Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
2.
Cancer Gene Ther ; 31(4): 599-611, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38272982

ABSTRACT

Irregularities in insulin signaling have significantly increased the risk of various cancers, yet the precise underlying mechanisms remain unclear. Within our study, we observed that inhibiting neddylation enhances cancer cell migration across different cancer types by activating both insulin receptor substrates 1 and 2 (IRS1 and IRS2), along with the PI3K/AKT signaling pathway. Notably, in the context of high-grade serous carcinoma (HGSC) patients, whether they had type 2 diabetes mellitus or not, IRS1 and IRS2 displayed a parallel relationship with each other while exhibiting an inverse relationship with NEDD8. We also identified C-CBL as an E3 ligase responsible for neddylating IRS1 and IRS2, with clinical evidence further confirming a reciprocal relationship between C-CBL and pAKT, thereby reinforcing the tumor suppressive role of C-CBL. Altogether, these findings suggest that neddylation genuinely participates in IRS1 and IRS2-dependent insulin signaling, effectively suppressing cancer cell migration. Thus, caution is advised when considering neddylation inhibitors as a treatment option for cancer patients, particularly those presenting with insulin signaling dysregulations linked to conditions like obesity-related type 2 diabetes or hyperinsulinemia.


Subject(s)
Diabetes Mellitus, Type 2 , Neoplasms , Humans , Insulin/metabolism , Receptor, Insulin/metabolism , Diabetes Mellitus, Type 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Neoplasms/genetics , Cell Movement
3.
Nat Commun ; 14(1): 6370, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37828054

ABSTRACT

Palmitic acid (PA) is the most common fatty acid in humans and mediates palmitoylation through its conversion into palmitoyl coenzyme A. Although palmitoylation affects many proteins, its pathophysiological functions are only partially understood. Here we demonstrate that PA acts as a molecular checkpoint of lipid reprogramming in HepG2 and Hep3B cells. The zinc finger DHHC-type palmitoyltransferase 23 (ZDHHC23) mediates the palmitoylation of plant homeodomain finger protein 2 (PHF2), subsequently enhancing ubiquitin-dependent degradation of PHF2. This study also reveals that PHF2 functions as a tumor suppressor by acting as an E3 ubiquitin ligase of sterol regulatory element-binding protein 1c (SREBP1c), a master transcription factor of lipogenesis. PHF2 directly destabilizes SREBP1c and reduces SREBP1c-dependent lipogenesis. Notably, SREBP1c increases free fatty acids in hepatocellular carcinoma (HCC) cells, and the consequent PA induction triggers the PHF2/SREBP1c axis. Since PA seems central to activating this axis, we suggest that levels of dietary PA should be carefully monitored in patients with HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/metabolism , Lipid Metabolism/physiology , Lipoylation , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Liver Neoplasms/metabolism , Ubiquitination , Homeodomain Proteins/metabolism
5.
Sci Rep ; 13(1): 1478, 2023 01 27.
Article in English | MEDLINE | ID: mdl-36707659

ABSTRACT

Dermal papilla cells (DPCs) play critical roles in hair follicle development, but the underlying mechanisms that contribute to hair regeneration have yet to be fully elucidated, particularly in terms of alterations in androgenetic alopecia patients. In this study, we demonstrated that hypoxia-inducible factor-1α (HIF-1α) is suppressed in scalp tissues of androgenetic alopecia patients and potentially associated with hair follicle development. Using RT-qPCR and western blot, we found that mRNA and protein levels of trichogenic genes, LEF1 and versican (VCAN), were attenuated in HIF-1α knockdown DPCs. Under an in vivo mimicked environment in a three-dimensional spheroid culture, HIF-1α-suppressed DPCs downregulated the expression of hair induction-related genes. Finally, treatment with a HIF-1α activator resulted in the elevated expression of trichogenic genes in DPCs. This study highlights the importance of dermal HIF-1α expression in regulating trichogenic genes and provides a promising therapeutic target and a fundamental tissue engineering approach for hair loss treatment.


Subject(s)
Hair Follicle , Hypoxia-Inducible Factor 1, alpha Subunit , Humans , Hair Follicle/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Alopecia/genetics , Alopecia/metabolism , Gene Expression , Hypoxia/metabolism , Cells, Cultured
6.
Acta Biomater ; 165: 168-179, 2023 Jul 15.
Article in English | MEDLINE | ID: mdl-36030051

ABSTRACT

The transplantation of pre-vascularized bone grafts is a promising strategy to improve the efficacy of engraftment and bone regeneration. We propose a hydrogel microbead-based approach for preparing vascularized and high-density tissue grafts. Mesenchymal stem cell-encapsulated collagen microgels (2 µL), termed bone beads, were prepared through spontaneous constriction, which improved the density of the mesenchymal stem cells and collagen molecules by more than 15-fold from the initial day of culture. Constriction was attributed to cell-attractive forces and involved better osteogenic differentiation of mesenchymal stem cells than that of spheroids. This approach was scalable, and ∼2000 bone beads were prepared semi-automatically using a liquid dispenser and spinner flask. The mechanical stimuli in the spinner flask further improved the osteogenic differentiation of the mesenchymal stem cells in the bone beads compared with that in static culture. Vascular endothelial cells readily attach to and cover the surface of bone beads. The in vitro assembly of the endothelial cell-enveloped bone beads resulted in microchannel formation in the interspaces between the bone beads. Significant effects of endothelialization on in vivo bone regeneration were shown in rats with cranial bone defects. The use of endothelialized bone beads may be a scalable and robust approach for treating large bone defects. STATEMENT OF SIGNIFICANCE: A unique aspect of this study is that the hMSC-encapsulated collagen microgels were prepared through spontaneous constriction, leading to the enrichment of collagen and cell density. This constriction resulted in favorable microenvironments for the osteogenic differentiation of hMSCs, which is superior to conventional spheroid culture. The microgel beads were then enveloped with vascular endothelial cells and assembled to fabricate a tissue graft with vasculature in the interspaces among the beads. The significant effects of endothelialization on in vivo bone regeneration were clearly demonstrated in rats with cranial bone defects. We believe that microgel beads covered with vascular endothelial cells provide a promising approach for engineering better tissue grafts for bone-regenerative medicine.


Subject(s)
Microgels , Regenerative Medicine , Rats , Animals , Osteogenesis , Endothelial Cells , Tissue Engineering/methods , Collagen/pharmacology , Cell Differentiation , Bone Regeneration
7.
Aesthetic Plast Surg ; 47(1): 412-422, 2023 02.
Article in English | MEDLINE | ID: mdl-35918436

ABSTRACT

BACKGROUND: Cell-assisted lipotransfer (CAL) is a novel technique for fat grafting that combines the grafting of autologous fat and adipose-derived stromal cells (ASCs) to enhance fat graft retention; however, its oncologic safety is controversial. METHODS: Herein, we investigated the oncologic safety of CAL for breast reconstruction using a murine model of residual breast cancer. Various concentrations of 4T1 cells (murine breast cancer cells) were injected into female mastectomized BALB/c mice to determine the appropriate concentration for injection. One week after injection, mice were divided into control (100 µL fat), low CAL (2.5 × 105 ASCs/100 µL fat), and high CAL (1.0 × 106 ASCs/100 µL fat) groups, and fat grafting was performed. The injection of 5.0 × 103 4T1 cells was appropriate to produce a murine model of residual breast cancer. RESULTS: The weight of the fat tumor mass was significantly higher in the high CAL group than in the other groups (p < 0.05). However, the estimated tumor weight was not significantly different between the groups. Additionally, the fat graft survival rate was significantly higher in the high CAL group than in the control and low CAL groups (p < 0.05). No significant difference was noted in the percentage of Ki-67-positive cells, suggesting that tumor proliferation was not significantly different between the groups. CONCLUSION: In summary, CAL significantly improved fat graft survival without affecting tumor size and proliferation in a murine model of residual breast cancer. These results highlight the oncologic safety of CAL for breast reconstruction. NO LEVEL ASSIGNED: This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .


Subject(s)
Mammaplasty , Neoplasms , Female , Animals , Mice , Adipose Tissue/transplantation , Disease Models, Animal , Adipocytes
8.
Pflugers Arch ; 474(6): 591-601, 2022 06.
Article in English | MEDLINE | ID: mdl-35348849

ABSTRACT

Cancer cells rewire metabolic processes to adapt to the nutrient- and oxygen-deprived tumour microenvironment, thereby promoting their proliferation and metastasis. Previous research has shown that modifying glucose metabolism, the Warburg effect, makes glycolytic cancer cells more invasive and aggressive. Lipid metabolism has also been receiving attention because lipids function as energy sources and signalling molecules. Because obesity is a risk factor for various cancer types, targeting lipid metabolism may be a promising cancer therapy. Here, we review the lipid metabolic reprogramming in cancer cells mediated by hypoxia-inducible factor-1 (HIF-1). HIF-1 is the master transcription factor for tumour growth and metastasis by transactivating genes related to proliferation, survival, angiogenesis, invasion, and metabolism. The glucose metabolic shift (the Warburg effect) is mediated by HIF-1. Recent research on HIF-1-related lipid metabolic reprogramming in cancer has confirmed that HIF-1 also modifies lipid accumulation, ß-oxidation, and lipolysis in cancer, triggering its progression. Therefore, targeting lipid metabolic alterations by HIF-1 has therapeutic potential for cancer. We summarize the role of the lipid metabolic shift mediated by HIF-1 in cancer and its putative applications for cancer therapy.


Subject(s)
Neoplasms , Tumor Microenvironment , Glycolysis , Humans , Hypoxia , Hypoxia-Inducible Factor 1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lipids , Neoplasms/metabolism
9.
J Gen Physiol ; 154(3)2022 03 07.
Article in English | MEDLINE | ID: mdl-35099502

ABSTRACT

Despite distinctive functional and anatomic differences, a precise understanding of the cardiac interventricular differences in excitation-contraction (E-C) coupling mechanisms is still lacking. Here, we directly compared rat right and left cardiomyocytes (RVCM and LVCM). Whole-cell patch clamp, the IonOptix system, and fura-2 fluorimetry were used to measure electrical properties (action potential and ionic currents), single-cell contractility, and cytosolic Ca2+ ([Ca2+]i), respectively. Myofilament proteins were analyzed by immunoblotting. RVCM showed significantly shorter action potential duration (APD) and higher density of transient outward K+ current (Ito). However, the triggered [Ca2+]i change (Ca2+ transient) was not different, while the decay rate of the Ca2+ transient was slower in RVCM. Although the relaxation speed was also slower, the sarcomere shortening amplitude (ΔSL) was smaller in RVCM. SERCA activity was ∼60% lower in RVCM, which is partly responsible for the slower decay of the Ca2+ transient. Immunoblot analysis revealed lower expression of the cardiac troponin complex (cTn) in RVCM, implying a smaller Ca2+ buffering capacity (κS), which was proved by in situ analysis. The introduction of these new levels of cTn, Ito, and SERCA into a mathematical model of rat LVCM reproduced the similar Ca2+ transient, slower Ca2+ decay, shorter APD, and smaller ΔSL of RVCM. Taken together, these data show reduced expression of cTn proteins in the RVCM, which provides an explanation for the interventricular difference in the E-C coupling kinetics.


Subject(s)
Heart Ventricles , Myocardial Contraction , Action Potentials , Animals , Calcium/metabolism , Heart Ventricles/metabolism , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Rats , Troponin/metabolism
10.
Cancer Genet ; 258-259: 151-156, 2021 11.
Article in English | MEDLINE | ID: mdl-34775260

ABSTRACT

Dysfunctional lipid metabolism is a known cause of cancer development and progression, yet little is known about the underlying molecular mechanisms that contribute to cancer progression. In this study, we demonstrate that fatty acid binding protein 5 (FABP5) is elevated in colon cancer tissue and this increased expression is linked to upregulation of the hypoxia-inducible factor-1 (HIF-1) signaling pathway. Under physiologically in vivo mimicked conditions via a polydimethylsiloxane (PDMS)-based three-dimensional (3D) culture chip, FABP5-knockdown colon cancer cells exhibited attenuated cell growth throughout the culture period. FABP5 was found to regulate HIF-1α protein levels and gene expression levels within the HIF-1α signaling pathway under hypoxic conditions. Our results provide evidence that supports the use of FABP5 as a prognostic factor in colon cancer. The FABP5/HIF-1α axis is a promising target for ameliorating fatty acid-triggered cancer progression.


Subject(s)
Colonic Neoplasms/pathology , Fatty Acid-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Apoptosis , Cell Proliferation , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Fatty Acid-Binding Proteins/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Signal Transduction , Tumor Cells, Cultured
11.
Lipids Health Dis ; 20(1): 163, 2021 Nov 14.
Article in English | MEDLINE | ID: mdl-34775964

ABSTRACT

Reprogramming of lipid metabolism has received increasing recognition as a hallmark of cancer cells because lipid dysregulation and the alteration of related enzyme profiles are closely correlated with oncogenic signals and malignant phenotypes, such as metastasis and therapeutic resistance. In this review, we describe recent findings that support the importance of lipids, as well as the transcription factors involved in cancer lipid metabolism. With recent advances in transcription factor analysis, including computer-modeling techniques, transcription factors are emerging as central players in cancer biology. Considering the limited number and the crucial role of transcription factors associated with lipid rewiring in cancers, transcription factor targeting is a promising potential strategy for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Lipid Metabolism/physiology , Neoplasms/drug therapy , Neoplasms/metabolism , Transcription Factors/metabolism , Molecular Targeted Therapy/methods , Neoplasms/immunology , Neoplasms/pathology
12.
Mol Brain ; 14(1): 106, 2021 07 03.
Article in English | MEDLINE | ID: mdl-34217333

ABSTRACT

Histone modifications are a key mechanism underlying the epigenetic regulation of gene expression, which is critically involved in the consolidation of multiple forms of memory. However, the roles of histone modifications in cerebellum-dependent motor learning and memory are not well understood. To test whether changes in histone methylation are involved in cerebellar learning, we used heterozygous Kdm3b knockout (Kdm3b+/-) mice, which show reduced lysine 9 on histone 3 (H3K9) demethylase activity. H3K9 di-methylation is significantly increased selectively in the granule cell layer of the cerebellum of Kdm3b+/- mice. In the cerebellum-dependent optokinetic response (OKR) learning, Kdm3b+/- mice show deficits in memory consolidation, whereas they are normal in basal oculomotor performance and OKR acquisition. In addition, RNA-seq analyses revealed that the expression levels of several plasticity-related genes were altered in the mutant cerebellum. Our study suggests that active regulation of histone methylation is critical for the consolidation of cerebellar motor memory.


Subject(s)
Cerebellum/physiology , Haploinsufficiency/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Memory Consolidation/physiology , Motor Activity/physiology , Animals , Gene Expression Regulation , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Lysine/metabolism , Male , Methylation , Mice, Inbred C57BL
13.
Cancers (Basel) ; 13(3)2021 Jan 30.
Article in English | MEDLINE | ID: mdl-33573293

ABSTRACT

The tumor suppressor protein p53 is frequently inactivated in human malignancies, in which it is associated with cancer aggressiveness and metastasis. Because p53 is heavily involved in epithelial-mesenchymal transition (EMT), a primary step in cell migration, p53 regulation is important for preventing cancer metastasis. p53 function can be modulated by diverse post-translational modifications including neddylation, a reversible process that conjugates NEDD8 to target proteins and inhibits the transcriptional activity of p53. However, the role of p53 in cancer migration by neddylation has not been fully elucidated. In this study, we reported that neddylation blockade induces cell migration depending on p53 status, specifically via the EMT-promoting transcription factor Slug. In cancer cell lines expressing wild type p53, neddylation blockade increased the transcriptional activity of p53 and expression of its downstream genes p21 and MDM2, eventually promoting proteasomal degradation of Slug. In the absence of p53, neddylation blockade increased cell migration by activating the PI3K/Akt/mTOR/Slug signaling axis. Because mutant p53 was transcriptionally inactivated but maintained the ability to bind to Slug, neddylation blockade did not affect the migration of cells expressing mutant p53. Our findings highlight how the p53 expression status influences neddylation-mediated cell migration in multiple cancer cell lines via Slug.

14.
Sci Rep ; 11(1): 624, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436760

ABSTRACT

Hair regenerative medicine has emerged as a promising approach for the treatment of severe hair loss. Recent advances in three-dimensional tissue engineering, such as formation of hair follicle germs (HFGs), have considerably improved hair regeneration after transplantation in animal models. Here, we proposed an approach for fabricating HFGs containing vascular endothelial cells. Epithelial, dermal papilla, and vascular endothelial cells initially formed a single aggregate, which subsequently became a dumbbell-shaped HFG, wherein the vascular endothelial cells localized in the region of dermal papilla cells. The HFGs containing vascular endothelial cells exhibited higher expression of hair morphogenesis-related genes in vitro, along with higher levels of hair shaft regeneration upon transplantation to the dorsal side of nude mice, than those without vascular endothelial cells. The generated hair follicles represented functional characteristics, such as piloerection, as well as morphological characteristics comparable to those of natural hair shafts. This approach may provide a promising strategy for fabricating tissue grafts with higher hair inductivity for hair regenerative medicine.


Subject(s)
Alopecia/therapy , Endothelium, Vascular/cytology , Hair Follicle/cytology , Regeneration , Regenerative Medicine , Stem Cells/cytology , Tissue Engineering/methods , Animals , Cells, Cultured , Female , Humans , Mice , Mice, Inbred ICR , Mice, Nude
15.
Biomaterials ; 269: 120622, 2021 02.
Article in English | MEDLINE | ID: mdl-33385686

ABSTRACT

Although obesity is a newly considered risk factor for cancer, the mechanisms by which adipocyte-derived metabolites accelerate cancer malignancy have yet to be elucidated. To identify the connection among heterogeneous cell types, conventional methods including Transwell assays or conditioned media (CM) have been used; however, these methods do not fully reflect niche effects in the tumor microenvironment (TME). Here, we established an oxygen permeable polydimethylsiloxane (PDMS)-based three-dimensional (3D) culture system to allow direct attachment between human adipocyte derived stem cells (ADSCs) and cancer cells. By doing so, a physiologically bioactive TME was created, which could be used to reveal further the relationships between different cell types. We found that co-culture of cancer cells with ADSCs resulted in a dispersion phenomenon, and the dispersed spheroid was well matched with the enhanced metastatic potential of cancer cells. Lipid profiling and in vitro migration assays suggested that lipids are the driving force for cancer cell migration via HIF-1α upregulation. In addition, the lipid/HIF-1α axis promoted tumor metastasis in a xenograft mouse model. This study presents an in vitro model of a biomimetic TME and provides new mechanistic insights into the effects of ADSC-released fatty acids on cancer cells as oncometabolites.


Subject(s)
Adipocytes , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lab-On-A-Chip Devices , Lipids , Neoplasms , Animals , Cell Line, Tumor , Cell Movement , Humans , Mice , Tumor Microenvironment
16.
Aesthet Surg J ; 41(6): NP653-NP661, 2021 05 18.
Article in English | MEDLINE | ID: mdl-33367475

ABSTRACT

BACKGROUND: The viable zone where adipocytes and/or adipose-derived stem cells survive is present at the surface of graft fat tissue; however, there is controversy regarding the zone thickness. Graft retention could be improved if more adipocytes are included in the zone. OBJECTIVES: We hypothesize that a temporary reduction in adipocyte size prior to grafting could increase the number of adipocytes in the viable zone. We reduced the adipocyte size by treatment with MLN4924, which controls lipid accumulation in adipocytes, and investigated the histological and microenvironmental changes in grafted fat. METHODS: Subcutaneous fat harvested from wild-type C57BL/6J mice was chopped into small pieces; treated with dimethyl sulfoxide (control group), 0.25 µM MLN4924, or 0.5 µM MLN4924 for 4 days; and grafted into recipient C57BL/6J mice at the supraperiosteal plane of the skull. RESULTS: The reduced adipocyte size in response to MLN4924 treatment was restored within 8 weeks after fat grafting. The MLN4924-treated groups exhibited substantially greater graft volume, lower tissue hypoxia, and higher production of M2 macrophages compared with the control group. CONCLUSIONS: Grafting with compact fat that had smaller adipocytes improved the microenvironment by modulating tissue hypoxia and macrophage polarization, leading to improved graft retention. Therefore, compact fat grafting may offer a new clinical strategy without the need for stem cell manipulation.


Subject(s)
Adipocytes , Graft Survival , Adipose Tissue , Animals , Mice , Mice, Inbred C57BL , Stem Cells
17.
Commun Biol ; 3(1): 638, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33128030

ABSTRACT

Hypoxia-inducible factor-1 alpha (HIF-1α) is a transcription factor essential for cancer cell survival. The reprogramming of lipid metabolism has emerged as a hallmark of cancer, yet the relevance of HIF-1α to this process remains elusive. In this study, we profile HIF-1α-interacting proteins using proteomics analysis and identify fatty acid-binding protein 5 (FABP5) as a critical HIF-1α-binding partner. In hepatocellular carcinoma (HCC) tissues, both FABP5 and HIF-1α are upregulated, and their expression levels are associated with poor prognosis. FABP5 enhances HIF-1α activity by promoting HIF-1α synthesis while disrupting FIH/HIF-1α interaction at the same time. Oleic-acid treatment activates the FABP5/HIF-1α axis, thereby promoting lipid accumulation and cell proliferation in HCC cells. Our results indicate that fatty-acid-induced FABP5 upregulation drives HCC progression through HIF-1-driven lipid metabolism reprogramming.


Subject(s)
Carcinoma, Hepatocellular/pathology , Fatty Acid-Binding Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/pathology , Ascorbic Acid/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytosol/metabolism , Fatty Acid-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lipid Metabolism/drug effects , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Mixed Function Oxygenases/metabolism , Oleic Acid/pharmacology , Repressor Proteins/metabolism
18.
Sci Rep ; 10(1): 18210, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33097763

ABSTRACT

Neddylation is a process by which NEDD8 is covalently conjugated to target proteins by sequential enzymatic reaction. Its role in cancer cell migration has only been recently acknowledged. Previously in cancer cell migration, the epithelial to mesenchymal transition (EMT) process has been well-known to play an important role in both invasion and metastasis by promoting mesenchymal phenotype in epithelial cells. However, the role of neddylation in the EMT process and its mechanistic details are yet to be elucidated. We recently reported that neddylation plays a crucial role in cancer cell migration through the PI3K-Akt pathway. Here, we report that inhibiting neddylation activates the hypoxia-inducible factor 1α (HIF-1α) through the PI3K-Akt pathway, which eventually regulates the EMT-activator ZEB1 (zinc finger E-box binding homeobox 1) in various cancer cell lines. As induction of HIF-1α is known to deteriorate the state of cancer and EMT process is one of the hallmarks of metastasis in cancer, our findings uncover the role of neddylation between HIF-1α and ZEB1.


Subject(s)
Cell Movement , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , NEDD8 Protein/metabolism , Neoplasms/pathology , Up-Regulation , Zinc Finger E-box-Binding Homeobox 1/metabolism , Cell Line , Epithelial-Mesenchymal Transition , Humans , Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , TOR Serine-Threonine Kinases/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics
19.
Cell Death Dis ; 11(4): 283, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32332706

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is a risk factor for progression of steatohepatitis, liver cirrhosis, and liver cancer. Although pathological condition of NAFLD, which arises from an excessive accumulation of triglyceride in the liver, is accompanied by elevated sterol regulatory element-binding protein 1c (SREBP1c) level, it is largely unknown which factors are involved in the modification of SREBP1c. In this study, we discovered that neddylation of SREBP1c competes with its ubiquitination and stabilizes SREBP1c protein level, and eventually promotes hepatic steatosis. We also demonstrated that human homolog of mouse double minute 2 (HDM2) acts as an E3 neddylation ligase of SREBP1c. Further, treatment with the neddylation inhibitor, MLN4924, attenuates high-fat diet-induced hepatic steatosis by reducing the levels of SREBP1c protein and hepatic triglyceride. Our results indicate that the blockade of SREBP1c neddylation could be a novel approach in the defense against NAFLD.


Subject(s)
NEDD8 Protein/genetics , Non-alcoholic Fatty Liver Disease/therapy , Sterol Regulatory Element Binding Protein 1/metabolism , Humans , NEDD8 Protein/metabolism , Risk Factors , Transfection , Trimeprazine
20.
Rev. cuba. invest. bioméd ; 38(3)Jul.-Sept. 2019.
Article in English | LILACS-Express | LILACS | ID: biblio-1508212

ABSTRACT

Maintaining health in microgravity and overcoming environmental hazards such as cosmic radiation are essential for long-term space flight. Recent studies have focused on the involvement of hypoxia-inducible factor (HIF)-1 in altered gravity using cell-based or in vivo mouse model systems. HIF-1alpha and its target downstream gene expression are differentially expressed in hypergravity and microgravity. Nevertheless, underlying molecular mechanism of HIF-1alpha involvement is still unclear. Herein, we analyzed the 2019 Science paper by Garrett-Bakelman and coauthors in which NASA performed multidimensional analyses of long-term human spaceflight in identical twin astronauts. Correlations were found between the expression of HIF-1alpha related cytokines and prolonged space flight. We hypothesize that HIF-1alpha is a molecular target for the development of therapeutics to prevent the detrimental effects of microgravity and cosmic radiation on astronauts during long-term space flight.


Mantener la salud en microgravedad y superar los peligros ambientales como la radiación cósmica son esenciales para los vuelos espaciales a largo plazo. Estudios recientes se han centrado en la participación del factor inducible por hipoxia (HIF) -1 en la gravedad alterada utilizando sistemas de modelos de ratones basados en células o in vivo. HIF-1alpha y su expresión génica secuencial objetivo se expresan diferencialmente en hipergravedad y microgravedad. Sin embargo, el mecanismo molecular subyacente de la participación de HIF-1alpha aún no está claro. Aquí, analizamos el artículo de Ciencia de 2019 de Garrett-Bakelman y coautores en el que la NASA realizó análisis multidimensionales de vuelos espaciales humanos a largo plazo en astronautas gemelos idénticos. Se encontraron correlaciones entre la expresión de citoquinas relacionadas con HIF-1alpha y el vuelo espacial prolongado. Presumimos que HIF-1alpha es un objetivo molecular para el desarrollo de terapias para prevenir los efectos perjudiciales de la microgravedad y la radiación cósmica en los astronautas durante los vuelos espaciales a largo plazo.

SELECTION OF CITATIONS
SEARCH DETAIL
...