Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Mol Cell ; 83(16): 3010-3026.e8, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37595559

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) is a master regulator of cell growth that stimulates macromolecule synthesis through transcription, RNA processing, and post-translational modification of metabolic enzymes. However, the mechanisms of how mTORC1 orchestrates multiple steps of gene expression programs remain unclear. Here, we identify family with sequence similarity 120A (FAM120A) as a transcription co-activator that couples transcription and splicing of de novo lipid synthesis enzymes downstream of mTORC1-serine/arginine-rich protein kinase 2 (SRPK2) signaling. The mTORC1-activated SRPK2 phosphorylates splicing factor serine/arginine-rich splicing factor 1 (SRSF1), enhancing its binding to FAM120A. FAM120A directly interacts with a lipogenic transcription factor SREBP1 at active promoters, thereby bridging the newly transcribed lipogenic genes from RNA polymerase II to the SRSF1 and U1-70K-containing RNA-splicing machinery. This mTORC1-regulated, multi-protein complex promotes efficient splicing and stability of lipogenic transcripts, resulting in fatty acid synthesis and cancer cell proliferation. These results elucidate FAM120A as a critical transcription co-factor that connects mTORC1-dependent gene regulation programs for anabolic cell growth.


Subject(s)
Arginine , Lipogenesis , Sterol Regulatory Element Binding Protein 1 , Lipogenesis/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , RNA Splicing Factors , Sterol Regulatory Element Binding Protein 1/metabolism , Humans , Sterol Regulatory Element Binding Proteins/metabolism
2.
Aging Cell ; 22(2): e13760, 2023 02.
Article in English | MEDLINE | ID: mdl-36567449

ABSTRACT

Aging-associated microbial dysbiosis exacerbates various disorders and dysfunctions, and is a major contributor to morbidity and mortality in the elderly, but the underlying cause of this aging-related syndrome is confusing. SIRT6 knockout (SIRT6 KO) mice undergo premature aging and succumb to death by 4 weeks, and are therefore useful as a premature aging research model. Here, fecal microbiota transplantation from SIRT6 KO mice into wild-type (WT) mice phenocopies the gut dysbiosis and premature aging observed in SIRT6 KO mice. Conversely, an expanded lifespan was observed in SIRT6 KO mice when transplanted with microbiota from WT mice. Antibiotic cocktail treatment attenuated inflammation and cell senescence in KO mice, directly suggesting that gut dysbiosis contributes to the premature aging of SIRT6 KO mice. Increased Enterobacteriaceae translocation, driven by the overgrowth of Escherichia coli, is the likely mechanism for the premature aging effects of microbiome dysregulation, which could be reversed by a high-fat diet. Our results provide a mechanism for the causal link between gut dysbiosis and aging, and support a beneficial effect of a high-fat diet for correcting gut dysbiosis and alleviating premature aging. This study provides a rationale for the integration of microbiome-based high-fat diets into therapeutic interventions against aging-associated diseases.


Subject(s)
Aging, Premature , Gastrointestinal Microbiome , Sirtuins , Animals , Mice , Aging, Premature/genetics , Diet, High-Fat , Dysbiosis/etiology , Enterobacteriaceae , Mice, Inbred C57BL
3.
Nat Commun ; 13(1): 6239, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36266345

ABSTRACT

The systemic metabolic shifts that occur during aging and the local metabolic alterations of a tumor, its stroma and their communication cooperate to establish a unique tumor microenvironment (TME) fostering cancer progression. Here, we show that methylmalonic acid (MMA), an aging-increased oncometabolite also produced by aggressive cancer cells, activates fibroblasts in the TME, which reciprocally secrete IL-6 loaded extracellular vesicles (EVs) that drive cancer progression, drug resistance and metastasis. The cancer-associated fibroblast (CAF)-released EV cargo is modified as a result of reactive oxygen species (ROS) generation and activation of the canonical and noncanonical TGFß signaling pathways. EV-associated IL-6 functions as a stroma-tumor messenger, activating the JAK/STAT3 and TGFß signaling pathways in tumor cells and promoting pro-aggressive behaviors. Our findings define the role of MMA in CAF activation to drive metastatic reprogramming, unveiling potential therapeutic avenues to target MMA at the nexus of aging, the tumor microenvironment and metastasis.


Subject(s)
Cancer-Associated Fibroblasts , Extracellular Vesicles , Neoplasms , Humans , Cancer-Associated Fibroblasts/metabolism , Reactive Oxygen Species/metabolism , Methylmalonic Acid/metabolism , Interleukin-6/metabolism , Tumor Microenvironment , Neoplasms/pathology , Extracellular Vesicles/metabolism , Transforming Growth Factor beta/metabolism
4.
Sci Adv ; 8(20): eabm8786, 2022 05 20.
Article in English | MEDLINE | ID: mdl-35594343

ABSTRACT

Serine/one-carbon metabolism provides critical resources for nucleotide biosynthesis and epigenetic maintenance and is thus necessary in cancer cell growth, although the detailed regulatory mechanisms remain unclear. We uncover a critical role of glycogen synthase kinase 3 (GSK3) in regulating the expression of serine/one-carbon metabolic enzymes. Nuclear enrichment of GSK3 significantly suppresses genes that mediate de novo serine synthesis, including PHGDH, PSAT1, PSPH, and one-carbon metabolism, including SHMT2 and MTHFD2. FRAT1 promotes nuclear exclusion of GSK3, enhances serine/one-carbon metabolism, and, as a result, confers cell vulnerability to inhibitors that target this metabolic process such as SHIN1, a specific SHMT1/2 inhibitor. Furthermore, pharmacological or genetic suppression of GSK3 promotes serine/one-carbon metabolism and exhibits a significant synergistic effect in combination with SHIN1 in suppressing cancer cell proliferation in cultured cells and in vivo. Our observations indicate that inhibition of nuclear GSK3 signaling creates a vulnerability, which results in enhanced efficacy of serine/one-carbon metabolism inhibitors for the treatment of cancer.


Subject(s)
Lung Neoplasms , Serine , Adaptor Proteins, Signal Transducing/metabolism , Carbon/metabolism , Glycogen Synthase Kinase 3 , Humans , Lung Neoplasms/genetics , Proto-Oncogene Proteins/metabolism , Signal Transduction
5.
Mol Cell ; 81(10): 2064-2075.e8, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33756105

ABSTRACT

Dysregulated mTORC1 signaling alters a wide range of cellular processes, contributing to metabolic disorders and cancer. Defining the molecular details of downstream effectors is thus critical for uncovering selective therapeutic targets. We report that mTORC1 and its downstream kinase S6K enhance eIF4A/4B-mediated translation of Wilms' tumor 1-associated protein (WTAP), an adaptor for the N6-methyladenosine (m6A) RNA methyltransferase complex. This regulation is mediated by 5' UTR of WTAP mRNA that is targeted by eIF4A/4B. Single-nucleotide-resolution m6A mapping revealed that MAX dimerization protein 2 (MXD2) mRNA contains m6A, and increased m6A modification enhances its degradation. WTAP induces cMyc-MAX association by suppressing MXD2 expression, which promotes cMyc transcriptional activity and proliferation of mTORC1-activated cancer cells. These results elucidate a mechanism whereby mTORC1 stimulates oncogenic signaling via m6A RNA modification and illuminates the WTAP-MXD2-cMyc axis as a potential therapeutic target for mTORC1-driven cancers.


Subject(s)
Adenosine/analogs & derivatives , Mechanistic Target of Rapamycin Complex 1/metabolism , RNA Stability , Adenosine/metabolism , Animals , Base Sequence , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Eukaryotic Initiation Factors/metabolism , HEK293 Cells , Humans , Male , Mice , Models, Biological , Protein Biosynthesis , Proto-Oncogene Proteins c-myc/metabolism , RNA Splicing Factors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction
6.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33649236

ABSTRACT

Mechanistic Target of Rapamycin Complex 1 (mTORC1) is a central regulator of cell growth and metabolism that senses and integrates nutritional and environmental cues with cellular responses. Recent studies have revealed critical roles of mTORC1 in RNA biogenesis and processing. Here, we find that the m6A methyltransferase complex (MTC) is a downstream effector of mTORC1 during autophagy in Drosophila and human cells. Furthermore, we show that the Chaperonin Containing Tailless complex polypeptide 1 (CCT) complex, which facilitates protein folding, acts as a link between mTORC1 and MTC. The mTORC1 activates the chaperonin CCT complex to stabilize MTC, thereby increasing m6A levels on the messenger RNAs encoding autophagy-related genes, leading to their degradation and suppression of autophagy. Altogether, our study reveals an evolutionarily conserved mechanism linking mTORC1 signaling with m6A RNA methylation and demonstrates their roles in suppressing autophagy.


Subject(s)
Autophagy , Drosophila Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Methyltransferases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/metabolism , Signal Transduction , Animals , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster , Humans , Mechanistic Target of Rapamycin Complex 1/genetics , Methylation , Methyltransferases/genetics , Orphan Nuclear Receptors , RNA Stability , Receptors, Cytoplasmic and Nuclear/genetics , Repressor Proteins/genetics
8.
Mol Cell ; 70(5): 949-960.e4, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29861159

ABSTRACT

The mammalian Target of Rapamycin Complex 1 (mTORC1)-signaling system plays a critical role in the maintenance of cellular homeostasis by sensing and integrating multiple extracellular and intracellular cues. Therefore, uncovering the effectors of mTORC1 signaling is pivotal to understanding its pathophysiological effects. Here we report that the transcription factor forkhead/winged helix family k1 (Foxk1) is a mediator of mTORC1-regulated gene expression. Surprisingly, Foxk1 phosphorylation is increased upon mTORC1 suppression, which elicits a 14-3-3 interaction, a reduction of DNA binding, and nuclear exclusion. Mechanistically, this occurs by mTORC1-dependent suppression of nuclear signaling by the Foxk1 kinase, Gsk3. This pathway then regulates the expression of multiple genes associated with glycolysis and downstream anabolic pathways directly modulated by Foxk1 and/or by Foxk1-regulated expression of Hif-1α. Thus, Foxk1 mediates mTORC1-driven metabolic rewiring, and it is likely to be critical for metabolic diseases where improper mTORC1 signaling plays an important role.


Subject(s)
Cellular Reprogramming , Energy Metabolism , Forkhead Transcription Factors/metabolism , Glycogen Synthase Kinase 3/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , 14-3-3 Proteins/metabolism , Active Transport, Cell Nucleus , Animals , Binding Sites , Cell Proliferation , Down-Regulation , Forkhead Transcription Factors/genetics , Glycogen Synthase Kinase 3/genetics , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Phosphorylation , Protein Binding , Signal Transduction
9.
Cell ; 171(7): 1545-1558.e18, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29153836

ABSTRACT

mTORC1 is a signal integrator and master regulator of cellular anabolic processes linked to cell growth and survival. Here, we demonstrate that mTORC1 promotes lipid biogenesis via SRPK2, a key regulator of RNA-binding SR proteins. mTORC1-activated S6K1 phosphorylates SRPK2 at Ser494, which primes Ser497 phosphorylation by CK1. These phosphorylation events promote SRPK2 nuclear translocation and phosphorylation of SR proteins. Genome-wide transcriptome analysis reveals that lipid biosynthetic enzymes are among the downstream targets of mTORC1-SRPK2 signaling. Mechanistically, SRPK2 promotes SR protein binding to U1-70K to induce splicing of lipogenic pre-mRNAs. Inhibition of this signaling pathway leads to intron retention of lipogenic genes, which triggers nonsense-mediated mRNA decay. Genetic or pharmacological inhibition of SRPK2 blunts de novo lipid synthesis, thereby suppressing cell growth. These results thus reveal a novel role of mTORC1-SRPK2 signaling in post-transcriptional regulation of lipid metabolism and demonstrate that SRPK2 is a potential therapeutic target for mTORC1-driven metabolic disorders.


Subject(s)
Gene Expression Regulation , Lipogenesis , RNA Processing, Post-Transcriptional , Signal Transduction , Animals , Cell Nucleus/metabolism , Cholesterol/metabolism , Fatty Acids/metabolism , Female , Heterografts , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Protein Serine-Threonine Kinases/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
10.
Arterioscler Thromb Vasc Biol ; 36(9): 1772-81, 2016 09.
Article in English | MEDLINE | ID: mdl-27417587

ABSTRACT

OBJECTIVE: Liver-enriched transcription factor cAMP-responsive element-binding protein H (CREBH) regulates plasma triglyceride clearance by inducing lipoprotein lipase cofactors, such as apolipoprotein A-IV (apoA-IV), apoA-V, and apoC-II. CREBH also regulates apoA-I transcription. This study aims to determine whether CREBH has a role in lipoprotein metabolism and development of atherosclerosis. APPROACH AND RESULTS: CREBH-deficient Creb3l3(-/-) mice were bred with Ldlr(-/-) mice creating Ldlr(-/-) Creb3l3(-/-) double knockout mice. Mice were fed on a high-fat and high-sucrose Western diet for 20 weeks. We showed that CREBH deletion in Ldlr(-/-) mice increased very low-density lipoprotein-associated triglyceride and cholesterol levels, consistent with the impairment of lipoprotein lipase-mediated triglyceride clearance in these mice. In contrast, high-density lipoprotein cholesterol levels were decreased in CREBH-deficient mice, which was associated with decreased production of apoA-I from the liver. The results indicate that CREBH directly activated Apoa1 gene transcription. Accompanied by the worsened atherogenic lipid profile, Ldlr(-/-) Creb3l3(-/-) mice developed significantly more atherosclerotic lesions in the aortas than Ldlr(-/-) mice. CONCLUSIONS: We identified CREBH as an important regulator of lipoprotein metabolism and suggest that increasing hepatic CREBH activity may be a novel strategy for prevention and treatment of atherosclerosis.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Cyclic AMP Response Element-Binding Protein/deficiency , Receptors, LDL/deficiency , Animals , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoprotein A-I/blood , Apolipoprotein A-I/genetics , Apolipoprotein B-100 , Apolipoproteins B/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Biomarkers/blood , Cell Line, Tumor , Cholesterol, HDL/blood , Cyclic AMP Response Element-Binding Protein/genetics , Diet, High-Fat , Dietary Sucrose , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Lipoprotein Lipase/metabolism , Lipoproteins, VLDL/blood , Liver/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Plaque, Atherosclerotic , Receptors, LDL/genetics , Severity of Illness Index , Transcription, Genetic , Transcriptional Activation , Triglycerides/blood
11.
Sci Rep ; 6: 27938, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27301791

ABSTRACT

Adipose tissue lipolysis produces glycerol and nonesterified fatty acids (NEFA) that serve as energy sources during nutrient scarcity. Adipose tissue lipolysis is tightly regulated and excessive lipolysis causes hepatic steatosis, as NEFA released from adipose tissue constitutes a major source of TG in the liver of patients with nonalcoholic fatty liver diseases. Here we show that the liver-enriched transcription factor CREBH is activated by TG accumulation and induces FGF21, which suppresses adipose tissue lipolysis, ameliorating hepatic steatosis. CREBH-deficient mice developed severe hepatic steatosis due to increased adipose tissue lipolysis, when fasted or fed a high-fat low-carbohydrate ketogenic diet. FGF21 production was impaired in CREBH-deficient mice, and adenoviral overexpression of FGF21 suppressed adipose tissue lipolysis and improved hepatic steatosis in these mice. Thus, our results uncover a negative feedback loop in which CREBH regulates NEFA flux from adipose tissue to the liver via FGF21.


Subject(s)
Adipose Tissue/pathology , Cyclic AMP Response Element-Binding Protein/metabolism , Fatty Liver/pathology , Fibroblast Growth Factors/metabolism , Lipolysis , Animals , Feedback, Physiological , Mice, Inbred C57BL , Mice, Knockout
12.
Mol Cell Biol ; 35(16): 2761-70, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26031337

ABSTRACT

The unfolded protein response (UPR) regulates endoplasmic reticulum (ER) homeostasis and protects cells from ER stress. IRE1α is a central regulator of the UPR that activates the transcription factor XBP1s through an unconventional splicing mechanism using its endoribonuclease activity. IRE1α also cleaves certain mRNAs containing XBP1-like secondary structures to promote the degradation of these mRNAs, a process known as regulated IRE1α-dependent decay (RIDD). We show here that the mRNA of CReP/Ppp1r15b, a regulatory subunit of eukaryotic translation initiation factor 2α (eIF2α) phosphatase, is a RIDD substrate. eIF2α plays a central role in the integrated stress response by mediating the translational attenuation to decrease the stress level in the cell. CReP expression was markedly suppressed in XBP1-deficient mice livers due to hyperactivated IRE1α. Decreased CReP expression caused the induction of eIF2α phosphorylation and the attenuation of protein synthesis in XBP1-deficient livers. ER stress also suppressed CReP expression in an IRE1α-dependent manner, which increased eIF2α phosphorylation and consequently attenuated protein synthesis. Taken together, the results of our study reveal a novel function of IRE1α in the regulation of eIF2α phosphorylation and the translational control.


Subject(s)
Endoribonucleases/metabolism , Eukaryotic Initiation Factor-2/metabolism , Protein Biosynthesis , Protein Phosphatase 1/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Stability , RNA, Messenger/metabolism , Animals , DNA-Binding Proteins/genetics , Down-Regulation , Endoplasmic Reticulum Stress , Gene Knockout Techniques , Liver/metabolism , Mice, Inbred C57BL , Phosphorylation , RNA, Messenger/chemistry , RNA, Messenger/genetics , Regulatory Factor X Transcription Factors , Transcription Factors/genetics , X-Box Binding Protein 1
13.
Proc Natl Acad Sci U S A ; 111(45): 15987-92, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25349418

ABSTRACT

Intrinsically disordered protein regions are widely distributed in the cytoplasmic domains of many transmembrane receptors. The cytoplasmic domain of a disintegrin and metalloprotease (ADAM)10, a transmembrane metalloprotease mediating ectodomain shedding of diverse membrane proteins, was recently suggested to mediate the homodimerization of ADAM10. Here we show that a recombinant cytoplasmic domain of ADAM10 (A10Cp) is unstructured as judged by its susceptibility to limited trypsin digestion and its circular dichroism spectrum. In comparison, recombinant transmembrane-cytoplasmic domain of ADAM10 (A10TmCp) reconstituted in dodecylphosphocholine (DPC) micelles exhibits much greater resistance to trypsin digestion, with its cytoplasmic domain taking on a significant ordered structure. FRET analysis demonstrates that, although A10Cp remains monomeric, A10TmCp forms a tight homodimer (K(d) ∼ 7 nM) in DPC micelles. Phospholipid-conjugated A10Cp dose-dependently inhibits formation of A10TmCp homodimer, whereas A10Cp achieves only limited inhibition. Placing the transmembrane and cytoplasmic domains of ADAM10, but not the transmembrane domain alone, in their native orientation in the inner membrane of Escherichia coli produces specific and strong dimerization signal in the AraC-based transcriptional reporter assay. A chimeric construct containing the otherwise monomeric transmembrane domain of L-selectin and the cytoplasmic domain of ADAM10 produces a similar dimerization signal. Overall, these results demonstrate that a transmembrane domain imparts a stable structure to the adjacent and intrinsically disordered cytoplasmic domain of ADAM10 to form a homodimer in the membrane. This finding advances our understanding of the regulatory mechanism of ADAMs and has general implications for membrane-protein interactions in the process of transmembrane signaling.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Cell Membrane/metabolism , Membrane Proteins/metabolism , Protein Multimerization/physiology , ADAM Proteins/chemistry , ADAM Proteins/genetics , ADAM10 Protein , Amyloid Precursor Protein Secretases/chemistry , Amyloid Precursor Protein Secretases/genetics , Cell Membrane/chemistry , Cell Membrane/genetics , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/physiology
14.
Bioorg Med Chem Lett ; 24(11): 2560-4, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24768446

ABSTRACT

Among many signals to regulate hypoxia inducible factor 1α (HIF-1α), sphingosine kinase 1 (SPHK1) is also involved in various biological activities such as cell growth, survival, invasion, angiogenesis, and carcinogenesis. Thus, in the present study, molecular mechanisms of coumestrol were investigated on the SPHK1 and HIF-1α signaling pathway in hypoxic PC-3 prostate cancer cells. Coumestrol significantly suppressed SPHK1 activity and accumulation of HIF-1α in a time- and concentration-dependent manner in hypoxic PC-3 cells. In addition, coumestrol inhibited the phosphorylation status of AKT and glycogen synthase kinase-3ß (GSK 3ß) signaling involved in cancer metabolism. Furthermore, SPHK1 siRNA transfection, sphigosine kinase inhibitor (SKI), reactive oxygen species (ROS) enhanced the inhibitory effect of coumestrol on the accumulation of HIF-1α and the expression of pAKT and pGSK 3ß in hypoxic PC-3 cells by combination index. Overall, our findings suggest that coumestrol suppresses the accumulation of HIF-1α via suppression of SPHK1 pathway in hypoxic PC-3 cells.


Subject(s)
Coumestrol/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Reactive Oxygen Species/antagonists & inhibitors , Cell Line, Tumor , Coumestrol/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Structure , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
15.
Blood ; 123(21): 3344-53, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24713928

ABSTRACT

Thrombotic thrombocytopenic purpura (TTP) is a life-threatening disease that presents with thrombocytopenia, disseminated thrombosis, hemolytic anemia, and organ dysfunction. The etiology of TTP has revealed that patients share a deficiency in plasma protease a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13), the enzyme responsible for cleaving ultra-large von Willebrand factor (VWF) multimers into nonthrombogenic fragments. Therefore, existing TTP mouse models were developed by targeted disruption of the ADAMTS13 gene. ADAMTS13(-/-) mice are mostly asymptomatic in the absence of a trigger, as redundant proteases appear to take on VWF processing. As an alternative approach to creating one such model, we devised a strategy based on the expression of a cleavage-resistant VWF mutant in mice. The creation of a disulfide bond within the A2 domain of VWF was found to render VWF multimers resistant to proteolysis by plasma proteases under flow. Furthermore, mice expressing the murine VWF/p.S1494C-p.A1534C mutant present with symptoms characteristics of acute TTP such as thrombocytopenia, red cell shredding, accumulation of VWF-rich thrombi in the microvasculature, and advanced TTP symptoms such as renal dysfunction and splenomegaly. Because this model appears to faithfully emulate the pathophysiology of TTP, it should prove most useful in the study of microangiopathic diseases and their treatment.


Subject(s)
Purpura, Thrombotic Thrombocytopenic/genetics , Purpura, Thrombotic Thrombocytopenic/metabolism , von Willebrand Factor/chemistry , von Willebrand Factor/genetics , Amino Acid Sequence , Animals , Disulfides/chemistry , Disulfides/metabolism , HEK293 Cells , Humans , Hydrolysis , Kidney/metabolism , Kidney/physiopathology , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Mutation , Peptide Hydrolases/metabolism , Protein Structure, Tertiary , Purpura, Thrombotic Thrombocytopenic/pathology , von Willebrand Factor/metabolism
16.
BMC Complement Altern Med ; 13: 189, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23883349

ABSTRACT

BACKGROUND: Though Mica, a thin and sheet like mineral, has been used as a mineral medicine for treatment of bleeding, dysentery and inflammation in traditional medicine including Ayurveda, the biological evidences of Mica were not clearly elucidated so far. Thus, in the present study, the antitumor mechanism of particled Mica (STB-HO) was examined in colorectal cancers. METHODS: Athymic nude mice were inoculated with HCT116 colon cancer cells and orally administered STB-HO daily for 41 days, and HCT116 and human umbilical vein endothelial cells (HUVECs) were treated with STB-HO for 0 ~ 24 hours to perform immunoblotting, cytotoxicity assay, FACs analysis and measurement of matrix metalloproteinase 9 (MMP-9) secretion and other experiments. Significant differences of all date were evaluated using Student's t-test and a Turkey-Kramer multiple-comparison post test. RESULTS: STB-HO significantly suppressed the tumor volume and weight in athymic nude mice inoculated with HCT116 cells at a dose of 100 mg/kg. Thus, the in vivo antitumor mechanism of STB-HO was to elucidated in vitro as well. STB-HO exerted cytotoxicity in HCT116, SW620 and HCT15 colorectal cancer cells. Also, STB-HO increased G1 cell population in a time and concentration dependent manner, enhanced the expression of p21, p27, p53 as cyclin dependent kinase (CDK) inhibitors, attenuated the expression of proliferating cell nuclear antigen (PCNA) and cyclin D1 and also reduced the production of vascular endothelial growth factor (VEGF) and matrix metalloproteinase 9 (MMP-9) in HCT116 cells. Consistently, STB-HO suppressed the phosphorylation of VEGFR2 in HCT116, SW620 and HCT15 cells. Also, STB-HO inhibited the VEGF mediated proliferation and also attenuated the phosphorylation of VEGFR2 and Akt in human umbilical vein endothelial cells (HUVECs). CONCLUSIONS: Collectively, these findings suggest that STB-HO has chemopreventive potential via G1 arrest and inhibition of proliferation and VEGFR2 in HCT116 colorectal cancer cells.


Subject(s)
Aluminum Silicates/pharmacology , Aluminum Silicates/therapeutic use , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , G1 Phase Cell Cycle Checkpoints/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation , Colorectal Neoplasms/metabolism , Female , HCT116 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Proto-Oncogene Proteins c-akt/metabolism , Umbilical Veins
17.
J Mol Biol ; 425(18): 3549-62, 2013 Sep 23.
Article in English | MEDLINE | ID: mdl-23796515

ABSTRACT

Moesin and calmodulin (CaM) jointly associate with the cytoplasmic domain of l-selectin in the cell to modulate the function and ectodomain shedding of l-selectin. Using fluorescence spectroscopy, we have examined the association of moesin FERM domain with the recombinant transmembrane and cytoplasmic domains of l-selectin (CLS) reconstituted in model phospholipid liposomes. The dissociation constant of moesin FERM domain to CLS in the phosphatidylcholine liposome is about 300nM. In contrast to disrupting the CaM association with CLS, inclusion of anionic phosphatidylserine lipids in the phosphatidylcholine liposome increased the apparent binding affinity of moesin FERM domain for CLS. Using the environmentally sensitive fluorescent probe attached to the cytoplasmic domain of CLS and the nitroxide quencher attached to the lipid bilayer, we showed that the association of moesin FERM domain induced the desorption of the basic-rich cytoplasmic domain of CLS from the anionic membrane surface, which enabled subsequent association of CaM to the cytoplasmic domain of CLS. These results have elucidated the molecular basis for the moesin/l-selectin/CaM ternary complex and suggested an important role of phospholipids in modulating l-selectin function and shedding.


Subject(s)
Amino Acids, Basic/chemistry , Cell Membrane/metabolism , Cytoplasm/metabolism , L-Selectin/chemistry , L-Selectin/metabolism , Microfilament Proteins/chemistry , Amino Acid Sequence , Anions/chemistry , Anions/metabolism , Calmodulin/chemistry , Calmodulin/metabolism , Cell Membrane/chemistry , Humans , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Liposomes/metabolism , Microfilament Proteins/metabolism , Microfilament Proteins/physiology , Models, Biological , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary/physiology , Spectrometry, Fluorescence
18.
Article in English | MEDLINE | ID: mdl-23589723

ABSTRACT

Here, antitumor mechanism of cinnamaldehyde derivative CB-PIC was elucidated in human SW620 colon cancer cells. CB-PIC significantly exerted cytotoxicity, increased sub-G1 accumulation, and cleaved PARP with apoptotic features, while it enhanced the phosphorylation of AMPK alpha and ACC as well as activated the ERK in hypoxic SW620 cells. Furthermore, CB-PIC suppressed the expression of HIF1 alpha, Akt, and mTOR and activated the AMPK phosphorylation in hypoxic SW620 cells. Conversely, silencing of AMPK α blocked PARP cleavage and ERK activation induced by CB-PIC, while ERK inhibitor PD 98059 attenuated the phosphorylation of AMPK α in hypoxic SW620 cells, implying cross-talk between ERK and AMPK α . Furthermore, cotreatment of CB-PIC and metformin enhanced the inhibition of HIF1 α and Akt/mTOR and the activation of AMPK α and pACC in hypoxic SW620 cells. In addition, CB-PIC suppressed the growth of SW620 cells inoculated in BALB/c athymic nude mice, and immunohistochemistry revealed that CB-PIC treatment attenuated the expression of Ki-67, CD34, and CAIX and increased the expression of pAMPK α in CB-PIC-treated group. Interestingly, CP-PIC showed better antitumor activity in SW620 colon cancer cells under hypoxia than under normoxia, since it may be applied to chemoresistance. Overall, our findings suggest that activation of AMPK α and ERK mediates CB-PIC-induced apoptosis in hypoxic SW620 colon cancer cells.

19.
FEBS Lett ; 585(20): 3289-96, 2011 Oct 20.
Article in English | MEDLINE | ID: mdl-21944868

ABSTRACT

We investigated hypocholesterolemic mechanisms of linalool, an aromatic anti-oxidative monoterpene, which is abundant in teas and essential oils. Oral administration of linalool to mice for 6 weeks significantly lowered total and low-density lipoprotein cholesterol concentrations, and HMG-CoA reductase protein expression (-46%; P<0.05) by both transcriptional and posttranscriptional mechanisms. Linalool suppressed the gene expression of HMG-CoA reductase by reducing the binding of SREBP-2 to its promoter, as assessed by qPCR and chromatin immunoprecipitation, and by inducing ubiquitin-dependent proteolysis of the HMG-CoA reductase. These findings suggest that food molecules with a pleasant scent could exert beneficial metabolic effects through multiple mechanisms.


Subject(s)
Anticholesteremic Agents/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Monoterpenes/pharmacology , Sterol Regulatory Element Binding Protein 2/metabolism , Ubiquitin/metabolism , Acyclic Monoterpenes , Animals , Cholesterol/blood , Gene Expression Regulation, Enzymologic/genetics , Hep G2 Cells , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Insecticides/pharmacology , Lipoproteins, LDL/blood , Male , Mice , Proteolysis/drug effects , Response Elements , Sterol Regulatory Element Binding Protein 2/genetics , Ubiquitin/genetics
20.
J Pineal Res ; 51(1): 87-93, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21392092

ABSTRACT

Sphingosine kinase 1 (SPHK1) is a newly discovered modulator of hypoxia inducible factor 1α (HIF-1α) with various biological activities such as cell growth, survival, invasion, angiogenesis, and carcinogenesis. Thus, in the present study, the biological mechanisms of melatonin were elucidated in association with SPHK1 pathway in PC-3 prostate cancer cells under hypoxia. Melatonin inhibited the stability of HIF-1α in a time- and concentration- dependent manners. Also, melatonin decreased SPHK1 activity in PC-3 cells during hypoxia. Furthermore, melatonin suppressed AKT/glycogen synthase kinase-3ß (GSK-3ß) signaling pathway, which stabilizes HIF-1α via inhibition of von Hippel-Lindau tumor suppressor protein. Consistently, siRNA-SPHK1 and sphingosine kinase inhibitor (SKI) effectively blocked the expression of HIF-1α, phospho-AKT and vascular endothelial growth factor (VEGF) production in PC-3 cells under hypoxia, suggesting the role of SPHK1 in melatonin-inhibited HIF-1α accumulation. Moreover, reactive oxygen species (ROS) scavenger N-acteylcysteine enhanced melatonin-inhibited HIF-1α expression and SPHK1 activity. Overall, our findings suggest that melatonin suppresses HIF-1α accumulation via inhibition of SPHK1 pathway and ROS generation in PC-3 cells under hypoxia.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Melatonin/pharmacology , Metabolic Networks and Pathways/drug effects , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Prostatic Neoplasms/metabolism , Cell Hypoxia/drug effects , Cell Line, Tumor , Gene Expression/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/pathology , Reactive Oxygen Species/metabolism , Statistics, Nonparametric , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...