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










Publication year range
1.
Cancer Metab ; 12(1): 6, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38365771

ABSTRACT

BACKGROUND: Metastasis is the leading cause of death among prostate cancer (PCa) patients. Obesity is associated with both PCa-specific and all-cause mortality. High-fat diet (HFD) is a risk factor contributing to obesity. However, the association of HFD with PCa metastasis and its underlying mechanisms are unclear. METHODS: Tumor xenografts were conducted by intrasplenic injections. The ability of migration or invasion was detected by transwell assay. The expression levels of RPS27 were detected by QRT-PCR and western blot. RESULTS: The present study verified the increase in PCa metastasis caused by HFD in mice. Bioinformatics analysis demonstrated increased RPS27 in the experimentally induced PCa in HFD mice, indicating that it is an unfavorable prognostic factor. Intrasplenic injections were used to demonstrate that RPS27 overexpression promotes, while RPS27 knockdown significantly reduces, PCa liver metastasis. Moreover, RPS27 inhibition suppresses the effects of HFD on PCa metastasis. Further mRNA sequencing analysis revealed that RPS27 promotes PCa metastasis by selectively enhancing the expression of various genes. CONCLUSION: Our findings indicate that HFD increases the risk of PCa metastasis by elevating RPS27 expression and, subsequently, the expression of genes involved in PRAD progression. Therefore, RPS27 may serve as a novel target for the diagnosis and treatment of metastatic PCa.

3.
Nat Chem Biol ; 20(7): 835-846, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38287154

ABSTRACT

Synchronized ferroptosis contributes to nephron loss in acute kidney injury (AKI). However, the propagation signals and the underlying mechanisms of the synchronized ferroptosis for renal tubular injury remain unresolved. Here we report that platelet-activating factor (PAF) and PAF-like phospholipids (PAF-LPLs) mediated synchronized ferroptosis and contributed to AKI. The emergence of PAF and PAF-LPLs in ferroptosis caused the instability of biomembranes and signaled the cell death of neighboring cells. This cascade could be suppressed by PAF-acetylhydrolase (II) (PAFAH2) or by addition of antibodies against PAF. Genetic knockout or pharmacological inhibition of PAFAH2 increased PAF production, augmented synchronized ferroptosis and exacerbated ischemia/reperfusion (I/R)-induced AKI. Notably, intravenous administration of wild-type PAFAH2 protein, but not its enzymatically inactive mutants, prevented synchronized tubular cell death, nephron loss and AKI. Our findings offer an insight into the mechanisms of synchronized ferroptosis and suggest a possibility for the preventive intervention of AKI.


Subject(s)
Acute Kidney Injury , Ferroptosis , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/drug therapy , Ferroptosis/drug effects , Animals , Mice , Mice, Inbred C57BL , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Platelet Activating Factor/metabolism , Mice, Knockout , Humans , Male
4.
Cancer Med ; 12(15): 16405-16415, 2023 08.
Article in English | MEDLINE | ID: mdl-37501397

ABSTRACT

BACKGROUND: Metastatic castration-resistant prostate cancer (mCRPC) remains fatal and incurable, despite a variety of treatments that can delay disease progression and prolong life. Immune checkpoint therapy is a promising treatment. However, emerging evidence suggests that exosomal programmed necrosis ligand 1 (PD-L1) directly binds to PD-1 on the surface of T cells in the drain lineage lymph nodes or neutralizes administered PD-L1 antibodies, resulting in poor response to anti-PD-L1 therapy in mCRPC. MATERIALS AND METHODS: Western blotting and immunofluorescence were performed to compare PD-L1 levels in exosomes derived from different prostate cancer cells. PC3 cells were subcutaneously injected into nude mice, and then ELISA assay was used to detect human specific PD-L1 in exosomes purified from mouse serum. The function of CD8+ T cells was detected by T cell mediated tumor cell killing assay and FACS analysis. A subcutaneous xenograft model was established using mouse prostate cancer cell RM1, exosomes with or without PD-L1 were injected every 3 days, and then tumor size and weight were analyzed to evaluate the effect of exosomal PD-L1. RESULTS: Herein, we found that exosomal-PD-L1 was taken up by tumor cells expressing low levels of PD-L1, thereby protecting them from T-cell killing. Higher levels of PD-L1 were detected in exosomes derived from the highly malignant prostate cancer PC3 and DU145 cell lines. Moreover, exosomal PD-L1 was taken up by the PD-L1-low-expressing LNCaP cell line and inhibited the killing function of CD8-T cells on tumor cells. The growth rate of RM1-derived subcutaneous tumors was decreased after knockdown of PD-L1 in tumor cells, whereas the growth rate recovered following exosomal PD-L1 tail vein injection. Furthermore, in the serum of mice with PCa subcutaneous tumors, PD-L1 was mainly present on exosomes. CONCLUSION: In summary, tumor cells share PD-L1 synergistically against T cells through exosomes. Inhibition of exosome secretion or prevention of PD-L1 sorting into exosomes may improve the therapeutic response of prostate tumors to anti-PD-L1 therapy.


Subject(s)
Exosomes , Prostatic Neoplasms, Castration-Resistant , Male , Humans , Animals , Mice , CD8-Positive T-Lymphocytes , Mice, Nude , Prostatic Neoplasms, Castration-Resistant/metabolism , Cell Line, Tumor , Disease Progression , Exosomes/metabolism , B7-H1 Antigen/metabolism
5.
EMBO J ; 42(16): e113258, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37409632

ABSTRACT

Mitochondrial biogenesis is the process of generating new mitochondria to maintain cellular homeostasis. Here, we report that viruses exploit mitochondrial biogenesis to antagonize innate antiviral immunity. We found that nuclear respiratory factor-1 (NRF1), a vital transcriptional factor involved in nuclear-mitochondrial interactions, is essential for RNA (VSV) or DNA (HSV-1) virus-induced mitochondrial biogenesis. NRF1 deficiency resulted in enhanced innate immunity, a diminished viral load, and morbidity in mice. Mechanistically, the inhibition of NRF1-mediated mitochondrial biogenesis aggravated virus-induced mitochondrial damage, promoted the release of mitochondrial DNA (mtDNA), increased the production of mitochondrial reactive oxygen species (mtROS), and activated the innate immune response. Notably, virus-activated kinase TBK1 phosphorylated NRF1 at Ser318 and thereby triggered the inactivation of the NRF1-TFAM axis during HSV-1 infection. A knock-in (KI) strategy that mimicked TBK1-NRF1 signaling revealed that interrupting the TBK1-NRF1 connection ablated mtDNA release and thereby attenuated the HSV-1-induced innate antiviral response. Our study reveals a previously unidentified antiviral mechanism that utilizes a NRF1-mediated negative feedback loop to modulate mitochondrial biogenesis and antagonize innate immune response.


Subject(s)
Antiviral Agents , Organelle Biogenesis , Animals , Mice , DNA, Mitochondrial/genetics , Immunity, Innate , Nuclear Respiratory Factor 1/genetics
6.
EMBO Rep ; 24(4): e54731, 2023 04 05.
Article in English | MEDLINE | ID: mdl-36847607

ABSTRACT

Ectopic lipid deposition and mitochondrial dysfunction are common etiologies of obesity and metabolic disorders. Excessive dietary uptake of saturated fatty acids (SFAs) causes mitochondrial dysfunction and metabolic disorders, while unsaturated fatty acids (UFAs) counterbalance these detrimental effects. It remains elusive how SFAs and UFAs differentially signal toward mitochondria for mitochondrial performance. We report here that saturated dietary fatty acids such as palmitic acid (PA), but not unsaturated oleic acid (OA), increase lysophosphatidylinositol (LPI) production to impact on the stability of the mitophagy receptor FUNDC1 and on mitochondrial quality. Mechanistically, PA shifts FUNDC1 from dimer to monomer via enhanced production of LPI. Monomeric FUNDC1 shows increased acetylation at K104 due to dissociation of HDAC3 and increased interaction with Tip60. Acetylated FUNDC1 can be further ubiquitinated by MARCH5 for proteasomal degradation. Conversely, OA antagonizes PA-induced accumulation of LPI, and FUNDC1 monomerization and degradation. A fructose-, palmitate-, and cholesterol-enriched (FPC) diet also affects FUNDC1 dimerization and promotes its degradation in a non-alcoholic steatohepatitis (NASH) mouse model. We thus uncover a signaling pathway that orchestrates lipid metabolism with mitochondrial quality.


Subject(s)
Fatty Acids , Mitophagy , Mice , Animals , Fatty Acids/metabolism , Dimerization , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Membrane Proteins/metabolism
7.
Elife ; 112022 08 01.
Article in English | MEDLINE | ID: mdl-35913115

ABSTRACT

DBC1 has been characterized as a key regulator of physiological and pathophysiological activities, such as DNA damage, senescence, and tumorigenesis. However, the mechanism by which the functional stability of DBC1 is regulated has yet to be elucidated. Here, we report that the ubiquitination-mediated degradation of DBC1 is regulated by the E3 ubiquitin ligase SIAH2 and deubiquitinase OTUD5 under hypoxic stress. Mechanistically, hypoxia promoted DBC1 to interact with SIAH2 but not OTUD5, resulting in the ubiquitination and subsequent degradation of DBC1 through the ubiquitin-proteasome pathway. SIAH2 knockout inhibited tumor cell proliferation and migration, which could be rescued by double knockout of SIAH2/CCAR2. Human tissue microarray analysis further revealed that the SIAH2/DBC1 axis was responsible for tumor progression under hypoxic stress. These findings define a key role of the hypoxia-mediated SIAH2-DBC1 pathway in the progression of human breast cancer and provide novel insights into the metastatic mechanism of breast cancer.


Subject(s)
Breast Neoplasms , Adaptor Proteins, Signal Transducing/metabolism , Breast/metabolism , Breast Neoplasms/pathology , Female , Humans , Hypoxia/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
8.
Cell Rep ; 40(7): 111195, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35977480

ABSTRACT

ATG9A is a highly conserved membrane protein required for autophagy initiation. It is trafficked from the trans-Golgi network (TGN) to the phagophore to act as a membrane source for autophagosome expansion. Here, we show that ATG9A is not just a passenger protein in the TGN but rather works in concert with GRASP55, a stacking factor for Golgi structure, to organize Golgi dynamics and integrity. Upon heat stress, the E3 ubiquitin ligase MARCH9 is promoted to ubiquitinate ATG9A in the form of K63 conjugation, and the nondegradable ubiquitinated ATG9A disperses from the Golgi apparatus to the cytoplasm more intensely, accompanied by inhibiting GRASP55 oligomerization, further resulting in Golgi fragmentation. Knockout of ATG9A or MARCH9 largely prevents Golgi fragmentation and protects Golgi functions under heat and other Golgi stresses. Our results reveal a noncanonical function of ATG9A for Golgi dynamics and suggest the pathway for sensing Golgi stress via the MARCH9/ATG9A axis.


Subject(s)
Autophagosomes , Golgi Apparatus , Autophagosomes/metabolism , Autophagy , Autophagy-Related Proteins/metabolism , Golgi Apparatus/metabolism , Protein Transport , Ubiquitin/metabolism , trans-Golgi Network/metabolism
9.
J Colloid Interface Sci ; 616: 759-768, 2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35248965

ABSTRACT

The unsatisfactory efficacy of conventional theranostic agents in ablating tumor poses urgent demands on the development of high-performance integrated theranostic agents utilizing rising nanotechnology. To cope with the existing limitations, here we presented an intelligent nanoplatform based on yolk-shell Fe3O4@polydopamine prepared by mussel-inspired polydopamine chemistry and sacrificial template method as well as subsequent incorporation of Pt nanoparticles and chlorine 6 (Ce6) by in situ reduction and electrostatic adsorption for photodynamic therapy (PDT) and photothermal (PTT). The resultant nanoplatform could effectively deliver photosensitizer Ce6 to tumor sites, then promoting the decomposition of endogenous H2O2 to oxygen, finally achieving enhanced PDT therapy, which is demonstrated by in vitro and in vivo evaluations. Importantly, the generated oxygen bubbles could improve the echogenicity signal of yolk-shell microspheres and thereby provide enhanced ultrasonic (US) signal for imaging solid tumors. Overall, the synergistic combination of magnetic Fe3O4, green polydopamine, catalytic Pt nanoparticles, photosensitive Ce6 enabled the hybrid nanoplatform to have good biocompatibility, efficient tumor accumulation, excellent phototherapy efficiency, high T2-weighted magnetic resonance imaging (MRI) and fluorescence imaging ability (FL). Our study integrating the merits of PDT/PTT and US/MRI/FL into a single nanoplatform will open an avenue of therapeutic strategy toward biomedical applications.


Subject(s)
Nanoparticles , Photochemotherapy , Cell Line, Tumor , Hydrogen Peroxide , Multimodal Imaging , Nanoparticles/chemistry , Oxygen , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Phototherapy
10.
Methods Cell Biol ; 164: 1-9, 2021.
Article in English | MEDLINE | ID: mdl-34225908

ABSTRACT

The transcription factor EB (TFEB) plays a critical role in autophagy induction and lysosomal biogenesis by orchestrating the expression of autophagy- and lysosome-related genes. In response to a series of stresses such as nutrient starvation, TFEB translocates from the cytoplasm to the nucleus, where it exerts its regulatory function. The activity of TFEB is tightly regulated by multiple phosphorylation and acetylation sites. Methods that rely on the analysis of posttranslational modification as a proxy for TFEB activation are often misleading. Here, we elaborate on protocols for monitoring nuclear translocation of TFEB by fluorescence microscopy.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Lysosomes , Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Lysosomes/metabolism , Phosphorylation , Protein Transport
11.
Elife ; 102021 05 04.
Article in English | MEDLINE | ID: mdl-33942716

ABSTRACT

Erythropoietin (EPO) drives erythropoiesis and is secreted mainly by the kidney upon hypoxic or anemic stress. The paucity of EPO production in renal EPO-producing cells (REPs) causes renal anemia, one of the most common complications of chronic nephropathies. Although mitochondrial dysfunction is commonly observed in several renal and hematopoietic disorders, the mechanism by which mitochondrial quality control impacts renal anemia remains elusive. In this study, we showed that FUNDC1, a mitophagy receptor, plays a critical role in EPO-driven erythropoiesis induced by stresses. Mechanistically, EPO production is impaired in REPs in Fundc1-/- mice upon stresses, and the impairment is caused by the accumulation of damaged mitochondria, which consequently leads to the elevation of the reactive oxygen species (ROS) level and triggers inflammatory responses by up-regulating proinflammatory cytokines. These inflammatory factors promote the myofibroblastic transformation of REPs, resulting in the reduction of EPO production. We therefore provide a link between aberrant mitophagy and deficient EPO generation in renal anemia. Our results also suggest that the mitochondrial quality control safeguards REPs under stresses, which may serve as a potential therapeutic strategy for the treatment of renal anemia.


Subject(s)
Anemia/prevention & control , Erythropoietin/metabolism , Gene Expression Regulation , Kidney Diseases/prevention & control , Membrane Proteins/genetics , Mitochondrial Proteins/genetics , Mitophagy/genetics , Animals , Erythropoiesis/genetics , Erythropoiesis/physiology , Erythropoietin/analysis , Erythropoietin/genetics , Kidney Diseases/classification , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Mitophagy/physiology , Reactive Oxygen Species
12.
Nanoscale Adv ; 3(13): 3708-3729, 2021 Jun 30.
Article in English | MEDLINE | ID: mdl-36133016

ABSTRACT

Bacterial infections and transmission threaten human health and well-being. Graphite carbon nitride (g-C3N4), a promising photocatalytic antibacterial nanomaterial, has attracted increasing attention to combat bacterial transmission, due to the outstanding stability, high efficiency and environmental sustainability of this material. However, the antibacterial efficiency of g-C3N4 is affected by several factors, including its specific surface area, rapid electron/hole recombination processes and optical absorption properties. To improve the efficiency of the antibacterial properties of g-C3N4 and extend its range of applications, various nanocomposites have been prepared and evaluated. In this review, the advances in amplifying the photocatalytic antibacterial efficiency of g-C3N4-based nanocomposites is discussed, including different topologies, noble metal decoration, non-noble metal doping and heterojunction construction. The enhancement mechanisms and synergistic effects in g-C3N4-based nanocomposites are highlighted. The remaining challenges and future perspectives of antibacterial g-C3N4-based nanocomposites are also discussed.

13.
Theranostics ; 10(16): 7287-7318, 2020.
Article in English | MEDLINE | ID: mdl-32641993

ABSTRACT

As an emerging antitumor strategy, photodynamic therapy (PDT) has attracted intensive attention for the treatment of various malignant tumors owing to its noninvasive nature and high spatial selectivity in recent years. However, the therapeutic effect is unsatisfactory on some occasions due to the presence of some unfavorable factors including nonspecific accumulation of PS towards malignant tissues, the lack of endogenous oxygen in tumors, as well as the limited light penetration depth, further hampering practical application. To circumvent these limitations and improve real utilization efficiency, various enhanced strategies have been developed and explored during the past years. In this review, we give an overview of the state-of-the-art advances progress on versatile nanoplatforms for enhanced PDT considering the enhancement from targeting or responsive, chemical and physical effect. Specifically, these effects mainly include organelle-targeting function, tumor microenvironment responsive release photosensitizers (PS), self-sufficient O2 (affinity oxygen and generating oxygen), photocatalytic water splitting, X-rays light stimulate, surface plasmon resonance enhancement, and the improvement by resonance energy transfer. When utilizing these strategies to improve the therapeutic effect, the advantages and limitations are addressed. Finally, the challenges and prospective will be discussed and demonstrated for the future development of advanced PDT with enhanced efficacy.


Subject(s)
Drug Carriers/chemistry , Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Theranostic Nanomedicine/methods , Animals , Disease Models, Animal , Humans , Nanoparticles/chemistry , Neoplasms/pathology , Photochemotherapy/trends , Theranostic Nanomedicine/trends , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
14.
Cell Death Differ ; 27(3): 1036-1051, 2020 03.
Article in English | MEDLINE | ID: mdl-31367011

ABSTRACT

Mitochondria are highly dynamic organelles and respond to stress by changing their fission-fusion cycle, undergoing mitophagy, or releasing apoptotic proteins to initiate cell death. The molecular mechanisms that sense different stresses and coordinate distinct effectors still await full characterization. Here, we show that PGAM5, which exists in an equilibrium between dimeric and multimeric states, dephosphorylates BCL-xL to inhibit apoptosis or FUNDC1 to activate mitofission and mitophagy in response to distinct stresses. In vinblastine-treated cells, PGAM5 dephosphorylates BCL-xL at Ser62 to restore BCL-xL sequestration of BAX and BAK and thereby resistance to apoptosis. Selenite-induced oxidative stress increases the multimerization of PGAM5, resulting in its dissociation from BCL-xL, which causes increased BCL-xL phosphorylation and apoptosis. Once freed, the more multimeric and active PGAM5 dephosphorylates FUNDC1 to initiate mitofission and mitophagy. The reciprocal interaction of PGAM5 with FUNDC1 and BCL-xL, controlled by PGAM5 multimerization, serves as a molecular switch between mitofission/mitophagy and apoptosis.


Subject(s)
Cell Lineage , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , bcl-X Protein/metabolism , Apoptosis/drug effects , Cell Lineage/drug effects , HeLa Cells , Humans , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , Mitophagy/drug effects , Models, Biological , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Multimerization/drug effects , Selenious Acid/pharmacology , Serine/metabolism , Vinblastine/pharmacology , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
15.
Nat Commun ; 10(1): 1034, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30833558

ABSTRACT

The interactions between tumor cells with their microenvironments, including hypoxia, acidosis and immune cells, lead to the tumor heterogeneity which promotes tumor progression. Here, we show that SIAH2-NRF1 axis remodels tumor microenvironment through regulating tumor mitochondrial function, tumor-associated macrophages (TAMs) polarization and cell death for tumor maintenance and progression. Mechanistically, low mitochondrial gene expression in breast cancers is associated with a poor clinical outcome. The hypoxia-activated E3 ligase SIAH2 spatially downregulates nuclear-encoded mitochondrial gene expression including pyruvate dehydrogenase beta via degrading NRF1 (Nuclear Respiratory Factor 1) through ubiquitination on lysine 230, resulting in enhanced Warburg effect, metabolic reprogramming and pro-tumor immune response. Dampening NRF1 degradation under hypoxia not only impairs the polarization of TAMs, but also promotes tumor cells to become more susceptible to apoptosis in a FADD-dependent fashion, resulting in secondary necrosis due to the impairment of efferocytosis. These data represent that inhibition of NRF1 degradation is a potential therapeutic strategy against cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Nuclear Proteins/metabolism , Nuclear Respiratory Factor 1/metabolism , Tumor Microenvironment , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Cellular Reprogramming , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Disease Models, Animal , Female , Gene Knockout Techniques , Humans , Hypoxia/metabolism , Macrophages/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/genetics , Nuclear Proteins/genetics , Nuclear Respiratory Factor 1/genetics , RNA, Small Interfering/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
16.
Cell Res ; 27(2): 184-201, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27934868

ABSTRACT

Autophagy requires diverse membrane sources and involves membrane trafficking of mATG9, the only membrane protein in the ATG family. However, the molecular regulation of mATG9 trafficking for autophagy initiation remains unclear. Here we identified two conserved classic adaptor protein sorting signals within the cytosolic N-terminus of mATG9, which mediate trafficking of mATG9 from the plasma membrane and trans-Golgi network (TGN) via interaction with the AP1/2 complex. Src phosphorylates mATG9 at Tyr8 to maintain its endocytic and constitutive trafficking in unstressed conditions. In response to starvation, phosphorylation of mATG9 at Tyr8 by Src and at Ser14 by ULK1 functionally cooperate to promote interactions between mATG9 and the AP1/2 complex, leading to redistribution of mATG9 from the plasma membrane and juxta-nuclear region to the peripheral pool for autophagy initiation. Our findings uncover novel mechanisms of mATG9 trafficking and suggest a coordination of basal and stress-induced autophagy.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Proteins/metabolism , Autophagy , Cell Membrane/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Starvation/metabolism , Starvation/pathology , Vesicular Transport Proteins/metabolism , src-Family Kinases/metabolism , Amino Acid Sequence , Autophagy/drug effects , Autophagy-Related Proteins/chemistry , Conserved Sequence , Epidermal Growth Factor/pharmacology , HEK293 Cells , HeLa Cells , Humans , Membrane Proteins/chemistry , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Transport/drug effects , Stress, Physiological/drug effects , Vesicular Transport Proteins/chemistry
17.
Nat Commun ; 7: 11123, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27030211

ABSTRACT

The evolutionarily conserved Hippo pathway is a regulator that controls organ size, cell growth and tissue homeostasis. Upstream signals of the Hippo pathway have been widely studied, but how microenvironmental factors coordinately regulate this pathway remains unclear. In this study, we identify LIM domain protein Zyxin, as a scaffold protein, that in response to hypoxia and TGF-ß stimuli, forms a ternary complex with Lats2 and Siah2 and stabilizes their interaction. This interaction facilitates Lats2 ubiquitination and degradation, Yap dephosphorylation and subsequently activation. We show that Zyxin is required for TGF-ß and hypoxia-induced Lats2 downregulation and deactivation of Hippo signalling in MDA-MB-231 cells. Depletion of Zyxin impairs the capability of cell migration, proliferation and tumourigenesis in a xenograft model. Zyxin is upregulated in human breast cancer and positively correlates with histological stages and metastasis. Our study demonstrates that Zyxin-Lats2-Siah2 axis may serve as a potential therapeutic target in cancer treatment.


Subject(s)
Nuclear Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Transforming Growth Factor beta/physiology , Tumor Suppressor Proteins/physiology , Ubiquitin-Protein Ligases/physiology , Zyxin/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carcinogenesis/genetics , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cellular Microenvironment , Female , HEK293 Cells , Heterografts/metabolism , Heterografts/pathology , Hippo Signaling Pathway , Humans , Mice , Mice, Inbred BALB C , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteolysis , Signal Transduction , Transcription Factors , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , YAP-Signaling Proteins , Zyxin/genetics , Zyxin/metabolism
18.
Autophagy ; 11(8): 1216-29, 2015.
Article in English | MEDLINE | ID: mdl-26018823

ABSTRACT

Mitochondria serve as membrane sources and signaling platforms for regulating autophagy. Accumulating evidence has also shown that damaged mitochondria are removed through both selective mitophagy and general autophagy in response to mitochondrial and oxidative stresses. Protein ubiquitination through mitochondrial E3 ligases plays an integrative role in mitochondrial outer membrane protein degradation, mitochondrial dynamics, and mitophagy. Here we showed that MUL1, a mitochondria-localized E3 ligase, regulates selenite-induced mitophagy in an ATG5 and ULK1-dependent manner. ULK1 partially translocated to mitochondria after selenite treatment and interacted with MUL1. We also demonstrated that ULK1 is a novel substrate of MUL1. These results suggest the association of mitochondria with autophagy regulation and provide a new mechanism for the beneficial effects of selenium as a chemopreventive agent.


Subject(s)
Autophagy , Intracellular Signaling Peptides and Proteins/chemistry , Mitochondrial Proteins/metabolism , Mitophagy , Protein Serine-Threonine Kinases/chemistry , Selenious Acid/chemistry , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Animals , Autophagy-Related Protein 5 , Autophagy-Related Protein-1 Homolog , Gene Expression Regulation, Enzymologic , Glutathione Transferase/metabolism , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice , Microtubule-Associated Proteins/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/pathology , Molecular Sequence Data , Oxidative Stress , RNA Interference , Reactive Oxygen Species/metabolism , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Substrate Specificity , Ubiquitination
19.
Nat Cell Biol ; 17(1): 95-103, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25438054

ABSTRACT

The Hippo signalling pathway plays important roles in animal development, physiology and tumorigenesis. Understanding how the activity of this pathway is regulated by the cellular microenvironment remains a major challenge. Here we elucidate a molecular mechanism by which hypoxia deactivates Hippo signalling. We demonstrate that the E3 ubiquitin ligase SIAH2 stimulates YAP by destabilizing LATS2, a critical component of the Hippo pathway, in response to hypoxia. Loss of SIAH2 suppresses tumorigenesis in a LATS2-dependent manner in a xenograft mouse model. We further show that YAP complexes with HIF1α and is essential for HIF1α stability and function in tumours in vivo. LATS2 is downregulated in human breast tumours and negatively correlates with SIAH2 expression levels, indicating that the SIAH2-LATS2 pathway may have a role in human cancer. Our data uncover oxygen availability as a microenvironment signal for the Hippo pathway and have implications for understanding the regulation of Hippo signalling in tumorigenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Hypoxia/physiology , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Down-Regulation , Female , HEK293 Cells , HeLa Cells , Hippo Signaling Pathway , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Phosphoproteins/biosynthesis , Phosphorylation , Protein Serine-Threonine Kinases/biosynthesis , RNA Interference , RNA, Small Interfering , Signal Transduction , Transcription Factors , Transplantation, Heterologous , Tumor Microenvironment , Tumor Suppressor Proteins/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics , YAP-Signaling Proteins
20.
Free Radic Biol Med ; 63: 485-94, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23732520

ABSTRACT

We have previously shown that the natural diterpenoid derivative S3 induced Bim upregulation and apoptosis in a Bax/Bak-independent manner. However, the exact molecular target(s) of S3 and the mechanism controlling Bim upregulation are still not clear. Here, we identify that S3 targets the selenoproteins TrxR1 and TrxR2 at the selenocysteine residue of the reactive center of the enzymes and inhibits their antioxidant activities. Consequently, cellular ROS is elevated, leading to the activation of FOXO3a, which contributes to Bim upregulation in Bax/Bak-deficient cells. Moreover, S3 retards tumor growth in subcutaneous xenograft tumors by inhibiting TrxR activity in vivo. Our studies delineate the signaling pathway controlling Bim upregulation, which results in Bax/Bak-independent apoptosis and provide evidence that the compounds can act as anticancer agents based on mammalian TrxRs inhibition.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Diterpenes/pharmacology , Neoplasms/metabolism , Selenocysteine/metabolism , Thioredoxin-Disulfide Reductase/metabolism , Animals , Catalytic Domain/drug effects , Fibroblasts/cytology , HCT116 Cells , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Selenocysteine/chemistry , Selenoproteins/antagonists & inhibitors , Selenoproteins/chemistry , Selenoproteins/metabolism , Thioredoxin Reductase 1/metabolism , Thioredoxin Reductase 2/metabolism , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/chemistry , Thioredoxins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...