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










Publication year range
1.
Adv Sci (Weinh) ; 11(22): e2310146, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38526153

ABSTRACT

Bladder cancer (BC) is one of the most common tumors characterized by a high rate of relapse and a lack of targeted therapy. Here, YEATS domain-containing protein 4 (YEATS4) is an essential gene for BC cell viability using CRISPR-Cas9 library screening is reported, and that HUWE1 is an E3 ligase responsible for YEATS4 ubiquitination and proteasomal degradation by the Protein Stability Regulators Screening Assay. KAT8-mediated acetylation of YEATS4 impaired its interaction with HUWE1 and consequently prevented its ubiquitination and degradation. The protein levels of YEATS4 and KAT8 are positively correlated and high levels of these two proteins are associated with poor overall survival in BC patients. Importantly, suppression of YEATS4 acetylation with the KAT8 inhibitor MG149 decreased YEATS4 acetylation, reduced cell viability, and sensitized BC cells to cisplatin treatment. The findings reveal a critical role of the KAT8/YEATS4 axis in both tumor growth and cisplatin sensitivity in BC cells, potentially generating a novel therapeutic strategy for BC patients.


Subject(s)
Cisplatin , Histone Acetyltransferases , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Humans , Cisplatin/pharmacology , Cell Line, Tumor , Mice , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Animals , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Acetylation/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics
3.
Sci Adv ; 9(47): eadi0889, 2023 11 24.
Article in English | MEDLINE | ID: mdl-37992172

ABSTRACT

Osteosarcoma is a highly aggressive cancer and lacks effective therapeutic targets. We found that L3MBTL2 acts as a tumor suppressor by transcriptionally repressing IFIT2 in osteosarcoma. L3MBTL2 recruits the components of Polycomb repressive complex 1.6 to form condensates via both Pho-binding pockets and polybasic regions within carboxyl-terminal intrinsically disordered regions; the L3MBTL2-induced condensates are required for its tumor suppression. Multi-monoubiquitination of L3MBTL2 by UBE2O results in its proteasomal degradation, and the UBE2O/L3MBTL2 axis was crucial for osteosarcoma growth. There is a reverse correlation between L3MBTL2 and UBE2O in osteosarcoma tissues, and higher UBE2O and lower L3MBTL2 are associated with poorer prognosis in osteosarcoma. Pharmacological blockage of UBE2O by arsenic trioxide can enhance L3MBTL2-induced condensates and consequently suppress osteosarcoma growth. Our findings unveil a crucial biological function of L3MBTL2-induced condensates in mediating tumor suppression, proposing the UBE2O-L3MBTL2 axis as a potential cancer therapeutic target in osteosarcoma.


Subject(s)
Bone Neoplasms , Osteosarcoma , Humans , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Cell Line, Tumor , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Osteosarcoma/metabolism , Polycomb Repressive Complex 1/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination
4.
Adv Sci (Weinh) ; 10(5): e2205483, 2023 02.
Article in English | MEDLINE | ID: mdl-36529692

ABSTRACT

Rab22a-NeoF fusion protein has recently been reported as a promising target for osteosarcoma lung metastasis. However, how this fusion protein is regulated in cells remains unknown. Here, using multiple screenings, it is reported that Rab22a-NeoF1 fusion protein is degraded by an E3 ligase STUB1 via the autophagy receptor NDP52-mediated lysosome pathway, which is facilitated by PINK1 kinase. Mechanistically, STUB1 catalyzes the K63-linked ubiquitin chains on lysine112 of Rab22a-NeoF1, which is responsible for the binding of Rab22a-NeoF1 to NDP52, resulting in lysosomal degradation of Rab22a-NeoF1. PINK1 is able to phosphorylate Rab22a-NeoF1 at serine120, which promotes ubiquitination and degradation of Rab22a-NeoF1. Consistently, by upregulating PINK1, Sorafenib and Regorafenib can inhibit osteosarcoma lung metastasis induced by Rab22a-NeoF1. These findings reveal that the lysosomal degradation of Rab22a-NeoF1 fusion protein is targetable for osteosarcoma lung metastasis, proposing that Sorafenib and Regorafenib may benefit cancer patients who are positive for the RAB22A-NeoF1 fusion gene.


Subject(s)
Lung Neoplasms , Oncogene Proteins, Fusion , Osteosarcoma , Humans , Lung Neoplasms/secondary , Lysosomes/metabolism , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Protein Kinases/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , Sorafenib/metabolism , Ubiquitin-Protein Ligases/metabolism , Oncogene Proteins, Fusion/metabolism , Oncogene Proteins, Fusion/therapeutic use
5.
Signal Transduct Target Ther ; 6(1): 59, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33568623

ABSTRACT

It remains unknown for decades how some of the therapeutic fusion proteins positive in a small percentage of cancer cells account for patient outcome. Here, we report that osteosarcoma Rab22a-NeoF1 fusion protein, together with its binding partner PYK2, is sorted into exosomes by HSP90 via its KFERQ-like motif (RVLFLN142). The exosomal Rab22a-NeoF1 fusion protein facilitates the pulmonary pre-metastatic niche formation by recruiting bone marrow-derived macrophages. The exosomal PYK2 activates RhoA in its negative recipient osteosarcoma cells and induces signal transducer and activator of transcription 3 activation in its recipient macrophages to increase M2 phenotype. Consequently, lung metastases of its recipient osteosarcoma cells are promoted by this exosomal Rab22a-NeoF1 fusion protein, and this event can be targeted by disrupting its interaction with PYK2 using a designed internalizing RGD peptide.


Subject(s)
Focal Adhesion Kinase 2/genetics , Lung Neoplasms/genetics , Osteosarcoma/genetics , rab GTP-Binding Proteins/genetics , Animals , Cell Line, Tumor , Exosomes/genetics , Gene Expression Regulation, Neoplastic/genetics , HSP90 Heat-Shock Proteins/genetics , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice , Oligopeptides/genetics , Oncogene Proteins, Fusion/genetics , Osteosarcoma/pathology , Protein Binding/genetics , RAW 264.7 Cells , rhoA GTP-Binding Protein/genetics
6.
Nat Cell Biol ; 22(7): 868-881, 2020 07.
Article in English | MEDLINE | ID: mdl-32483387

ABSTRACT

Osteosarcoma is a type of aggressive malignant bone tumour that frequently metastasizes to lungs, resulting in poor prognosis. However, the molecular mechanisms of lung metastasis of osteosarcoma remain poorly understood. Here we identify exon-intron fusion genes in osteosarcoma cell lines and tissues. These fusion genes are derived from chromosomal translocations that juxtapose the coding region for amino acids 1-38 of Rab22a (Rab22a1-38) with multiple inverted introns and untranslated regions of chromosome 20. The resulting translation products, designated Rab22a-NeoFs, acquire the ability to drive lung metastasis of osteosarcoma. The Rab22a1-38 moiety governs the function of Rab22a-NeoFs by binding to SmgGDS-607, a GTP-GDP exchange factor of RhoA. This association facilitates the release of GTP-bound RhoA from SmgGDS-607, which induces increased activity of RhoA and promotes metastasis. Disrupting the interaction between Rab22a-NeoF1 and SmgGDS-607 with a synthetic peptide prevents lung metastasis in an orthotopic model of osteosarcoma. Our findings may provide a promising strategy for a subset of osteosarcoma patients with lung metastases.


Subject(s)
Biomarkers, Tumor/metabolism , Bone Neoplasms/pathology , Lung Neoplasms/secondary , Osteosarcoma/pathology , Translocation, Genetic , rab GTP-Binding Proteins/metabolism , Adult , Animals , Apoptosis , Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Osteosarcoma/genetics , Osteosarcoma/metabolism , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Young Adult , rab GTP-Binding Proteins/genetics
7.
8.
Cancer Lett ; 442: 252-261, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30423408

ABSTRACT

N6-methyladenosin (m6A) is one of the most pervasive modification of mRNA in eukaryotes and the m6A methyltransferases and demethylases play critical roles in many types of cancer. However the role of m6A-binding proteins in cancer remains elusive. Here we report that the down-regulation of YTHDF2 was specifically induced by hypoxia in hepatocellular carcinoma (HCC) cells, and that overexpression of YTHDF2 suppressed cell proliferation, tumor growth and activation of MEK and ERK in HCC cells. Mechanistically, YTHDF2 directly bound the m6A modification site of EGFR 3'-UTR to promote the degradation of EGFR mRNA in HCC cells. This is the first report showing that YTHDF2 may act as a tumor suppressor to repress cell proliferation and growth via destabilizing the EGFR mRNA in HCC.


Subject(s)
Adenosine/analogs & derivatives , Cell Proliferation , Liver Neoplasms/metabolism , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Tumor Suppressor Proteins/metabolism , 3' Untranslated Regions , Adenosine/metabolism , Animals , Binding Sites , Cell Line, Tumor , Enzyme Activation , ErbB Receptors/genetics , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinase Kinases/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Signal Transduction , Tumor Burden , Tumor Hypoxia , Tumor Suppressor Proteins/genetics
9.
Sci Rep ; 6: 27819, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27292155

ABSTRACT

Drug repurposing of non-cancer drugs represents an attractive approach to develop new cancer therapy. Using the TRAMP transgenic mouse model, glipizide, a widely used drug for type 2 diabetes mellitus, has been identified to suppress prostate cancer (PC) growth and metastasis. Angiogenesis is intimately associated with various human cancer developments. Intriguingly, glipizide significantly reduces microvessel density in PC tumor tissues, while not inhibiting prostate cancer cell proliferation from the MTT assay and flow cytometry investigation. Moreover, glipizide inhibits the tubular structure formation of human umbilical vein endothelial cells by regulating the HMGIY/Angiopoietin-1 signaling pathway. Taken together, these results demonstrate that glipizide has the potential to be repurposed as an effective therapeutic for the treatment of PC by targeting tumor-induced angiogenesis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antigens, Polyomavirus Transforming/genetics , Glipizide/pharmacology , Neovascularization, Physiologic/drug effects , Angiogenesis Inhibitors/therapeutic use , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Down-Regulation/drug effects , Glipizide/therapeutic use , Human Umbilical Vein Endothelial Cells , Humans , Liver Neoplasms/pathology , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
10.
Atherosclerosis ; 244: 179-87, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26649902

ABSTRACT

OBJECTIVE: Serum amyloid P conpoent (SAP), a member of the pentraxin family, interact with pathogens and cell debris to promote their removal by macrophages and neutrophils and is co-localized with atherosclerotic plaques in patients. However, the exact mechanism of SAP in atherogenesis is still unclear. We investigated whether SAP influence macrophage recruitment and foam cell formation and ultimately affect atherosclerotic progression. METHODS: we generated apoE(-/-); SAP(-/-) (DKO) mice and fed them western diet for 4 and 8 weeks to characterize atherosclerosis development. RESULTS: SAP deficiency effectively reduced plaque size both in the aorta (p = 0.0006 for 4 wks; p = 0.0001 for 8 wks) and the aortic root (p = 0.0061 for 4 wks; p = 0.0079 for 8wks) compared with apoE(-/-) mice. Meanwhile, SAP deficiency inhibited oxLDL-induced foam cell formation (p = 0.0004) compared with apoE(-/-) mice and SAP treatment increases oxLDL-induced foam cell formation (p = 0.002) in RAW cells. Besides, SAP deficiency reduced macrophages recruitment (p = 0.035) in vivo and in vitro (p = 0.026). Furthermore, SAP treatment enhanced CD36 (p = 0.007) and FcγRI (p = 0.031) expression induced by oxLDL through upregulating JNK and p38 MAPK phosphorylation whereas specific JNK1/2 inhibitor reduced CD36 (p = 0.0005) and FcγRI (P = 0.0007) expression in RAW cell. SAP deficiency also significantly decreased the expression of M1 and M2 macrophage markers and inflammatory cytokines in oxLDL-induced macrophages. CONCLUSION: SAP deficiency mitigated foam cell formation and atherosclerotic development in apoE(-/-) mice, due to reduction in macrophages recruitment, polarization and pro-inflammatory cytokines and inhibition the CD36/FcγR-dependent signaling pathway.


Subject(s)
Aorta, Thoracic/pathology , Atherosclerosis/genetics , Gene Expression Regulation , Hyperlipidemias/complications , RNA/genetics , Serum Amyloid P-Component/deficiency , Serum Amyloid P-Component/genetics , Animals , Aorta, Thoracic/metabolism , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/etiology , Atherosclerosis/metabolism , Blotting, Western , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Humans , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Immunohistochemistry , Male , Mice , Mice, Knockout , Phenotype , Real-Time Polymerase Chain Reaction
11.
Arterioscler Thromb Vasc Biol ; 35(2): 368-77, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25503990

ABSTRACT

OBJECTIVE: An aneurysm is an inflammatory vascular condition. Phosphatidylinositol 3-kinases δ is highly expressed in leukocytes, and play a key role in innate immunity. However, the link between phosphatidylinositol 3-kinases δ and aneurysm development has not yet been elucidated. APPROACH AND RESULTS: Carotid ligation unexpectedly induced characteristic aneurysm formation beneath the ligation point in p110δ(D910A/D910A) mice (n=25; P<0.001 versus wild-type). Besides, p110δ inactivation exacerbated CaCl2-induced abdominal aortic aneurysms development. A reverse transcription polymerase chain reaction microarray revealed significant extracellular matrix components degradation and matrix metalloproteinases (MMPs) upregulation in the abdominal aorta of p110δ(D910A/D910A) mice. Similarly, the expression of both collagen I and IV was significantly decreased (n=10; P<0.05 versus wild-type) in carotid artery. Western blot assay confirmed that MMP-12 was significantly upregulated in arteries of p110δ(D910A/D910A) mice (n=10; P<0.01 versus wild-type). In vitro, p110δ inactivation marked increase peritoneal macrophages recruitment and synergistically enhance tumor necrosis factor-α-induced recruitment. A specific phosphatidylinositol 3-kinases δ inhibitor (IC87114) or genetic p110δ inactivation upregulated MMP-12 expression and c-Jun phosphorylation (n=6; P<0.05 versus wild-type macrophages). IC87114 also increased activator protein-1 DNA-binding activity (n=6; P<0.001 versus control) and enhanced the effect of tumor necrosis factor-α on activator protein-1-binding activity (n=5; P<0.01 versus tumor necrosis factor-α treatment groups). Knockdown of c-Jun suppressed the effect of the IC87114 and tumor necrosis factor-α on MMP-12 mRNA expression (n=5 in each group; P<0.01 versus scrRNA treatment groups). CONCLUSIONS: Our findings demonstrate that p110δ inactivation leads to extracellular matrix degradation in vessels and promotes aneurysm development by inducing macrophages migration and upregulating the activator protein-1/MMP-12 pathway in macrophages.


Subject(s)
Aorta, Abdominal/enzymology , Aortic Aneurysm, Abdominal/enzymology , Carotid Artery Injuries/enzymology , Carotid Artery, Common/enzymology , Macrophages, Peritoneal/enzymology , Matrix Metalloproteinase 12/metabolism , Phosphatidylinositol 3-Kinases/deficiency , Transcription Factor AP-1/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Aorta, Abdominal/drug effects , Aorta, Abdominal/pathology , Aorta, Abdominal/surgery , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/pathology , Calcium Chloride , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Carotid Artery, Common/drug effects , Carotid Artery, Common/pathology , Carotid Artery, Common/surgery , Cell Line , Class I Phosphatidylinositol 3-Kinases , Disease Models, Animal , Enzyme Activation , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation, Enzymologic , Ligation , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , RNA Interference , Signal Transduction , Transcription Factor AP-1/genetics , Transfection , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...