Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Hepatol Commun ; 8(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38780316

ABSTRACT

BACKGROUND: Previous reports suggest that lipid droplets (LDs) in the hepatocyte can be catabolized by a direct engulfment from nearby endolysosomes (microlipophagy). Further, it is likely that this process is compromised by chronic ethanol (EtOH) exposure leading to hepatic steatosis. This study investigates the hepatocellular machinery supporting microlipophagy and EtOH-induced alterations in this process with a focus on the small, endosome-associated, GTPase Rab5. METHODS AND RESULTS: Here we report that this small Ras-related GTPase is a resident component of LDs, and its activity is important for hepatocellular LD-lysosome proximity and physical interactions. We find that Rab5 siRNA knockdown causes an accumulation of LDs in hepatocytes by inhibiting lysosome dependent LD catabolism. Importantly, Rab5 appears to support this process by mediating the recruitment of early endosomal and or multivesicular body compartments to the LD surface before lysosome fusion. Interestingly, while wild-type or a constituently active GTPase form (Q79L) of Rab5 supports LD-lysosome transport, this process is markedly reduced in cells expressing a GTPase dead (S34N) Rab5 protein or in hepatocytes exposed to chronic EtOH. CONCLUSIONS: These findings support the novel premise of an early endosomal/multivesicular body intermediate compartment on the LD surface that provides a "docking" site for lysosomal trafficking, not unlike the process that occurs during the hepatocellular degradation of endocytosed ligands that is also known to be compromised by EtOH exposure.


Subject(s)
Ethanol , Hepatocytes , Lysosomes , rab5 GTP-Binding Proteins , rab5 GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/genetics , Lysosomes/metabolism , Lysosomes/drug effects , Ethanol/pharmacology , Hepatocytes/metabolism , Hepatocytes/drug effects , Humans , Lipid Droplets/metabolism , Autophagy/drug effects , Animals , Endosomes/metabolism
2.
Cell Rep ; 42(9): 113042, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37651233

ABSTRACT

Amplified lysosome activity is a hallmark of pancreatic ductal adenocarcinoma (PDAC) orchestrated by oncogenic KRAS that mediates tumor growth and metastasis, though the mechanisms underlying this phenomenon remain unclear. Using comparative proteomics, we found that oncogenic KRAS significantly enriches levels of the guanine nucleotide exchange factor (GEF) dedicator of cytokinesis 8 (DOCK8) on lysosomes. Surprisingly, DOCK8 is aberrantly expressed in a subset of PDAC, where it promotes cell invasion in vitro and in vivo. DOCK8 associates with lysosomes and regulates lysosomal morphology and motility, with loss of DOCK8 leading to increased lysosome size. DOCK8 promotes actin polymerization at the surface of lysosomes while also increasing the proteolytic activity of the lysosomal protease cathepsin B. Critically, depletion of DOCK8 significantly reduces cathepsin-dependent extracellular matrix degradation and impairs the invasive capacity of PDAC cells. These findings implicate ectopic expression of DOCK8 as a key driver of KRAS-driven lysosomal regulation and invasion in pancreatic cancer cells.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Cytokinesis , Ectopic Gene Expression , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Pancreatic Neoplasms/pathology , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Carcinoma, Pancreatic Ductal/pathology , Lysosomes/metabolism
3.
JCI Insight ; 7(2)2022 01 25.
Article in English | MEDLINE | ID: mdl-34874916

ABSTRACT

Approximately 80% of pancreatic cancer patients suffer from cachexia, and one-third die due to cachexia-related complications such as respiratory failure and cardiac arrest. Although there has been considerable research into cachexia mechanisms and interventions, there are, to date, no FDA-approved therapies. A major contributing factor for the lack of therapy options could be the failure of animal models to accurately recapitulate the human condition. In this study, we generated an aged model of pancreatic cancer cachexia to compare cachexia progression in young versus aged tumor-bearing mice. Comparative skeletal muscle transcriptome analyses identified 3-methyladenine (3-MA) as a candidate antiwasting compound. In vitro analyses confirmed antiwasting capacity, while in vivo analysis revealed potent antitumor effects. Transcriptome analyses of 3-MA-treated tumor cells implicated Perp as a 3-MA target gene. We subsequently (a) observed significantly higher expression of Perp in cancer cell lines compared with control cells, (b) noted a survival disadvantage associated with elevated Perp, and (c) found that 3-MA-associated Perp reduction inhibited tumor cell growth. Finally, we have provided in vivo evidence that survival benefits conferred by 3-MA administration are independent of its effect on tumor progression. Taken together, we report a mechanism linking 3-MA to Perp inhibition, and we further implicate Perp as a tumor-promoting factor in pancreatic cancer.


Subject(s)
Adenine/analogs & derivatives , Cachexia , Membrane Proteins , Muscle, Skeletal , Pancreatic Neoplasms , Adenine/metabolism , Adenine/pharmacology , Age Factors , Animals , Autophagy/drug effects , Cachexia/etiology , Cachexia/metabolism , Cachexia/therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Gene Expression Profiling/methods , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/therapy
4.
Hepatol Commun ; 5(7): 1238-1251, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34278172

ABSTRACT

Currently, the hepatocellular trafficking pathways that are used by the hepatitis B virus (HBV) during viral infection and shedding are poorly defined. It is known that the HBV uses late endosomal and multivesicular body (MVB) compartments for assembly and release. The intraluminal vesicles (ILVs) generated within MVBs have also been implicated in the late synthesis stages of a variety of pathogenic viruses. We recently observed that the HBV within infected hepatocytes appears to associate with the tetraspanin protein CD63, known to be a prominent and essential component of ILVs. Immunofluorescence microscopy of HBV-expressing cells showed that CD63 colocalized with HBV proteins (large hepatitis B surface antigens [LHBs] and hepatitis B core) and labeled an exceptionally large number of secreted extracellular vesicles of uniform size. Small interfering RNA (siRNA)-mediated depletion of CD63 induced a substantial accumulation of intracellular LHBs protein but did not alter the levels of either intracellular or extracellular HBV DNA, nor pregenomic RNA. Consistent with these findings, we found that markedly less LHBs protein was associated with the released HBV particles from CD63 siRNA-treated cells. Importantly, the HBV viral particles that were shed from CD63-depleted cells were substantially less infective than those collected from control cells with normal CD63 levels. Conclusion: These findings implicate the tetraspanin protein CD63 as a marker and an important component in the formation and release of infectious HBV particles.

5.
Sci Adv ; 7(25)2021 06.
Article in English | MEDLINE | ID: mdl-34144977

ABSTRACT

53BP1 activates nonhomologous end joining (NHEJ) and inhibits homologous recombination (HR) repair of DNA double-strand breaks (DSBs). Dissociation of 53BP1 from DSBs and consequent activation of HR, a less error-prone pathway than NHEJ, helps maintain genome integrity during DNA replication; however, the underlying mechanisms are not fully understood. Here, we demonstrate that E3 ubiquitin ligase SPOP promotes HR during S phase of the cell cycle by excluding 53BP1 from DSBs. In response to DNA damage, ATM kinase-catalyzed phosphorylation of SPOP causes a conformational change in SPOP, revealed by x-ray crystal structures, that stabilizes its interaction with 53BP1. 53BP1-bound SPOP induces polyubiquitination of 53BP1, eliciting 53BP1 extraction from chromatin by a valosin-containing protein/p97 segregase complex. Our work shows that SPOP facilitates HR repair over NHEJ during DNA replication by contributing to 53BP1 removal from chromatin. Cancer-derived SPOP mutations block SPOP interaction with 53BP1, inducing HR defects and chromosomal instability.


Subject(s)
Chromatin , DNA Breaks, Double-Stranded , Chromatin/genetics , DNA End-Joining Repair , DNA Replication , Nuclear Proteins , Recombinational DNA Repair , Repressor Proteins , Tumor Suppressor p53-Binding Protein 1
6.
Mol Biol Cell ; 32(15): 1393-1407, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34010028

ABSTRACT

The α-actinin family of actin cross-linking proteins have been implicated in driving tumor cell metastasis through regulation of the actin cytoskeleton; however, there has been little investigation into whether these proteins can influence tumor cell growth. We demonstrate that α-actinin 1 and 4 are essential for nutrient uptake through the process of macropinocytosis in pancreatic ductal adenocarcinoma (PDAC) cells, and inhibition of these proteins decreases tumor cell survival in the presence of extracellular protein. The α-actinin proteins play essential roles throughout the macropinocytic process, where α-actinin 4 stabilizes the actin cytoskeleton on the plasma membrane to drive membrane ruffling and macropinosome internalization and α-actinin 1 localizes to actin tails on macropinosomes to facilitate trafficking to the lysosome for degradation. In addition to tumor cell growth, we also observe that the α-actinin proteins can influence uptake of chemotherapeutics and extracellular matrix proteins through macropinocytosis, suggesting that the α-actinin proteins can regulate multiple tumor cell properties through this endocytic process. In summary, these data demonstrate a critical role for the α-actinin isoforms in tumor cell macropinocytosis, thereby affecting the growth and invasive potential of PDAC tumors.


Subject(s)
Actinin/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/metabolism , Pinocytosis , Actin Cytoskeleton/metabolism , Carcinoma, Pancreatic Ductal/physiopathology , Cell Line, Tumor , Endosomes , Humans , Pancreatic Neoplasms/physiopathology
7.
Proc Natl Acad Sci U S A ; 117(51): 32443-32452, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33288726

ABSTRACT

Hepatocytes metabolize energy-rich cytoplasmic lipid droplets (LDs) in the lysosome-directed process of autophagy. An organelle-selective form of this process (macrolipophagy) results in the engulfment of LDs within double-membrane delimited structures (autophagosomes) before lysosomal fusion. Whether this is an exclusive autophagic mechanism used by hepatocytes to catabolize LDs is unclear. It is also unknown whether lysosomes alone might be sufficient to mediate LD turnover in the absence of an autophagosomal intermediate. We performed live-cell microscopy of hepatocytes to monitor the dynamic interactions between lysosomes and LDs in real-time. We additionally used a fluorescent variant of the LD-specific protein (PLIN2) that exhibits altered fluorescence in response to LD interactions with the lysosome. We find that mammalian lysosomes and LDs undergo interactions during which proteins and lipids can be transferred from LDs directly into lysosomes. Electron microscopy (EM) of primary hepatocytes or hepatocyte-derived cell lines supports the existence of these interactions. It reveals a dramatic process whereby the lipid contents of the LD can be "extruded" directly into the lysosomal lumen under nutrient-limited conditions. Significantly, these interactions are not affected by perturbations to crucial components of the canonical macroautophagy machinery and can occur in the absence of double-membrane lipoautophagosomes. These findings implicate the existence of an autophagic mechanism used by mammalian cells for the direct transfer of LD components into the lysosome for breakdown. This process further emphasizes the critical role of lysosomes in hepatic LD catabolism and provides insights into the mechanisms underlying lipid homeostasis in the liver.


Subject(s)
Autophagy/physiology , Hepatocytes/metabolism , Lipid Droplets/metabolism , Lysosomes/metabolism , Animals , Autophagosomes/metabolism , Cell Line , Lipid Metabolism , Mice , Microscopy, Confocal , Protein Transport , Rats, Sprague-Dawley
8.
Mol Biol Cell ; 31(6): 439-451, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31967944

ABSTRACT

The large GTPase Dynamin 2 (Dyn2) is known to increase the invasiveness of pancreatic cancer tumor cells, but the mechanisms by which Dyn2 regulates changes in the actin cytoskeleton to drive cell migration are still unclear. Here we report that a direct interaction between Dyn2 and the actin-bundling protein alpha-actinin (α-actinin) 4 is critical for tumor cell migration and remodeling of the extracellular matrix in pancreatic ductal adenocarcinoma (PDAC) cells. The direct interaction is mediated through the C-terminal tails of both Dyn2 and α-actinin 4, and these proteins interact at invasive structures at the plasma membrane. While Dyn2 binds directly to both α-actinin 1 and α-actinin 4, only the interaction with α-actinin 4 is required to promote tumor cell invasion. Specific disruption of the Dyn2-α-actinin 4 interaction blocks the ability of PDAC cells to migrate in either two dimensions or invade through extracellular matrix as a result of impaired invadopodia stability. Analysis of human PDAC tumor tissue additionally reveals that elevated α-actinin 4 or Dyn2 expression are predictive of poor survival. Overall, these data demonstrate that Dyn2 regulates cytoskeletal dynamics, in part, by interacting with the actin-binding protein α-actinin 4 during tumor cell invasion.


Subject(s)
Actinin/metabolism , Dynamin II/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement , Humans , Neoplasm Invasiveness , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Binding , Pseudopodia/metabolism , Pancreatic Neoplasms
9.
Hepatology ; 72(2): 486-502, 2020 08.
Article in English | MEDLINE | ID: mdl-31808574

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.


Subject(s)
Autophagy/physiology , Dynamin II/physiology , Hepatocytes/ultrastructure , Organelles/physiology , rab GTP-Binding Proteins/physiology , Animals , Cells, Cultured , Lipid Droplets , Rats , Rats, Sprague-Dawley
10.
J Cell Biol ; 218(10): 3320-3335, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31391210

ABSTRACT

Lipid droplet (LD) catabolism in hepatocytes is mediated by a combination of lipolysis and a selective autophagic mechanism called lipophagy, but the relative contributions of these seemingly distinct pathways remain unclear. We find that inhibition of lipolysis, lipophagy, or both resulted in similar overall LD content but dramatic differences in LD morphology. Inhibition of the lipolysis enzyme adipose triglyceride lipase (ATGL) resulted in large cytoplasmic LDs, whereas lysosomal inhibition caused the accumulation of numerous small LDs within the cytoplasm and degradative acidic vesicles. Combined inhibition of ATGL and LAL resulted in large LDs, suggesting that lipolysis targets these LDs upstream of lipophagy. Consistent with this, ATGL was enriched in larger-sized LDs, whereas lipophagic vesicles were restricted to small LDs as revealed by immunofluorescence, electron microscopy, and Western blot of size-separated LDs. These findings provide new evidence indicating a synergistic relationship whereby lipolysis targets larger-sized LDs to produce both size-reduced and nascently synthesized small LDs that are amenable for lipophagic internalization.


Subject(s)
Hepatocytes/cytology , Hepatocytes/metabolism , Lipid Droplets/metabolism , Lipolysis , Animals , Female , Rats , Rats, Sprague-Dawley , Triglycerides/analysis , Triglycerides/metabolism
11.
J Cell Biol ; 218(1): 317-332, 2019 01 07.
Article in English | MEDLINE | ID: mdl-30487181

ABSTRACT

The process by which tumor cells mechanically invade through surrounding stroma into peripheral tissues is an essential component of metastatic dissemination. The directed recruitment of the metalloproteinase MT1-MMP to invadopodia plays a critical role in this invasive process. Here, we provide mechanistic insight into MT1-MMP cytoplasmic tail binding protein 1 (MTCBP-1) with respect to invadopodia formation, matrix remodeling, and invasion by pancreatic tumor cells. MTCBP-1 localizes to invadopodia and interacts with MT1-MMP. We find that this interaction displaces MT1-MMP from invadopodia, thereby attenuating their number and function and reducing the capacity of tumor cells to degrade matrix. Further, we observe an inverse correlation between MTCBP-1 and MT1-MMP expression both in cultured cell lines and human pancreatic tumors. Consistently, MTCBP-1-expressing cells show decreased ability to invade in vitro and metastasize in vivo. These findings implicate MTCBP-1 as an inhibitor of the metastatic process.


Subject(s)
Dioxygenases/genetics , Gene Expression Regulation, Neoplastic , Intestinal Neoplasms/genetics , Matrix Metalloproteinase 14/genetics , Pancreatic Neoplasms/genetics , Podosomes/genetics , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Dioxygenases/antagonists & inhibitors , Dioxygenases/metabolism , Humans , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/mortality , Intestinal Neoplasms/secondary , Intestine, Small/metabolism , Intestine, Small/pathology , Lymphatic Metastasis , Matrix Metalloproteinase 14/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Podosomes/metabolism , Podosomes/pathology , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , Xenograft Model Antitumor Assays
12.
Sci Adv ; 2(12): e1601470, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28028537

ABSTRACT

The autophagic digestion of lipid droplets (LDs) through lipophagy is an essential process by which most cells catabolize lipids as an energy source. However, the cellular machinery used for the envelopment of LDs during autophagy is poorly understood. We report a novel function for a small Rab guanosine triphosphatase (GTPase) in the recruitment of adaptors required for the engulfment of LDs by the growing autophagosome. In hepatocytes stimulated to undergo autophagy, Rab10 activity is amplified significantly, concomitant with its increased recruitment to nascent autophagic membranes at the LD surface. Disruption of Rab10 function by small interfering RNA knockdown or expression of a GTPase-defective variant leads to LD accumulation. Finally, Rab10 activation during autophagy is essential for LC3 recruitment to the autophagosome and stimulates its increased association with the adaptor protein EHBP1 (EH domain binding protein 1) and the membrane-deforming adenosine triphosphatase EHD2 (EH domain containing 2) that, together, are essential in driving the activated "engulfment" of LDs during lipophagy in hepatocytes.

13.
Oncogene ; 35(9): 1099-1110, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-25982272

ABSTRACT

Metastatic invasion of tumors into peripheral tissues is known to rely upon protease-mediated degradation of the surrounding stroma. This remodeling process uses complex, actin-based, specializations of the plasma membrane termed invadopodia that act both to sequester and release matrix metalloproteinases. Here we report that cells of mesenchymal origin, including tumor-associated fibroblasts, degrade substantial amounts of surrounding matrix by a mechanism independent of conventional invadopodia. These degradative sites lack the punctate shape of conventional invadopodia to spread along the cell base and are reticular and/or fibrous in character. In marked contrast to invadopodia, this degradation does not require the action of Src kinase, Cdc42 or Dyn2. Rather, inhibition of Dyn2 causes a marked upregulation of stromal matrix degradation. Further, expression and activity of matrix metalloproteinases are differentially regulated between tumor cells and stromal fibroblasts. This matrix remodeling by fibroblasts increases the invasive capacity of tumor cells, thereby illustrating how the tumor microenvironment can contribute to metastasis. These findings provide evidence for a novel matrix remodeling process conducted by stromal fibroblasts that is substantially more effective than conventional invadopodia, distinct in structural organization and regulated by disparate molecular mechanisms.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Pancreatic Ductal/genetics , Dynamins/genetics , Podosomes/genetics , cdc42 GTP-Binding Protein/genetics , src-Family Kinases/genetics , Breast Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Dynamin II , Dynamins/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Matrix Metalloproteinases/genetics , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Podosomes/metabolism , Proteolysis , Stromal Cells/metabolism , Stromal Cells/pathology , Xenograft Model Antitumor Assays , cdc42 GTP-Binding Protein/metabolism , src-Family Kinases/metabolism
14.
J Cell Sci ; 128(9): 1696-706, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25770103

ABSTRACT

The cellular mechanisms by which hepatitis B virus (HBV) is assembled and exported are largely undefined. Recently, it has been suggested that these steps require the multivesicular body (MVB) and the autophagic machinery. However, the mechanisms by which HBV might regulate these compartments are unclear. In this study, we have found that by activating Rab7a, HBV alters its own secretion by inducing dramatic changes in the morphology of MVB and autophagic compartments. These changes are characterized by the formation of numerous tubules that are dependent upon the increase in Rab7 activity observed in the HBV-expressing HepG2.2.15 cells compared to HepG2 cells. Interestingly, transfection-based expression of the five individual viral proteins indicated that the precore protein, which is a precursor of HBeAg, was largely responsible for the increased Rab7 activity. Finally, small interfering RNA (siRNA)-mediated depletion of Rab7 significantly increased the secretion of virions, suggesting that reduced delivery of the virus to the lysosome facilitates viral secretion. These findings provide novel evidence indicating that HBV can regulate its own secretion through an activation of the endo-lysosomal and autophagic pathway mediated by Rab7 activation.


Subject(s)
Autophagy , Cell Compartmentation , Endocytosis , Hepatitis B virus/metabolism , rab GTP-Binding Proteins/metabolism , Antigens, Viral/metabolism , Hep G2 Cells , Hepatitis B/pathology , Hepatitis B/virology , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Hepatocytes/virology , Humans , Lysosomes/metabolism , Lysosomes/ultrastructure , Membrane Fusion , Models, Biological , Phagosomes/metabolism , Phagosomes/ultrastructure , rab7 GTP-Binding Proteins
15.
J Cell Biol ; 203(2): 315-26, 2013 Oct 28.
Article in English | MEDLINE | ID: mdl-24145164

ABSTRACT

Lipid droplets (LDs) are lipid storage organelles that in hepatocytes may be catabolized by autophagy for use as an energy source, but the membrane-trafficking machinery regulating such a process is poorly characterized. We hypothesized that the large GTPase dynamin 2 (Dyn2), well known for its involvement in membrane deformation and cellular protein trafficking, could orchestrate autophagy-mediated LD breakdown. Accordingly, depletion or pharmacologic inhibition of Dyn2 led to a substantial accumulation of LDs in hepatocytes. Strikingly, the targeted disruption of Dyn2 induced a dramatic four- to fivefold increase in the size of autolysosomes. Chronic or acute Dyn2 inhibition combined with nutrient deprivation stimulated the excessive tubulation of these autolysosomal compartments. Importantly, Dyn2 associated with these tubules along their length, and the tubules vesiculated and fragmented in the presence of functional Dyn2. These findings provide new evidence for the participation of the autolysosome in LD metabolism and demonstrate a novel role for dynamin in the function and maturation of an autophagic compartment.


Subject(s)
Autophagy , Dynamin II/metabolism , Hepatocytes/enzymology , Lipolysis , Lysosomes/enzymology , Animals , Autophagy/drug effects , Cell Line, Tumor , Dynamin II/antagonists & inhibitors , Dynamin II/deficiency , Dynamin II/genetics , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Lipolysis/drug effects , Lysosomes/drug effects , Lysosomes/pathology , Mice , Mice, Knockout , Microscopy, Fluorescence , RNA Interference , Time Factors , Time-Lapse Imaging , Transfection , Video Recording
16.
Dev Cell ; 24(6): 573-85, 2013 Mar 25.
Article in English | MEDLINE | ID: mdl-23537630

ABSTRACT

The large GTPase Dynamin 2 (Dyn2) is markedly upregulated in pancreatic cancer, is a potent activator of metastatic migration, and is required for Rac1-mediated formation of lamellipodia. Here we demonstrate an unexpected mechanism of Dyn2 action in these contexts via direct binding to the Rac1 guanine nucleotide exchange factor (GEF) Vav1. Surprisingly, disruption of the Dyn2-Vav1 interaction targets Vav1 to the lysosome for degradation via an interaction with the cytoplasmic chaperone Hsc70, resulting in a dramatic reduction of Vav1 protein stability. Importantly, a specific mutation in Vav1 near its Dyn2-binding C-terminal Src homology 3 (SH3) domain prevents Hsc70 binding, resulting in a stabilization of Vav1 levels. Dyn2 binding regulates the interaction of Vav1 with Hsc70 to control the stability and subsequent activity of this oncogenic GEF. These findings elucidate how Dyn2 activates Rac1, lamellipod protrusion, and invasive cellular migration and provide insight into how this specific Vav is ectopically expressed in pancreatic tumors.


Subject(s)
Dynamin II/metabolism , HSC70 Heat-Shock Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-vav/metabolism , rac1 GTP-Binding Protein/metabolism , Binding Sites , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement , Dynamin II/chemistry , Dynamin II/genetics , Gene Expression Regulation, Neoplastic , Humans , Lysosomal-Associated Membrane Protein 2 , Lysosomal Membrane Proteins/genetics , Lysosomes/metabolism , Pancreatic Neoplasms/genetics , Protein Binding , Protein Stability , Protein Structure, Tertiary , Proto-Oncogene Proteins c-vav/genetics , Pseudopodia/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Up-Regulation
17.
PLoS One ; 7(12): e52603, 2012.
Article in English | MEDLINE | ID: mdl-23285107

ABSTRACT

Heterologous expression of HIV-1 Gag in a variety of host cells results in its packaging into virus-like particles (VLPs) that are subsequently released into the extracellular milieu. This phenomenon represents a useful tool for probing cellular factors required for viral budding and has contributed to the discovery of roles for ubiquitin ligases and the endosomal sorting complexes required for transport (ESCRTs) in viral budding. These factors are highly conserved throughout eukaryotes and have been studied extensively in the yeast Saccharomyces cerevisiae, a model eukaryote previously utilized as a host for the production of VLPs. We used heterologous expression of HIV Gag in yeast spheroplasts to examine the role of ESCRTs and associated factors (Rsp5, a HECT ubiquitin ligase of the Nedd4 family; Bro1, a homolog of Alix; and Vps4, the AAA-ATPase required for ESCRT function in all contexts/organisms investigated) in the generation of VLPs. Our data reveal: 1) characterized Gag-ESCRT interaction motifs (late domains) are not required for VLP budding, 2) loss of function alleles of the essential HECT ubiquitin ligase Rsp5 do not display defects in VLP formation, and 3) ESCRT function is not required for VLP formation from spheroplasts. These results suggest that the egress of HIV Gag from yeast cells is distinct from the most commonly described mode of exit from mammalian cells, instead mimicking ESCRT-independent VLP formation observed in a subset of mammalian cells. As such, budding of Gag from yeast cells appears to represent ESCRT-independent budding relevant to viral replication in at least some situations. Thus the myriad of genetic and biochemical tools available in the yeast system may be of utility in the study of this aspect of viral budding.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Saccharomyces cerevisiae/metabolism , Spheroplasts/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism , Gene Expression , Humans , Protein Interaction Domains and Motifs , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Virus Release , gag Gene Products, Human Immunodeficiency Virus/genetics
18.
Hepatology ; 54(5): 1819-29, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21793030

ABSTRACT

UNLABELLED: Clathrin-mediated endocytosis in mammalian epithelial cells is believed to require the synergistic action of structural coat proteins and mechanochemical enzymes to deform and sever the plasma membrane (PM) into discreet vesicles. It is generally believed that the formation of clathrin-coated pits in epithelial cells occurs randomly along the apical and basolateral plasma membranes. In this study we visualized the endocytic machinery in living hepatocytes using green fluorescent protein (GFP)-tagged dynamin, a large mechanochemical guanosine triphosphate (GTP)ase implicated in the liberation of nascent vesicles from the plasma membrane and a variety of internal membrane compartments. Confocal microscopy of living cells expressing the epithelial isoform of GFP-tagged dynamin [Dyn2-GFP] revealed a distribution along the ventral PM in discrete vesicle-like puncta or in large (2-10 µm) tubuloreticular plaques. Remarkably, these large structures are dynamic as they form and then disappear, while generating large numbers of motile endocytic vesicles with which dynamin associates. Inhibiting dynamin function by microinjection of purified dynamin antibodies increases the number and size of the tubuloreticular plaques. Importantly, these "hot spots" sequester specific trophic receptors and cognate ligands such as transferrin receptor 1 (TfR1), but not TfR2. CONCLUSION: These findings suggest that hepatocytes sequester or prerecruit both structural and enzymatic components of the clathrin-based endocytic machinery to functional hot spots, from which large numbers of coated pits form and vesicles are generated. This process may mimic the endocytic organization found at the synapse in neuronal cells.


Subject(s)
Endocytosis/physiology , Hepatocytes/cytology , Hepatocytes/metabolism , Protein Transport/physiology , Antigens, CD/metabolism , Biomarkers/metabolism , Cell Membrane/metabolism , Clathrin/metabolism , Clathrin-Coated Vesicles/metabolism , Dynamin II/genetics , Dynamin II/metabolism , Endosomes/metabolism , Green Fluorescent Proteins/genetics , Guanosine Triphosphate/metabolism , Hep G2 Cells , Humans , Receptors, Transferrin/metabolism , Transfection/methods
19.
Mol Cell Biol ; 30(3): 781-92, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19995918

ABSTRACT

The mechanisms by which epithelial cells regulate clathrin-mediated endocytosis (CME) of transferrin are poorly defined and generally viewed as a constitutive process that occurs continuously without regulatory constraints. In this study, we demonstrate for the first time that endocytosis of the transferrin receptor is a regulated process that requires activated Src kinase and, subsequently, phosphorylation of two important components of the endocytic machinery, namely, the large GTPase dynamin 2 (Dyn2) and its associated actin-binding protein, cortactin (Cort). To our knowledge these findings are among the first to implicate an Src-mediated endocytic cascade in what was previously presumed to be a nonregulated internalization process.


Subject(s)
Cortactin/metabolism , Dynamin II/metabolism , Endocytosis , Epithelial Cells/metabolism , Receptors, Transferrin/metabolism , src-Family Kinases/metabolism , Animals , Cell Line , Cortactin/agonists , Dynamin II/agonists , Mice , Phosphorylation/drug effects , Phosphorylation/physiology , Rats , Transferrin/metabolism , src-Family Kinases/drug effects
20.
Mol Biol Cell ; 20(19): 4140-52, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19641024

ABSTRACT

Remodeling of cell-cell contacts through the internalization of adherens junction proteins is an important event during both normal development and the process of tumor cell metastasis. Here we show that the integrity of tumor cell-cell contacts is disrupted after epidermal growth factor (EGF) stimulation through caveolae-mediated endocytosis of the adherens junction protein E-cadherin. Caveolin-1 and E-cadherin closely associated at cell borders and in internalized structures upon stimulation with EGF. Furthermore, preventing caveolae assembly through reduction of caveolin-1 protein or expression of a caveolin-1 tyrosine phospho-mutant resulted in the accumulation of E-cadherin at cell borders and the formation of tightly adherent cells. Most striking was the fact that exogenous expression of caveolin-1 in tumor cells that contain tight, well-defined, borders resulted in a dramatic dispersal of these cells. Together, these findings provide new insights into how cells might disassemble cell-cell contacts to help mediate the remodeling of adherens junctions, and tumor cell metastasis and invasion.


Subject(s)
Adherens Junctions/drug effects , Cadherins/metabolism , Caveolae/metabolism , Caveolin 1/metabolism , Epidermal Growth Factor/pharmacology , Adherens Junctions/metabolism , Animals , Blotting, Western , Cadherins/genetics , Caveolae/ultrastructure , Caveolin 1/genetics , Cell Adhesion/drug effects , Cell Communication/drug effects , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Endocytosis/drug effects , Endosomes/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Microscopy, Confocal , Microscopy, Electron , Mutation , RNA Interference , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...