Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 299(4): 103071, 2023 04.
Article in English | MEDLINE | ID: mdl-36849008

ABSTRACT

Lipid droplets (LDs) are fat-storing organelles enclosed by a phospholipid monolayer, which harbors membrane-associated proteins that regulate distinct LD functions. LD proteins are degraded by the ubiquitin-proteasome system (UPS) and/or by lysosomes. Because chronic ethanol (EtOH) consumption diminishes the hepatic functions of the UPS and lysosomes, we hypothesized that continuous EtOH consumption slows the breakdown of lipogenic LD proteins targeted for degradation, thereby causing LD accumulation. Here, we report that LDs from livers of EtOH-fed rats exhibited higher levels of polyubiquitylated-proteins, linked at either lysine 48 (directed to proteasome) or lysine 63 (directed to lysosomes) than LDs from pair-fed control rats. MS proteomics of LD proteins, immunoprecipitated with UB remnant motif antibody (K-ε-GG), identified 75 potential UB proteins, of which 20 were altered by chronic EtOH administration. Among these, hydroxysteroid 17ß-dehydrogenase 11 (HSD17ß11) was prominent. Immunoblot analyses of LD fractions revealed that EtOH administration enriched HSD17ß11 localization to LDs. When we overexpressed HSD17ß11 in EtOH-metabolizing VA-13 cells, the steroid dehydrogenase 11 became principally localized to LDs, resulting in elevated cellular triglycerides (TGs). Ethanol exposure augmented cellular TG, while HSD17ß11 siRNA decreased both control and EtOH-induced TG accumulation. Remarkably, HSD17ß11 overexpression lowered the LD localization of adipose triglyceride lipase. EtOH exposure further reduced this localization. Reactivation of proteasome activity in VA-13 cells blocked the EtOH-induced rises in both HSD17ß11 and TGs. Our findings indicate that EtOH exposure blocks HSD17ß11 degradation by inhibiting the UPS, thereby stabilizing HSD17ß11 on LD membranes, to prevent lipolysis by adipose triglyceride lipase and promote cellular LD accumulation.


Subject(s)
17-Hydroxysteroid Dehydrogenases , Ethanol , Fatty Liver , Animals , Rats , Ethanol/pharmacology , Ethanol/metabolism , Fatty Liver/metabolism , Lipase/genetics , Lipid Droplets/metabolism , Lipid Metabolism , Lysine/metabolism , Proteasome Endopeptidase Complex/metabolism , 17-Hydroxysteroid Dehydrogenases/metabolism
2.
J Cell Sci ; 135(5)2022 03 01.
Article in English | MEDLINE | ID: mdl-35260889

ABSTRACT

Lipophagy is a central cellular process for providing the cell with a readily utilized, high energy source of neutral lipids. Since its discovery over a decade ago, we are just starting to understand the molecular components that drive lipophagy, how it is activated in response to nutrient availability, and its potential as a therapeutic target in disease. In this Cell Science at a Glance article and the accompanying poster, we first provide a brief overview of the different structural and enzymatic proteins that comprise the lipid droplet (LD) proteome and reside within the limiting phospholipid monolayer of this complex organelle. We then highlight key players in the catabolic breakdown of LDs during the functionally linked lipolysis and lipophagy processes. Finally, we discuss what is currently known about macro- and micro-lipophagy based on findings in yeast, mammalian and other model systems, and how impairment of these important functions can lead to disease states.


Subject(s)
Lipid Droplets , Lipolysis , Animals , Autophagy/physiology , Lipid Droplets/metabolism , Lipid Metabolism/physiology , Mammals/metabolism , Phospholipids/metabolism , Proteins/metabolism , Saccharomyces cerevisiae/metabolism
3.
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
4.
Am J Hum Genet ; 105(1): 108-121, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31204009

ABSTRACT

Pediatric acute liver failure (ALF) is life threatening with genetic, immunologic, and environmental etiologies. Approximately half of all cases remain unexplained. Recurrent ALF (RALF) in infants describes repeated episodes of severe liver injury with recovery of hepatic function between crises. We describe bi-allelic RINT1 alterations as the cause of a multisystem disorder including RALF and skeletal abnormalities. Three unrelated individuals with RALF onset ≤3 years of age have splice alterations at the same position (c.1333+1G>A or G>T) in trans with a missense (p.Ala368Thr or p.Leu370Pro) or in-frame deletion (p.Val618_Lys619del) in RINT1. ALF episodes are concomitant with fever/infection and not all individuals have complete normalization of liver function testing between episodes. Liver biopsies revealed nonspecific liver damage including fibrosis, steatosis, or mild increases in Kupffer cells. Skeletal imaging revealed abnormalities affecting the vertebrae and pelvis. Dermal fibroblasts showed splice-variant mediated skipping of exon 9 leading to an out-of-frame product and nonsense-mediated transcript decay. Fibroblasts also revealed decreased RINT1 protein, abnormal Golgi morphology, and impaired autophagic flux compared to control. RINT1 interacts with NBAS, recently implicated in RALF, and UVRAG, to facilitate Golgi-to-ER retrograde vesicle transport. During nutrient depletion or infection, Golgi-to-ER transport is suppressed and autophagy is promoted through UVRAG regulation by mTOR. Aberrant autophagy has been associated with the development of similar skeletal abnormalities and also with liver disease, suggesting that disruption of these RINT1 functions may explain the liver and skeletal findings. Clarifying the pathomechanism underlying this gene-disease relationship may inform therapeutic opportunities.


Subject(s)
Autophagy , Bone Diseases, Developmental/etiology , Cell Cycle Proteins/genetics , Fibroblasts/pathology , Liver Failure, Acute/etiology , Mutation , Age of Onset , Alleles , Amino Acid Sequence , Bone Diseases, Developmental/metabolism , Bone Diseases, Developmental/pathology , Cell Cycle Proteins/metabolism , Child , Child, Preschool , Female , Fibroblasts/metabolism , Golgi Apparatus/metabolism , Golgi Apparatus/pathology , Humans , Infant , Liver Failure, Acute/metabolism , Liver Failure, Acute/pathology , Male , Pedigree , Protein Transport , Recurrence , Sequence Homology
5.
J Cell Sci ; 133(11)2020 06 11.
Article in English | MEDLINE | ID: mdl-32295849

ABSTRACT

Epithelial cells, such as liver-resident hepatocytes, rely heavily on the Rab family of small GTPases to perform membrane trafficking events that dictate cell physiology and metabolism. Not surprisingly, disruption of several Rab proteins can manifest in metabolic diseases or cancer. Rab32 is expressed in many secretory epithelial cells but its role in cellular metabolism is virtually unknown. In this study, we find that Rab32 associates with lysosomes and regulates proliferation and cell size of Hep3B hepatoma and HeLa cells. Specifically, we identify that Rab32 supports the mechanistic target of rapamycin complex 1 (mTORC1) signaling under basal and amino acid-stimulated conditions. Consistent with inhibited mTORC1, an increase in nuclear TFEB localization and lysosome biogenesis is also observed in Rab32-depleted cells. Finally, we find that Rab32 interacts with mTOR kinase, and that loss of Rab32 reduces the association of mTOR and mTORC1 pathway proteins with lysosomes, suggesting that Rab32 regulates lysosomal mTOR trafficking. In summary, these findings suggest that Rab32 functions as a novel regulator of cellular metabolism through supporting mTORC1 signaling.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Monomeric GTP-Binding Proteins , HeLa Cells , Humans , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Signal Transduction , rab GTP-Binding Proteins
6.
J Lipid Res ; 62: 100049, 2021.
Article in English | MEDLINE | ID: mdl-33617872

ABSTRACT

Lipid droplets (LDs) are composed of neutral lipids enclosed in a phospholipid monolayer, which harbors membrane-associated proteins that regulate LD functions. Despite the crucial role of LDs in lipid metabolism, remodeling of LD protein composition in disease contexts, such as steatosis, remains poorly understood. We hypothesized that chronic ethanol consumption, subsequent abstinence from ethanol, or fasting differentially affects the LD membrane proteome content and that these changes influence how LDs interact with other intracellular organelles. Here, male Wistar rats were pair-fed liquid control or ethanol diets for 6 weeks, and then, randomly chosen animals from both groups were either refed a control diet for 7 days or fasted for 48 h before euthanizing. From all groups, LD membrane proteins from purified liver LDs were analyzed immunochemically and by MS proteomics. Liver LD numbers and sizes were greater in ethanol-fed rats than in pair-fed control, 7-day refed, or fasted rats. Compared with control rats, ethanol feeding markedly altered the LD membrane proteome, enriching LD structural perilipins and proteins involved in lipid biosynthesis, while lowering LD lipase levels. Ethanol feeding also lowered LD-associated mitochondrial and lysosomal proteins. In 7-day refed (i.e., ethanol-abstained) or fasted-ethanol-fed rats, we detected distinct remodeling of the LD proteome, as judged by lower levels of lipid biosynthetic proteins, and enhanced LD interaction with mitochondria and lysosomes. Our study reveals evidence of significant remodeling of the LD membrane proteome that regulates ethanol-induced steatosis, its resolution after withdrawal and abstinence, and changes in LD interactions with other intracellular organelles.


Subject(s)
Lipid Droplets
7.
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
8.
J Virol ; 93(15)2019 08 01.
Article in English | MEDLINE | ID: mdl-31118260

ABSTRACT

Viruses are considered to use vesicular trafficking in infected cells, but the details of assembly/release pathways of hepatitis B virus (HBV) are still unknown. To identify key regulators of HBV production, we performed short interfering RNA (siRNA) screening for Rab proteins, which are considered to act as molecular switches in vesicular trafficking using HepG2.2.15 cells. Among 62 Rab proteins, the suppression of Rab5B most significantly increased HBV DNA in the culture supernatant. Surprisingly, 5 days after the transfection of Rab5B siRNA, HBV DNA in the supernatant was increased more than 30-fold, reflecting the increase of infectious HBV particles. Northern blotting showed that transcription of 2.4/2.1-kb mRNA coding envelope proteins containing large hepatitis B surface protein (LHBs) was increased. Analysis of hepatocyte nuclear factors (HNFs) showed that transcription of HNF4α, which is known to enhance 2.4-kb mRNA transcription, was regulated by Rab5B. Also, it was revealed that LHBs had accumulated in the endoplasmic reticulum (ER) after Rab5B depletion but not in the multivesicular body (MVB), which is thought to be an organelle utilized for HBV envelope formation. Therefore, it was considered that Rab5B is required for the transport of LHBs from the ER to MVB. Immunofluorescent microscopy showed that HBs proteins, including LHBs, colocalized with HBc in the ER of Rab5B-depleted cells, suggesting that HBV envelopment occurs not only in the MVB but also in the ER. In conclusion, Rab5B is a key regulator of HBV production and could be a target of antiviral therapy.IMPORTANCE HBV infection is a worldwide health problem, but the mechanisms of how HBV utilizes cellular machinery for its life cycle are poorly understood. In particular, it has been unclear how the viral components and virions are transported among the organelles. The HBV budding site has been reported to be the ER or MVB, but it has not been clearly determined. In this study, siRNA-based screening of Rab proteins using HBV-expressing cells showed that Rab5B, one of the Rab5 isoforms, has important roles in late steps of the HBV life cycle. Although Rab5 is known to work on early endosomes, this study showed that Rab5B plays a role in the transport of LHBs between the ER and MVB. Furthermore, it affects the transcription of LHBs. This is the first report on the mechanisms of HBV envelope protein transport among the organelles, and the results provide important insights into the therapeutic control of HBV infection.


Subject(s)
Hepatitis B Surface Antigens/metabolism , Hepatitis B virus/growth & development , Hepatocytes/virology , Host Microbial Interactions , rab5 GTP-Binding Proteins/metabolism , Cell Line , Culture Media/chemistry , DNA, Viral/analysis , Endoplasmic Reticulum/metabolism , Gene Silencing , Genetic Testing , Humans , Multivesicular Bodies/metabolism , Protein Transport , RNA, Small Interfering , rab5 GTP-Binding Proteins/genetics
9.
Semin Liver Dis ; 39(3): 283-290, 2019 07.
Article in English | MEDLINE | ID: mdl-31041790

ABSTRACT

Lipid droplets (LDs) are key sites of neutral lipid storage that can be found in all cells. Metabolic imbalances between the synthesis and degradation of LDs can result in the accumulation of significant amounts of lipid deposition, a characteristic feature of hepatocytes in patients with fatty liver disease, a leading indication for liver transplant in the United States. In this review, the authors highlight new literature related to the synthesis and autophagic catabolism of LDs, discussing key proteins and machinery involved in these processes. They also discuss recent findings that have revealed novel genetic risk factors associated with LD biology that contribute to lipid retention in the diseased liver.


Subject(s)
Autophagy , Fatty Liver/metabolism , Lipid Droplets/metabolism , Animals , Fatty Liver/physiopathology , Humans , Non-alcoholic Fatty Liver Disease/genetics
10.
J Biol Chem ; 293(28): 11143-11153, 2018 07 13.
Article in English | MEDLINE | ID: mdl-29853638

ABSTRACT

Inflammation is a major driver of tumor progression and metastasis, although the mechanisms by which proinflammatory cytokines drive metastatic invasion are unknown. Interleukin-6 (IL-6) is a potent proinflammatory cytokine that is elevated in individuals with pancreatic cancer (PDAC), is required for PDAC progression in mice, and increases tumor cell invasion in vitro Here, we provide insights into the mechanisms by which IL-6 activates tumor cell invasion. We found that IL-6 stimulation rapidly and robustly activates the small GTPase cell division cycle 42 (CDC42) in human PDAC cells and promotes the formation of premigratory filopodia. The CDC42 activation was required for IL-6-induced invasion as blocking CDC42 activity rendered the cells insensitive to IL-6's proinvasive effects. Loss of Janus kinase 2 (JAK2) or signal transducer and activator of transcription 3 (STAT3) prevented IL-6-mediated CDC42 activation, indicating that IL-6 activates CDC42 through both JAK2 and STAT3. However, the rapid activation of CDC42 suggested that this activation may be distinct from canonical STAT3-mediated transcriptional activation. Importantly, we observed an interaction between STAT3 and IQ motif-containing GTPase-activating protein 1 (IQGAP1), a scaffolding platform that binds CDC42. STAT3 colocalized with CDC42 and IQGAP1 at the plasma membrane, suggesting cross-talk between IL-6-mediated STAT3 signaling and CDC42 activation. These results suggest that IL-6 promotes metastatic invasion, at least partially, through CDC42 and that, along with its pleiotropic effects on tumor growth and progression, IL-6 signaling also activates proinvasive GTPase signaling, priming tumor cells for metastatic invasion.


Subject(s)
Cell Movement , Gene Expression Regulation, Neoplastic , Interleukin-6/pharmacology , Pancreatic Neoplasms/pathology , cdc42 GTP-Binding Protein/metabolism , Cell Proliferation , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Tumor Cells, Cultured , cdc42 GTP-Binding Protein/genetics , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
11.
Am J Physiol Gastrointest Liver Physiol ; 316(4): G509-G518, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30714813

ABSTRACT

We are investigating the changes in hepatic lipid catabolism that contribute to alcohol-induced fatty liver. Following chronic ethanol (EtOH) exposure, abstinence from alcohol resolves steatosis. Here, we investigated the hepatocellular events that lead to this resolution by quantifying specific catabolic parameters that returned to control levels after EtOH was withdrawn. We hypothesized that, after its chronic consumption, EtOH withdrawal reactivates lipid catabolic processes that restore lipostasis. Male Wistar rats were fed control and EtOH liquid diets for 6 wk. Randomly chosen EtOH-fed rats were then fed control diet for 7 days. Liver triglycerides (TG), lipid peroxides, key markers of fatty acid (FA) metabolism, lipophagy, and autophagy were quantified. Compared with controls, EtOH-fed rats had higher hepatic triglycerides, lipid peroxides, and serum free fatty acids (FFA). The latter findings were associated with higher levels of FA transporters (FATP 2, 4, and 5) but lower quantities of peroxisome proliferator-activated receptor-α (PPAR-α), which governs FA oxidation. EtOH-fed animals also had lower nuclear levels of the autophagy-regulating transcription factor EB (TFEB), associated with lower hepatic lipophagy and autophagy. After EtOH-fed rats were refed control diet for 7 days, their serum FFA levels and those of FATPs fell to control (normal) levels, whereas PPAR-α levels rose to normal. Hepatic TG and malondialdehyde levels in EtOH-withdrawn rats declined to near control levels. EtOH withdrawal restored nuclear TFEB content, hepatic lipophagy, and autophagy activity to control levels. EtOH withdrawal reversed aberrant FA metabolism and restored lysosomal function to promote resolution of alcohol-induced fatty liver. NEW & NOTEWORTHY Here, using an animal model, we show mechanisms of reversal of fatty liver and injury following EtOH withdrawal. Our data indicate that reactivation of autophagy and lysosome function through the restoration of transcription factor EB contribute to reversal of fatty liver and injury following EtOH withdrawal.


Subject(s)
Autophagy-Related Proteins/metabolism , Ethanol/pharmacokinetics , Fatty Liver, Alcoholic , Hepatocytes/metabolism , Liver Regeneration/physiology , Alcohol Abstinence , Animals , Autophagy/physiology , Central Nervous System Depressants/pharmacokinetics , Cyclic AMP Receptor Protein/metabolism , Fatty Acid Transport Proteins/metabolism , Fatty Liver, Alcoholic/metabolism , Fatty Liver, Alcoholic/pathology , Peroxisome Proliferator-Activated Receptors/metabolism , Rats , Rats, Wistar
12.
Hepatology ; 68(2): 751-762, 2018 08.
Article in English | MEDLINE | ID: mdl-29331069

ABSTRACT

While the life cycles of hepatitis viruses (A, B, C, D, and E) have been modestly characterized, recent intensive studies have provided new insights. Because these viruses "hijack" the membrane trafficking of the host cell machinery during replicative propagation, it is essential to determine and understand these specific cellular pathways. Hepatitis B virus (HBV) and hepatitis C virus are well known as leading causes of liver cirrhosis and hepatocellular carcinoma. While substantial inroads toward treating hepatitis C virus patients have recently been made, patients with HBV continue to require lifelong treatment, which makes a thorough understanding of the HBV life cycle essential. Importantly, these viruses have been observed to "hijack" the secretory and endocytic membrane trafficking machineries of the hepatocyte. These can include the canonical clathrin-mediated endocytic process that internalizes virus through cell surface receptors. While these receptors are encoded by the host genome for normal hepatocellular functions, they also exhibit virus-specific recognition. Further, functions provided by the multivesicular body, which include endosomal sorting complexes required for transport, are now known to envelope a variety of different hepatitis viruses. In this review, we summarize the recent findings regarding the cellular membrane trafficking machineries used by HBV in the context of other hepatitis viruses. (Hepatology 2018; 00:000-000).


Subject(s)
Cell Membrane/physiology , Hepatitis Viruses/pathogenicity , Hepatocytes/virology , Protein Transport/physiology , Hepatocytes/physiology , Humans
13.
J Biol Chem ; 292(28): 11815-11828, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28515323

ABSTRACT

In liver steatosis (i.e. fatty liver), hepatocytes accumulate many large neutral lipid storage organelles known as lipid droplets (LDs). LDs are important in the maintenance of energy homeostasis, but the signaling mechanisms that stimulate LD metabolism in hepatocytes are poorly defined. In adipocytes, catecholamines target the ß-adrenergic (ß-AR)/cAMP pathway to activate cytosolic lipases and induce their recruitment to the LD surface. Therefore, the goal of this study was to determine whether hepatocytes, like adipocytes, also undergo cAMP-mediated lipolysis in response to ß-AR stimulation. Using primary rat hepatocytes and human hepatoma cells, we found that treatment with the ß-AR agent isoproterenol caused substantial LD loss via activation of cytosolic lipases adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL). ß-Adrenergic stimulation rapidly activated PKA, which led to the phosphorylation of ATGL and HSL and their recruitment to the LD surface. To test whether this ß-AR-dependent lipolysis pathway was altered in a model of alcoholic fatty liver, primary hepatocytes from rats fed a 6-week EtOH-containing Lieber-DeCarli diet were treated with cAMP agonists. Compared with controls, EtOH-exposed hepatocytes showed a drastic inhibition in ß-AR/cAMP-induced LD breakdown and the phosphorylation of PKA substrates, including HSL. This observation was supported in VA-13 cells, an EtOH-metabolizing human hepatoma cell line, which displayed marked defects in both PKA activation and isoproterenol-induced ATGL translocation to the LD periphery. In summary, these findings suggest that ß-AR stimulation mobilizes cytosolic lipases for LD breakdown in hepatocytes, and perturbation of this pathway could be a major consequence of chronic EtOH insult leading to fatty liver.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Cyclic AMP/agonists , Fatty Liver, Alcoholic/metabolism , Hepatocytes/drug effects , Lipolysis/drug effects , Receptors, Adrenergic, beta/metabolism , Second Messenger Systems/drug effects , Animals , Cell Line, Tumor , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/drug effects , Fatty Liver, Alcoholic/pathology , Female , Hepatocytes/cytology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Lipase/chemistry , Lipase/metabolism , Lipid Droplets/drug effects , Lipid Droplets/metabolism , Lipid Droplets/pathology , Male , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Rats , Receptors, Adrenergic, beta/chemistry , Sterol Esterase/chemistry , Sterol Esterase/metabolism
14.
Blood ; 128(9): 1234-45, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27297792

ABSTRACT

Peripheral T-cell lymphomas (PTCLs) represent a heterogeneous group of T-cell malignancies that generally demonstrate aggressive clinical behavior, often are refractory to standard therapy, and remain significantly understudied. The most common World Health Organization subtype is PTCL, not otherwise specified (NOS), essentially a "wastebasket" category because of inadequate understanding to assign cases to a more specific diagnostic entity. Identification of novel fusion genes has contributed significantly to improving the classification, biologic understanding, and therapeutic targeting of PTCLs. Here, we integrated mate-pair DNA and RNA next-generation sequencing to identify chromosomal rearrangements encoding expressed fusion transcripts in PTCL, NOS. Two of 11 cases had novel fusions involving VAV1, encoding a truncated form of the VAV1 guanine nucleotide exchange factor important in T-cell receptor signaling. Fluorescence in situ hybridization studies identified VAV1 rearrangements in 10 of 148 PTCLs (7%). These were observed exclusively in PTCL, NOS (11%) and anaplastic large cell lymphoma (11%). In vitro, ectopic expression of a VAV1 fusion promoted cell growth and migration in a RAC1-dependent manner. This growth was inhibited by azathioprine, a clinically available RAC1 inhibitor. We also identified novel kinase gene fusions, ITK-FER and IKZF2-ERBB4, as candidate therapeutic targets that show similarities to known recurrent oncogenic ITK-SYK fusions and ERBB4 transcript variants in PTCLs, respectively. Additional novel and potentially clinically relevant fusions also were discovered. Together, these findings identify VAV1 fusions as recurrent and targetable events in PTCLs and highlight the potential for clinical sequencing to guide individualized therapy approaches for this group of aggressive malignancies.


Subject(s)
Lymphoma, T-Cell, Peripheral/genetics , Oncogene Proteins, Fusion/genetics , Aged , Animals , Female , High-Throughput Nucleotide Sequencing , Humans , Jurkat Cells , Lymphoma, T-Cell, Peripheral/metabolism , Male , Mice , Middle Aged , NIH 3T3 Cells , Oncogene Proteins, Fusion/metabolism
15.
J Biol Chem ; 291(32): 16424-37, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27226592

ABSTRACT

Clathrin-mediated endocytosis of transferrin (Tf) and its cognate receptor (TfR1) is a central pathway supporting the uptake of trophic iron. It has generally been assumed that this is a constitutive process. However, we have reported that the non-receptor tyrosine kinase, Src, is activated by Tf to facilitate the internalization of the Tf-TfR1 ligand-receptor complex. As an extension of these findings, we have tested whether subsequent trafficking steps might be regulated by additional kinase-dependent cascades, and we observed a significant endocytic block by inhibiting c-Abl kinase by a variety of methods. Importantly, Tf internalization was reduced significantly in all of these cell models and could be restored by re-expression of WT c-Abl. Surprisingly, this attenuated Tf-TfR1 endocytosis was due to a substantial drop in both the surface and total cellular receptor levels. Additional studies with the LDL receptor showed a similar effect. Surprisingly, immunofluorescence microscopy of imatinib-treated cells revealed a marked colocalization of internalized TfR1 with late endosomes/lysosomes, whereas attenuating the lysosome function with several inhibitors reduced this receptor loss. Importantly, inhibition of c-Abl resulted in a striking redistribution of the chaperone Hsc70 from a diffuse cytosolic localization to an association with the TfR1 at the late endosome-lysosome. Pharmacological inhibition of Hsc70 ATPase activity in cultured cells by the drug VER155008 prevents this chaperone-receptor interaction, resulting in an accumulation of the TfR1 in the early endosome. Thus, inhibition of c-Abl minimizes receptor recycling pathways and results in chaperone-dependent trafficking of the TfR1 to the lysosome for degradation. These findings implicate a novel role for c-Abl and Hsc70 as an unexpected regulator of Hsc70-mediated transport of trophic receptor cargo between the early and late endosomal compartments.


Subject(s)
Antigens, CD/metabolism , Endocytosis/physiology , Proto-Oncogene Proteins c-abl/metabolism , Receptors, Transferrin/metabolism , Animals , Antigens, CD/genetics , Endosomes/genetics , Endosomes/metabolism , HSC70 Heat-Shock Proteins/genetics , HSC70 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Lysosomes/genetics , Lysosomes/metabolism , Protein Transport/physiology , Proto-Oncogene Proteins c-abl/genetics , Rats , Receptors, Transferrin/genetics
16.
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
17.
Hepatology ; 73 Suppl 1: 1-3, 2021 01.
Article in English | MEDLINE | ID: mdl-33616970
18.
Hepatology ; 61(6): 1896-907, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25565581

ABSTRACT

UNLABELLED: Autophagy is a central mechanism by which hepatocytes catabolize lipid droplets (LDs). Currently, the regulatory mechanisms that control this important process are poorly defined. The small guanosine triphosphatase (GTPase) Rab7 has been implicated in the late endocytic pathway and is known to associate with LDs, although its role in LD breakdown has not been tested. In this study, we demonstrate that Rab7 is indispensable for LD breakdown ("lipophagy") in hepatocytes subjected to nutrient deprivation. Importantly, Rab7 is dramatically activated in cells placed under nutrient stress; this activation is required for the trafficking of both multivesicular bodies and lysosomes to the LD surface during lipophagy, resulting in the formation of a lipophagic "synapse." Depletion of Rab7 leads to gross morphological changes of multivesicular bodies, lysosomes, and autophagosomes, consequently leading to attenuation of hepatocellular lipophagy. CONCLUSION: These findings provide additional support for the role of autophagy in hepatocellular LD catabolism while implicating the small GTPase Rab7 as a key regulatory component of this essential process.


Subject(s)
Autophagy , Hepatocytes/metabolism , Lipid Droplets/metabolism , Lipolysis , rab GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Humans , Lysosomes/physiology , Multivesicular Bodies/physiology , rab7 GTP-Binding Proteins
19.
BMC Gastroenterol ; 16: 27, 2016 Feb 29.
Article in English | MEDLINE | ID: mdl-26924554

ABSTRACT

BACKGROUND: Non-alcoholic and alcoholic fatty liver disease (NAFLD and AFLD, respectively) are major health problems, as patients with either condition can progress to hepatitis, fibrosis, and cirrhosis. Although histologically similar, key differences likely exist in these two models. For example, altered content of several vesicle trafficking proteins have been identified in AFLD, but their content in NAFLD is unknown. In this study, we compared select parameters in NAFLD and AFLD in a rat model. METHODS: We fed either Lieber- DeCarli liquid control or alcohol-containing (35 % as calories) diet (AFLD model) or lean or high-fat (12 or 60 % derived from fat, respectively) pellets (NAFLD model) for 8-10 weeks, n = 8 in each model. Serum, hepatocytes and liver tissue were analyzed. Liver injury markers were measured in serum, triglyceride content and endocytosis (binding and internalization of (125)I- asialoorosomucoid) was measured in isolated hepatocytes, and content of selected trafficking proteins (Rab3D, Rab7 and Rab18) were determined in whole liver tissue. RESULTS: Although liver injury markers and triglyceride content were similar in both models, binding and internalization of (125)I- asialoorosomucoid was significantly impaired in the hepatocytes from AFLD, but not NAFLD, animals. In addition, protein content of the asialoglycoprotein receptor (ASGPR) and three trafficking proteins, Rab3D, Rab7and Rab18, were significantly decreased after alcohol, but not high-fat feeding. Levels of protein carbonylation, amount of glutathione stores, and lipid peroxidation were similar irrespective of the insult to the livers that resulted in fatty liver. CONCLUSION: Impairments in protein trafficking in AFLD are likely a direct result of alcohol administration, and not a function of fatty liver.


Subject(s)
Endocytosis/physiology , Fatty Liver, Alcoholic/metabolism , Hepatocytes/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , RNA, Messenger/metabolism , Transport Vesicles/metabolism , Alanine Transaminase/metabolism , Alkaline Phosphatase/metabolism , Animals , Asialoglycoprotein Receptor/genetics , Asialoglycoprotein Receptor/metabolism , Bile Acids and Salts/metabolism , Blotting, Western , Cholesterol/metabolism , Diet, High-Fat , Disease Models, Animal , Ethanol/toxicity , Fatty Liver, Alcoholic/etiology , Immunohistochemistry , Membrane Proteins/metabolism , Perilipin-2 , Rats , Serum Albumin/metabolism , Solvents/toxicity , Triglycerides/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab3 GTP-Binding Proteins/genetics , rab3 GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL