Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 883
Filter
1.
Traffic ; 25(9): e12956, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39313937

ABSTRACT

Limited nutrient availability in the tumor microenvironment can cause the rewiring of signaling and metabolic networks to confer cancer cells with survival advantages. We show here that the limitation of glucose, glutamine and serum from the culture medium resulted in the survival of a population of cancer cells with high viability and capacity to form tumors in vivo. These cells also displayed a remarkable increase in the abundance and size of lysosomes. Moreover, lysosomes were located mainly in the perinuclear region in nutrient-limited cells; this translocation was mediated by a rapid post-transcriptional increase in the key endolysosomal trafficking protein Rab7a. The acidic lysosomes in nutrient-limited cells could trap weakly basic drugs such as doxorubicin, mediating resistance of the cells to the drug, which could be partially reversed with the lysosomal inhibitor bafilomycin A1. An in vivo chorioallantoic membrane (CAM) assay indicated a remarkable decrease in microtumor volume when nutrient-limited cells were treated with 5-Fluorouracil (5-FU) and bafilomycin A1 compared to cells treated with either agent alone. Overall, our data indicate the activation of complementary pathways with nutrient limitation that can enable cancer cells to survive, proliferate and acquire drug resistance.


Subject(s)
Drug Resistance, Neoplasm , Lysosomes , Macrolides , rab7 GTP-Binding Proteins , Humans , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Fluorouracil/pharmacology , Lysosomes/metabolism , Macrolides/pharmacology , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/drug therapy , Nutrients/metabolism , rab7 GTP-Binding Proteins/metabolism
2.
Curr Biol ; 34(19): 4476-4494.e11, 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39276769

ABSTRACT

Kinesin and dynein-dynactin motors move endosomes and other vesicles bidirectionally along microtubules, a process mainly studied under in vitro conditions. Here, we provide a physiological bidirectional transport model following color-coded, endogenously tagged transport-related proteins as they move through a crowded cellular environment. Late endosomes (LEs) surf bidirectionally on Protrudin-enriched endoplasmic reticulum (ER) membrane contact sites, while hopping and gliding along microtubules and bypassing cellular obstacles, such as mitochondria. During bidirectional transport, late endosomes do not switch between opposing Rab7 GTPase effectors, RILP and FYCO1, or their associated dynein and KIF5B motor proteins, respectively. In the endogenous setting, far fewer motors associate with endosomal membranes relative to effectors, implying coordination of transport with other aspects of endosome physiology through GTPase-regulated mechanisms. We find that directionality of transport is provided in part by various microtubule-associated proteins (MAPs), including MID1, EB1, and CEP169, which recruit Lis1-activated dynein motors to microtubule plus ends for transport of early and late endosomal populations. At these microtubule plus ends, activated dynein motors encounter the dynactin subunit p150glued and become competent for endosomal capture and minus-end movement in collaboration with membrane-associated Rab7-RILP. We show that endosomes surf over the ER through the crowded cell and move bidirectionally under the control of MAPs for motor activation and through motor replacement and capture by endosomal anchors.


Subject(s)
Endosomes , Microtubules , Endosomes/metabolism , Humans , Microtubules/metabolism , Dyneins/metabolism , Biological Transport , Microtubule-Associated Proteins/metabolism , HeLa Cells , Endoplasmic Reticulum/metabolism , Kinesins/metabolism , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins , Protein Transport
3.
EMBO J ; 43(18): 3948-3967, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39103493

ABSTRACT

Lysosomes play a pivotal role in coordinating macromolecule degradation and regulating cell growth and metabolism. Despite substantial progress in identifying lysosomal signaling proteins, understanding the pathways that synchronize lysosome functions with changing cellular demands remains incomplete. This study uncovers a role for TANK-binding kinase 1 (TBK1), well known for its role in innate immunity and organelle quality control, in modulating lysosomal responsiveness to nutrients. Specifically, we identify a pool of TBK1 that is recruited to lysosomes in response to elevated amino acid levels. This lysosomal TBK1 phosphorylates Rab7 on serine 72. This is critical for alleviating Rab7-mediated inhibition of amino acid-dependent mTORC1 activation. Furthermore, a TBK1 mutant (E696K) associated with amyotrophic lateral sclerosis and frontotemporal dementia constitutively accumulates at lysosomes, resulting in elevated Rab7 phosphorylation and increased mTORC1 activation. This data establishes the lysosome as a site of amino acid regulated TBK1 signaling that is crucial for efficient mTORC1 activation. This lysosomal pool of TBK1 has broader implications for lysosome homeostasis, and its dysregulation could contribute to the pathogenesis of ALS-FTD.


Subject(s)
Amino Acids , Lysosomes , Mechanistic Target of Rapamycin Complex 1 , Protein Serine-Threonine Kinases , rab7 GTP-Binding Proteins , Humans , Amino Acids/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , HEK293 Cells , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Signal Transduction
4.
FEBS J ; 291(18): 4142-4154, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39097908

ABSTRACT

DABMA is a chemical molecule optimized from the parent compound ABMA and exhibits broad-spectrum antipathogenic activity by modulating the host's endolysosomal and autophagic pathways. Both DABMA and ABMA inhibit severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in a cellular assay, which further expands their anti-pathogen spectrum in vitro. However, their precise mechanism of action has not yet been resolved. TMEM175 is a newly characterized endolysosomal channel which plays an essential role in the homeostasis of endosomes and lysosomes as well as organelle fusion. Here, we show that DABMA increases the endosomal TMEM175 current through organelle patch clamping with an EC50 of 17.9 µm. Depletion of TMEM175 protein significantly decreases the antitoxin activity of DABMA and affects its action on acidic- and Rab7-positive endosomes as well as on endolysosomal trafficking. Thus, TMEM175 is necessary for DABMA's activity and may represent a druggable target for the development of anti-infective drugs. Moreover, DABMA, as an activator of the TMEM175 channel, may be useful for the in-depth characterization of the physiological and pathological roles of this endolysosomal channel.


Subject(s)
Endosomes , Lysosomes , SARS-CoV-2 , Humans , Endosomes/metabolism , Endosomes/drug effects , Lysosomes/metabolism , Lysosomes/drug effects , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , HEK293 Cells , rab7 GTP-Binding Proteins , Antiviral Agents/pharmacology , Ion Channels/metabolism , Ion Channels/genetics , Animals , COVID-19 Drug Treatment , HeLa Cells , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , COVID-19/virology , COVID-19/metabolism
5.
Curr Biol ; 34(18): 4170-4183.e9, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39181128

ABSTRACT

Proteome maintenance in contracting skeletal and cardiac muscles depends on the chaperone-regulating protein BAG3. Reduced BAG3 activity leads to muscle weakness and heart failure in animal models and patients. BAG3 and its chaperone partners recognize mechanically damaged muscle proteins and initiate their disposal through chaperone-assisted selective autophagy (CASA). However, molecular details of the force-dependent regulation of BAG3 have remained elusive so far. Here, we demonstrate that mechanical stress triggers the dephosphorylation of BAG3 in human muscle and in isolated cells. We identify force-regulated phospho-switches in BAG3 that control CASA complex assembly and CASA activity. Differential proteomics reveal RAB GTPases, which organize membrane traffic and fusion, as dephosphorylation-dependent interactors of BAG3. In fact, RAB7A and RAB11B are shown here to be essential for CASA in skeletal muscle cells. Moreover, BAG3 dephosphorylation is also observed upon induction of mitophagy, suggesting an involvement of the cochaperone in the RAB7A-dependent autophagic engulfment of damaged mitochondria in exercised muscle. Cooperation of BAG3 with RAB7A relies on a direct interaction of both proteins, which is regulated by the nucleotide state of the GTPase and by association with the autophagosome membrane protein LC3B. Finally, we provide evidence that BAG3 and RAB7A also cooperate in non-muscle cells and propose that overactivation of CASA in RAB7A-L129F patients contributes to the loss of peripheral neurons in Charcot-Marie-Tooth neuropathy.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins , rab GTP-Binding Proteins , rab7 GTP-Binding Proteins , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Phosphorylation , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins/metabolism , Proteostasis , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Muscle, Skeletal/metabolism , Autophagy/physiology , Animals , Mice , Protein Transport
6.
Cells ; 13(16)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39195203

ABSTRACT

Endoplasmic reticulum (ER)-endolysosome interactions regulate cholesterol exchange between the ER and the endolysosome. ER-endolysosome membrane contact sites mediate the ER-endolysosome interaction. VAP-ORP1L (vesicle-associated membrane protein-associated protein- OSBP-related protein 1L) interaction forms the major contact site between the ER and the lysosome, which is regulated by Rab7. RILP (Rab7-interacting lysosomal protein) is the downstream effector of Rab7, but its role in the organelle interaction between the ER and the lysosome is not clear. In this study, we found RILP interacts with ORP1L to competitively inhibit the formation of the VAP-ORP1L contact site. Immunofluorescence microscopy revealed that RILP induces late endosome/lysosome clustering, which reduces the contact of endolysosomes with the ER, interfering with the ER-endolysosome interaction. Further examination demonstrated that over-expression of RILP results in the accumulation of cholesterol in the clustered endolysosomes, which triggers cellular autophagy depending on RILP. Our results suggest that RILP interferes with the ER-endolysosome interaction to inhibit cholesterol flow from the endolysosome to the ER, which feedbacks to trigger autophagy.


Subject(s)
Cholesterol , Endoplasmic Reticulum , Endosomes , Lysosomes , Lysosomes/metabolism , Cholesterol/metabolism , Endoplasmic Reticulum/metabolism , Humans , Endosomes/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Autophagy , HeLa Cells , Receptors, Steroid/metabolism , Vesicular Transport Proteins/metabolism , Protein Binding , rab7 GTP-Binding Proteins , HEK293 Cells
7.
Dev Cell ; 59(17): 2287-2301.e6, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39111309

ABSTRACT

Autophagy is a universal degradation system in eukaryotic cells. In plants, although autophagosome biogenesis has been extensively studied, the mechanism of how autophagosomes are transported to the vacuole for degradation remains largely unexplored. In this study, we demonstrated that upon autophagy induction, Arabidopsis homotypic fusion and protein sorting (HOPS) subunit VPS41 converts first from condensates to puncta, then to ring-like structures, termed VPS41-associated phagic vacuoles (VAPVs), which enclose autophagy-related gene (ATG)8s for vacuolar degradation. This process is initiated by ADP ribosylation factor (ARF)-like GTPases ARLA1s and occurs concurrently with autophagy progression through coupling with the synaptic-soluble N-ethylmaleimide-sensitive factor attachment protein rmleceptor (SNARE) proteins. Unlike in other eukaryotes, autophagy degradation in Arabidopsis is largely independent of the RAB7 pathway. By contrast, dysfunction in the condensates-to-VAPVs conversion process impairs autophagosome structure and disrupts their vacuolar transport, leading to a significant reduction in autophagic flux and plant survival rate. Our findings suggest that the conversion pathway might be an integral part of the autophagy program unique to plants.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Autophagosomes , Autophagy , Vacuoles , Arabidopsis/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Vacuoles/metabolism , Autophagosomes/metabolism , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Autophagy-Related Protein 8 Family/metabolism , Autophagy-Related Protein 8 Family/genetics , SNARE Proteins/metabolism , SNARE Proteins/genetics , rab7 GTP-Binding Proteins , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics
8.
J Extracell Vesicles ; 13(7): e12494, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39051763

ABSTRACT

Microvesicles (MVs) containing proteins, nucleic acid or organelles are shed from the plasma membrane. Although the mechanisms of MV budding are well elucidated, the connection between endosomal trafficking and MV formation remains poorly understood. In this report, RAB22A is revealed to be crucial for EGFR-containing MVs formation by the RAB GTPase family screening. RAB22A recruits TBC1D2B, a GTPase-activating protein (GAP) of RAB7A, to inactivate RAB7A, thus preventing EGFR from being transported to late endosomes and lysosomes. RAB22A also engages SH3BP5L, a guanine-nucleotide exchange factor (GEF) of RAB11A, to activate RAB11A on early endosomes. Consequently, EGFR is recycled to the cell surface and packaged into MVs. Furthermore, EGFR can phosphorylate RAB22A at Tyr136, which in turn promotes EGFR-containing MVs formation. Our findings illustrate that RAB22A acts as a sorter on early endosomes to sort EGFR to recycling endosomes for MV shedding by both activating RAB11A and inactivating RAB7A.


Subject(s)
Endosomes , ErbB Receptors , rab GTP-Binding Proteins , ErbB Receptors/metabolism , rab GTP-Binding Proteins/metabolism , Endosomes/metabolism , Humans , Protein Transport , Cell-Derived Microparticles/metabolism , rab7 GTP-Binding Proteins/metabolism , HeLa Cells , GTPase-Activating Proteins/metabolism , Lysosomes/metabolism
9.
Nat Commun ; 15(1): 6311, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39060258

ABSTRACT

Respiratory syncytial virus (RSV) hijacks cholesterol or autophagy pathways to facilitate optimal replication. However, our understanding of the associated molecular mechanisms remains limited. Here, we show that RSV infection blocks cholesterol transport from lysosomes to the endoplasmic reticulum by downregulating the activity of lysosomal acid lipase, activates the SREBP2-LDLR axis, and promotes uptake and accumulation of exogenous cholesterol in lysosomes. High cholesterol levels impair the VAP-A-binding activity of ORP1L and promote the recruitment of dynein-dynactin, PLEKHM1, or HOPS VPS39 to Rab7-RILP, thereby facilitating minus-end transport of autophagosomes and autolysosome formation. Acidification inhibition and dysfunction of cholesterol-rich lysosomes impair autophagy flux by inhibiting autolysosome degradation, which promotes the accumulation of RSV fusion protein. RSV-F storage is nearly abolished after cholesterol depletion or knockdown of LDLR. Most importantly, the knockout of LDLR effectively inhibits RSV infection in vivo. These findings elucidate the molecular mechanism of how RSV co-regulates lysosomal cholesterol reprogramming and autophagy and reveal LDLR as a novel target for anti-RSV drug development.


Subject(s)
Autophagy , Cholesterol , Lysosomes , Receptors, LDL , Respiratory Syncytial Virus Infections , Vesicular Transport Proteins , Virus Replication , rab GTP-Binding Proteins , rab7 GTP-Binding Proteins , Lysosomes/metabolism , Cholesterol/metabolism , Humans , Animals , Receptors, LDL/metabolism , Receptors, LDL/genetics , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/virology , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Mice , Dynactin Complex/metabolism , Endoplasmic Reticulum/metabolism , Dyneins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Respiratory Syncytial Virus, Human/physiology , Autophagosomes/metabolism , Viral Fusion Proteins/metabolism , Viral Fusion Proteins/genetics , HeLa Cells , A549 Cells
10.
Plant Physiol Biochem ; 214: 108928, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39033652

ABSTRACT

The RING-type E3 ligases play a significant role in stress signaling, primarily through post-translational regulation. Ubiquitination is a crucial post-translational modification that regulates the turnover and activity of proteins. The overexpression of AlRabring7, RING-HC E3 Ub ligase in tobacco provides insights into the regulation of salinity and ABA signaling in transgenic tobacco. The seed germination potential of AlRabring7 transgenics was higher than WT, with NaCl and ABA treatments. The transgenics showed improved morpho-physio-biochemical parameters in response to salinity and ABA treatments. The photosynthetic pigments, soluble sugars, reducing sugars and proline increased in transgenics in response to NaCl and ABA treatments. The decreased ROS accumulation in transgenics on NaCl and ABA treatments can be co-related to improved activity of enzymatic and non-enzymatic antioxidants. The potential of transgenics to maintain ABA levels with ABA treatment, highlights the active participation of ABA feedback loop mechanism. Interestingly, the ability of AlRabring7 transgenics to upregulate Rab7 protein, suggests its role in facilitating vacuolar transport. Furthermore, the improved potassium accumulation and reduced sodium content indicate an efficient ion regulation mechanism in transgenic plants facilitating higher stomatal opening. The expression of downstream ion transporter (NbNHX1 and NbVHA1), ABA signaling (NbABI2 and NbABI5) and vesicle trafficking (NbMON1) responsive genes were upregulated with stress. The present study, reports that AlRabring7 participates in maintaining vacuolar transport, ion balance, ROS homeostasis, stomatal regulation through activation of Rab7 protein and regulation of downstream stress-responsive during stress. This emphasizes the potential of AlRabring7 gene for improved performance and resilience in challenging environments.


Subject(s)
Nicotiana , Plant Proteins , Plants, Genetically Modified , Salt Tolerance , Signal Transduction , rab GTP-Binding Proteins , Nicotiana/genetics , Nicotiana/metabolism , Salt Tolerance/genetics , Plant Proteins/metabolism , Plant Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins , Abscisic Acid/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Gene Expression Regulation, Plant , Stress, Physiological/genetics , Reactive Oxygen Species/metabolism
11.
Int J Mol Sci ; 25(11)2024 May 31.
Article in English | MEDLINE | ID: mdl-38892241

ABSTRACT

Glutamate functions as the major excitatory neurotransmitter for primary sensory neurons and has a crucial role in sensitizing peripheral nociceptor terminals producing sensitization. Glutaminase (GLS) is the synthetic enzyme that converts glutamine to glutamate. GLS-immunoreactivity (-ir) and enzyme activity are elevated in dorsal root ganglion (DRG) neuronal cell bodies during chronic peripheral inflammation, but the mechanism for this GLS elevation is yet to be fully characterized. It has been well established that, after nerve growth factor (NGF) binds to its high-affinity receptor tropomyosin receptor kinase A (TrkA), a retrograde signaling endosome is formed. This endosome contains the late endosomal marker Rab7GTPase and is retrogradely transported via axons to the cell soma located in the DRG. This complex is responsible for regulating the transcription of several critical nociceptive genes. Here, we show that this retrograde NGF signaling mediates the expression of GLS in DRG neurons during the process of peripheral inflammation. We disrupted the normal NGF/TrkA signaling in adjuvant-induced arthritic (AIA) Sprague Dawley rats by the pharmacological inhibition of TrkA or blockade of Rab7GTPase, which significantly attenuated the expression of GLS in DRG cell bodies. The results indicate that NGF/TrkA signaling is crucial for the production of glutamate and has a vital role in the development of neurogenic inflammation. In addition, our pain behavioral data suggest that Rab7GTPase can be a potential target for attenuating peripheral inflammatory pain.


Subject(s)
Ganglia, Spinal , Glutaminase , Inflammation , Nerve Growth Factor , Rats, Sprague-Dawley , Receptor, trkA , Signal Transduction , Animals , Ganglia, Spinal/metabolism , Nerve Growth Factor/metabolism , Glutaminase/metabolism , Rats , Receptor, trkA/metabolism , Inflammation/metabolism , Inflammation/pathology , Male , Neurons/metabolism , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins
12.
J Alzheimers Dis ; 100(1): 279-296, 2024.
Article in English | MEDLINE | ID: mdl-38848175

ABSTRACT

Background: Mitochondrial dysfunction exists in Alzheimer's disease (AD) brain, and damaged mitochondria need to be removed by mitophagy. Small GTPase Rab7 regulates the fusion of mitochondria and lysosome, while TBC1D5 inhibits Rab7 activation. However, it is not clear whether the regulation of Rab7 activity by TBC1D5 can improve mitophagy and inhibit AD progression. Objective: To investigate the role of TBC1D5 in mitophagy and its regulatory mechanism for Rab7, and whether activation of mitophagy can inhibit the progression of AD. Methods: Mitophagy was determined by western blot and immunofluorescence. The morphology and quantity of mitochondria were tracked by TEM. pCMV-Mito-AT1.03 was employed to detect the cellular ATP. Amyloid-ß secreted by AD cells was detected by ELISA. Co-immunoprecipitation was used to investigate the binding partner of the target protein. Golgi-cox staining was applied to observe neuronal morphology of mice. The Morris water maze test and Y-maze were performed to assess spatial learning and memory, and the open field test was measured to evaluate motor function and anxiety-like phenotype of experimental animals. Results: Mitochondrial morphology was impaired in AD models, and TBC1D5 was highly expressed. Knocking down TBC1D5 increased the expression of active Rab7, promoted the fusion of lysosome and autophagosome, thus improving mitophagy, and improved the morphology of hippocampal neurons and the impaired behavior in AD mice. Conclusions: Knocking down TBC1D5 increased Rab7 activity and promoted the fusion of autophagosome and lysosome. Our study provided insights into the mechanisms that bring new possibilities for AD therapy targeting mitophagy.


Subject(s)
Alzheimer Disease , Disease Models, Animal , GTPase-Activating Proteins , Mitochondria , Mitophagy , rab GTP-Binding Proteins , rab7 GTP-Binding Proteins , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Mitophagy/physiology , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Mice , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Humans , Mitochondria/metabolism , Male , Mice, Transgenic , Amyloid beta-Peptides/metabolism , Neurons/metabolism , Neurons/pathology
13.
Adv Sci (Weinh) ; 11(30): e2402030, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38837686

ABSTRACT

Cadmium (Cd) is a neurotoxic contaminant that induces cognitive decline similar to that observed in Alzheimer's disease (AD). Autophagic flux dysfunction is attributed to the pathogenesis of AD, and this study aimed to investigate the effect of autophagy on environmental Cd-induced AD progression and the underlying mechanism. Here, Cd exposure inhibited autophagosome-lysosome fusion and impaired lysosomal function, leading to defects in autophagic clearance and then to APP accumulation and nerve cell death. Proteomic analysis coupled with Ingenuity Pathway Analysis (IPA) identified SIRT5 as an essential molecular target in Cd-impaired autophagic flux. Mechanistically, Cd exposure hampered the expression of SIRT5, thus increasing the succinylation of RAB7A at lysine 31 and inhibiting RAB7A activity, which contributed to autophagic flux blockade. Importantly, SIRT5 overexpression led to the restoration of autophagic flux blockade, the alleviation of Aß deposition and memory deficits, and the desuccinylation of RAB7A in Cd-exposed FAD4T mice. Additionally, SIRT5 levels decrease mainly in neurons but not in other cell clusters in the brains of AD patients according to single-nucleus RNA sequencing data from the public dataset GSE188545. This study reveals that SIRT5-catalysed RAB7A desuccinylation is an essential adaptive mechanism for the amelioration of Cd-induced autophagic flux blockade and AD-like pathogenesis.


Subject(s)
Alzheimer Disease , Autophagy , Cadmium , Disease Models, Animal , Sirtuins , rab GTP-Binding Proteins , rab7 GTP-Binding Proteins , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Animals , Mice , Cadmium/metabolism , Cadmium/toxicity , Autophagy/drug effects , Sirtuins/metabolism , Sirtuins/genetics , rab7 GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Humans , Male
14.
Biochem Biophys Res Commun ; 725: 150232, 2024 09 17.
Article in English | MEDLINE | ID: mdl-38897042

ABSTRACT

Molecular processes are orchestrated by various proteins that promote early endosomes to become late endosomes and eventually fuse with lysosomes, guaranteeing the degradation of the content. Rab7, which is localized to late endosomes, is one of the most well-known GTPases. ORP1L is recruited by Rab7 to facilitate the fusion of late endosomes and lysosomes. Here, we present the structure of GDP-bound Rab7 Q67L with ORP1L. Structural analysis, supported by biochemical and ITC binding experiments, not only provides structural insight into the interactions between the ORP1L ANK domain and Rab7 but also suggests that the GTPase activity of Rab7 does not interfere with its ORP1L-binding capacity.


Subject(s)
Guanosine Diphosphate , Protein Binding , rab GTP-Binding Proteins , rab7 GTP-Binding Proteins , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins/metabolism , Guanosine Diphosphate/metabolism , Guanosine Diphosphate/chemistry , Humans , Models, Molecular , Receptors, Steroid/metabolism , Receptors, Steroid/chemistry , Protein Conformation , Binding Sites
15.
Biochem Biophys Res Commun ; 724: 150198, 2024 09 10.
Article in English | MEDLINE | ID: mdl-38852504

ABSTRACT

Autophagy is a critical catabolic pathway that enables cells to survive and adapt to stressful conditions, especially nutrient deprivation. The fusion of autophagic vacuoles with lysosomes is the final step of autophagy, which degrades the engulfed contents into metabolic precursors for re-use by the cell. O-GlcNAc transferase (OGT) plays a crucial role in regulating autophagy flux in response to nutrient stress, particularly by targeting key proteins involved in autophagosome-lysosome fusion. However, the role of OGT in basal autophagy, which occurs at a low and constitutive levels under growth conditions, remains poorly understood. Silencing or inhibition of OGT was used to compare the effect of OGT downregulation on autophagy flux in the non-cancerous CCD841CoN and cancerous HCT116 human colon cell lines under nutrient-rich conditions. We provide evidence that the reduction of OGT activity impairs the maturation of autophagosomes, thereby blocking the completion of basal autophagy in both cell lines. Additionally, OGT inhibition results in the accumulation of lysosomes and enlarged late endosomes in the perinuclear region, as demonstrated by confocal imaging. This is associated with a defect in the localization of the small GTPase Rab7 to these organelles. The regulation of transport and fusion events between the endosomal and lysosomal compartments is crucial for maintaining the autophagic flux. These findings suggest an interplay between OGT and the homeostasis of the endolysosomal network in human cells.


Subject(s)
Autophagy , Down-Regulation , Endosomes , Lysosomes , N-Acetylglucosaminyltransferases , Nutrients , rab7 GTP-Binding Proteins , Humans , N-Acetylglucosaminyltransferases/metabolism , N-Acetylglucosaminyltransferases/genetics , Endosomes/metabolism , Lysosomes/metabolism , Nutrients/metabolism , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Colon/metabolism , Colon/pathology , HCT116 Cells , Autophagosomes/metabolism
16.
Cell Mol Life Sci ; 81(1): 249, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38836877

ABSTRACT

Protein ubiquitination is one of the most important posttranslational modifications (PTMs) in eukaryotes and is involved in the regulation of almost all cellular signaling pathways. The intracellular bacterial pathogen Legionella pneumophila translocates at least 26 effectors to hijack host ubiquitination signaling via distinct mechanisms. Among these effectors, SidC/SdcA are novel E3 ubiquitin ligases with the adoption of a Cys-His-Asp catalytic triad. SidC/SdcA are critical for the recruitment of endoplasmic reticulum (ER)-derived vesicles to the Legionella-containing vacuole (LCV). However, the ubiquitination targets of SidC/SdcA are largely unknown, which restricts our understanding of the mechanisms used by these effectors to hijack the vesicle trafficking pathway. Here, we demonstrated that multiple Rab small GTPases and target soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNARE) proteins are bona fide ubiquitination substrates of SidC/SdcA. SidC/SdcA-mediated ubiquitination of syntaxin 3 and syntaxin 4 promotes their unconventional pairing with the vesicle-SNARE protein Sec22b, thereby contributing to the membrane fusion of ER-derived vesicles with the phagosome. In addition, our data reveal that ubiquitination of Rab7 by SidC/SdcA is critical for its association with the LCV membrane. Rab7 ubiquitination could impair its binding with the downstream effector Rab-interacting lysosomal protein (RILP), which partially explains why LCVs avoid fusion with lysosomes despite the acquisition of Rab7. Taken together, our study reveals the biological mechanisms employed by SidC/SdcA to promote the maturation of the LCVs.


Subject(s)
Legionella pneumophila , Phagosomes , SNARE Proteins , Ubiquitination , rab GTP-Binding Proteins , Legionella pneumophila/metabolism , Humans , Phagosomes/metabolism , Phagosomes/microbiology , SNARE Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Animals , Qa-SNARE Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Vacuoles/metabolism , Vacuoles/microbiology , HEK293 Cells , Mice , rab7 GTP-Binding Proteins/metabolism , Monomeric GTP-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism
17.
Adv Sci (Weinh) ; 11(29): e2401676, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38837607

ABSTRACT

Diabetic cardiomyopathy (DbCM) is characterized by diastolic dysfunction, which progresses into heart failure and aberrant electrophysiology in diabetic patients. Dyslipidemia in type 2 diabetic patients leads to the accumulation of lipid droplets (LDs) in cardiomyocytes and results in lipid toxicity which has been suggested to drive DbCM. It is aimed to explore potential pathways that may boost LDs degradation in DbCM and restore cardiac function. LDs accumulation resulted in an increase in lipid toxicity in DbCM hearts is confirmed. Microlipophagy pathway, rather than traditional macrolipophagy, is activated in DbCM hearts. RNA-Seq data and Rab7-CKO mice implicate that Rab7 is a major modulator of the microlipophagy pathway. Mechanistically, Rab7 is phosphorylated at Tyrosine 183, which allows the recruitment of Rab-interacting lysosome protein (Rilp) to proceed LDs degradation by lysosome. Treating DbCM mice with Rab7 activator ML-098 enhanced Rilp level and rescued the observed cardiac dysfunction. Overall, Rab7-Rilp-mediated microlipophagy may be a promising target in the treatment of lipid toxicity in DbCM is suggested.


Subject(s)
Diabetic Cardiomyopathies , rab GTP-Binding Proteins , rab7 GTP-Binding Proteins , Animals , Male , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Autophagy , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/genetics , Disease Models, Animal , Lipid Droplets/metabolism , Lipid Metabolism , Mice, Knockout , Myocytes, Cardiac/metabolism , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins/metabolism
18.
J Cell Biochem ; 125(8): e30621, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38924128

ABSTRACT

Activating transcription factor 6 (ATF6) and its downstream genes are involved in progression of hepatocellular carcinoma (HCC). Herein, we demonstrated that sulfhydration of Ras-related protein Rab-7a (RAB7A) was regulated by ATF6. High expression of RAB7A indicated poor prognosis of HCC patients. RAB7A overexpression contributed to proliferation, colony formation, migration, and invasion of HepG2 and Hep3B cells. Furthermore, we found that RAB7A enhanced aerobic glycolysis in HepG2 cells, indicating a higher degree of tumor malignancy. Mechanistically, RAB7A suppressed Yes-associated protein 1 (YAP1) binding to 14-3-3 and conduced to YAP1 nuclear translocation and activation, promoting its downstream gene expression, thereby promoting growth and metastasis of liver cancer cells. In addition, knocking down RAB7A attenuated the progression of orthotopic liver tumors in mice. These findings illustrate the important role of RAB7A in regulating HCC progression. Thus, RAB7A may be a potential innovative target for HCC treatment.


Subject(s)
Adaptor Proteins, Signal Transducing , Carcinoma, Hepatocellular , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glycolysis , Liver Neoplasms , Transcription Factors , YAP-Signaling Proteins , rab7 GTP-Binding Proteins , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/genetics , YAP-Signaling Proteins/metabolism , Animals , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Mice , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Prognosis , Transcription Factors/metabolism , Transcription Factors/genetics , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Mice, Nude , Hep G2 Cells , Cell Movement , Neoplasm Metastasis , Mice, Inbred BALB C
19.
mBio ; 15(7): e0168423, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38874413

ABSTRACT

Lymphocytic choriomeningitis virus (LCMV) is an enveloped and segmented negative-sense RNA virus classified within the Arenaviridae family of the Bunyavirales order. LCMV is associated with fatal disease in immunocompromised populations and, as the prototypical arenavirus member, acts as a model for the many highly pathogenic members of the Arenaviridae family, such as Junín, Lassa, and Lujo viruses, all of which are associated with devastating hemorrhagic fevers. To enter cells, the LCMV envelope fuses with late endosomal membranes, for which two established requirements are low pH and interaction between the LCMV glycoprotein (GP) spike and secondary receptor CD164. LCMV subsequently uncoats, where the RNA genome-associated nucleoprotein (NP) separates from the Z protein matrix layer, releasing the viral genome into the cytosol. To further examine LCMV endosome escape, we performed an siRNA screen which identified host cell potassium ion (K+) channels as important for LCMV infection, with pharmacological inhibition confirming K+ channel involvement during the LCMV entry phase completely abrogating productive infection. To better understand the K+-mediated block in infection, we tracked incoming virions along their entry pathway under physiological conditions, where uncoating was signified by separation of NP and Z proteins. In contrast, K+ channel blockade prevented uncoating, trapping virions within Rab7 and CD164-positive endosomes, identifying K+ as a third LCMV entry requirement. K+ did not increase GP-CD164 binding or alter GP-CD164-dependent fusion. Thus, we propose that K+ mediates uncoating by modulating NP-Z interactions within the virion interior. These results suggest K+ channels represent a potential anti-arenaviral target.IMPORTANCEArenaviruses can cause fatal human disease for which approved preventative or therapeutic options are not available. Here, using the prototypical LCMV, we identified K+ channels as critical for arenavirus infection, playing a vital role during the entry phase of the infection cycle. We showed that blocking K+ channel function resulted in entrapment of LCMV particles within late endosomal compartments, thus preventing productive replication. Our data suggest K+ is required for LCMV uncoating and genome release by modulating interactions between the viral nucleoprotein and the matrix protein layer inside the virus particle.


Subject(s)
Endosomes , Lymphocytic choriomeningitis virus , Potassium , Virus Internalization , Virus Uncoating , Endosomes/virology , Endosomes/metabolism , Lymphocytic choriomeningitis virus/physiology , Lymphocytic choriomeningitis virus/genetics , Humans , Potassium/metabolism , rab7 GTP-Binding Proteins , Cell Line , Animals , Potassium Channels/metabolism , Potassium Channels/genetics
20.
Cell Mol Life Sci ; 81(1): 207, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38709385

ABSTRACT

The co-localization of the lysosomal protease cathepsin B (CTSB) and the digestive zymogen trypsinogen is a prerequisite for the initiation of acute pancreatitis. However, the exact molecular mechanisms of co-localization are not fully understood. In this study, we investigated the role of lysosomes in the onset of acute pancreatitis by using two different experimental approaches. Using an acinar cell-specific genetic deletion of the ras-related protein Rab7, important for intracellular vesicle trafficking and fusion, we analyzed the subcellular distribution of lysosomal enzymes and the severity of pancreatitis in vivo and ex vivo. Lysosomal permeabilization was performed by the lysosomotropic agent Glycyl-L-phenylalanine 2-naphthylamide (GPN). Acinar cell-specific deletion of Rab7 increased endogenous CTSB activity and despite the lack of re-distribution of CTSB from lysosomes to the secretory vesicles, the activation of CTSB localized in the zymogen compartment still took place leading to trypsinogen activation and pancreatic injury. Disease severity was comparable to controls during the early phase but more severe at later time points. Similarly, GPN did not prevent CTSB activation inside the secretory compartment upon caerulein stimulation, while lysosomal CTSB shifted to the cytosol. Intracellular trypsinogen activation was maintained leading to acute pancreatitis similar to controls. Our results indicate that initiation of acute pancreatitis seems to be independent of the presence of lysosomes and that fusion of lysosomes and zymogen granules is dispensable for the disease onset. Intact lysosomes rather appear to have protective effects at later disease stages.


Subject(s)
Cathepsin B , Lysosomes , Pancreatitis , Secretory Vesicles , rab GTP-Binding Proteins , rab7 GTP-Binding Proteins , Animals , Lysosomes/metabolism , Pancreatitis/metabolism , Pancreatitis/pathology , Pancreatitis/genetics , Cathepsin B/metabolism , Cathepsin B/genetics , Mice , Secretory Vesicles/metabolism , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins/metabolism , Acute Disease , Acinar Cells/metabolism , Acinar Cells/pathology , Trypsinogen/metabolism , Trypsinogen/genetics , Ceruletide , Enzyme Precursors/metabolism , Enzyme Precursors/genetics , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL