Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nat Immunol ; 25(2): 268-281, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38195702

ABSTRACT

Melanoma cells, deriving from neuroectodermal melanocytes, may exploit the nervous system's immune privilege for growth. Here we show that nerve growth factor (NGF) has both melanoma cell intrinsic and extrinsic immunosuppressive functions. Autocrine NGF engages tropomyosin receptor kinase A (TrkA) on melanoma cells to desensitize interferon γ signaling, leading to T and natural killer cell exclusion. In effector T cells that upregulate surface TrkA expression upon T cell receptor activation, paracrine NGF dampens T cell receptor signaling and effector function. Inhibiting NGF, either through genetic modification or with the tropomyosin receptor kinase inhibitor larotrectinib, renders melanomas susceptible to immune checkpoint blockade therapy and fosters long-term immunity by activating memory T cells with low affinity. These results identify the NGF-TrkA axis as an important suppressor of anti-tumor immunity and suggest larotrectinib might be repurposed for immune sensitization. Moreover, by enlisting low-affinity T cells, anti-NGF reduces acquired resistance to immune checkpoint blockade and prevents melanoma recurrence.


Subject(s)
Melanoma , Receptor, Nerve Growth Factor , Humans , Receptor, Nerve Growth Factor/genetics , Receptor, Nerve Growth Factor/metabolism , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Tropomyosin , Melanoma/therapy , Receptor, trkA/genetics , Receptor, trkA/metabolism , Cytoprotection , Immune Checkpoint Inhibitors , Memory T Cells , Immunosuppression Therapy , Immunotherapy , Receptors, Antigen, T-Cell
2.
Cell Res ; 33(7): 516-532, 2023 07.
Article in English | MEDLINE | ID: mdl-37169907

ABSTRACT

Cellular senescence is a stress-induced, stable cell cycle arrest phenotype which generates a pro-inflammatory microenvironment, leading to chronic inflammation and age-associated diseases. Determining the fundamental molecular pathways driving senescence instead of apoptosis could enable the identification of senolytic agents to restore tissue homeostasis. Here, we identify thrombomodulin (THBD) signaling as a key molecular determinant of the senescent cell fate. Although normally restricted to endothelial cells, THBD is rapidly upregulated and maintained throughout all phases of the senescence program in aged mammalian tissues and in senescent cell models. Mechanistically, THBD activates a proteolytic feed-forward signaling pathway by stabilizing a multi-protein complex in early endosomes, thus forming a molecular basis for the irreversibility of the senescence program and ensuring senescent cell viability. Therapeutically, THBD signaling depletion or inhibition using vorapaxar, an FDA-approved drug, effectively ablates senescent cells and restores tissue homeostasis in liver fibrosis models. Collectively, these results uncover proteolytic THBD signaling as a conserved pro-survival pathway essential for senescent cell viability, thus providing a pharmacologically exploitable senolytic target for senescence-associated diseases.


Subject(s)
Endothelial Cells , Thrombomodulin , Animals , Cellular Senescence , Liver Cirrhosis/drug therapy , Signal Transduction , Apoptosis , Mammals
3.
J Biol Chem ; 298(9): 102297, 2022 09.
Article in English | MEDLINE | ID: mdl-35872017

ABSTRACT

Insulin signaling in blood vessels primarily functions to stimulate angiogenesis and maintain vascular homeostasis through the canonical PI3K and MAPK signaling pathways. However, angiogenesis is a complex process coordinated by multiple other signaling events. Here, we report a distinct crosstalk between the insulin receptor and endoglin/activin receptor-like kinase 1 (ALK1), an endothelial cell-specific TGF-ß receptor complex essential for angiogenesis. While the endoglin-ALK1 complex normally binds to TGF-ß or bone morphogenetic protein 9 (BMP9) to promote gene regulation via transcription factors Smad1/5, we show that insulin drives insulin receptor oligomerization with endoglin-ALK1 at the cell surface to trigger rapid Smad1/5 activation. Through quantitative proteomic analysis, we identify ependymin-related protein 1 (EPDR1) as a major Smad1/5 gene target induced by insulin but not by TGF-ß or BMP9. We found endothelial EPDR1 expression is minimal at the basal state but is markedly enhanced upon prolonged insulin treatment to promote cell migration and formation of capillary tubules. Conversely, we demonstrate EPDR1 depletion strongly abrogates these angiogenic effects, indicating that EPDR1 is a crucial mediator of insulin-induced angiogenesis. Taken together, these results suggest important therapeutic implications for EPDR1 and the TGF-ß pathways in pathologic angiogenesis during hyperinsulinemia and insulin resistance.


Subject(s)
Endoglin , Growth Differentiation Factor 2 , Insulin , Neovascularization, Pathologic , Nerve Tissue Proteins , Receptors, Transforming Growth Factor beta , Animals , Humans , Mice , Activin Receptors, Type II/metabolism , Chlorocebus aethiops , COS Cells , Endoglin/genetics , Endoglin/metabolism , Growth Differentiation Factor 2/genetics , Insulin/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phosphatidylinositol 3-Kinases , Proteomics , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Transforming Growth Factor beta/metabolism
4.
Nat Commun ; 13(1): 1326, 2022 03 14.
Article in English | MEDLINE | ID: mdl-35288568

ABSTRACT

Defective angiogenesis underlies over 50 malignant, ischemic and inflammatory disorders yet long-term therapeutic applications inevitably fail, thus highlighting the need for greater understanding of the vast crosstalk and compensatory mechanisms. Based on proteomic profiling of angiogenic endothelial components, here we report ßIV-spectrin, a non-erythrocytic cytoskeletal protein, as a critical regulator of sprouting angiogenesis. Early loss of endothelial-specific ßIV-spectrin promotes embryonic lethality in mice due to hypervascularization and hemorrhagic defects whereas neonatal depletion yields higher vascular density and tip cell populations in developing retina. During sprouting, ßIV-spectrin expresses in stalk cells to inhibit their tip cell potential by enhancing VEGFR2 turnover in a manner independent of most cell-fate determining mechanisms. Rather, ßIV-spectrin recruits CaMKII to the plasma membrane to directly phosphorylate VEGFR2 at Ser984, a previously undefined phosphoregulatory site that strongly induces VEGFR2 internalization and degradation. These findings support a distinct spectrin-based mechanism of tip-stalk cell specification during vascular development.


Subject(s)
Spectrin , Vascular Endothelial Growth Factor A , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Mice , Neovascularization, Physiologic , Proteomics , Signal Transduction , Spectrin/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
5.
EMBO Rep ; 19(6)2018 06.
Article in English | MEDLINE | ID: mdl-29777051

ABSTRACT

Cellular senescence is a unique cell fate characterized by stable proliferative arrest and the extensive production and secretion of various inflammatory proteins, a phenomenon known as the senescence-associated secretory phenotype (SASP). The molecular mechanisms responsible for generating a SASP in response to senescent stimuli remain largely obscure. Here, using unbiased gene expression profiling, we discover that the scavenger receptor CD36 is rapidly upregulated in multiple cell types in response to replicative, oncogenic, and chemical senescent stimuli. Moreover, ectopic CD36 expression in dividing mammalian cells is sufficient to initiate the production of a large subset of the known SASP components via activation of canonical Src-p38-NF-κB signaling, resulting in the onset of a full senescent state. The secretome is further shown to be ligand-dependent, as amyloid-beta (Aß) is sufficient to drive CD36-dependent NF-κB and SASP activation. Finally, loss-of-function experiments revealed a strict requirement for CD36 in secretory molecule production during conventional senescence reprogramming. Taken together, these results uncover the Aß-CD36-NF-κB signaling axis as an important regulator of the senescent cell fate via induction of the SASP.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , CD36 Antigens/physiology , Cellular Senescence/physiology , NF-kappa B/metabolism , CD36 Antigens/genetics , Cells, Cultured , Cellular Senescence/genetics , Fibroblasts/metabolism , Humans , Loss of Function Mutation , Signal Transduction
6.
Nat Commun ; 9(1): 1696, 2018 04 27.
Article in English | MEDLINE | ID: mdl-29703898

ABSTRACT

Acetylation of microtubules (MT) confers mechanical stability necessary for numerous functions including cell cycle and intracellular transport. Although αTAT1 is a major MT acetyltransferase, how this enzyme is regulated remains much less clear. Here we report TGF-ß-activated kinase 1 (TAK1) as a key activator of αTAT1. TAK1 directly interacts with and phosphorylates αTAT1 at Ser237 to critically enhance its catalytic activity, as mutating this site to alanine abrogates, whereas a phosphomimetic induces MT hyperacetylation across cell types. Using a custom phospho-αTAT1-Ser237 antibody, we screen various mouse tissues to discover that brain contains some of the highest TAK1-dependent αTAT1 activity, which, accordingly, is diminished rapidly upon intra-cerebral injection of a TAK1 inhibitor. Lastly, we show that TAK1 selectively inhibits AKT to suppress mitogenic and metabolism-related pathways through MT-based mechanisms in culture and in vivo. Collectively, our findings support a fundamental new role for TGF-ß signaling in MT-related functions and disease.


Subject(s)
Acetyltransferases/metabolism , Cell Proliferation/physiology , MAP Kinase Kinase Kinases/metabolism , Microtubule Proteins/metabolism , Microtubules/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Acetylation/drug effects , Animals , Benzamides/pharmacology , COS Cells , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line , Cell Proliferation/drug effects , Chlorocebus aethiops , Dioxoles/pharmacology , Gene Knockdown Techniques , HeLa Cells , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/genetics , Male , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/physiology , Mice , Mice, Inbred ICR , Microtubules/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Zearalenone/analogs & derivatives , Zearalenone/pharmacology
7.
Oncotarget ; 8(8): 12675-12685, 2017 Feb 21.
Article in English | MEDLINE | ID: mdl-27177332

ABSTRACT

Angiogenesis is the formation of new blood vessels from existing vasculature critical for embryonic development and vascular remodeling. Its dysregulation underlies numerous pathologic states ranging from ischemia to tumor growth and as such identifying new targeted- therapies is of significant interest for angiogenesis-based medicine. Here we evaluated the potential angiostatic properties of capsicodendrin (CPCD), a natural compound isolated from Cinnamosma macrocarpa, a plant belonging to the Malagasy Cinnamosma. CPCD potently inhibits endothelial proliferation, migration and capillary tube formation at nanomolar to low micromolar concentrations without inducing cytotoxic effects. We show that CPCD directly inactivates VEGFR2 and downstream AKT signaling, thereby strongly inducing autophagy as determined by increased expression of beclin1, autophagy-related gene (Atg) 3, Atg5 and LC3 cleavage. Ectopic AKT overexpression counteracts the inhibitory effects of CPCD on proliferation and capillary tubule formation. Importantly, CPCD treatment in vivo inhibits sprouting angiogenesis as evidenced by strongly reduced intersegmental vessel (ISV) sprouting and subintestinal vessel (SIV) formation during zebrafish embryonic development, and correlates with increased presence of LC3II along the ISVs despite overall reduced vasculature. These findings demonstrate CPCD as a potent inhibitor of the VEGFR2/AKT pathway at nanomolar concentrations and inducer of autophagy-related angiostatic effects.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neovascularization, Physiologic/drug effects , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Autophagy/drug effects , Cell Line , Endothelial Cells/drug effects , Fluorescent Antibody Technique , Humans , Magnoliaceae , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Zebrafish
8.
Mol Cell ; 62(4): 520-31, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27184078

ABSTRACT

Smads are nuclear-shuttling transcriptional mediators of transforming growth factor-ß (TGF-ß) signaling. Although their essential nuclear roles in gene regulation during development and carcinogenesis are well established, whether they have important cytoplasmic functions remains unclear. Here we report that Smad2 is a critical determinant of mitochondrial dynamics. We identified mitofusin2 (MFN2) and Rab and Ras Interactor 1 (RIN1) as new Smad2 binding partners required for mitochondrial fusion. Unlike TGF-ß-induced Smad2/3 transcriptional responses underlying mitochondrial fragmentation and apoptosis, inactive cytoplasmic Smad2 rapidly promotes mitochondrial fusion by recruiting RIN1 into a complex with MFN2. We demonstrate that Smad2 is a key scaffold, allowing RIN1 to act as a GTP exchange factor for MFN2-GTPase activation to promote mitochondrial ATP synthesis and suppress superoxide production. These results reveal functional implications between Smads and mitochondrial dysfunction in cancer and metabolic and neurodegenerative disorders.


Subject(s)
GTP Phosphohydrolases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondria/enzymology , Mitochondrial Dynamics , Mitochondrial Proteins/metabolism , Smad2 Protein/metabolism , A549 Cells , Adenosine Triphosphate/metabolism , Animals , COS Cells , Chlorocebus aethiops , Energy Metabolism , GTP Phosphohydrolases/genetics , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/genetics , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , RNA Interference , Signal Transduction , Smad2 Protein/genetics , Superoxides/metabolism , Transfection
9.
J Biol Chem ; 290(24): 14884-92, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25931117

ABSTRACT

Autophagy is the targeted degradation of proteins and organelles critical for homeostasis and cell survival. Transforming growth factor ß (TGF-ß) differentially regulates autophagy in a context-specific manner, although the precise intracellular mechanisms remain less clear. Importantly, how TGF-ß controls autophagic responses in endothelial cells (EC) during angiogenesis is unknown. Here we identified endoglin, an EC-specific TGF-ß co-receptor essential for angiogenesis, as a key determinant of autophagy. Among the two opposing TGF-ß Smad pathways in the EC system (Smad1/5/8 and Smad2/3), we found Smad2 as the major transcriptional regulator of autophagy that targets beclin1 (BECN1) gene expression. Smad2, but not Smad3, acts as a repressor upstream of the BECN1 promoter region. Overall, endoglin promotes autophagy by impeding Smad2 transcriptional repressor activity. Notably, increased beclin1 levels upon Smad2 knockdown directly correlated with enhanced autophagy during angiogenesis. Taken together, these results establish endoglin as a critical mediator of autophagy and demonstrate a new transcriptional mechanism by which Smad2 inhibits angiogenesis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , Endothelium/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Smad2 Protein/physiology , Animals , Base Sequence , Beclin-1 , Cells, Cultured , Chromatin Immunoprecipitation , DNA Primers , Endoglin , Endothelium/cytology , Mice , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Smad2 Protein/genetics
10.
J Biol Chem ; 289(37): 25486-96, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25070888

ABSTRACT

Endoglin is a transforming growth factor ß (TGF-ß) co-receptor essential for angiogenesis and tumor vascularization. Endoglin modulates the crucial balance between pro- and anti-angiogenic signaling by activin receptor-like kinase (ALK) 1, 5, and TGF-ß type II (TßRII) receptors. Despite its established role in physiology and disease, the mechanism of endoglin down-regulation remains unknown. Here we report that the conserved juxtamembrane cytoplasmic tyrosine motif ((612)YIY(614)) is a critical determinant of angiogenesis. Src directly phosphorylates this motif to induce endoglin internalization and degradation via the lysosome. We identified epidermal growth factor (EGF) and vascular endothelial growth factor (VEGF) as Src-activators that induce endoglin turnover following (612)YIY(614) phosphorylation. Interestingly, Src phosphorylation of endoglin-(612)YIY(614) was also an important process for receptor down-regulation by TRACON105 (TRC105), an endoglin-targeting antibody currently in clinical trials. The regulation of (612)YIY(614) phosphorylation was critical for angiogenesis, as both the phosphomimetic and unphosphorylatable mutants impaired endothelial functions including proliferation, migration, and capillary tube formation. Collectively, these findings establish Src and pro-angiogenic mitogens as critical mediators of endoglin stability and function.


Subject(s)
Amino Acid Motifs/genetics , Antigens, CD/genetics , Neovascularization, Physiologic/genetics , Receptors, Cell Surface/genetics , src-Family Kinases/metabolism , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Cell Proliferation/genetics , Endoglin , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Stability , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
11.
Biochem Biophys Res Commun ; 424(3): 620-3, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22789855

ABSTRACT

Endoglin is an endothelial-specific transforming growth factor beta (TGF-ß) co-receptor essential for angiogenesis and vascular remodeling. Endoglin regulates a wide range of cellular processes, including cell adhesion, migration, and proliferation, through TGF-ß signaling to canonical Smad and Smad-independent pathways. Despite its overall pro-angiogenic role in the vasculature, the underlying mechanism of endoglin action is poorly characterized. We previously identified ß-arrestin2 as a binding partner that causes endoglin internalization from the plasma membrane and inhibits ERK signaling towards endothelial migration. In the present study, we examined the mechanistic role of endoglin and ß-arrestin2 in endothelial cell proliferation. We show that endoglin impedes cell growth through sustained inhibition of ERK-induced c-Myc and cyclin D1 expression in a TGF-ß-independent manner. The down-regulation of c-Myc and cyclin D1, along with growth-inhibition, are reversed when the endoglin/ß-arrestin2 interaction is disrupted. Given that TGF-ß-induced Smad signaling potently represses c-Myc in most cell types, our findings here show a novel mechanism by which endoglin augments growth-inhibition by targeting ERK and key downstream mitogenic substrates.


Subject(s)
Antigens, CD/physiology , Cyclin D1/biosynthesis , Endothelium, Vascular/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Neovascularization, Physiologic , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Receptors, Cell Surface/physiology , Cell Line , Cell Proliferation , Endoglin , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Transforming Growth Factor alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...