrophage polarization and delayed inflammation resolution. These findings identify a mechanism of persistent inflammation and a potential therapy during ALI/ARDS., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Zhang, Wang, Li, Xu, Wang, Wu, Gao, Qian, You, Zhang, He and Wang.)
- Published
- 2021
- Full Text
- View/download PDF
5. RAB39B-mediated trafficking of the GluA2-AMPAR subunit controls dendritic spine maturation and intellectual disability-related behaviour.
- Author
-
Mignogna ML, Musardo S, Ranieri G, Gelmini S, Espinosa P, Marra P, Belloli S, Murtaj V, Moresco RM, Bellone C, and D'Adamo P
- Subjects
- Animals, Calcium, Mice, Neuronal Plasticity, Neurons physiology, Receptors, Glutamate physiology, Dendritic Spines physiology, Intellectual Disability, rab GTP-Binding Proteins physiology
- Abstract
Mutations in the RAB39B gene cause X-linked intellectual disability (XLID), comorbid with autism spectrum disorders or early Parkinson's disease. One of the functions of the neuronal small GTPase RAB39B is to drive GluA2/GluA3 α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) maturation and trafficking, determining AMPAR subunit composition at glutamatergic postsynaptic neuronal terminals. Taking advantage of the Rab39b knockout murine model, we show that a lack of RAB39B affects neuronal dendritic spine refinement, prompting a more Ca
2+ -permeable and excitable synaptic network, which correlates with an immature spine arrangement and behavioural and cognitive alterations in adult mice. The persistence of immature circuits is triggered by increased hypermobility of the spine, which is restored by the Ca2+ -permeable AMPAR antagonist NASPM. Together, these data confirm that RAB39B controls AMPAR trafficking, which in turn plays a pivotal role in neuronal dendritic spine remodelling and that targeting Ca2+ -permeable AMPARs may highlight future pharmaceutical interventions for RAB39B-associated disease conditions., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
6. RABL2A-CCDC34 Axis Promotes Sorafenib Resistance in Hepatocellular Carcinoma.
- Author
-
Zhou M, Chen X, Bai H, Sun Y, Zhang Z, Li S, Wang X, and Zeng M
- Subjects
- Antigens, Neoplasm physiology, Antineoplastic Agents pharmacology, Carcinoma, Hepatocellular drug therapy, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, China, Drug Resistance, Neoplasm physiology, Gene Expression drug effects, Gene Expression genetics, Gene Expression Regulation, Neoplastic drug effects, Humans, Liver Neoplasms drug therapy, Liver Neoplasms metabolism, Neoplasm Proteins physiology, Signal Transduction drug effects, Sorafenib pharmacology, rab GTP-Binding Proteins metabolism, rab GTP-Binding Proteins physiology, Antigens, Neoplasm metabolism, Carcinoma, Hepatocellular metabolism, Neoplasm Proteins metabolism, Sorafenib metabolism
- Abstract
In this study, we examined the regulatory role of CCDC34 in the sorafenib sensitivity of hepatocellular carcinoma (HCC) and its functional partners. Wide-type Huh7 and Hep3B and induced sorafenib-resistant (SR) Huh7/SR and Hep3B/SR cells were used as in vitro cell models. Immunofluorescent staining and coimmunoprecipitation were performed to check protein-protein interaction. Cell Counting Kit-8 (CCK-8), terminal deoxynucleotidyl transferase-mediated nick end labeling (TUNEL), PI/Annexin V staining, and western blot analysis were performed to assess cell response to sorafenib. The results showed that CCDC34 upregulation in HCC was associated with poor survival. Huh7/SR and Hep3B/SR cells had significantly higher CCDC34 expression than the parental cell lines. RABL2A expression was significantly upregulated in SR HCC cells and interacted with CCDC34 in its GTP-bound state in Huh7/SR and Hep3B/SR cells. RABL2A depletion sensitized Huh7/SR and Hep3B/SR cells to sorafenib. RABL2A Q80L mutant (GTP-bound state locked), but not S35N mutant (GDP-bound state locked) overexpression increased sorafenib IC50 of Huh7 and Hep3B cells. CCDC34 depletion nearly abrogated the protective effects of RABL2A Q80L overexpression both in vitro and in vivo . RABL2A Q80L overexpression significantly increased the expression of p-p38 and p-JNK, the effects of which were significantly attenuated by CCDC34 depletion. In summary, we infer that the RABL2A-CCDC34 axis plays an important role in mediating p38/MAPK and JNK/MAPK signaling, thereby contributing to acquired sorafenib resistance in HCC.
- Published
- 2021
- Full Text
- View/download PDF
7. Tombusviruses Target a Major Crossroad in the Endocytic and Recycling Pathways via Co-opting Rab7 Small GTPase.
- Author
-
Feng Z, Inaba JI, and Nagy PD
- Subjects
- 1-Phosphatidylinositol 4-Kinase metabolism, Endosomes metabolism, Gene Knockdown Techniques, Guanine Nucleotide Exchange Factors physiology, Host Microbial Interactions, Organelles metabolism, Plant Diseases virology, Protein Binding, Protein Transport, Saccharomyces cerevisiae Proteins metabolism, Saccharomyces cerevisiae Proteins physiology, Sorting Nexins metabolism, Organelles virology, Saccharomyces cerevisiae virology, Nicotiana virology, Tombusvirus physiology, Viral Proteins metabolism, Virus Replication, rab GTP-Binding Proteins physiology
- Abstract
Positive-strand RNA viruses induce the biogenesis of unique membranous organelles called viral replication organelles (VROs), which perform virus replication in infected cells. Tombusviruses have been shown to rewire cellular trafficking and metabolic pathways, remodel host membranes, and recruit multiple host factors to support viral replication. In this work, we demonstrate that tomato bushy stunt virus (TBSV) and the closely related carnation Italian ringspot virus (CIRV) usurp Rab7 small GTPase to facilitate building VROs in the surrogate host yeast and in plants. Depletion of Rab7 small GTPase, which is needed for late endosome and retromer biogenesis, strongly inhibits TBSV and CIRV replication in yeast and in planta. The viral p33 replication protein interacts with Rab7 small GTPase, which results in the relocalization of Rab7 into the large VROs. Similar to the depletion of Rab7, the deletion of either MON1 or CCZ1 heterodimeric GEFs (guanine nucleotide exchange factors) of Rab7 inhibited TBSV RNA replication in yeast. This suggests that the activated Rab7 has proviral functions. We show that the proviral function of Rab7 is to facilitate the recruitment of the retromer complex and the endosomal sorting nexin-BAR proteins into VROs. We demonstrate that TBSV p33-driven retargeting of Rab7 into VROs results in the delivery of several retromer cargos with proviral functions. These proteins include lipid enzymes, such as Vps34 PI3K (phosphatidylinositol 3-kinase), PI4Kα-like Stt4 phosphatidylinositol 4-kinase, and Psd2 phosphatidylserine decarboxylase. In summary, based on these and previous findings, we propose that subversion of Rab7 into VROs allows tombusviruses to reroute endocytic and recycling trafficking to support virus replication. IMPORTANCE The replication of positive-strand RNA viruses depends on the biogenesis of viral replication organelles (VROs). However, the formation of membranous VROs is not well understood yet. Using tombusviruses and the model host yeast, we discovered that the endosomal Rab7 small GTPase is critical for the formation of VROs. Interaction between Rab7 and the TBSV p33 replication protein leads to the recruitment of Rab7 into VROs. TBSV-driven usurping of Rab7 has proviral functions through facilitating the delivery of the co-opted retromer complex, sorting nexin-BAR proteins, and lipid enzymes into VROs to create an optimal milieu for virus replication. These results open up the possibility that controlling cellular Rab7 activities in infected cells could be a target for new antiviral strategies.
- Published
- 2021
- Full Text
- View/download PDF
8. Leucine zipper transcription factor-like 1 (LZTFL1), an intraflagellar transporter protein 27 (IFT27) associated protein, is required for normal sperm function and male fertility.
- Author
-
Huang Q, Li W, Zhou Q, Awasthi P, Cazin C, Yap Y, Mladenovic-Lucas L, Hu B, Jeyasuria P, Zhang L, Granneman JG, Hess RA, Ray PF, Kherraf ZE, Natarajan V, and Zhang Z
- Subjects
- Animals, CHO Cells, COS Cells, Chlorocebus aethiops, Cricetulus, Female, Fertility genetics, HEK293 Cells, Humans, Male, Mice, Knockout, Protein Binding, RNA, Messenger metabolism, Spermatogenesis genetics, Spermatogenesis physiology, Transcription Factors genetics, rab GTP-Binding Proteins physiology, Mice, Fertility physiology, Spermatozoa physiology, Transcription Factors physiology
- Abstract
Intraflagellar transport (IFT) is an evolutionarily conserved mechanism essential for the assembly and maintenance of most eukaryotic cilia and flagella, including mammalian sperm tails. Depletion of IFT27, a component of the IFT complex, in male germ cells results in infertility associated with disrupted sperm flagella structure and motility. Leucine zipper transcription factor-like 1 (LZTFL1) is an IFT27 associated protein. LZTFL1, also known as BBS17, is a Bardet-Biedl syndrome (BBS) associated protein. Patients carrying biallelic variants of LZTFL1 gene exhibit the common BBS phenotypes. The global Lztfl1 knockout mice showed abnormal growth rate and retinal degeneration, typical of BBS phenotype. However, it is not clear if Lztfl1 has a role in male fertility. The LZTFL1 protein is highly and predominantly expressed in mouse testis. During the first wave of spermatogenesis, the protein is only expressed during spermiogenesis phase from the round spermatid stage and displays a cytoplasmic localization with a vesicular distribution pattern. At the elongated spermatid stage, LZTFL1 is present in the developing flagella and appears also close to the manchette. Fertility of Lztfl1 knockout mice was significantly reduced and associated with low sperm motility and a high level of abnormal sperm (astheno-teratozoospermia). In vitro assessment of fertility revealed reduced fertilization and embryonic development when using sperm from homozygous mutant mice. In addition, we observed a significant decrease of the testicular IFT27 protein level in Lztfl1 mutant mice contrasting with a stable expression levels of other IFT proteins, including IFT20, IFT81, IFT88 and IFT140. Overall, our results support strongly the important role of LZTFL1 in mouse spermatogenesis and male fertility., Competing Interests: Declaration of competing interest The authors declare that they have no known competing interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
9. SopD from Salmonella specifically inactivates Rab8.
- Author
-
Savitskiy S and Itzen A
- Subjects
- Bacterial Proteins chemistry, Bacterial Proteins physiology, Protein Binding physiology, Protein Interaction Mapping methods, Protein Transport, Salmonella metabolism, Virulence, rab GTP-Binding Proteins physiology, Bacterial Proteins metabolism, rab GTP-Binding Proteins metabolism
- Abstract
Salmonella outer protein D (SopD) is secreted into a host during the first stages of the Salmonella infection and contributes to the systemic virulence of the bacterium. SopD2 is a SopD homolog and possesses GTPase activating protein (GAP) activity towards Rab32. Here, we identified Rab-proteins as putative SopD-targets using a yeast two-hybrid approach. In vitro investigations subsequently revealed Rab8a as an exclusive SopD substrate in contrast to SopD2, which has a broader specificity targeting Rab29, Rab32 and Rab38 in vitro. Additionally, we determined the catalytic efficiencies of SopD and SopD2 towards their physiologically relevant substrates. Moreover, mutagenesis studies provided insights into possible key residues of the Rab-protein and the GAP involved in the conversion of active to inactive GTPase. In conclusion, we demonstrate that Salmonella SopD and SopD2 act as RabGAPs and can inactivate Rab signaling., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
10. FOXO1-Mediated Downregulation of RAB27B Leads to Decreased Exosome Secretion in Diabetic Kidneys.
- Author
-
Zeng M, Wen J, Ma Z, Xiao L, Liu Y, Kwon S, Liu Y, and Dong Z
- Subjects
- Animals, Cells, Cultured, Down-Regulation, Glomerular Filtration Rate, Male, Mice, Mice, Inbred C57BL, Phosphorylation, Diabetic Nephropathies etiology, Exosomes physiology, Forkhead Box Protein O1 physiology, rab GTP-Binding Proteins physiology
- Abstract
Exosomes have been implicated in diabetic kidney disease (DKD), but the regulation of exosomes in DKD is largely unknown. Here, we have verified the decrease of exosome secretion in DKD and unveiled the underlying mechanism. In Boston University mouse proximal tubule (BUMPT) cells, high-glucose (HG) treatment led to a significant decrease in exosome secretion, which was associated with specific downregulation of RAB27B, a key guanosine-5'-triphosphatase in exosome secretion. Overexpression of RAB27B restored exosome secretion in HG-treated cells, suggesting a role of RAB27B downregulation in the decrease of exosome secretion in DKD. To understand the mechanism of RAB27B downregulation, we conducted bioinformatics analysis that identified FOXO1 binding sites in the Rab27b gene promoter. Consistently, HG induced phosphorylation of FOXO1 in BUMPT cells, preventing FOXO1 accumulation and activation in the nucleus. Overexpression of nonphosphorylatable, constitutively active FOXO1 led to the upregulation of RAB27B and an increase in exosome secretion in HG-treated cells. In vivo, compared with normal mice, diabetic mice showed increased FOXO1 phosphorylation, decreased RAB27B expression, and reduced exosome secretion. Collectively, these results unveil the mechanism of exosome dysfunction in DKD where FOXO1 is phosphorylated and inactivated in DKD, resulting in RAB27B downregulation and the decrease of exosome secretion., (© 2021 by the American Diabetes Association.)
- Published
- 2021
- Full Text
- View/download PDF
11. The calcium sensing receptor (CaSR) promotes Rab27B expression and activity to control secretion in breast cancer cells.
- Author
-
Zavala-Barrera C, Del-Río-Robles JE, García-Jiménez I, Egusquiza-Alvarez CA, Hernández-Maldonado JP, Vázquez-Prado J, and Reyes-Cruz G
- Subjects
- Calcium metabolism, Cell Line, Tumor, Chemokines metabolism, Chemotaxis, Cyclic AMP-Dependent Protein Kinases, Cytokines metabolism, Death Domain Receptor Signaling Adaptor Proteins metabolism, Female, Gene Expression genetics, Gene Expression Regulation, Neoplastic genetics, Guanine Nucleotide Exchange Factors metabolism, Humans, Phosphatidylinositol 3-Kinase, Receptors, Calcium-Sensing physiology, Secretory Pathway physiology, TOR Serine-Threonine Kinases, Tumor Microenvironment, rab GTP-Binding Proteins physiology, Breast Neoplasms metabolism, Receptors, Calcium-Sensing metabolism, rab GTP-Binding Proteins metabolism
- Abstract
Chemotactic and angiogenic factors secreted within the tumor microenvironment eventually facilitate the metastatic dissemination of cancer cells. Calcium-sensing receptor (CaSR) activates secretory pathways in breast cancer cells via a mechanism driven by vesicular trafficking of this receptor. However, it remains to be elucidated how endosomal proteins in secretory vesicles are controlled by CaSR. In the present study, we demonstrate that CaSR promotes expression of Rab27B and activates this secretory small GTPase via PI3K, PKA, mTOR and MADD, a guanine nucleotide exchange factor, also known as DENN/Rab3GEP. Active Rab27B leads secretion of various cytokines and chemokines, including IL-6, IL-1β, IL-8, IP-10 and RANTES. Expression of Rab27B is stimulated by CaSR in MDA-MB-231 and MCF-7 breast epithelial cancer cells, but not in non-cancerous MCF-10A cells. This regulatory mechanism also occurs in HeLa and PC3 cells. Our findings provide insightful information regarding how CaSR activates a Rab27B-dependent mechanism to control secretion of factors known to intervene in paracrine communication circuits within the tumor microenvironment., (Copyright © 2021. Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
12. Arf1 orchestrates Rab GTPase conversion at the trans -Golgi network.
- Author
-
Thomas LL, Highland CM, and Fromme JC
- Subjects
- ADP-Ribosylation Factor 1 genetics, GTPase-Activating Proteins metabolism, Golgi Apparatus metabolism, Monomeric GTP-Binding Proteins metabolism, Protein Transport physiology, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins metabolism, Vesicular Transport Proteins metabolism, rab GTP-Binding Proteins physiology, trans-Golgi Network metabolism, ADP-Ribosylation Factor 1 metabolism, rab GTP-Binding Proteins metabolism, trans-Golgi Network physiology
- Abstract
Rab family GTPases are key organizers of membrane trafficking and function as markers of organelle identity. Accordingly, Rab GTPases often occupy specific membrane domains, and mechanisms exist to prevent the inappropriate mixing of distinct Rab domains. The yeast Golgi complex can be divided into two broad Rab domains: Ypt1 (Rab1) and Ypt6 (Rab6) are present at the early/medial Golgi and sharply transition to Ypt31/32 (Rab11) at the late Golgi/ trans -Golgi network (TGN). This Rab conversion has been attributed to GTPase-activating protein (GAP) cascades in which Ypt31/32 recruits the Rab-GAPs Gyp1 and Gyp6 to inactivate Ypt1 and Ypt6, respectively. Here we report that Rab transition at the TGN involves additional layers of regulation. We provide new evidence confirming the TRAPPII complex as an important regulator of Ypt6 inactivation and uncover an unexpected role of the Arf1 GTPase in recruiting Gyp1 to drive Ypt1 inactivation at the TGN. Given its established role in directly recruiting TRAPPII to the TGN, Arf1 is therefore a master regulator of Rab conversion on maturing Golgi compartments.
- Published
- 2021
- Full Text
- View/download PDF
13. JRAB/MICAL-L2 undergoes liquid-liquid phase separation to form tubular recycling endosomes.
- Author
-
Sakane A, Yano TA, Uchihashi T, Horikawa K, Hara Y, Imoto I, Kurisu S, Yamada H, Takei K, and Sasaki T
- Subjects
- Cytoskeletal Proteins metabolism, Endocytosis physiology, Endosomes physiology, HEK293 Cells, HeLa Cells, Humans, Microfilament Proteins physiology, Protein Binding physiology, Protein Transport physiology, Tight Junctions physiology, rab GTP-Binding Proteins metabolism, rab GTP-Binding Proteins physiology, Endosomes metabolism, Microfilament Proteins metabolism
- Abstract
Elongated tubular endosomes play essential roles in diverse cellular functions. Multiple molecules have been implicated in tubulation of recycling endosomes, but the mechanism of endosomal tubule biogenesis has remained unclear. In this study, we found that JRAB/MICAL-L2 induces endosomal tubulation via activated Rab8A. In association with Rab8A, JRAB/MICAL-L2 adopts its closed form, which functions in the tubulation of recycling endosomes. Moreover, JRAB/MICAL-L2 induces liquid-liquid phase separation, initiating the formation of tubular recycling endosomes upon overexpression. Between its N-terminal and C-terminal globular domains, JRAB/MICAL-L2 contains an intrinsically disordered region, which contributes to the formation of JRAB/MICAL-L2 condensates. Based on our findings, we propose that JRAB/MICAL-L2 plays two sequential roles in the biogenesis of tubular recycling endosomes: first, JRAB/MICAL-L2 organizes phase separation, and then the closed form of JRAB/MICAL-L2 formed by interaction with Rab8A promotes endosomal tubulation.
- Published
- 2021
- Full Text
- View/download PDF
14. Regulation of apical constriction via microtubule- and Rab11-dependent apical transport during tissue invagination.
- Author
-
Le TP and Chung S
- Subjects
- Actin Cytoskeleton physiology, Actomyosin physiology, Animals, Biological Transport, Cadherins physiology, Drosophila Proteins genetics, Dyneins physiology, Gastrulation, Gene Knockdown Techniques, Intercellular Junctions physiology, Myosins physiology, Nuclear Proteins physiology, rab GTP-Binding Proteins genetics, Drosophila embryology, Drosophila Proteins physiology, Microtubules physiology, Salivary Glands embryology, rab GTP-Binding Proteins physiology
- Abstract
The formation of an epithelial tube is a fundamental process for organogenesis. During Drosophila embryonic salivary gland (SG) invagination, Folded gastrulation (Fog)-dependent Rho-associated kinase (Rok) promotes contractile apical myosin formation to drive apical constriction. Microtubules (MTs) are also crucial for this process and are required for forming and maintaining apicomedial myosin. However, the underlying mechanism that coordinates actomyosin and MT networks still remains elusive. Here, we show that MT-dependent intracellular trafficking regulates apical constriction during SG invagination. Key components involved in protein trafficking, such as Rab11 and Nuclear fallout (Nuf), are apically enriched near the SG invagination pit in a MT-dependent manner. Disruption of the MT networks or knockdown of Rab11 impairs apicomedial myosin formation and apical constriction. We show that MTs and Rab11 are required for apical enrichment of the Fog ligand and the continuous distribution of the apical determinant protein Crumbs (Crb) and the key adherens junction protein E-Cadherin (E-Cad) along junctions. Targeted knockdown of crb or E-Cad in the SG disrupts apical myosin networks and results in apical constriction defects. Our data suggest a role of MT- and Rab11-dependent intracellular trafficking in regulating actomyosin networks and cell junctions to coordinate cell behaviors during tubular organ formation.
- Published
- 2021
- Full Text
- View/download PDF
15. A unique AtSar1D-AtRabD2a nexus modulates autophagosome biogenesis in Arabidopsis thaliana .
- Author
-
Zeng Y, Li B, Ji C, Feng L, Niu F, Deng C, Chen S, Lin Y, Cheung KCP, Shen J, Wong KB, and Jiang L
- Subjects
- Arabidopsis metabolism, Arabidopsis Proteins physiology, Autophagosomes metabolism, Autophagy physiology, COP-Coated Vesicles physiology, Endoplasmic Reticulum metabolism, Golgi Apparatus metabolism, Phagosomes metabolism, Protein Transport physiology, Proteomics methods, R-SNARE Proteins physiology, Vacuoles metabolism, Vesicular Transport Proteins metabolism, rab GTP-Binding Proteins physiology, Arabidopsis Proteins metabolism, COP-Coated Vesicles metabolism, R-SNARE Proteins metabolism, rab GTP-Binding Proteins metabolism
- Abstract
In eukaryotes, secretory proteins traffic from the endoplasmic reticulum (ER) to the Golgi apparatus via coat protein complex II (COPII) vesicles. Intriguingly, during nutrient starvation, the COPII machinery acts constructively as a membrane source for autophagosomes during autophagy to maintain cellular homeostasis by recycling intermediate metabolites. In higher plants, essential roles of autophagy have been implicated in plant development and stress responses. Nonetheless, the membrane sources of autophagosomes, especially the participation of the COPII machinery in the autophagic pathway and autophagosome biogenesis, remains elusive in plants. Here, we provided evidence in support of a novel role of a specific Sar1 homolog AtSar1d in plant autophagy in concert with a unique Rab1/Ypt1 homolog AtRabD2a. First, proteomic analysis of the plant ATG (autophagy-related gene) interactome uncovered the mechanistic connections between ATG machinery and specific COPII components including AtSar1d and Sec23s, while a dominant negative mutant of AtSar1d exhibited distinct inhibition on YFP-ATG8 vacuolar degradation upon autophagic induction. Second, a transfer DNA insertion mutant of AtSar1d displayed starvation-related phenotypes. Third, AtSar1d regulated autophagosome progression through specific recognition of ATG8e by a noncanonical motif. Fourth, we demonstrated that a plant-unique Rab1/Ypt1 homolog AtRabD2a coordinates with AtSar1d to function as the molecular switch in mediating the COPII functions in the autophagy pathway. AtRabD2a appears to be essential for bridging the specific AtSar1d-positive COPII vesicles to the autophagy initiation complex and therefore contributes to autophagosome formation in plants. Taken together, we identified a plant-specific nexus of AtSar1d-AtRabD2a in regulating autophagosome biogenesis., Competing Interests: The authors declare no competing interest.
- Published
- 2021
- Full Text
- View/download PDF
16. Rab14/MACF2 complex regulates endosomal targeting during cytokinesis.
- Author
-
Gibieža P, Peterman E, Hoffman HK, Van Engeleburg S, Skeberdis VA, and Prekeris R
- Subjects
- Actins metabolism, Endosomal Sorting Complexes Required for Transport metabolism, Endosomes metabolism, Endosomes physiology, HeLa Cells, Humans, Microtubule-Associated Proteins physiology, Microtubules physiology, rab GTP-Binding Proteins physiology, Cytokinesis physiology, Microtubule-Associated Proteins metabolism, rab GTP-Binding Proteins metabolism
- Abstract
Abscission is a complex cellular process that is required for mitotic division. It is well established that coordinated and localized changes in actin and microtubule dynamics are vital for cytokinetic ring formation, as well as establishment of the abscission site. Actin cytoskeleton reorganization during abscission would not be possible without the interplay between Rab11- and Rab35-containing endosomes and their effector proteins, whose roles in regulating endocytic pathways at the cleavage furrow have now been studied extensively. Here, we identified Rab14 as a novel regulator of cytokinesis. We demonstrate that depletion of Rab14 causes either cytokinesis failure or significantly prolongs division time. We show that Rab14 contributes to the efficiency of recruiting Rab11-endosomes to the thin intracellular bridge (ICB) microtubules and that Rab14 knockout leads to inhibition of actin clearance at the abscission site. Finally, we demonstrate that Rab14 binds to microtubule minus-end interacting MACF2/CAMSAP3 complex and that this binding affects targeting of endosomes to the ICB microtubules. Collectively, our data identified Rab14 and MACF2/CAMSAP3 as proteins that regulate actin depolymerization and endosome targeting during cytokinesis.
- Published
- 2021
- Full Text
- View/download PDF
17. A role for Rab30 in retrograde trafficking and maintenance of endosome-TGN organization.
- Author
-
Zulkefli KL, Mahmoud IS, Williamson NA, Gosavi PK, Houghton FJ, and Gleeson PA
- Subjects
- Antigens, CD metabolism, Endosomes genetics, Golgi Apparatus metabolism, HeLa Cells, Humans, Protein Transport genetics, Receptors, Transferrin metabolism, rab GTP-Binding Proteins genetics, trans-Golgi Network metabolism, Endosomes metabolism, rab GTP-Binding Proteins physiology, trans-Golgi Network genetics
- Abstract
Rab30 is a poorly characterized small GTPase. Here we show that Rab30 is localised primarily to the TGN and recycling endosomes in a range of cell types, including primary neurons; minor levels of Rab30 were also detected throughout the Golgi stack and early endosomes. Silencing of Rab30 resulted in the dispersal of both early and recycling endosomes and TGN compartments in HeLa cells. By analyzing cargo trafficking in Rab30-silenced and Rab30-overexpressing HeLa cells, we demonstrate that Rab30 plays a role in retrograde trafficking of TGN38 from endosomes to the Golgi, but has no apparent role in the endocytic recycling of the transferrin receptor to the plasma membrane. Five interactive partners with Rab30 were identified by pull-down and MS analysis using GFP-tagged Rab30 mutant, Rab30(Q68L). Two of the interactive partners identified were Arf1 and Arf4, known regulators of endosome to TGN retrograde transport. Knockdown of Arf1 and Arf4 results in GFP-Rab30 decorated tubules arising from the recycling endosomes, suggesting association of Rab30 with tubular carriers. Overall our data demonstrates a role for Rab30 in regulating retrograde transport to the TGN and maintenance of endosomal-TGN organization., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
18. RAB7L1 Participates in Secondary Brain Injury Induced by Experimental Intracerebral Hemorrhage in Rats.
- Author
-
Tan X, Wei Y, Cao J, Wu D, Lai N, Deng R, Li H, Shen H, Peng Y, Li X, and Chen G
- Subjects
- Animals, Apoptosis, Basal Ganglia, Blood, Brain Damage, Chronic genetics, Cerebral Hemorrhage complications, Cognition Disorders etiology, Cognition Disorders genetics, Disease Models, Animal, Gene Expression Regulation, Injections, Injections, Intraventricular, Learning Disabilities etiology, Learning Disabilities genetics, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 metabolism, Male, Morris Water Maze Test, Nerve Tissue Proteins antagonists & inhibitors, Nerve Tissue Proteins biosynthesis, Nerve Tissue Proteins genetics, Neurodegenerative Diseases etiology, Neurodegenerative Diseases genetics, Neurons pathology, Phosphorylation, Protein Processing, Post-Translational, RNA, Small Interfering administration & dosage, Random Allocation, Rats, Rats, Sprague-Dawley, Stereotaxic Techniques, Up-Regulation, rab GTP-Binding Proteins antagonists & inhibitors, rab GTP-Binding Proteins biosynthesis, rab GTP-Binding Proteins genetics, Brain Damage, Chronic etiology, Cerebral Hemorrhage genetics, Nerve Tissue Proteins physiology, rab GTP-Binding Proteins physiology
- Abstract
RAB7, a member of RAS oncogene family-like 1 (RAB7L1), is a GTPase belonging to the Rab family and acts as an upstream regulator to regulate the kinase activity of leucine-rich repeat kinase 2 (LRRK2). Although LRRK2 has been shown to aggravate secondary brain injury (SBI) after intracerebral hemorrhage (ICH), it is unknown whether RAB7L1 is also involved in this process. The purpose of the present study was to investigate the role of RAB7L1 in ICH-induced SBI in vivo. Autologous blood was injected into adult male Sprague-Dawley rats to induce an ICH model in vivo. The results showed that the protein levels of RAB7L1 increased after ICH. Overexpression of RAB7L1 induced neuronal apoptosis and damage, as demonstrated by TUNEL-positive and FJB-positive cells, and exacerbated ICH-induced learning and cognitive dysfunctions; in contrast, downregulation of RAB7L1 via RNA interference yielded comparatively opposite changes in these parameters. In summary, this study demonstrates that RAB7L1 promotes SBI after ICH and may represent a potential target for ICH therapy.
- Published
- 2021
- Full Text
- View/download PDF
19. Variants in RABL2A causing male infertility and ciliopathy.
- Author
-
Ding X, Fragoza R, Singh P, Zhang S, Yu H, and Schimenti JC
- Subjects
- Animals, Ciliopathies pathology, Female, Humans, Infertility, Male pathology, Male, Mice, Phenotype, Ciliopathies etiology, Infertility, Male etiology, Polymorphism, Single Nucleotide, rab GTP-Binding Proteins genetics, rab GTP-Binding Proteins physiology
- Abstract
Approximately 7% of men worldwide suffer from infertility, with sperm abnormalities being the most common defect. Though genetic causes are thought to underlie a substantial fraction of idiopathic cases, the actual molecular bases are usually undetermined. Because the consequences of most genetic variants in populations are unknown, this complicates genetic diagnosis even after genome sequencing of patients. Some patients with ciliopathies, including primary ciliary dyskinesia and Bardet-Biedl syndrome, also suffer from infertility because cilia and sperm flagella share several characteristics. Here, we identified two deleterious alleles of RABL2A, a gene essential for normal function of cilia and flagella. Our in silico predictions and in vitro assays suggest that both alleles destabilize the protein. We constructed and analyzed mice homozygous for these two single-nucleotide polymorphisms, Rabl2L119F (rs80006029) and Rabl2V158F (rs200121688), and found that they exhibit ciliopathy-associated disorders including male infertility, early growth retardation, excessive weight gain in adulthood, heterotaxia, pre-axial polydactyly, neural tube defects and hydrocephalus. Our study provides a paradigm for triaging candidate infertility variants in the population for in vivo functional validation, using computational, in vitro and in vivo approaches., (© The Author(s) 2020. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
20. RAB25 confers resistance to chemotherapy by altering mitochondrial apoptosis signaling in ovarian cancer cells.
- Author
-
Temel SG, Giray A, Karakas B, Gul O, Kozanoglu I, Celik H, Basaga H, Acikbas U, Sucularli C, Oztop S, Aka Y, and Kutuk O
- Subjects
- Adult, Aged, Cell Cycle, Cell Line, Tumor, Cell Movement, Cell Proliferation, Female, Humans, Middle Aged, Mitochondria, Neoplasm Invasiveness, Ovarian Neoplasms drug therapy, Ovarian Neoplasms pathology, Proto-Oncogene Proteins c-bcl-2 metabolism, Signal Transduction, rab GTP-Binding Proteins metabolism, Apoptosis, Drug Resistance, Neoplasm, Ovarian Neoplasms metabolism, rab GTP-Binding Proteins physiology
- Abstract
Ovarian cancer remains one of the most frequent causes of cancer-related death in women. Many patients with ovarian cancer suffer from de novo or acquired resistance to chemotherapy. Here, we report that RAB25 suppresses chemotherapy-induced mitochondrial apoptosis signaling in ovarian cancer cell lines and primary ovarian cancer cells. RAB25 blocks chemotherapy-induced apoptosis upstream of mitochondrial outer membrane permeabilization by either increasing antiapoptotic BCL-2 proteins or decreasing proapoptotic BCL-2 proteins. In particular, BAX expression negatively correlates with RAB25 expression in ovarian cancer cells. BH3 profiling assays corroborated that RAB25 decreases mitochondrial cell death priming. Suppressing RAB25 by means of RNAi or RFP14 inhibitory hydrocarbon-stapled peptide sensitizes ovarian cancer cells to chemotherapy as well as RAB25-mediated proliferation, invasion and migration. Our data suggest that RAB25 is a potential therapeutic target for ovarian cancer.
- Published
- 2020
- Full Text
- View/download PDF
21. The Rab5-Rab11 Endosomal Pathway is Required for BDNF-Induced CREB Transcriptional Regulation in Hippocampal Neurons.
- Author
-
González-Gutiérrez A, Lazo OM, and Bronfman FC
- Subjects
- Animals, Cyclic AMP Response Element-Binding Protein genetics, Dendrites drug effects, Gene Expression Regulation drug effects, Gene Expression Regulation physiology, MAP Kinase Signaling System physiology, Phosphorylation, Primary Cell Culture, Rats, Brain-Derived Neurotrophic Factor pharmacology, Cyclic AMP Response Element-Binding Protein biosynthesis, Hippocampus metabolism, Neurons metabolism, Signal Transduction physiology, rab GTP-Binding Proteins physiology, rab5 GTP-Binding Proteins physiology
- Abstract
Brain-derived neurotrophic factor (BDNF) is a key regulator of the morphology and connectivity of central neurons. We have previously shown that BDNF/TrkB signaling regulates the activity and mobility of the GTPases Rab5 and Rab11, which in turn determine the postendocytic sorting of signaling TrkB receptors. Moreover, decreased Rab5 or Rab11 activity inhibits BDNF-induced dendritic branching. Whether Rab5 or Rab11 activity is important for local events only or for regulating nuclear signaling and gene expression is unknown. Here, we investigated, in rat hippocampal neuronal cultures derived from embryos of unknown sex, whether BDNF-induced signaling cascades are altered when early and recycling endosomes are disrupted by the expression of dominant-negative mutants of Rab5 and Rab11. The activity of both Rab5 and Rab11 was required for sustained activity of Erk1/2 and nuclear CREB phosphorylation, and increased transcription of a BDNF-dependent program of gene expression containing CRE binding sites, which includes activity-regulated genes such as Arc , Dusp1 , c-fos , Egr1 , and Egr2 , and growth and survival genes such as Atf3 and Gem Based on our results, we propose that early and recycling endosomes provide a platform for the integration of neurotrophic signaling from the plasma membrane to the nucleus in neurons, and that this mechanism is likely to regulate neuronal plasticity and survival. SIGNIFICANCE STATEMENT BDNF is a neurotrophic factor that regulates plastic changes in the brain, including dendritic growth. The cellular and molecular mechanisms underlying this process are not completely understood. Our results uncover the cellular requirements that central neurons possess to integrate the plasma membrane into nuclear signaling in neurons. Our results indicate that the endosomal pathway is required for the signaling cascade initiated by BDNF and its receptors at the plasma membrane to modulate BDNF-dependent gene expression and neuronal dendritic growth mediated by the CREB transcription factor. CREB is a key transcription factor regulating circuit development and learning and memory., (Copyright © 2020 the authors.)
- Published
- 2020
- Full Text
- View/download PDF
22. Rab6 is required for rapid, cisternal-specific, intra-Golgi cargo transport.
- Author
-
Dickson LJ, Liu S, and Storrie B
- Subjects
- Endoplasmic Reticulum metabolism, Golgi Apparatus physiology, Golgi Apparatus ultrastructure, HeLa Cells, Humans, Isoenzymes metabolism, Microscopy, Electron, Golgi Apparatus genetics, Golgi Apparatus metabolism, Protein Transport genetics, rab GTP-Binding Proteins physiology
- Abstract
Rab6, the most abundant Golgi associated small GTPase, consists of 2 equally common isoforms, Rab6A and Rab6A', that differ in 3 amino acids and localize to trans Golgi cisternae. The two isoforms are largely redundant in function and hence are often referred to generically as Rab6. Rab6 loss-of-function inhibits retrograde Golgi trafficking, induces an increase in Golgi cisternal number in HeLa cells and delays the cell surface appearance of the anterograde cargo protein, VSVG. We hypothesized that these effects are linked and might be explained by a cisternal-specific delay in cargo transport. In pulse chase experiments using a deconvolved, confocal line scanning approach to score the distribution of the tsO45 mutant of VSVG protein in Rab6 depleted cells, we found that anterograde transport at 32 °C, permissive conditions, through the Golgi apparatus was locally delayed, almost tenfold, between medial and trans Golgi cisterna. Cis to medial transport was nearly normal as was trans Golgi to TGN transport. TGN exit was unaffected by Rab6 depletion. These effects were the same with either of two siRNAs. Similar intra-Golgi transport delays were seen at 37 °C with RUSH VSVG or a RUSH GPI-anchored construct using a biotin pulse to release the marker proteins from the ER. Using 3D-SIM, a super resolution approach, we found that RUSH VSVG transport was delayed pre-trans Golgi. These visual approaches suggest a selective slowing of anterograde transport relative to 3 different marker proteins downstream of the trans Golgi. Using a biochemical approach, we found that the onset of VSVG endoglycosidase H resistance in Rab6 depleted cells was delayed. Depletion of neither Rab6A or Rab6A' isoforms alone had any effect on anterograde transport through the Golgi suggesting that Rab6A and Rab6A' act coordinately. Delayed cargo transport conditions correlate strongly with a proliferation of Golgi cisternae observed in earlier electron microscopy. Our results strongly indicate that Rab6 is selectively required for rapid anterograde transport from the medial to trans Golgi. We suggest that the observed correlation with localized cisternal proliferation fits best with a cisternal progression model of Golgi function.
- Published
- 2020
- Full Text
- View/download PDF
23. Maturation of Lipophagic Organelles in Hepatocytes Is Dependent Upon a Rab10/Dynamin-2 Complex.
- Author
-
Li Z, Weller SG, Drizyte-Miller K, Chen J, Krueger EW, Mehall B, Casey CA, Cao H, and McNiven MA
- Subjects
- Animals, Cells, Cultured, Lipid Droplets, Rats, Rats, Sprague-Dawley, Autophagy physiology, Dynamin II physiology, Hepatocytes ultrastructure, Organelles physiology, rab GTP-Binding Proteins physiology
- Abstract
Background and Aims: Hepatocytes play a central role in storage and utilization of fat by the liver. Selective breakdown of lipid droplets (LDs) by autophagy (also called lipophagy) is a key process utilized to catabolize these lipids as an energy source. How the autophagic machinery is selectively targeted to LDs, where it mediates membrane engulfment and subsequent degradation, is unclear. Recently, we have reported that two distinct GTPases, the mechanoenzyme, dynamin2 (Dyn2), and the small regulatory Rab GTPase, Rab10, work independently at distinct steps of lipophagy in hepatocytes., Approach and Results: In an attempt to understand how these proteins are regulated and recruited to autophagic organelles, we performed a nonbiased biochemical screen for Dyn2-binding partners and found that Dyn2 actually binds Rab10 directly through a defined effector domain of Rab10 and the middle domain of Dyn2. These two GTPases can be observed to interact transiently on membrane tubules in hepatoma cells and along LD-centric autophagic membranes. Most important, we found that a targeted disruption of this interaction leads to an inability of cells to trim tubulated cytoplasmic membranes, some of which extend from lipophagic organelles, resulting in LD accumulation., Conclusions: This study identifies a functional, and direct, interaction between Dyn2 and a regulatory Rab GTPase that may play an important role in hepatocellular metabolism., (© 2020 by the American Association for the Study of Liver Diseases.)
- Published
- 2020
- Full Text
- View/download PDF
24. Structural basis for autophagy inhibition by the human Rubicon-Rab7 complex.
- Author
-
Bhargava HK, Tabata K, Byck JM, Hamasaki M, Farrell DP, Anishchenko I, DiMaio F, Im YJ, Yoshimori T, and Hurley JH
- Subjects
- Crystallography, X-Ray, HeLa Cells, Humans, Models, Molecular, Protein Binding, Protein Domains physiology, rab7 GTP-Binding Proteins, Autophagy physiology, Autophagy-Related Proteins chemistry, Autophagy-Related Proteins metabolism, Autophagy-Related Proteins physiology, rab GTP-Binding Proteins chemistry, rab GTP-Binding Proteins metabolism, rab GTP-Binding Proteins physiology
- Abstract
Rubicon is a potent negative regulator of autophagy and a potential target for autophagy-inducing therapeutics. Rubicon-mediated inhibition of autophagy requires the interaction of the C-terminal Rubicon homology (RH) domain of Rubicon with Rab7-GTP. Here we report the 2.8-Å crystal structure of the Rubicon RH domain in complex with Rab7-GTP. Our structure reveals a fold for the RH domain built around four zinc clusters. The switch regions of Rab7 insert into pockets on the surface of the RH domain in a mode that is distinct from those of other Rab-effector complexes. Rubicon residues at the dimer interface are required for Rubicon and Rab7 to colocalize in living cells. Mutation of Rubicon RH residues in the Rab7-binding site restores efficient autophagic flux in the presence of overexpressed Rubicon, validating the Rubicon RH domain as a promising therapeutic target., Competing Interests: Competing interest statement: The authors declare a competing interest. J.H.H. is a cofounder of Casma Therapeutics., (Copyright © 2020 the Author(s). Published by PNAS.)
- Published
- 2020
- Full Text
- View/download PDF
25. Loss of Rab6a in the small intestine causes lipid accumulation and epithelial cell death from lactation.
- Author
-
Iwaki A, Moriwaki K, Sobajima T, Taniguchi M, Yoshimura SI, Kunii M, Kanda S, Kamada Y, Miyoshi E, and Harada A
- Subjects
- Animals, Epithelial Cells metabolism, Female, Glycosylation, Inflammation etiology, Inflammation metabolism, Intestine, Small metabolism, Mice, Mice, Knockout, Cell Death, Epithelial Cells pathology, Inflammation pathology, Intestine, Small pathology, Lactation, Lipids chemistry, rab GTP-Binding Proteins physiology
- Abstract
Intestinal epithelial cells (IECs) are not only responsible for the digestion and absorption of dietary substrates but also function as a first line of host defense against commensal and pathogenic luminal bacteria. Disruption of the epithelial layer causes malnutrition and enteritis. Rab6 is a small GTPase localized to the Golgi, where it regulates anterograde and retrograde transport by interacting with various effector proteins. Here, we generated mice with IEC-specific deletion of Rab6a (Rab6a
∆IEC mice). While Rab6aΔIEC mice were born at the Mendelian ratio, they started to show IEC death, inflammation, and bleeding in the small intestine shortly after birth, and these changes culminated in early postnatal death. We further found massive lipid accumulation in the IECs of Rab6a∆IEC neonates. In contrast to Rab6a∆IEC neonates, knockout embryos did not show any of these abnormalities. Lipid accumulation and IEC death became evident when Rab6a∆IEC embryos were nursed by a foster mother, suggesting that dietary milk-derived lipids accumulated in Rab6a-deficient IECs and triggered IEC death. These results indicate that Rab6a plays a crucial role in regulating the lipid transport and maintaining tissue integrity., (© 2020 Federation of American Societies for Experimental Biology.)- Published
- 2020
- Full Text
- View/download PDF
26. Zygote morphogenesis but not the establishment of cell polarity in Plasmodium berghei is controlled by the small GTPase, RAB11A.
- Author
-
Patil H, Hughes KR, Lemgruber L, Philip N, Dickens N, Starnes GL, and Waters AP
- Subjects
- Animals, Cell Polarity physiology, Culicidae parasitology, Malaria parasitology, Morphogenesis, Plasmodium berghei growth & development, Protozoan Proteins metabolism, Zygote metabolism, rab GTP-Binding Proteins physiology, Plasmodium berghei metabolism, Zygote growth & development, rab GTP-Binding Proteins metabolism
- Abstract
Plasmodium species are apicomplexan parasites whose zoites are polarized cells with a marked apical organisation where the organelles associated with host cell invasion and colonization reside. Plasmodium gametes mate in the mosquito midgut to form the spherical and presumed apolar zygote that morphs during the following 24 hours into a polarized, elongated and motile zoite form, the ookinete. Endocytosis-mediated protein transport is generally necessary for the establishment and maintenance of polarity in epithelial cells and neurons, and the small GTPase RAB11A is an important regulator of protein transport via recycling endosomes. PbRAB11A is essential in blood stage asexual of Plasmodium. Therefore, a promoter swap strategy was employed to down-regulate PbRAB11A expression in gametocytes and zygotes of the rodent malaria parasite, Plasmodium berghei which demonstrated the essential role of RAB11A in ookinete development. The approach revealed that lack of PbRAB11A had no effect on gamete production and fertility rates however, the zygote to ookinete transition was almost totally inhibited and transmission through the mosquito was prevented. Lack of PbRAB11A did not prevent meiosis and mitosis, nor the establishment of polarity as indicated by the correct formation and positioning of the Inner Membrane Complex (IMC) and apical complex. However, morphological maturation was prevented and parasites remained spherical and immotile and furthermore, they were impaired in the secretion and distribution of microneme cargo. The data are consistent with the previously proposed model of RAB11A endosome mediated delivery of plasma membrane in Toxoplasma gondii if not its role in IMC formation and implicate it in microneme function., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
27. Rab11A regulates dense granule transport and secretion during Toxoplasma gondii invasion of host cells and parasite replication.
- Author
-
Venugopal K, Chehade S, Werkmeister E, Barois N, Periz J, Lafont F, Tardieux I, Khalife J, Langsley G, Meissner M, and Marion S
- Subjects
- Animals, Cell Adhesion, Cell Line, Cell Membrane metabolism, Cytoskeleton metabolism, Host-Parasite Interactions physiology, Humans, Membrane Proteins metabolism, Microtubules metabolism, Parasites metabolism, Protein Transport, Protozoan Proteins, Toxoplasma metabolism, Toxoplasmosis metabolism, rab GTP-Binding Proteins physiology, Transport Vesicles metabolism, Vacuoles metabolism, rab GTP-Binding Proteins metabolism
- Abstract
Toxoplasma gondii possesses an armada of secreted virulent factors that enable parasite invasion and survival into host cells. These factors are contained in specific secretory organelles, the rhoptries, micronemes and dense granules that release their content upon host cell recognition. Dense granules are secreted in a constitutive manner during parasite replication and play a crucial role in modulating host metabolic and immune responses. While the molecular mechanisms triggering rhoptry and microneme release upon host cell adhesion have been well studied, constitutive secretion remains a poorly explored aspect of T. gondii vesicular trafficking. Here, we investigated the role of the small GTPase Rab11A, a known regulator of exocytosis in eukaryotic cells. Our data revealed an essential role of Rab11A in promoting the cytoskeleton driven transport of dense granules and the release of their content into the vacuolar space. Rab11A also regulates transmembrane protein trafficking and localization during parasite replication, indicating a broader role of Rab11A in cargo exocytosis at the plasma membrane. Moreover, we found that Rab11A also regulates extracellular parasite motility and adhesion to host cells. In line with these findings, MIC2 secretion was altered in Rab11A-defective parasites, which also exhibited severe morphological defects. Strikingly, by live imaging we observed a polarized accumulation of Rab11A-positive vesicles and dense granules at the apical pole of extracellular motile and invading parasites suggesting that apically polarized Rab11A-dependent delivery of cargo regulates early secretory events during parasite entry into host cells., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
28. Actions of Rab27B-GTPase on mammalian central excitatory synaptic transmission.
- Author
-
Arias-Hervert ER, Xu N, Njus M, Murphy GG, Hou Y, Williams JA, Lentz SI, Ernst SA, and Stuenkel EL
- Subjects
- Animals, Long-Term Potentiation physiology, Mice, Mice, Inbred C57BL, Mice, Knockout, rab3A GTP-Binding Protein metabolism, Hippocampus metabolism, Presynaptic Terminals metabolism, Synapses metabolism, Synaptic Transmission physiology, rab GTP-Binding Proteins physiology
- Abstract
Members of the Rab3 gene family are considered central to membrane trafficking of synaptic vesicles at mammalian central excitatory synapses. Recent evidence, however, indicates that the Rab27B-GTPase, which is highly homologous to the Rab3 family, is also enriched on SV membranes and co-localize with Rab3A and Synaptotagmin at presynaptic terminals. While functional roles of Rab3A have been well-established, little functional information exists on the role of Rab27B in synaptic transmission. Here we report on functional effects of Rab27B at SC-CA1 and DG-MF hippocampal synapses. The data establish distinct functional actions of Rab27B and demonstrate functions of Rab27B that differ between SC-CA1 and DG-MF synapses. Rab27B knockout reduced frequency facilitation compared to wild-type (WT) controls at the DG/MF-CA3 synaptic region, while increasing facilitation at the SC-CA1 synaptic region. Remarkably, Rab27B KO resulted in a complete elimination of LTP at the MF-CA3 synapse with no effect at the SC-CA1 synapse. These actions are similar to those previously reported for Rab3A KO. Specificity of action on LTP to Rab27B was confirmed as LTP was rescued in response to lentiviral infection and expression of human Rab27B, but not to GFP, in the DG in the Rab27B KO mice. Notably, the effect of Rab27B KO on MF-CA3 LTP occurred in spite of continued expression of Rab3A in the Rab27B KO. Overall, the results provide a novel perspective in suggesting that Rab27B and Rab3A act synergistically, perhaps via sequential effector recruitment or signaling for presynaptic LTP expression in this hippocampal synaptic region., (© 2020 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of The Physiological Society and the American Physiological Society.)
- Published
- 2020
- Full Text
- View/download PDF
29. Genetic and structural studies of RABL3 reveal an essential role in lymphoid development and function.
- Author
-
Zhong X, Su L, Yang Y, Nair-Gill E, Tang M, Anderton P, Li X, Wang J, Zhan X, Tomchick DR, Brautigam CA, Moresco EMY, Choi JH, and Beutler B
- Subjects
- Animals, B-Lymphocytes metabolism, B-Lymphocytes pathology, Crystallography, X-Ray, Female, Herpesviridae Infections immunology, Herpesviridae Infections virology, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, Killer Cells, Natural pathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Muromegalovirus immunology, Mutant Proteins chemistry, Mutant Proteins genetics, Mutation, Protein Conformation, T-Lymphocytes metabolism, T-Lymphocytes pathology, B-Lymphocytes immunology, Lymphopoiesis, Mutant Proteins metabolism, Receptors, G-Protein-Coupled metabolism, T-Lymphocytes immunology, rab GTP-Binding Proteins chemistry, rab GTP-Binding Proteins physiology
- Abstract
The small GTPase RABL3 is an oncogene of unknown physiological function. Homozygous knockout alleles of mouse Rabl3 were embryonic lethal, but a viable hypomorphic allele ( xiamen [ xm ]) causing in-frame deletion of four amino acids from the interswitch region resulted in profound defects in lymphopoiesis. Impaired lymphoid progenitor development led to deficiencies of B cells, T cells, and natural killer (NK) cells in Rabl3
xm/xm mice. T cells and NK cells exhibited impaired cytolytic activity, and mice infected with mouse cytomegalovirus (MCMV) displayed elevated titers in the spleen. Myeloid cells were normal in number and function. Biophysical and crystallographic studies demonstrated that RABL3 formed a homodimer in solution via interactions between the effector binding surfaces on each subunit; monomers adopted a typical small G protein fold. RABL3xm displayed a large compensatory alteration in switch I, which adopted a β-strand configuration normally provided by the deleted interswitch residues, thereby permitting homodimer formation. Dysregulated effector binding due to conformational changes in the switch I-interswitch-switch II module likely underlies the xm phenotype. One such effector may be GPR89, putatively an ion channel or G protein-coupled receptor (GPCR). RABL3, but not RABL3xm , strongly associated with and stabilized GPR89, and an N -ethyl- N -nitrosourea (ENU)-induced mutation ( explorer ) in Gpr89 phenocopied Rabl3xm ., Competing Interests: The authors declare no competing interest.- Published
- 2020
- Full Text
- View/download PDF
30. Double deletion of the active zone proteins CAST/ELKS in the mouse forebrain causes high mortality of newborn pups.
- Author
-
Hagiwara A, Hamada S, Hida Y, and Ohtsuka T
- Subjects
- Animals, Animals, Newborn, Animals, Suckling, Body Weight, Cytoskeletal Proteins genetics, Cytoskeletal Proteins physiology, Female, Hippocampus abnormalities, Male, Mice, Mice, Knockout, Nerve Tissue Proteins genetics, Nerve Tissue Proteins physiology, Open Field Test, Synapses physiology, Trigeminal Nuclei abnormalities, Vibrissae abnormalities, rab GTP-Binding Proteins genetics, rab GTP-Binding Proteins physiology, Cytoskeletal Proteins deficiency, Exploratory Behavior physiology, Hippocampus metabolism, Nerve Tissue Proteins deficiency, Sucking Behavior physiology, rab GTP-Binding Proteins deficiency
- Abstract
Presynaptic active zone cytomatrix proteins are essential elements of neurotransmitter release machinery that govern neural transmission. Among active zone proteins, cytomatrix at the active zone-associated structural protein (CAST) is known to regulate active zone size in retinal photoreceptors and neurotransmitter release by recruiting Ca
2+ channels at various synapses. However, the role of ELKS-a protein from the same family as CAST-and the synergistic roles of CAST/ELKS have not been thoroughly investigated, particularly with regard to mouse behavior. Here, we generated ELKS conditional KO in mouse forebrain synapses by crossing ELKS flox mice with a CaMKII promoter-induced Cre line. Results showed that CAST is dominant at these synapses and that ELKS can support CAST function, but is less effective in the ELKS single KO. Pups of CAST/ELKS double KO in the forebrain were born in Mendelian rations but resulted in eventual death right after the birth. Anatomically, the forebrain neuronal compositions of CAST KO and CAST/ELKS double KO mice were indistinguishable, and the sensory neural network from whiskers on the face was identified as barrelette-like patches in the spinal trigeminal nucleus. Therefore, depletion of CAST and ELKS disrupts neurotransmission from sensory to motor networks, which can lead to deficits in exploration and failure to suckle.- Published
- 2020
- Full Text
- View/download PDF
31. CCB is Involved in Actin-Based Axonal Transport of Selected Synaptic Proteins.
- Author
-
Martin-Peña A and Ferrus A
- Subjects
- Animals, Animals, Genetically Modified, Axons physiology, Drosophila Proteins genetics, Drosophila melanogaster, Endoplasmic Reticulum metabolism, Female, Growth Cones physiology, Male, Membrane Proteins genetics, Mutation genetics, rab GTP-Binding Proteins genetics, rab GTP-Binding Proteins physiology, Actins physiology, Axonal Transport genetics, Axonal Transport physiology, Drosophila Proteins physiology, Membrane Proteins physiology, Synapses physiology
- Abstract
Synapse formation, maturation, and turnover require a finely regulated transport system that delivers selected cargos to specific synapses. However, the supporting mechanisms of this process are not fully understood. The present study unravels a new molecular system for vesicle-based axonal transport of proteins in male and female flies ( Drosophila melanogaster ). Here, we identify the gene CG14579 as the transcription unit corresponding to the regulatory mutations known as central complex broad ( ccb ). These mutations were previously isolated for their morphological phenotype in R-neurons of the ellipsoid body, a component of the central complex. Mutant axons from R-neurons fail to cross the midline, which is indicative of an aberrant composition of the growth cone. However, the molecular mechanism remained to be deciphered. In this manuscript, we show that CCB is involved in axonal trafficking of FasII and synaptobrevin, but not syntaxin. These results suggest that axonal transport of certain proteins is required for the correct pathfinding of R-neurons. We further investigated the molecular network supporting the CCB system and found that CCB colocalizes and coimmunoprecipitates with Rab11. Epistasis studies indicated that Rab11 is positioned downstream of CCB within this axonal transport system. Interestingly, ccb also interacts with actin and the actin nucleator spire The data revealed that this interaction plays a key role in the development of axonal connections within the ellipsoid body. We propose that the CCB/Rab11/SPIRE system regulates axonal trafficking of synaptic proteins required for proper connectivity and synaptic function. SIGNIFICANCE STATEMENT Proper function of the nervous system requires the establishment of mature, functional synapses. Differential protein composition in the synapse enables optimal performance of cognitive tasks. Therefore, it is critical to have a finely regulated transport system to deliver selected synaptic proteins to synapses. Remarkably, impairments in cytoskeleton-based protein-transport systems often underlie cognitive deficits, such as those associated with aging and neurodegenerative diseases. This study reveals that CCB is part of a novel transport system that delivers certain synaptic proteins via the actin cytoskeleton within the Rab11-related domain of slow recycling endosomes., (Copyright © 2020 the authors.)
- Published
- 2020
- Full Text
- View/download PDF
32. The GTPase Rab39a promotes phagosome maturation into MHC-I antigen-presenting compartments.
- Author
-
Cruz FM, Colbert JD, and Rock KL
- Subjects
- Animals, Endoplasmic Reticulum metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Phagocytosis, Protein Transport, Antigen Presentation immunology, CD8-Positive T-Lymphocytes immunology, Dendritic Cells immunology, Endoplasmic Reticulum immunology, Histocompatibility Antigens Class I immunology, Phagosomes physiology, rab GTP-Binding Proteins physiology
- Abstract
For CD8 T lymphocytes to mount responses to cancer and virally-infected cells, dendritic cells must capture antigens present in tissues and display them as peptides bound to MHC-I molecules. This is most often accomplished through a pathway called antigen cross-presentation (XPT). Here, we report that the vesicular trafficking protein Rab39a is needed for optimal cross-presentation by dendritic cells in vitro and cross-priming of CD8 T cells in vivo. Without Rab39a, MHC-I presentation of intraphagosomal peptides is inhibited, indicating that Rab39a converts phagosomes into peptide-loading compartments. In this process, Rab39a promotes the delivery of MHC-I molecules from the endoplasmic reticulum (ER) to phagosomes, and increases the levels of peptide-empty MHC-I conformers that can be loaded with peptide in this compartment. Rab39a also increases the levels of Sec22b and NOX2, previously recognized to participate in cross-presentation, on phagosomes, thereby filling in a missing link into how phagosomes mature into cross-presenting vesicles., (© 2019 The Authors.)
- Published
- 2020
- Full Text
- View/download PDF
33. Regulated resurfacing of a somatostatin receptor storage compartment fine-tunes pituitary secretion.
- Author
-
Alshafie W, Francis V, Bednarz K, Pan YE, Stroh T, and McPherson PS
- Subjects
- Animals, Corticotropin-Releasing Hormone, Humans, Mice, Inbred C57BL, Mice, Knockout, Pituitary Gland cytology, Qa-SNARE Proteins genetics, Receptors, Somatostatin genetics, Signal Transduction, Corticotrophs metabolism, Human Growth Hormone metabolism, Pituitary Gland metabolism, Qa-SNARE Proteins metabolism, Receptors, Somatostatin metabolism, rab GTP-Binding Proteins physiology
- Abstract
The surfacing of the glucose transporter GLUT4 driven by insulin receptor activation provides the prototypic example of a homeostasis response dependent on mobilization of an intracellular storage compartment. Here, we generalize this concept to a G protein-coupled receptor, somatostatin receptor subtype 2 (SSTR2), in pituitary cells. Following internalization in corticotropes, SSTR2 moves to a juxtanuclear syntaxin-6-positive compartment, where it remains until the corticotropes are stimulated with corticotropin releasing factor (CRF), whereupon SSTR2 exits the compartment on syntaxin-6-positive vesicular/tubular carriers that depend on Rab10 for their fusion with the plasma membrane. As SSTR2 activation antagonizes CRF-mediated hormone release, this storage/resurfacing mechanism may allow for a physiological homeostatic feedback system. In fact, we find that SSTR2 moves from an intracellular compartment to the cell surface in pituitary gland somatotropes, concomitant with increasing levels of serum growth hormone (GH) during natural GH cycles. Our data thus provide a mechanism by which signaling-mediated plasma membrane resurfacing of SSTR2 can fine-tune pituitary hormone release., (© 2019 McGill University.)
- Published
- 2020
- Full Text
- View/download PDF
34. The ERC1 scaffold protein implicated in cell motility drives the assembly of a liquid phase.
- Author
-
Sala K, Corbetta A, Minici C, Tonoli D, Murray DH, Cammarota E, Ribolla L, Ramella M, Fesce R, Mazza D, Degano M, and de Curtis I
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Animals, COS Cells, Cell Line, Tumor, Cell Membrane metabolism, Chlorocebus aethiops, Cytoplasm metabolism, Humans, Nerve Tissue Proteins physiology, Nuclear Matrix-Associated Proteins metabolism, Nuclear Matrix-Associated Proteins physiology, rab GTP-Binding Proteins physiology, Cell Movement physiology, Nerve Tissue Proteins metabolism, rab GTP-Binding Proteins metabolism
- Abstract
Several cellular processes depend on networks of proteins assembled at specific sites near the plasma membrane. Scaffold proteins assemble these networks by recruiting relevant molecules. The scaffold protein ERC1/ELKS and its partners promote cell migration and invasion, and assemble into dynamic networks at the protruding edge of cells. Here by electron microscopy and single molecule analysis we identify ERC1 as an extended flexible dimer. We found that ERC1 scaffolds form cytoplasmic condensates with a behavior that is consistent with liquid phases that are modulated by a predicted disordered region of ERC1. These condensates specifically host partners of a network relevant to cell motility, including liprin-α1, which was unnecessary for the formation of condensates, but influenced their dynamic behavior. Phase separation at specific sites of the cell periphery may represent an elegant mechanism to control the assembly and turnover of dynamic scaffolds needed for the spatial localization and processing of molecules.
- Published
- 2019
- Full Text
- View/download PDF
35. RAB38 promotes bladder cancer growth by promoting cell proliferation and motility.
- Author
-
Tian DW, Liu SL, Jiang LM, Wu ZL, Gao J, Hu HL, and Wu CL
- Subjects
- Animals, Humans, Mice, Tumor Cells, Cultured, Cell Movement physiology, Cell Proliferation physiology, Urinary Bladder Neoplasms pathology, rab GTP-Binding Proteins physiology
- Abstract
Background: Bladder cancer is the most common malignancy of urinary system with high morbidity and mortality. In general, the development and progression of bladder cancer are complicated pathological processes, and the treatment methods mainly include surgical resection, radiotherapy, chemotherapy, and combined therapy. In recent years, targeted therapy has made progress in the treatment of bladder cancer. Therefore, to improve survival rates of patients with advanced bladder cancer, novel therapeutic targets are still urgently needed., Methods and Results: In this study, we found that RAB38 expressed in tumor tissues of patients with bladder cancer was linked to clinical features including pTNM stage and tumor recurrence, and positively correlated with the poor prognosis of bladder cancer. Notably, further results indicated that depletion of RAB38 could significantly inhibit the proliferation and motility of two types of human bladder cancer cells, T24 and 5637 cells. In addition, RAB38 ablation obviously blocked tumor growth and development in mice compared with control., Conclusion: In conclusion, this study provides significant evidence that RAB38 promotes the development of bladder cancer and provides a novel therapeutic target of bladder cancer.
- Published
- 2019
- Full Text
- View/download PDF
36. Rab GTPases: Switching to Human Diseases.
- Author
-
Guadagno NA and Progida C
- Subjects
- Animals, Biological Transport, Cell Membrane metabolism, Humans, Signal Transduction, Immune System Diseases metabolism, Infections metabolism, Neoplasms metabolism, Neurodegenerative Diseases metabolism, rab GTP-Binding Proteins physiology
- Abstract
Rab proteins compose the largest family of small GTPases and control the different steps of intracellular membrane traffic. More recently, they have been shown to also regulate cell signaling, division, survival, and migration. The regulation of these processes generally occurs through recruitment of effectors and regulatory proteins, which control the association of Rab proteins to membranes and their activation state. Alterations in Rab proteins and their effectors are associated with multiple human diseases, including neurodegeneration, cancer, and infections. This review provides an overview of how the dysregulation of Rab-mediated functions and membrane trafficking contributes to these disorders. Understanding the altered dynamics of Rabs and intracellular transport defects might thus shed new light on potential therapeutic strategies.
- Published
- 2019
- Full Text
- View/download PDF
37. A Chemical Genetics Screen Reveals Influence of p38 Mitogen-Activated Protein Kinase and Autophagy on Phagosome Development and Intracellular Replication of Brucella neotomae in Macrophages.
- Author
-
Kang YS and Kirby JE
- Subjects
- Animals, Cell Line, Female, Humans, Imidazoles pharmacology, Mice, Mice, Inbred BALB C, Pyridines pharmacology, rab GTP-Binding Proteins physiology, rab7 GTP-Binding Proteins, Autophagy physiology, Brucella growth & development, Macrophages microbiology, Phagosomes physiology, p38 Mitogen-Activated Protein Kinases physiology
- Abstract
Brucella is an intracellular bacterial pathogen that causes chronic systemic infection in domesticated livestock and poses a zoonotic infectious risk to humans. The virulence of Brucella is critically dependent on its ability to replicate and survive within host macrophages. Brucella modulates host physiological pathways and cell biology in order to establish a productive intracellular replicative niche. Conversely, the host cell presumably activates pathways that limit infection. To identify host pathways contributing to this yin and yang during host cell infection, we performed a high-throughput chemical genetics screen of known inhibitors and agonists of host cell targets to identify host factors that contribute to intracellular growth of the model pathogen Brucella neotomae Using this approach, we identified the p38 mitogen-activated protein (MAP) kinase pathway and autophagy machinery as both a linchpin and an Achilles' heel in B. neotomae 's ability to coopt host cell machinery and replicate within macrophages. Specifically, B. neotomae induced p38 MAP kinase phosphorylation and autophagy in a type IV secretion system-dependent fashion. Both p38 MAP kinase stimulation and an intact autophagy machinery in turn were required for phagosome maturation and intracellular replication. These findings contrasted with those for Legionella pneumophila , where chemical inhibition of the p38 MAP kinase pathway and autophagy factor depletion failed to block intracellular replication. Therefore, results from a chemical genetics screen suggest that intersections of the MAP kinase pathways and autophagy machinery are critical components of Brucella 's intracellular life cycle., (Copyright © 2019 American Society for Microbiology.)
- Published
- 2019
- Full Text
- View/download PDF
38. Intrinsic lipid binding activity of ATG16L1 supports efficient membrane anchoring and autophagy.
- Author
-
Dudley LJ, Cabodevilla AG, Makar AN, Sztacho M, Michelberger T, Marsh JA, Houston DR, Martens S, Jiang X, and Gammoh N
- Subjects
- Animals, Autophagy-Related Proteins physiology, Cells, Cultured, Embryo, Mammalian cytology, Embryo, Mammalian metabolism, Endosomes metabolism, Fibroblasts cytology, Fibroblasts metabolism, Mice, Mice, Knockout, Phosphate-Binding Proteins physiology, Ubiquitin-Conjugating Enzymes physiology, rab GTP-Binding Proteins physiology, Autophagy, Autophagy-Related Proteins metabolism, Cell Membrane metabolism, Phosphatidylinositol Phosphates metabolism
- Abstract
Membrane targeting of autophagy-related complexes is an important step that regulates their activities and prevents their aberrant engagement on non-autophagic membranes. ATG16L1 is a core autophagy protein implicated at distinct phases of autophagosome biogenesis. In this study, we dissected the recruitment of ATG16L1 to the pre-autophagosomal structure (PAS) and showed that it requires sequences within its coiled-coil domain (CCD) dispensable for homodimerisation. Structural and mutational analyses identified conserved residues within the CCD of ATG16L1 that mediate direct binding to phosphoinositides, including phosphatidylinositol 3-phosphate (PI3P). Mutating putative lipid binding residues abrogated the localisation of ATG16L1 to the PAS and inhibited LC3 lipidation. On the other hand, enhancing lipid binding of ATG16L1 by mutating negatively charged residues adjacent to the lipid binding motif also resulted in autophagy inhibition, suggesting that regulated recruitment of ATG16L1 to the PAS is required for its autophagic activity. Overall, our findings indicate that ATG16L1 harbours an intrinsic ability to bind lipids that plays an essential role during LC3 lipidation and autophagosome maturation., (© 2019 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2019
- Full Text
- View/download PDF
39. Coordination between Rac1 and Rab Proteins: Functional Implications in Health and Disease.
- Author
-
Margiotta A and Bucci C
- Subjects
- Actin Cytoskeleton metabolism, Actins metabolism, Cell Movement physiology, DNA Repair physiology, Humans, Immunity, Innate physiology, Neoplasms physiopathology, Nervous System Diseases physiopathology, Reactive Oxygen Species metabolism, Transcription, Genetic physiology, rab GTP-Binding Proteins physiology, rac1 GTP-Binding Protein physiology
- Abstract
The small GTPases of the Rho family regulate many aspects of actin dynamics, but are functionally connected to many other cellular processes. Rac1, a member of this family, besides its known function in the regulation of actin cytoskeleton, plays a key role in the production of reactive oxygen species, in gene transcription, in DNA repair, and also has been proven to have specific roles in neurons. This review focuses on the cooperation between Rac1 and Rab proteins, analyzing how the coordination between these GTPases impact on cells and how alterations of their functions lead to disease.
- Published
- 2019
- Full Text
- View/download PDF
40. Drosophila Arl8 is a general positive regulator of lysosomal fusion events.
- Author
-
Boda A, Lőrincz P, Takáts S, Csizmadia T, Tóth S, Kovács AL, and Juhász G
- Subjects
- ADP-Ribosylation Factors genetics, ADP-Ribosylation Factors metabolism, Animals, Autophagosomes physiology, Drosophila Proteins genetics, Drosophila Proteins metabolism, Drosophila melanogaster physiology, Drosophila melanogaster ultrastructure, Lysosomes metabolism, Lysosomes ultrastructure, Membrane Fusion, Protein Subunits physiology, rab GTP-Binding Proteins physiology, ADP-Ribosylation Factors physiology, Drosophila Proteins physiology, Lysosomes physiology
- Abstract
The small GTPase Arl8 is known to be involved in the periphery-directed motility of lysosomes. However, the overall importance of moving these vesicles is still poorly understood. Here we show that Drosophila Arl8 is required not only for the proper distribution of lysosomes, but also for autophagosome-lysosome fusion in starved fat cells, endosome-lysosome fusion in garland nephrocytes, and developmentally programmed secretory granule degradation (crinophagy) in salivary gland cells. Moreover, proper Arl8 localization to lysosomes depends on the shared subunits of the BLOC-1 and BORC complexes, which also promote autophagy and crinophagy. In conclusion, we demonstrate that Arl8 is responsible not only for positioning lysosomes but also acts as a general lysosomal fusion factor., (Copyright © 2018 Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
41. Exosomes from MiR-21-5p-Increased Neurons Play a Role in Neuroprotection by Suppressing Rab11a-Mediated Neuronal Autophagy In Vitro After Traumatic Brain Injury.
- Author
-
Li D, Huang S, Zhu J, Hu T, Han Z, Zhang S, Zhao J, Chen F, and Lei P
- Subjects
- Animals, Autophagy genetics, Autophagy physiology, Brain pathology, Brain Injuries, Traumatic physiopathology, Cells, Cultured, Disease Models, Animal, Exosomes genetics, Male, Mice, Mice, Inbred C57BL, MicroRNAs metabolism, Neurons metabolism, Neurons physiology, Neuroprotection drug effects, rab GTP-Binding Proteins metabolism, rab GTP-Binding Proteins physiology, Brain Injuries, Traumatic genetics, MicroRNAs genetics, MicroRNAs physiology
- Abstract
BACKGROUND Traumatic brain injury (TBI) produces a series of pathological processes. Recent studies have indicated that autophagy pathway is persistently activated after TBI, which may lead to deterioration of nerve injury. Our preliminary work found miR-21-5p was upregulated in both in vivo and in vitro TBI models. MicroRNAs (miRNAs) could be loaded into exosomes to perform cell-to-cell interactions. This research aimed to evaluate the therapeutic effect of neuron-derived exosomes enriched with miR-21-5p on the TBI in vitro and to further explore the possible mechanisms. MATERIAL AND METHODS Brain extracts harvested from an rTBI mouse model were added to cultured HT-22 neurons to imitate the microenvironment of injured brain on in vitro cultured cells. Ultracentrifugation was performed to isolate exosomes. Transmission electron microscopy and Nano sight technology were used to examine exosomes. An in vitro model of TBI was established to study the effect of exosomal miR-21-5p on nerve injury and on neuronal autophagy regulation. RESULTS The expression of miR-21-5p was increased in exosomes derived from HT-22 neurons after treatment with rTBI mouse brain extracts. Autophagy was activated in HT-22 neurons after scratch injury. Exosomal miR-21-5p produced a protective effect by suppressing autophagy in a TBI model in vitro. MiR-21-5p could directly target the Rab11a 3'UTR region to reduce its translation and further suppressed Rab11a-mediated neuronal autophagy. CONCLUSIONS The levels of miR-21-5p in neuronal exosomes increased from the acute to the chronic phase of TBI. Neuronal exosomes enriched with miR-21-5p can inhibit the activity of neuronal autophagy by targeting Rab11a, thus attenuating trauma-induced, autophagy-mediated nerve injury in vitro.
- Published
- 2019
- Full Text
- View/download PDF
42. Porcine sapelovirus enters PK-15 cells via caveolae-dependent endocytosis and requires Rab7 and Rab11.
- Author
-
Zhao T, Cui L, Yu X, Zhang Z, Shen X, and Hua X
- Subjects
- Animals, Cell Line, Cell Survival, Hydrogen-Ion Concentration, Swine, rab7 GTP-Binding Proteins, Caveolae physiology, Endocytosis physiology, Picornaviridae physiology, Virus Internalization, rab GTP-Binding Proteins physiology
- Abstract
To comprehensively understand the endocytosis of Sapelovirus A (PSV) entry into PK-15 cells, we studied PSV infection in the context of cell perturbations through drug inhibition, siRNA silencing and overexpression of dominant negative (DN) mutants. We showed here that PSV infection of PK-15 cells was unaffected by pretreated with chlorpromazine, EIPA, knockdown of the clathrin heavy chain or overexpression of Eps15 DN mutant. Conversely, PSV infection was sensitive to NH
4 Cl, chloroquine, dynasore, nystatin, MβCD and wortmannin with reduced PSV VP1 expression levels and virus titer. Additionally, PSV invasion leaded to rapid actin rearrangement and disruption of the cellular actin network enhanced PSV infection. After internalization the virus was transported to late endosomes and/or cycling endosomes that requires the participation of Rab7 and Rab11. Our findings demonstrate that PSV uses caveolae-dependent endocytosis as the predominant entry portal into PK-15 cells which requires low pH, dynamin, Rab7 and Rab11., (Copyright © 2019 Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
43. Role of Rab GTPases in Alzheimer's Disease.
- Author
-
Zhang X, Huang TY, Yancey J, Luo H, and Zhang YW
- Subjects
- Alzheimer Disease drug therapy, Alzheimer Disease pathology, Amyloid beta-Peptides metabolism, Animals, Brain drug effects, Brain pathology, Enzyme Inhibitors pharmacology, Enzyme Inhibitors therapeutic use, Humans, Plaque, Amyloid drug therapy, Plaque, Amyloid enzymology, Plaque, Amyloid pathology, Protein Transport drug effects, rab GTP-Binding Proteins antagonists & inhibitors, tau Proteins metabolism, Alzheimer Disease enzymology, Brain enzymology, Protein Transport physiology, rab GTP-Binding Proteins physiology
- Abstract
Alzheimer's disease (AD) comprises two major pathological hallmarks: extraneuronal deposition of β-amyloid (Aβ) peptides ("senile plaques") and intraneuronal aggregation of the microtubule-associated protein tau ("neurofibrillary tangles"). Aβ is derived from sequential cleavage of the β-amyloid precursor protein by β- and γ-secretases, while aggregated tau is hyperphosphorylated in AD. Mounting evidence suggests that dysregulated trafficking of these AD-related proteins contributes to AD pathogenesis. Rab proteins are small GTPases that function as master regulators of vesicular transport and membrane trafficking. Multiple Rab GTPases have been implicated in AD-related protein trafficking, and their expression has been observed to be altered in postmortem AD brain. Here we review current implicated roles of Rab GTPase dysregulation in AD pathogenesis. Further elucidation of the pathophysiological role of Rab GTPases will likely reveal novel targets for AD therapeutics.
- Published
- 2019
- Full Text
- View/download PDF
44. Late Endosomes Act as mRNA Translation Platforms and Sustain Mitochondria in Axons.
- Author
-
Cioni JM, Lin JQ, Holtermann AV, Koppers M, Jakobs MAH, Azizi A, Turner-Bridger B, Shigeoka T, Franze K, Harris WA, and Holt CE
- Subjects
- Animals, Axons metabolism, Endosomes metabolism, Mitochondria genetics, Mitochondria metabolism, RNA metabolism, RNA, Messenger metabolism, RNA, Messenger physiology, Retinal Ganglion Cells metabolism, Retinal Ganglion Cells physiology, Ribosomes metabolism, Xenopus Proteins metabolism, Xenopus laevis metabolism, rab GTP-Binding Proteins genetics, rab GTP-Binding Proteins physiology, rab7 GTP-Binding Proteins, Endosomes physiology, Protein Biosynthesis physiology, rab GTP-Binding Proteins metabolism
- Abstract
Local translation regulates the axonal proteome, playing an important role in neuronal wiring and axon maintenance. How axonal mRNAs are localized to specific subcellular sites for translation, however, is not understood. Here we report that RNA granules associate with endosomes along the axons of retinal ganglion cells. RNA-bearing Rab7a late endosomes also associate with ribosomes, and real-time translation imaging reveals that they are sites of local protein synthesis. We show that RNA-bearing late endosomes often pause on mitochondria and that mRNAs encoding proteins for mitochondrial function are translated on Rab7a endosomes. Disruption of Rab7a function with Rab7a mutants, including those associated with Charcot-Marie-Tooth type 2B neuropathy, markedly decreases axonal protein synthesis, impairs mitochondrial function, and compromises axonal viability. Our findings thus reveal that late endosomes interact with RNA granules, translation machinery, and mitochondria and suggest that they serve as sites for regulating the supply of nascent pro-survival proteins in axons., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
45. Rab-Effector-Kinase Interplay Modulates Intralumenal Fragment Formation during Vacuole Fusion.
- Author
-
Karim MA, McNally EK, Samyn DR, Mattie S, and Brett CL
- Subjects
- Casein Kinase I metabolism, Homeostasis, Lipid Bilayers, Lipid Metabolism, Lipids physiology, Lysosomes, Permeability, Phosphorylation, Phosphotransferases, Proteolysis, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism, Vacuoles metabolism, Vesicular Transport Proteins genetics, Vesicular Transport Proteins metabolism, Vesicular Transport Proteins physiology, rab GTP-Binding Proteins genetics, rab GTP-Binding Proteins metabolism, Membrane Fusion physiology, Saccharomyces cerevisiae Proteins physiology, Vacuoles physiology, rab GTP-Binding Proteins physiology
- Abstract
Upon vacuolar lysosome (or vacuole) fusion in S. cerevisiae, a portion of membrane is internalized and catabolized. Formation of this intralumenal fragment (ILF) is important for organelle protein and lipid homeostasis and remodeling. But how ILF formation is optimized for membrane turnover is not understood. Here, we show that fewer ILFs form when the interaction between the Rab-GTPase Ypt7 and its effector Vps41 (a subunit of the tethering complex HOPS) is interrupted by a point mutation (Ypt7-D44N). Subsequent phosphorylation of Vps41 by the casein kinase Yck3 prevents stabilization of trans-SNARE complexes needed for lipid bilayer pore formation. Impairing ILF formation prevents clearance of misfolded proteins from vacuole membranes and promotes organelle permeability and cell death. We propose that HOPS coordinates Rab, kinase, and SNARE cycles to modulate ILF size during vacuole fusion, regulating lipid and protein turnover important for quality control and membrane integrity., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
46. Rab GTPases in Osteoclastic Endomembrane Systems.
- Author
-
Roy M and Roux S
- Subjects
- Bone Resorption, Bone and Bones, Endosomes, Protein Transport, Osteoclasts physiology, rab GTP-Binding Proteins physiology
- Abstract
Osteoclasts (OCs) are bone-resorbing cells that maintain bone homeostasis. OC differentiation, survival, and activity are regulated by numerous small GTPases, including those of the Rab family, which are involved in plasma membrane delivery and lysosomal and autophagic degradation pathways. In resorbing OCs, polarized vesicular trafficking pathways also result in formation of the ruffled membrane, the resorbing organelle, and in transcytosis.
- Published
- 2018
- Full Text
- View/download PDF
47. Characterization of the Populus Rab family genes and the function of PtRabE1b in salt tolerance.
- Author
-
Zhang J, Li Y, Liu B, Wang L, Zhang L, Hu J, Chen J, Zheng H, and Lu M
- Subjects
- Chromosomes, Plant genetics, Conserved Sequence genetics, Genes, Plant physiology, Phylogeny, Plant Proteins physiology, Populus physiology, Promoter Regions, Genetic genetics, Salt Tolerance, Salt-Tolerant Plants physiology, Transcriptome, rab GTP-Binding Proteins physiology, Genes, Plant genetics, Plant Proteins genetics, Populus genetics, Salt-Tolerant Plants genetics, rab GTP-Binding Proteins genetics
- Abstract
Background: Rab proteins form the largest family of the Ras superfamily of small GTP-binding proteins and regulate intracellular trafficking pathways. However, the function of the Rab proteins in woody species is still an open question., Results: Here, a total of 67 PtRabs were identified in Populus trichocarpa and categorized into eight subfamilies (RabA-RabH). Based on their chromosomal distribution and duplication blocks in the Populus genome, a total of 27 PtRab paralogous pairs were identified and all of them were generated by whole-genome duplication events. Combined the expression correlation and duplication date, the PtRab paralogous pairs that still keeping highly similar expression patterns were generated around the latest large-scale duplication (~ 13 MYA). The cis-elements and co-expression network of unique expanded PtRabs suggest their potential roles in poplar development and environmental responses. Subcellular localization of PtRabs from each subfamily indicates each subfamily shows a localization pattern similar to what is revealed in Arabidopsis but RabC shows a localization different from their counterparts. Furthermore, we characterized PtRabE1b by overexpressing its constitutively active mutant PtRabE1b(Q74L) in poplar and found that PtRabE1b(Q74L) enhanced the salt tolerance., Conclusions: These findings provide new insights into the functional divergence of PtRabs and resources for genetic engineering resistant breeding in tree species.
- Published
- 2018
- Full Text
- View/download PDF
48. Single-shot super-resolution total internal reflection fluorescence microscopy.
- Author
-
Guo M, Chandris P, Giannini JP, Trexler AJ, Fischer R, Chen J, Vishwasrao HD, Rey-Suarez I, Wu Y, Wu X, Waterman CM, Patterson GH, Upadhyaya A, Taraska JW, and Shroff H
- Subjects
- Cell Line, Tumor, Humans, Microtubules, Osteosarcoma, rab GTP-Binding Proteins physiology, Microscopy, Fluorescence methods
- Abstract
We combined instant structured illumination microscopy (iSIM) with total internal reflection fluorescence microscopy (TIRFM) in an approach referred to as instant TIRF-SIM, thereby improving the lateral spatial resolution of TIRFM to 115 ± 13 nm without compromising speed, and enabling imaging frame rates up to 100 Hz over hundreds of time points. We applied instant TIRF-SIM to multiple live samples and achieved rapid, high-contrast super-resolution imaging close to the coverslip surface.
- Published
- 2018
- Full Text
- View/download PDF
49. Role of Rab GTPases in HSV-1 infection: Molecular understanding of viral maturation and egress.
- Author
-
Raza S, Alvisi G, Shahin F, Husain U, Rabbani M, Yaqub T, Anjum AA, Sheikh AA, Nawaz M, and Ali MA
- Subjects
- Glycoproteins metabolism, Herpesvirus 1, Human pathogenicity, Humans, Protein Transport physiology, Viral Envelope Proteins physiology, Viral Proteins genetics, Virus Assembly physiology, rab1 GTP-Binding Proteins physiology, rab27 GTP-Binding Proteins physiology, rab5 GTP-Binding Proteins physiology, Herpes Simplex metabolism, Herpesvirus 1, Human physiology, Virus Physiological Phenomena, Virus Release physiology, rab GTP-Binding Proteins physiology
- Abstract
Most enveloped viruses exploit complex cellular pathways for assembly and egress from the host cell, and the large DNA virus Herpes simplex virus 1 (HSV-1) makes no exception, hijacking several cellular transport pathways for its glycoprotein trafficking and maturation, as well as for viral morphogenesis and egress according to the envelopment, de-envelopment and re-envelopment model. Importantly Rab GTPases, widely distributed master regulators of intracellular membrane trafficking pathways, have recently being tightly implicated in such process. Indeed, siRNA-mediated genetic ablation of specific Rab proteins differently affected HSV-1 production, suggesting a complex role of different Rab proteins in HSV-1 life cycle. In this review, we discuss how different Rabs can regulate HSV-1 assembly/egress and the potential therapeutic applications of such findings for the management of HSV-1 infections., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
50. Electrical pulse stimulation induces GLUT4 translocation in C 2 C 12 myotubes that depends on Rab8A, Rab13, and Rab14.
- Author
-
Li Z, Yue Y, Hu F, Zhang C, Ma X, Li N, Qiu L, Fu M, Chen L, Yao Z, Bilan PJ, Klip A, and Niu W
- Subjects
- Animals, Cells, Cultured, Electric Stimulation, Glucose metabolism, Mice, Muscle Contraction physiology, Protein Transport genetics, Signal Transduction genetics, rab GTP-Binding Proteins genetics, Glucose Transporter Type 4 metabolism, Muscle Fibers, Skeletal metabolism, rab GTP-Binding Proteins physiology
- Abstract
The signals mobilizing GLUT4 to the plasma membrane in response to muscle contraction are less known than those elicited by insulin. This disparity is undoubtedly due to lack of suitable in vitro models to study skeletal muscle contraction. We generated C
2 C12 myotubes stably expressing HA-tagged GLUT4 (C2C12-GLUT4 HA) that contract in response to electrical pulse stimulation (EPS) and investigated molecular mechanisms regulating GLUT4 HA. EPS (60 min, 20 V, 1 Hz, 24-ms pulses at 976-ms intervals) elicited a gain in surface GLUT4 HA (GLUT4 translocation) comparably to insulin or 5-amino imidazole-4-carboxamide ribonucleotide (AICAR). A myosin II inhibitor prevented EPS-stimulated myotube contraction and reduced surface GLUT4 by 56%. EPS stimulated AMPK and CaMKII phosphorylation, and EPS-stimulated GLUT4 translocation was reduced in part by small interfering (si)RNA-mediated AMPKα1/α2 knockdown, compound C, siRNA-mediated Ca2+ /calmodulin-dependent protein kinase (CaMKII)δ knockdown, or CaMKII inhibitor KN93. Key regulatory residues on the Rab-GAPs AS160 and TBC1D1 were phosphorylated in response to EPS. Stable expression of an activated form of the Rab-GAP AS160 (AS160-4A) diminished EPS- and insulin-stimulated GLUT4 translocation, suggesting regulation of GLUT4 vesicle traffic by Rab GTPases. Knockdown of each Rab8a, Rab13, or Rab14 reduced, in part, GLUT4 translocation induced by EPS, whereas only Rab8a, or Rab14 knockdown reduced the AICAR response. In conclusion, EPS involves Rab8a, Rab13, and Rab14 to elicit GLUT4 translocation but not Rab10; moreover, Rab10 and Rab13 are not engaged by AMPK activation alone. C2C12-GLUT4 HA cultures constitute a valuable in vitro model to investigate molecular mechanisms of contraction-stimulated GLUT4 translocation.- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.