Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cell Metab ; 36(6): 1335-1350.e8, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38701775

ABSTRACT

Perivascular collagen deposition by activated fibroblasts promotes vascular stiffening and drives cardiovascular diseases such as pulmonary hypertension (PH). Whether and how vascular fibroblasts rewire their metabolism to sustain collagen biosynthesis remains unknown. Here, we found that inflammation, hypoxia, and mechanical stress converge on activating the transcriptional coactivators YAP and TAZ (WWTR1) in pulmonary arterial adventitial fibroblasts (PAAFs). Consequently, YAP and TAZ drive glutamine and serine catabolism to sustain proline and glycine anabolism and promote collagen biosynthesis. Pharmacologic or dietary intervention on proline and glycine anabolic demand decreases vascular stiffening and improves cardiovascular function in PH rodent models. By identifying the limiting metabolic pathways for vascular collagen biosynthesis, our findings provide guidance for incorporating metabolic and dietary interventions for treating cardiopulmonary vascular disease.


Subject(s)
Glutamine , Serine , Vascular Stiffness , Animals , Glutamine/metabolism , Serine/metabolism , Male , Mice , Mice, Inbred C57BL , Fibroblasts/metabolism , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Humans , Collagen/metabolism , Rats
2.
Sci Transl Med ; 16(729): eadd2029, 2024 01 10.
Article in English | MEDLINE | ID: mdl-38198571

ABSTRACT

Hypoxic reprogramming of vasculature relies on genetic, epigenetic, and metabolic circuitry, but the control points are unknown. In pulmonary arterial hypertension (PAH), a disease driven by hypoxia inducible factor (HIF)-dependent vascular dysfunction, HIF-2α promoted expression of neighboring genes, long noncoding RNA (lncRNA) histone lysine N-methyltransferase 2E-antisense 1 (KMT2E-AS1) and histone lysine N-methyltransferase 2E (KMT2E). KMT2E-AS1 stabilized KMT2E protein to increase epigenetic histone 3 lysine 4 trimethylation (H3K4me3), driving HIF-2α-dependent metabolic and pathogenic endothelial activity. This lncRNA axis also increased HIF-2α expression across epigenetic, transcriptional, and posttranscriptional contexts, thus promoting a positive feedback loop to further augment HIF-2α activity. We identified a genetic association between rs73184087, a single-nucleotide variant (SNV) within a KMT2E intron, and disease risk in PAH discovery and replication patient cohorts and in a global meta-analysis. This SNV displayed allele (G)-specific association with HIF-2α, engaged in long-range chromatin interactions, and induced the lncRNA-KMT2E tandem in hypoxic (G/G) cells. In vivo, KMT2E-AS1 deficiency protected against PAH in mice, as did pharmacologic inhibition of histone methylation in rats. Conversely, forced lncRNA expression promoted more severe PH. Thus, the KMT2E-AS1/KMT2E pair orchestrates across convergent multi-ome landscapes to mediate HIF-2α pathobiology and represents a key clinical target in pulmonary hypertension.


Subject(s)
Hypertension, Pulmonary , RNA, Long Noncoding , Humans , Rats , Animals , Mice , Alleles , Hypertension, Pulmonary/genetics , Histones , RNA, Long Noncoding/genetics , Rodentia , Lysine , Familial Primary Pulmonary Hypertension , Hypoxia/genetics , Methyltransferases , Basic Helix-Loop-Helix Transcription Factors/genetics
3.
bioRxiv ; 2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37546967

ABSTRACT

Biomolecular condensates regulate a wide range of cellular functions from signaling to RNA metabolism 1, 2 , yet, the physiologic conditions regulating their formation remain largely unexplored. Biomolecular condensate assembly is tightly regulated by the intracellular environment. Changes in the chemical or physical conditions inside cells can stimulate or inhibit condensate formation 3-5 . However, whether and how the external environment of cells can also regulate biomolecular condensation remain poorly understood. Increasing our understanding of these mechanisms is paramount as failure to control condensate formation and dynamics can lead to many diseases 6, 7 . Here, we provide evidence that matrix stiffening promotes biomolecular condensation in vivo . We demonstrate that the extracellular matrix links mechanical cues with the control of glucose metabolism to sorbitol. In turn, sorbitol acts as a natural crowding agent to promote biomolecular condensation. Using in silico simulations and in vitro assays, we establish that variations in the physiological range of sorbitol, but not glucose, concentrations, are sufficient to regulate biomolecular condensates. Accordingly, pharmacologic and genetic manipulation of intracellular sorbitol concentration modulates biomolecular condensates in breast cancer - a mechano-dependent disease. We propose that sorbitol is a mechanosensitive metabolite enabling protein condensation to control mechano-regulated cellular functions. Altogether, we uncover molecular driving forces underlying protein phase transition and provide critical insights to understand the biological function and dysfunction of protein phase separation.

4.
Trends Cell Biol ; 32(7): 624-636, 2022 07.
Article in English | MEDLINE | ID: mdl-35177291

ABSTRACT

Altered metabolic programs and corruption of tissue architecture are hallmarks of disease. The spatiotemporal control of cell behavior requires transmission of information from the complex structure of tissues to their constituent cells. Cytoskeletal mechanotransduction enables this transmission by sensing mechanical environments and adapting cellular behaviors. However, this process requires energy. Recent findings have shed light on the bidirectional relationship between mechanical forces and upstream and downstream metabolic cues. We discuss recent advances in the reciprocal regulation ('metabo-reciprocity') that allows cells to adapt their metabolic needs to their mechanically constrained environment but can also contribute to adjustable feedback that promotes disease progression.


Subject(s)
Cytoskeleton , Mechanotransduction, Cellular , Humans , Mechanotransduction, Cellular/physiology
5.
Sci Adv ; 7(43): eabh3794, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34669463

ABSTRACT

Cancer therapies are being considered for treating rare noncancerous diseases like pulmonary hypertension (PH), but effective computational screening is lacking. Via transcriptomic differential dependency analyses leveraging parallels between cancer and PH, we mapped a landscape of cancer drug functions dependent upon rewiring of PH gene clusters. Bromodomain and extra-terminal motif (BET) protein inhibitors were predicted to rely upon several gene clusters inclusive of galectin-8 (LGALS8). Correspondingly, LGALS8 was found to mediate the BET inhibitor­dependent control of endothelial apoptosis, an essential role for PH in vivo. Separately, a piperlongumine analog's actions were predicted to depend upon the iron-sulfur biogenesis gene ISCU. Correspondingly, the analog was found to inhibit ISCU glutathionylation, rescuing oxidative metabolism, decreasing endothelial apoptosis, and improving PH. Thus, we identified crucial drug-gene axes central to endothelial dysfunction and therapeutic priorities for PH. These results establish a wide-ranging, network dependency platform to redefine cancer drugs for use in noncancerous conditions.

6.
Cell Metab ; 33(7): 1342-1357.e10, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34102109

ABSTRACT

Mechanical signals from the tumor microenvironment modulate cell mechanics and influence cell metabolism to promote cancer aggressiveness. Cells withstand external forces by adjusting the stiffness of their cytoskeleton. Microtubules (MTs) act as compression-bearing elements. Yet how cancer cells regulate MT dynamic in response to the locally constrained environment has remained unclear. Using breast cancer as a model of a disease in which mechanical signaling promotes disease progression, we show that matrix stiffening rewires glutamine metabolism to promote MT glutamylation and force MT stabilization, thereby promoting cell invasion. Pharmacologic inhibition of glutamine metabolism decreased MT glutamylation and affected their mechanical stabilization. Similarly, decreased MT glutamylation by overexpressing tubulin mutants lacking glutamylation site(s) decreased MT stability, thereby hampering cancer aggressiveness in vitro and in vivo. Together, our results decipher part of the enigmatic tubulin code that coordinates the fine-tunable properties of MT and link cell metabolism to MT dynamics and cancer aggressiveness.


Subject(s)
Glutamic Acid/metabolism , Mechanotransduction, Cellular/physiology , Microtubules/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Animals , Cells, Cultured , Energy Metabolism/physiology , Female , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Protein Processing, Post-Translational , Tubulin/metabolism , Tumor Microenvironment/physiology
7.
Elife ; 102021 04 22.
Article in English | MEDLINE | ID: mdl-33884955

ABSTRACT

To adapt in an ever-changing environment, cells must integrate physical and chemical signals and translate them into biological meaningful information through complex signaling pathways. By combining lipidomic and proteomic approaches with functional analysis, we have shown that ubiquitin domain-containing protein 1 (UBTD1) plays a crucial role in both the epidermal growth factor receptor (EGFR) self-phosphorylation and its lysosomal degradation. On the one hand, by modulating the cellular level of ceramides through N-acylsphingosine amidohydrolase 1 (ASAH1) ubiquitination, UBTD1 controls the ligand-independent phosphorylation of EGFR. On the other hand, UBTD1, via the ubiquitination of Sequestosome 1 (SQSTM1/p62) by RNF26 and endolysosome positioning, participates in the lysosomal degradation of EGFR. The coordination of these two ubiquitin-dependent processes contributes to the control of the duration of the EGFR signal. Moreover, we showed that UBTD1 depletion exacerbates EGFR signaling and induces cell proliferation emphasizing a hitherto unknown function of UBTD1 in EGFR-driven human cell proliferation.


Subject(s)
Ceramides/metabolism , Lysosomes/enzymology , Prostatic Neoplasms/enzymology , Ubiquitins/metabolism , Acid Ceramidase/genetics , Acid Ceramidase/metabolism , Cell Line, Tumor , Cell Proliferation , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Kinetics , Lysosomes/genetics , Male , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proteolysis , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Signal Transduction , Ubiquitination , Ubiquitins/genetics
8.
Commun Biol ; 4(1): 166, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33547392

ABSTRACT

Polo-like kinase 1 (Plk1) expression is inversely correlated with survival advantages in many cancers. However, molecular mechanisms that underlie Plk1 expression are poorly understood. Here, we uncover a hypoxia-regulated mechanism of Plk1-mediated cancer metastasis and drug resistance. We demonstrated that a HIF-2-dependent regulatory pathway drives Plk1 expression in clear cell renal cell carcinoma (ccRCC). Mechanistically, HIF-2 transcriptionally targets the hypoxia response element of the Plk1 promoter. In ccRCC patients, high expression of Plk1 was correlated to poor disease-free survival and overall survival. Loss-of-function of Plk1 in vivo markedly attenuated ccRCC growth and metastasis. High Plk1 expression conferred a resistant phenotype of ccRCC to targeted therapeutics such as sunitinib, in vitro, in vivo, and in metastatic ccRCC patients. Importantly, high Plk1 expression was defined in a subpopulation of ccRCC patients that are refractory to current therapies. Hence, we propose a therapeutic paradigm for improving outcomes of ccRCC patients.


Subject(s)
Carcinoma, Renal Cell , Cell Cycle Proteins/physiology , Drug Resistance, Neoplasm/genetics , Kidney Neoplasms , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cohort Studies , Embryo, Nonmammalian , Female , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mice , Mice, Nude , Neoplasm Metastasis , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Up-Regulation/genetics , Zebrafish , Polo-Like Kinase 1
9.
Circ Res ; 127(5): 677-692, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32493166

ABSTRACT

RATIONALE: Unproven theories abound regarding the long-range uptake and endocrine activity of extracellular blood-borne microRNAs into tissue. In pulmonary hypertension (PH), microRNA-210 (miR-210) in pulmonary endothelial cells promotes disease, but its activity as an extracellular molecule is incompletely defined. OBJECTIVE: We investigated whether chronic and endogenous endocrine delivery of extracellular miR-210 to pulmonary vascular endothelial cells promotes PH. METHODS AND RESULTS: Using miR-210 replete (wild-type [WT]) and knockout mice, we tracked blood-borne miR-210 using bone marrow transplantation and parabiosis (conjoining of circulatory systems). With bone marrow transplantation, circulating miR-210 was derived predominantly from bone marrow. Via parabiosis during chronic hypoxia to induce miR-210 production and PH, miR-210 was undetectable in knockout-knockout mice pairs. However, in plasma and lung endothelium, but not smooth muscle or adventitia, miR-210 was observed in knockout mice of WT-knockout pairs. This was accompanied by downregulation of miR-210 targets ISCU (iron-sulfur assembly proteins)1/2 and COX10 (cytochrome c oxidase assembly protein-10), indicating endothelial import of functional miR-210. Via hemodynamic and histological indices, knockout-knockout pairs were protected from PH, whereas knockout mice in WT-knockout pairs developed PH. In particular, pulmonary vascular engraftment of miR-210-positive interstitial lung macrophages was observed in knockout mice of WT-knockout pairs. To address whether engrafted miR-210-positive myeloid or lymphoid cells contribute to paracrine miR-210 delivery, we studied miR-210 knockout mice parabiosed with miR-210 WT; Cx3cr1 knockout mice (deficient in myeloid recruitment) or miR-210 WT; Rag1 knockout mice (deficient in lymphocytes). In both pairs, miR-210 knockout mice still displayed miR-210 delivery and PH, thus demonstrating a pathogenic endocrine delivery of extracellular miR-210. CONCLUSIONS: Endogenous blood-borne transport of miR-210 into pulmonary vascular endothelial cells promotes PH, offering fundamental insight into the systemic physiology of microRNA activity. These results also describe a platform for RNA-mediated crosstalk in PH, providing an impetus for developing blood-based miR-210 technologies for diagnosis and therapy in this disease.


Subject(s)
Endothelium, Vascular/metabolism , Hypertension, Pulmonary/metabolism , Lung/blood supply , MicroRNAs/metabolism , Animals , Bone Marrow Transplantation , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/physiopathology , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Hypoxia/complications , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/blood , MicroRNAs/genetics , Parabiosis , Signal Transduction
10.
Sci Rep ; 10(1): 7998, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32409676

ABSTRACT

EHD2 is a mechanotransducing ATPase localized in caveolae invaginations at the plasma membrane. EHD2 has recently been associated with several human cancers, however the significance of EHD2 transcript levels in cancer prognosis remains debated. Breast cancer is the most commonly occurring cancer in women and prognosis is variable depending on the subtypes. Triple negative breast cancer (TNBC) often has a poor therapeutic response. The aim of this study was to assess the prognostic significance of EHD2 transcripts and protein expression levels in breast carcinomas. We found that low EHD2 levels were associated with enhanced proliferation, migration and invasion of TNBC cells. EHD2 expression was significantly reduced in TNBC tissues and the loss of EHD2 led to higher expression of the pro-tumoral cytokine IL-8. In apparent contradiction with in vitro data, multivariate analysis of two independent cohorts of breast cancer patients revealed that low EHD2 was in fact associated with good prognosis in the highly proliferative TNBC subtype. Accordingly, TNBC low EHD2 expressers were found to benefit the most from chemotherapy when compared to all subtypes of breast cancers. Our study validates EHD2 expression level as an independent prognostic factor of metastasis-free survival and as a new predictive marker of chemotherapy efficacy in TNBC patients.


Subject(s)
Biomarkers, Tumor , Carrier Proteins/metabolism , Triple Negative Breast Neoplasms/metabolism , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carrier Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Treatment Outcome , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology
12.
Cancer Res ; 79(13): 3268-3280, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31064849

ABSTRACT

Although tumorigenesis is dependent on the reprogramming of cellular metabolism, the metabolic pathways engaged in the formation of metastases remain largely unknown. The transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) plays a pleiotropic role in the control of cancer cell metabolism and has been associated with a good prognosis in prostate cancer. Here, we show that PGC1α represses the metastatic properties of prostate cancer cells via modulation of the polyamine biosynthesis pathway. Mechanistically, PGC1α inhibits the expression of c-MYC and ornithine decarboxylase 1 (ODC1), the rate-limiting enzyme for polyamine synthesis. Analysis of in vivo metastases and clinical data from patients with prostate cancer support the proposition that the PGC1α/c-MYC/ODC1 axis regulates polyamine biosynthesis and prostate cancer aggressiveness. In conclusion, downregulation of PGC1α renders prostate cancer cells dependent on polyamine to promote metastasis. SIGNIFICANCE: These findings show that a major regulator of mitochondrial metabolism controls polyamine synthesis and prostate cancer aggressiveness, with potential applications in therapy and identification of new biomarkers.


Subject(s)
Biomarkers, Tumor/metabolism , Dicarboxylic Acid Transporters/metabolism , Gene Expression Regulation, Neoplastic , Mitochondrial Membrane Transport Proteins/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Polyamines/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-myc/metabolism , Aged , Aged, 80 and over , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Dicarboxylic Acid Transporters/genetics , Follow-Up Studies , Humans , Male , Mice , Mice, Nude , Middle Aged , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Membrane Transport Proteins/genetics , Neoplasm Metastasis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Prognosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
EMBO Rep ; 20(4)2019 04.
Article in English | MEDLINE | ID: mdl-30804013

ABSTRACT

Ubiquitin domain-containing protein 1 (UBTD1) is highly evolutionary conserved and has been described to interact with E2 enzymes of the ubiquitin-proteasome system. However, its biological role and the functional significance of this interaction remain largely unknown. Here, we demonstrate that depletion of UBTD1 drastically affects the mechanical properties of epithelial cancer cells via RhoA activation and strongly promotes their aggressiveness. On a stiff matrix, UBTD1 expression is regulated by cell-cell contacts, and the protein is associated with ß-catenin at cell junctions. Yes-associated protein (YAP) is a major cell mechano-transducer, and we show that UBTD1 is associated with components of the YAP degradation complex. Interestingly, UBTD1 promotes the interaction of YAP with its E3 ubiquitin ligase ß-TrCP Consequently, in cancer cells, UBTD1 depletion decreases YAP ubiquitylation and triggers robust ROCK2-dependent YAP activation and downstream signaling. Data from lung and prostate cancer patients further corroborate the in cellulo results, confirming that low levels of UBTD1 are associated with poor patient survival, suggesting that biological functions of UBTD1 could be beneficial in limiting cancer progression.


Subject(s)
Disease Susceptibility , Insulin-Like Growth Factor I/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Ubiquitins/metabolism , Cell Adhesion , Cell Cycle Proteins/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Humans , Mechanotransduction, Cellular , Models, Biological , Neoplasms/mortality , Neoplasms/pathology , Prognosis , Protein Binding , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factors/metabolism , beta Catenin/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
14.
Angiology ; 70(3): 249-256, 2019 03.
Article in English | MEDLINE | ID: mdl-30009628

ABSTRACT

Angioplasty with or without stenting has become a well-established procedure to treat transplant renal artery stenosis (TRAS). We evaluated our experience on postoperative outcomes following the intervention and identified potential predictive factors of TRAS recurrence. Consecutive patients who underwent endovascular treatment of TRAS were retrospectively reviewed. The study end points were the technical success, 30-day postoperative complications, and the estimated glomerular filtration rate (eGFR). Thirty-two patients underwent endovascular treatment for TRAS. The technical success rate was 96.6%. Complications were observed for 7 (21.9%) patients: 4 had a dissection, 2 a pseudoaneurysm, and 1 (3.1%) patient developed an acute pulmonary edema. The mean eGFR significantly increased at 7 days, 3 months, and 6 months postintervention (43.1, 44.9, and 44.3 vs 33.9 mL/min/1.73 m2 preoperatively, P < .05). The TRAS recurrence was observed in 7 (21.9%) patients. These patients had significantly higher preoperative peak systolic velocity and systolic rise time (5 vs 4 m/s, P = .0383 and 103 vs 80 milliseconds, P = .0148, respectively). Endovascular treatment of TRAS is associated with high technical success and significant improvement in renal function. Further studies are required to confirm predictive factors of TRAS recurrence following endovascular treatment.


Subject(s)
Antihypertensive Agents/therapeutic use , Kidney Transplantation , Postoperative Complications/surgery , Renal Artery Obstruction/surgery , Adult , Angioplasty, Balloon/adverse effects , Female , Glomerular Filtration Rate/physiology , Humans , Kidney Transplantation/methods , Male , Middle Aged , Postoperative Complications/etiology , Recurrence , Risk Factors , Treatment Outcome
15.
J Cell Biol ; 217(12): 4092-4105, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30348749

ABSTRACT

Caveolae are small invaginated pits that function as dynamic mechanosensors to buffer tension variations at the plasma membrane. Here we show that under mechanical stress, the EHD2 ATPase is rapidly released from caveolae, SUMOylated, and translocated to the nucleus, where it regulates the transcription of several genes including those coding for caveolae constituents. We also found that EHD2 is required to maintain the caveolae reservoir at the plasma membrane during the variations of membrane tension induced by mechanical stress. Metal-replica electron microscopy of breast cancer cells lacking EHD2 revealed a complete absence of caveolae and a lack of gene regulation under mechanical stress. Expressing EHD2 was sufficient to restore both functions in these cells. Our findings therefore define EHD2 as a central player in mechanotransduction connecting the disassembly of the caveolae reservoir with the regulation of gene transcription under mechanical stress.


Subject(s)
Carrier Proteins/metabolism , Caveolae/metabolism , Mechanotransduction, Cellular , Stress, Mechanical , Transcription, Genetic , Carrier Proteins/genetics , HeLa Cells , Humans
16.
Biomed J ; 38(5): 367-79, 2015.
Article in English | MEDLINE | ID: mdl-26345539

ABSTRACT

Caveolae are small invaginations of the plasma membrane in cells. In addition to their classically described functions in cell signaling and membrane trafficking, it was recently shown that caveolae act also as plasma membrane sensors that respond immediately to acute mechanical stresses. Caveolin 1 (Cav1), the main component of caveolae, is a multifunctional scaffolding protein that can remodel the extracellular environment. Caveolae dysfunction, due to mutations in  caveolins, has been linked to several human diseases called "caveolinopathies," including muscular dystrophies, cardiac disease, infection, osteoporosis, and cancer. The role of caveolae and/or Cav1 remains controversial particularly in tumor progression. Cav1 function has been associated with several steps of cancerogenesis such as tumor growth, cell migration, metastasis, and angiogenesis, yet it was observed that Cav1 could affect these steps in a positive or negative manner. Here, we discuss the possible function of caveolae and Cav1 in tumor progression in the context of their recently discovered role in cell mechanics.


Subject(s)
Caveolae/metabolism , Caveolin 1/metabolism , Cell Membrane/metabolism , Neoplasms/metabolism , Neoplasms/therapy , Animals , Disease Progression , Humans , Signal Transduction/physiology
17.
Nat Commun ; 4: 2180, 2013.
Article in English | MEDLINE | ID: mdl-23864022

ABSTRACT

The Hace1-HECT E3 ligase is a tumor suppressor that ubiquitylates the activated GTP-bound form of the Rho family GTPase Rac1, leading to Rac1 proteasomal degradation. Here we show that, in vertebrates, Hace1 targets Rac1 for degradation when Rac1 is localized to the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase holoenzyme. This event blocks de novo reactive oxygen species generation by Rac1-dependent NADPH oxidases, and thereby confers cellular protection from reactive oxygen species-induced DNA damage and cyclin D1-driven hyper-proliferation. Genetic inactivation of Hace1 in mice or zebrafish, as well as Hace1 loss in human tumor cell lines or primary murine or human tumors, leads to chronic NADPH oxidase-dependent reactive oxygen species elevation, DNA damage responses and enhanced cyclin D1 expression. Our data reveal a conserved ubiquitin-dependent molecular mechanism that controls the activity of Rac1-dependent NADPH oxidase complexes, and thus constitutes the first known example of a tumor suppressor protein that directly regulates reactive oxygen species production in vertebrates.


Subject(s)
NADPH Oxidases/genetics , Neuropeptides/genetics , Protein Isoforms/genetics , Reactive Oxygen Species/metabolism , Ubiquitin-Protein Ligases/genetics , rac1 GTP-Binding Protein/genetics , Animals , Cell Line, Tumor , Cyclin D1/genetics , Cyclin D1/metabolism , DNA Damage , Fibroblasts/cytology , Fibroblasts/metabolism , HEK293 Cells , Humans , Mice , Mice, Knockout , NADPH Oxidases/metabolism , Neuropeptides/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/deficiency , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/deficiency , Zebrafish , rac1 GTP-Binding Protein/metabolism
18.
Dev Cell ; 21(5): 959-65, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22036506

ABSTRACT

Rac1 small GTPase controls essential aspects of cell biology and is a direct target of numerous bacterial virulence factors. The CNF1 toxin of pathogenic Escherichia coli addresses Rac1 to ubiquitin-proteasome system (UPS). We report the essential role of the tumor suppressor HACE1, a HECT-domain containing E3 ubiquitin-ligase, in the targeting of Rac1 to UPS. HACE1 binds preferentially GTP-bound Rac1 and catalyzes its polyubiquitylation. HACE1 expression increases the ubiquitylation of Rac1, when the GTPase is activated by point mutations or by the GEF-domain of Dbl. RNAi-mediated depletion of HACE1 blocks the ubiquitylation of active Rac1 and increases GTP-bound Rac1 cellular levels. HACE1 antagonizes cell isotropic spreading, a hallmark of Rac1 activation, and is required for endothelial cell monolayer invasion by bacteria. Together, these data establish the role of the HACE1 E3 ubiquitin-ligase in controlling Rac1 ubiquitylation and activity.


Subject(s)
Biocatalysis , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , rac1 GTP-Binding Protein/metabolism , Animals , CHO Cells , Cells, Cultured , Cricetinae , HEK293 Cells , Humans , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/biosynthesis
19.
Cell Host Microbe ; 10(5): 464-74, 2011 Nov 17.
Article in English | MEDLINE | ID: mdl-22100162

ABSTRACT

RhoA-inhibitory bacterial toxins, such as Staphylococcus aureus EDIN toxin, induce large transendothelial cell macroaperture (TEM) tunnels that rupture the host endothelium barrier and promote bacterial dissemination. Host cells repair these tunnels by extending actin-rich membrane waves from the TEM edges. We reveal that cyclic-AMP signaling produced by Bacillus anthracis edema toxin (ET) also induces TEM formation, which correlates with increased vascular permeability. We show that ET-induced TEM formation resembles liquid dewetting, a physical process of nucleation and growth of holes within a thin liquid film. We also identify the cellular mechanisms of tunnel closure and reveal that the I-BAR domain protein Missing in Metastasis (MIM) senses de novo membrane curvature generated by the TEM, accumulates at the TEM edge, and triggers Arp2/3-dependent actin polymerization, which induces actin-rich membrane waves that close the TEM. Thus, the balance between ET-induced TEM formation and resealing likely determines the integrity of the host endothelium barrier.


Subject(s)
Actin-Related Protein 2-3 Complex/chemistry , Anthrax/metabolism , Antigens, Bacterial/metabolism , Bacillus anthracis/metabolism , Bacterial Toxins/metabolism , Cyclic AMP/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Microfilament Proteins/metabolism , Neoplasm Proteins/metabolism , Signal Transduction , Actin-Related Protein 2-3 Complex/genetics , Actin-Related Protein 2-3 Complex/metabolism , Animals , Anthrax/microbiology , Bacillus anthracis/genetics , Human Umbilical Vein Endothelial Cells/microbiology , Humans , Mice , Mice, Inbred BALB C , Microfilament Proteins/genetics , Neoplasm Proteins/genetics , Polymerization
20.
Traffic ; 12(5): 579-90, 2011 May.
Article in English | MEDLINE | ID: mdl-21291504

ABSTRACT

Rho GTPases, which are master regulators of both the actin cytoskeleton and membrane trafficking, are often hijacked by pathogens to enable their invasion of host cells. Here we report that the cytotoxic necrotizing factor-1 (CNF1) toxin of uropathogenic Escherichia coli (UPEC) promotes Rac1-dependent entry of bacteria into host cells. Our screen for proteins involved in Rac1-dependent UPEC entry identifies the Toll-interacting protein (Tollip) as a new interacting protein of Rac1 and its ubiquitinated forms. We show that knockdown of Tollip reduces CNF1-induced Rac1-dependent UPEC entry. Tollip depletion also reduces the Rac1-dependent entry of Listeria monocytogenes expressing InlB invasion protein. Moreover, knockdown of Tollip, Tom1 and clathrin, decreases CNF1 and Rac1-dependent internalization of UPEC. Finally, we show that Tollip, Tom1 and clathrin associate with Rac1 and localize at the site of bacterial entry. Collectively, these findings reveal a new link between Rac1 and Tollip, Tom1 and clathrin membrane trafficking components hijacked by pathogenic bacteria to allow their efficient invasion of host cells.


Subject(s)
Bacterial Infections/metabolism , Bacterial Toxins/metabolism , Escherichia coli Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Uropathogenic Escherichia coli/metabolism , Uropathogenic Escherichia coli/pathogenicity , rac1 GTP-Binding Protein/metabolism , Animals , Cell Line , Endocytosis/physiology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Proteins/metabolism , Uropathogenic Escherichia coli/cytology , rac1 GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...