1
|
Chang S, Moon R, Nam D, Lee SW, Yoon I, Lee DS, Choi S, Paek E, Hwang D, Hur JK, Nam Y, Chang R, Park H. Hypoxia increases methylated histones to prevent histone clipping and heterochromatin redistribution during Raf-induced senescence. Nucleic Acids Res 2025; 53:gkae1210. [PMID: 39660649 PMCID: PMC11797049 DOI: 10.1093/nar/gkae1210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 11/18/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024] Open
Abstract
Hypoxia enhances histone methylation by inhibiting oxygen- and α-ketoglutarate-dependent demethylases, resulting in increased methylated histones. This study reveals how hypoxia-induced methylation affects histone clipping and the reorganization of heterochromatin into senescence-associated heterochromatin foci (SAHF) during oncogene-induced senescence (OIS) in IMR90 human fibroblasts. Notably, using top-down proteomics, we discovered specific cleavage sites targeted by Cathepsin L (CTSL) in H3, H2B and H4 during Raf activation, identifying novel sites in H2B and H4. Hypoxia counteracts CTSL-mediated histone clipping by promoting methylation without affecting CTSL's activity. This increase in methylation under hypoxia protects against clipping, reshaping the epigenetic landscape and influencing chromatin accessibility, as shown by ATAC-seq analysis. These insights underscore the pivotal role of hypoxia-induced histone methylation in protecting chromatin from significant epigenetic shifts during cellular aging.
Collapse
Affiliation(s)
- Soojeong Chang
- Department of Life Science, University of Seoul, Seoul 02504, Republic of Korea
| | - Ramhee Moon
- Department of Life Science, University of Seoul, Seoul 02504, Republic of Korea
| | - Dowoon Nam
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Sang-Won Lee
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Insoo Yoon
- Department of Life Science, University of Seoul, Seoul 02504, Republic of Korea
| | - Dong-Sung Lee
- Department of Life Science, University of Seoul, Seoul 02504, Republic of Korea
| | - Seunghyuk Choi
- Department of Computer Science, Hanyang University, Seoul 04763, Republic of Korea
| | - Eunok Paek
- Department of Computer Science, Hanyang University, Seoul 04763, Republic of Korea
| | - Daehee Hwang
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Junho K Hur
- Department of Genetics, College of Medicine, Hanyang University, Seoul 04763, Republic of Korea
| | - Youhyun Nam
- Department of Applied Chemistry, University of Seoul, Seoul 02504, Republic of Korea
| | - Rakwoo Chang
- Department of Applied Chemistry, University of Seoul, Seoul 02504, Republic of Korea
| | - Hyunsung Park
- Department of Life Science, University of Seoul, Seoul 02504, Republic of Korea
- Department of Applied Chemistry, University of Seoul, Seoul 02504, Republic of Korea
| |
Collapse
|
2
|
Pourdashti S, Faridi N, Monem-Homaie F, Yaghooti SH, Soroush A, Bathaie SZ. The size of human subcutaneous adipocytes, but not adiposity, is associated with inflammation, endoplasmic reticulum stress, and insulin resistance markers. Mol Biol Rep 2023; 50:5755-5765. [PMID: 37219669 PMCID: PMC10289932 DOI: 10.1007/s11033-023-08460-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/13/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND The fat storage capacity of the adipose tissue prevents ectopic lipid deposition, which is one of the risk factors for metabolic abnormalities in obesity. This capacity depends upon the adipogenic gene expression and blood supply provision for tissue expansion through angiogenesis. Here, we studied hyperplasia/hypertrophy of subcutaneous white adipose tissue (scWAT) concerning adipogenic gene expression, angiogenic status, and metabolic parameters in non-obese and different classes of obese individuals. METHODS The scWAT samples were collected from 80 individuals. The anthropometric parameters, adipose tissue cell size, serum biochemistry, ER stress-induced XBP1 splicing, PPARγ2, SFRP1, WNT10B, and VEGFA gene expression levels were studied. In addition, the CD31 level was investigated by Western blotting. RESULTS The obese individuals had greater waist circumferences and higher serum TG, TC, insulin, and HOMA-IR than the non-obese group. However, the largest adipocyte size, increased TNFα, insulin, and HOMA-IR, and the highest expression level of sXBP1, WNT10B, and VEGFA were observed in Class I obese individuals. It means that inflammation, insulin resistance, and ER stress accompany hypertrophic scWAT adipocytes with limited adipose tissue expansion ability. Furthermore, the Class II + III obese individuals showed high PPARγ2 expression and CD31 levels. There is adipogenesis through hyperplasia in this group. The SFRP1 expression was not significantly different in the studied groups. CONCLUSION The results suggest that the capability of adipogenesis with inadequate angiogenesis is related to the metabolic status, inflammation, and ER function. Therefore, therapeutic strategies that support both angiogenesis and adipogenesis can effectively prevent the complications of obesity.
Collapse
Affiliation(s)
- Sara Pourdashti
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University (TMU), P.O. Box: 14155-331, Tehran, Iran
| | - Nassim Faridi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University (TMU), P.O. Box: 14155-331, Tehran, Iran
| | - Forouzandeh Monem-Homaie
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University (TMU), P.O. Box: 14155-331, Tehran, Iran
| | - S Hamid Yaghooti
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University (TMU), P.O. Box: 14155-331, Tehran, Iran
| | - Ahmadreza Soroush
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular- Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - S Zahra Bathaie
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University (TMU), P.O. Box: 14155-331, Tehran, Iran.
- UCLA-DOE Institute, University of California, Los Angeles (UCLA), CA, USA.
| |
Collapse
|
3
|
Perkins RS, Singh R, Abell AN, Krum SA, Miranda-Carboni GA. The role of WNT10B in physiology and disease: A 10-year update. Front Cell Dev Biol 2023; 11:1120365. [PMID: 36814601 PMCID: PMC9939717 DOI: 10.3389/fcell.2023.1120365] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/16/2023] [Indexed: 02/09/2023] Open
Abstract
WNT10B, a member of the WNT family of secreted glycoproteins, activates the WNT/β-catenin signaling cascade to control proliferation, stemness, pluripotency, and cell fate decisions. WNT10B plays roles in many tissues, including bone, adipocytes, skin, hair, muscle, placenta, and the immune system. Aberrant WNT10B signaling leads to several diseases, such as osteoporosis, obesity, split-hand/foot malformation (SHFM), fibrosis, dental anomalies, and cancer. We reviewed WNT10B a decade ago, and here we provide a comprehensive update to the field. Novel research on WNT10B has expanded to many more tissues and diseases. WNT10B polymorphisms and mutations correlate with many phenotypes, including bone mineral density, obesity, pig litter size, dog elbow dysplasia, and cow body size. In addition, the field has focused on the regulation of WNT10B using upstream mediators, such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs). We also discussed the therapeutic implications of WNT10B regulation. In summary, research conducted during 2012-2022 revealed several new, diverse functions in the role of WNT10B in physiology and disease.
Collapse
Affiliation(s)
- Rachel S. Perkins
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Rishika Singh
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Amy N. Abell
- Department of Biological Sciences, University of Memphis, Memphis, TN, United States
| | - Susan A. Krum
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, TN, United States,Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Gustavo A. Miranda-Carboni
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, United States,Department of Medicine, Division of Hematology and Oncology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States,*Correspondence: Gustavo A. Miranda-Carboni,
| |
Collapse
|
4
|
Martini T, Naef F, Tchorz JS. Spatiotemporal Metabolic Liver Zonation and Consequences on Pathophysiology. ANNUAL REVIEW OF PATHOLOGY 2023; 18:439-466. [PMID: 36693201 DOI: 10.1146/annurev-pathmechdis-031521-024831] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Hepatocytes are the main workers in the hepatic factory, managing metabolism of nutrients and xenobiotics, production and recycling of proteins, and glucose and lipid homeostasis. Division of labor between hepatocytes is critical to coordinate complex complementary or opposing multistep processes, similar to distributed tasks at an assembly line. This so-called metabolic zonation has both spatial and temporal components. Spatial distribution of metabolic function in hepatocytes of different lobular zones is necessary to perform complex sequential multistep metabolic processes and to assign metabolic tasks to the right environment. Moreover, temporal control of metabolic processes is critical to align required metabolic processes to the feeding and fasting cycles. Disruption of this complex spatiotemporal hepatic organization impairs key metabolic processes with both local and systemic consequences. Many metabolic diseases, such as nonalcoholic steatohepatitis and diabetes, are associated with impaired metabolic liver zonation. Recent technological advances shed new light on the spatiotemporal gene expression networks controlling liver function and how their deregulation may be involved in a large variety of diseases. We summarize the current knowledge about spatiotemporal metabolic liver zonation and consequences on liver pathobiology.
Collapse
Affiliation(s)
- Tomaz Martini
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland;
| | - Felix Naef
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland;
| | - Jan S Tchorz
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland;
| |
Collapse
|
5
|
Roumiguié M, Estève D, Manceau C, Toulet A, Gilleron J, Belles C, Jia Y, Houël C, Pericart S, LeGonidec S, Valet P, Cormont M, Tanti JF, Malavaud B, Bouloumié A, Milhas D, Muller C. Periprostatic Adipose Tissue Displays a Chronic Hypoxic State that Limits Its Expandability. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:926-942. [PMID: 35358473 DOI: 10.1016/j.ajpath.2022.03.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/15/2022] [Accepted: 03/08/2022] [Indexed: 06/14/2023]
Abstract
White adipose tissue accumulates at various sites throughout the body, some adipose tissue depots exist near organs whose function they influence in a paracrine manner. Prostate gland is surrounded by a poorly characterized adipose depot called periprostatic adipose tissue (PPAT), which plays emerging roles in prostate-related disorders. Unlike all other adipose depots, PPAT secretes proinflammatory cytokines even in lean individuals and does not increase in volume during obesity. These unique features remain unexplained because of the poor structural and functional characterization of this tissue. This study characterized the structural organization of PPAT in patients compared with abdominopelvic adipose tissue (APAT), an extraperitoneal adipose depot, the accumulation of which is correlated to body mass index. Confocal microscopy followed by three-dimensional reconstructions showed a sparse vascular network in PPAT when compared with that in APAT, suggesting that this tissue is hypoxic. Unbiased comparisons of PPAT and APAT transcriptomes found that most differentially expressed genes were related to the hypoxia response. High levels of the hypoxia-inducible factor 2α confirmed the presence of an adaptive response to hypoxia in PPAT. This chronic hypoxic state was associated with inflammation and fibrosis, which were not further up-regulated by obesity. This fibrosis and inflammation explain the failure of PPAT to expand in obesity and open new mechanistic avenues to explain its role in prostate-related disorders, including cancer.
Collapse
Affiliation(s)
- Mathieu Roumiguié
- Institut de Pharmacologie et Biologie Structurale (Equipe Labélisée Ligue Nationale contre le Cancer), Université de Toulouse, Centre national de la recherche scientifique, Toulouse, France; Département d'Urologie, Institut Universitaire du Cancer, Toulouse, France
| | - David Estève
- Institut de Pharmacologie et Biologie Structurale (Equipe Labélisée Ligue Nationale contre le Cancer), Université de Toulouse, Centre national de la recherche scientifique, Toulouse, France
| | - Cécile Manceau
- Institut de Pharmacologie et Biologie Structurale (Equipe Labélisée Ligue Nationale contre le Cancer), Université de Toulouse, Centre national de la recherche scientifique, Toulouse, France; Département d'Urologie, Institut Universitaire du Cancer, Toulouse, France
| | - Aurélie Toulet
- Institut de Pharmacologie et Biologie Structurale (Equipe Labélisée Ligue Nationale contre le Cancer), Université de Toulouse, Centre national de la recherche scientifique, Toulouse, France
| | - Jérôme Gilleron
- Université Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire, Team Cellular and Molecular Pathophysiology of Obesity, Nice, France
| | - Chloé Belles
- Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, INSERM, Toulouse, France
| | - Yiyue Jia
- Institut de Pharmacologie et Biologie Structurale (Equipe Labélisée Ligue Nationale contre le Cancer), Université de Toulouse, Centre national de la recherche scientifique, Toulouse, France
| | - Cynthia Houël
- Institut de Pharmacologie et Biologie Structurale (Equipe Labélisée Ligue Nationale contre le Cancer), Université de Toulouse, Centre national de la recherche scientifique, Toulouse, France
| | - Sarah Pericart
- Département d'Anatomo-Pathologie, Institut Universitaire du Cancer, Toulouse, France
| | - Sophie LeGonidec
- Institut RESTORE, Université de Toulouse, Centre national de la recherche scientifique U-5070, Etablissement Français du Sang, Ecole Nationale Vétérinaire de Toulouse, INSERM U1301, Toulouse, France
| | - Philippe Valet
- Institut RESTORE, Université de Toulouse, Centre national de la recherche scientifique U-5070, Etablissement Français du Sang, Ecole Nationale Vétérinaire de Toulouse, INSERM U1301, Toulouse, France
| | - Mireille Cormont
- Université Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire, Team Cellular and Molecular Pathophysiology of Obesity, Nice, France
| | - Jean-François Tanti
- Université Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire, Team Cellular and Molecular Pathophysiology of Obesity, Nice, France
| | - Bernard Malavaud
- Département d'Urologie, Institut Universitaire du Cancer, Toulouse, France
| | - Anne Bouloumié
- Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, INSERM, Toulouse, France
| | - Delphine Milhas
- Institut de Pharmacologie et Biologie Structurale (Equipe Labélisée Ligue Nationale contre le Cancer), Université de Toulouse, Centre national de la recherche scientifique, Toulouse, France.
| | - Catherine Muller
- Institut de Pharmacologie et Biologie Structurale (Equipe Labélisée Ligue Nationale contre le Cancer), Université de Toulouse, Centre national de la recherche scientifique, Toulouse, France.
| |
Collapse
|
6
|
YAP-dependent Wnt5a induction in hypertrophic adipocytes restrains adiposity. Cell Death Dis 2022; 13:407. [PMID: 35478181 PMCID: PMC9046197 DOI: 10.1038/s41419-022-04847-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 02/02/2022] [Accepted: 04/07/2022] [Indexed: 12/22/2022]
Abstract
Wnt5a, a prototypic non-canonical Wnt, is an inflammatory factor elevated in the sera of obese humans and mice. In the present study, fat-specific knockout of Wnt5a (Wnt5a-FKO) prevented HFD-induced increases in serum Wnt5a levels in male C57BL/6 J mice, which suggested adipocytes are primarily responsible for obesity-induced increases in Wnt5a levels. Mouse subcutaneous white adipose tissues (WATs) more sensitively responded to HFD, in terms of cell size increases and Wnt5a levels than epididymal WATs. Furthermore, adipocyte sizes were positively correlated with Wnt5a levels in vitro and in vivo. In hypertrophic adipocytes, enlarged lipid droplets increased cell stiffness and rearranged the f-actin stress fibers from the cytoplasm to the cortical region. The activities of YAP (Yes-associated protein) and TAZ (transcriptional co-activator with PDZ-binding motif) increased in response to these mechanical changes in hypertrophic adipocytes, and inhibition or knock-down of YAP and TAZ reduced Wnt5a expression. ChIP (chromatin immunoprecipitation) analyses revealed that YAP was recruited by Wnt5a-1 gene promoter and increased Wnt5a expression. These results suggested that YAP responds to mechanical stress in hypertrophic adipocytes to induce the expression Wnt5a. When 8-week-old Wnt5a-FKO mice were fed an HFD for 20 weeks, the fat mass increased, especially in subcutaneous WATs, as compared with that observed in floxed mice, without significant changes in food intake or activity. Furthermore, Wnt5a-FKO mice showed impaired glucose tolerance regardless of diet type. Our findings show that hypertrophy/YAP/Wnt5a signaling constitutes a negative-feedback loop that retrains adipose tissue hypertrophy.
Collapse
|
7
|
Pourdashti S, Faridi N, Yaghooti H, Jalali MT, Soroush A, Bathaie SZ. Possible role of WNT10B in increased proliferation and tubule formation of human umbilical vein endothelial cell cultures treated with hypoxic conditioned medium from human adipocytes. Biotech Histochem 2021; 97:168-179. [PMID: 34044678 DOI: 10.1080/10520295.2021.1923801] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Regulation of angiogenesis plays an important role in adipose tissue expansion and function. The Wnt pathway and WNT10B, the main member of Wnt family, participate in angiogenesis in cancer tumors, but there is limited evidence to support the regulatory role of WNT10B in human adipose tissue angiogenesis. Subcutaneous white adipose tissue (scWAT) of 80 participants including obese and non-obese subjects was obtained and the expression of WNT10B and VEGFA genes were evaluated using qPCR. Human adipose-derived stem cells (hADSC) were differentiated to adipocytes and incubated under either hypoxic or normoxic conditions. The conditioned media of these adipocytes were collected and used as growth media for human umbilical vein endothelial cells (HUVEC) in Matrigel. We evaluated the proliferation, cell cycle phases, tubule formation and β-catenin activation of these treated cells. We found a significant correlation between WNT10B and VEGFA expression in the scWAT of both obese and non-obese subjects. Proliferation and tubule formation of HUVEC treated with conditioned media of hypoxic adipocytes (hCM) in the S-phase were increased significantly compared to the HUVEC treated with the conditioned media of normoxic adipocytes (nCM). The expression of WNT10B and VEGFA was enhanced in hypoxic adipocytes compared to normoxic adipocytes; also, activation and nuclear translocation of β-catenin was enhanced in the HUVEC treated with hCM compared to nCM. WNT10B acts as an angiogenic protein in scWAT under hypoxic conditions. Hypoxia induced WNT10B increases VEGFA expression and causes tube formation by HUVECs and angiogenesis in adipose tissue via the canonical Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Sara Pourdashti
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University (TMU), Tehran, Iran
| | - Nassim Faridi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University (TMU), Tehran, Iran
| | - Hamid Yaghooti
- Cellular and Molecular Research Center and Hyperlipidemia Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad-Taha Jalali
- Hyperlipidemia Research Center and Diabetes Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
| | - Ahmadreza Soroush
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - S Zahra Bathaie
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University (TMU), Tehran, Iran
| |
Collapse
|
8
|
Moon Y, Moon R, Roh H, Chang S, Lee S, Park H. HIF-1α-Dependent Induction of Carboxypeptidase A4 and Carboxypeptidase E in Hypoxic Human Adipose-Derived Stem Cells. Mol Cells 2020; 43:945-952. [PMID: 33203807 PMCID: PMC7700838 DOI: 10.14348/molcells.2020.0100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 08/22/2020] [Accepted: 10/29/2020] [Indexed: 12/29/2022] Open
Abstract
Hypoxia induces the expression of several genes through the activation of a master transcription factor, hypoxia-inducible factor (HIF)-1α. This study shows that hypoxia strongly induced the expression of two carboxypeptidases (CP), CPA4 and CPE, in an HIF-1α-dependent manner. The hypoxic induction of CPA4 and CPE gene was accompanied by the recruitment of HIF-1α and upregulation in the active histone modification, H3K4me3, at their promoter regions. The hypoxic responsiveness of CPA4 and CPE genes was observed in human adipocytes, human adipose-derived stem cells, and human primary fibroblasts but not mouse primary adipocyte progenitor cells. CPA4 and CPE have been identified as secreted exopeptidases that degrade and process other secreted proteins and matrix proteins. This finding suggests that hypoxia changes the microenvironment of the obese hypoxic adipose tissue by inducing the expression of not only adipokines but also peptidases such as CPA4 and CPE.
Collapse
Affiliation(s)
- Yunwon Moon
- Department of Life Science, University of Seoul, Seoul 02504, Korea
- Present address: Cell Therapy Research Center, GC LabCell, Yongin 16924, Korea
- These authors contributed equally to this work
| | - Ramhee Moon
- Department of Life Science, University of Seoul, Seoul 02504, Korea
- These authors contributed equally to this work
| | - Hyunsoo Roh
- Department of Life Science, University of Seoul, Seoul 02504, Korea
| | - Soojeong Chang
- Department of Life Science, University of Seoul, Seoul 02504, Korea
| | - Seongyeol Lee
- Department of Life Science, University of Seoul, Seoul 02504, Korea
| | - Hyunsung Park
- Department of Life Science, University of Seoul, Seoul 02504, Korea
| |
Collapse
|
9
|
Moon Y, Kim I, Chang S, Park B, Lee S, Yoo S, Chae S, Hwang D, Park H. Hypoxia regulates allele-specific histone modification of the imprinted H19 gene. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2020; 1863:194643. [DOI: 10.1016/j.bbagrm.2020.194643] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/29/2020] [Accepted: 10/02/2020] [Indexed: 01/20/2023]
|
10
|
Abstract
Obesity is characterized by increased adipose tissue mass and has been associated with a strong predisposition towards metabolic diseases and cancer. Thus, it constitutes a public health issue of major proportion. The expansion of adipose depots can be driven either by the increase in adipocyte size (hypertrophy) or by the formation of new adipocytes from precursor differentiation in the process of adipogenesis (hyperplasia). Notably, adipocyte expansion through adipogenesis can offset the negative metabolic effects of obesity, and the mechanisms and regulators of this adaptive process are now emerging. Over the past several years, we have learned a considerable amount about how adipocyte fate is determined and how adipogenesis is regulated by signalling and systemic factors. We have also gained appreciation that the adipogenic niche can influence tissue adipogenic capability. Approaches aimed at increasing adipogenesis over adipocyte hypertrophy can now be explored as a means to treat metabolic diseases.
Collapse
|
11
|
Huang X, Fu C, Liu W, Liang Y, Li P, Liu Z, Sheng Q, Liu P. Chemerin-induced angiogenesis and adipogenesis in 3 T3-L1 preadipocytes is mediated by lncRNA Meg3 through regulating Dickkopf-3 by sponging miR-217. Toxicol Appl Pharmacol 2019; 385:114815. [DOI: 10.1016/j.taap.2019.114815] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/29/2019] [Accepted: 11/08/2019] [Indexed: 01/06/2023]
|
12
|
Efficiently Anti-Obesity Effects of Unsaturated Alginate Oligosaccharides (UAOS) in High-Fat Diet (HFD)-Fed Mice. Mar Drugs 2019; 17:md17090540. [PMID: 31533255 PMCID: PMC6780860 DOI: 10.3390/md17090540] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 12/15/2022] Open
Abstract
Obesity and its related complications have become one of the leading problems affecting human health. However, current anti-obesity treatments are limited by high cost and numerous adverse effects. In this study, we investigated the use of a non-toxic green food additive, known as unsaturated alginate oligosaccharides (UAOS) from the enzymatic degradation of Laminaria japonicais, which showed effective anti-obesity effects in a high-fat diet (HFD) mouse model. Compared with acid hydrolyzed saturated alginate oligosaccharides (SAOS), UAOS significantly reduced body weight, serum lipid, including triacylglycerol (TG), total cholesterol (TC) and free fatty acids (FFA), liver weight, liver TG and TC, serum alanine aminotransferase (ALT), and aspartate aminotransferase (AST) levels, adipose mass, reactive oxygen species (ROS) formation, and accumulation induced in HFD mice. Moreover, the structural differences in β-d-mannuronate (M) and its C5 epimer α-l-guluronate (G) did not cause significant functional differences. Meanwhile, UAOS significantly increased both AMP-activated protein kinase α (AMPKα) and acetyl-CoA carboxylase (ACC) phosphorylation in adipocytes, which indicated that UAOS had an anti-obesity effect mainly through AMPK signaling. Our results indicate that UAOS has the potential for further development as an adjuvant treatment for many metabolic diseases such as fatty liver, hypertriglyceridemia, and possibly diabetes.
Collapse
|
13
|
Wnt3a disrupts GR-TEAD4-PPARγ2 positive circuits and cytoskeletal rearrangement in a β-catenin-dependent manner during early adipogenesis. Cell Death Dis 2019; 10:16. [PMID: 30622240 PMCID: PMC6325140 DOI: 10.1038/s41419-018-1249-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/25/2018] [Accepted: 11/30/2018] [Indexed: 11/09/2022]
Abstract
Adipogenesis is a process which induces or represses many genes in a way to drive irreversible changes of cell phenotypes; lipid accumulation, round cell-shape, secreting many adipokines. As a master transcription factor (TF), PPARγ2 induces several target genes to orchestrate these adipogenic changes. Thus induction of Pparg2 gene is tightly regulated by many adipogenic and also anti-adipogenic factors. Four hours after the treatment of adipogenic hormones, more than fifteen TFs including glucocorticoid receptor (GR), C/EBPβ and AP-1 cooperatively bind the promoter of Pparg2 gene covering 400 bps, termed "hotspot". In this study, we show that TEA domain family transcription factor (TEAD)4 reinforces occupancy of both GR and C/EBPβ on the hotspot of Pparg2 during early adipogenesis. Our findings that TEAD4 requires GR for its expression and for the ability to bind its own promoter and the hotspot region of Pparg2 gene indicate that GR is a common component of two positive circuits, which regulates the expression of both Tead4 and Pparg2. Wnt3a disrupts these mutually related positive circuits by limiting the nuclear location of GR in a β-catenin dependent manner. The antagonistic effects of β-catenin extend to cytoskeletal remodeling during the early phase of adipogenesis. GR is necessary for the rearrangements of both cytoskeleton and chromatin of Pparg2, whereas Wnt3a inhibits both processes in a β-catenin-dependent manner. Our results suggest that hotspot formation during early adipogenesis is related to cytoskeletal remodeling, which is regulated by the antagonistic action of GR and β-catenin, and that Wnt3a reinforces β-catenin function.
Collapse
|
14
|
Sun S, Zhou L, Yu Y, Zhang T, Wang M. Knocking down clock control gene CRY1 decreases adipogenesis via canonical Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun 2018; 506:746-753. [DOI: 10.1016/j.bbrc.2018.10.134] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 10/22/2018] [Indexed: 12/29/2022]
|
15
|
Yan Y, Liu F, Han L, Zhao L, Chen J, Olopade OI, He M, Wei M. HIF-2α promotes conversion to a stem cell phenotype and induces chemoresistance in breast cancer cells by activating Wnt and Notch pathways. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:256. [PMID: 30340507 PMCID: PMC6194720 DOI: 10.1186/s13046-018-0925-x] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/02/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Hypoxic tumor microenvironment and maintenance of stemness contribute to drug resistance in breast cancer. However, whether Hypoxia-inducible factor-2α (HIF-2α) in hypoxic tumor microenvironment mediates conversion to a stem cell phenotype and chemoresistance of breast tumors has not been elucidated. METHODS The mRNA and protein expressions of HIF-1α, HIF-2α, Wnt and Notch pathway were determined using qRT-PCR and western blot. Cell viability and renew ability were assessed by MTT, Flow cytometric analysis and soft agar colony formation. RESULTS In our study, acute hypoxia (6-12 h) briefly increased HIF-1α expression, while chronic hypoxia (48 h) continuously enhanced HIF-2α expression and induced the resistance of breast cancer cells to Paclitaxel (PTX). Furthermore, HIF-2α overexpression induced a stem cell phenotype, the resistance to PTX and enhanced protein expression of stem cell markers, c-Myc, OCT4 and Nanog. Most importantly, Wnt and Notch signaling, but not including Shh, pathways were both activated by HIF-2α overexpression. Dickkopf-1 (DKK-1), a Wnt pathway inhibitor, and L685,458, an inhibitor of the Notch pathway, reversed the resistance to PTX and stem phenotype conversion induced by HIF-2α overexpression. In addition, HIF-2α overexpression enhanced tumorigenicity and resistance of xenograft tumors to PTX, increased activation of the Wnt and Notch pathways and induced a stem cell phenotype in vivo. CONCLUSION In conclusion, HIF-2α promoted stem phenotype conversion and induced resistance to PTX by activating Wnt and Notch pathways.
Collapse
Affiliation(s)
- Yuanyuan Yan
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang North Area, Shenyang City, 110122, Liaoning, China
| | - Fangxiao Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang North Area, Shenyang City, 110122, Liaoning, China
| | - Li Han
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang North Area, Shenyang City, 110122, Liaoning, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang North Area, Shenyang City, 110122, Liaoning, China
| | - Jianjun Chen
- Department of Systems Biology, City of hope, Los Angeles, CA, USA
| | - Olufunmilayo I Olopade
- Center for Clinical Cancer Genetics, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China. .,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang North Area, Shenyang City, 110122, Liaoning, China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China. .,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang North Area, Shenyang City, 110122, Liaoning, China.
| |
Collapse
|
16
|
Fainberg HP, Birtwistle M, Alagal R, Alhaddad A, Pope M, Davies G, Woods R, Castellanos M, May ST, Ortori CA, Barrett DA, Perry V, Wiens F, Stahl B, van der Beek E, Sacks H, Budge H, Symonds ME. Transcriptional analysis of adipose tissue during development reveals depot-specific responsiveness to maternal dietary supplementation. Sci Rep 2018; 8:9628. [PMID: 29941966 PMCID: PMC6018169 DOI: 10.1038/s41598-018-27376-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 05/30/2018] [Indexed: 01/23/2023] Open
Abstract
Brown adipose tissue (BAT) undergoes pronounced changes after birth coincident with the loss of the BAT-specific uncoupling protein (UCP)1 and rapid fat growth. The extent to which this adaptation may vary between anatomical locations remains unknown, or whether the process is sensitive to maternal dietary supplementation. We, therefore, conducted a data mining based study on the major fat depots (i.e. epicardial, perirenal, sternal (which possess UCP1 at 7 days), subcutaneous and omental) (that do not possess UCP1) of young sheep during the first month of life. Initially we determined what effect adding 3% canola oil to the maternal diet has on mitochondrial protein abundance in those depots which possessed UCP1. This demonstrated that maternal dietary supplementation delayed the loss of mitochondrial proteins, with the amount of cytochrome C actually being increased. Using machine learning algorithms followed by weighted gene co-expression network analysis, we demonstrated that each depot could be segregated into a unique and concise set of modules containing co-expressed genes involved in adipose function. Finally using lipidomic analysis following the maternal dietary intervention, we confirmed the perirenal depot to be most responsive. These insights point at new research avenues for examining interventions to modulate fat development in early life.
Collapse
Affiliation(s)
- Hernan P Fainberg
- Division of Child Health, Obstetrics & Gynaecology, The University of Nottingham, Nottingham, United Kingdom
| | - Mark Birtwistle
- Division of Child Health, Obstetrics & Gynaecology, The University of Nottingham, Nottingham, United Kingdom
| | - Reham Alagal
- Division of Child Health, Obstetrics & Gynaecology, The University of Nottingham, Nottingham, United Kingdom.,Princess Nourah Bint Abdulrahman University, Department of Nutrition and food science, College of Home Economics, Riyadh, BOX: 84428, Saudi Arabia
| | - Ahmad Alhaddad
- Division of Child Health, Obstetrics & Gynaecology, The University of Nottingham, Nottingham, United Kingdom
| | - Mark Pope
- Division of Child Health, Obstetrics & Gynaecology, The University of Nottingham, Nottingham, United Kingdom
| | - Graeme Davies
- Division of Child Health, Obstetrics & Gynaecology, The University of Nottingham, Nottingham, United Kingdom
| | - Rachel Woods
- Division of Child Health, Obstetrics & Gynaecology, The University of Nottingham, Nottingham, United Kingdom
| | - Marcos Castellanos
- Nottingham Arabidopsis Stock Centre, School of Biosciences, The University of Nottingham, Nottingham, United Kingdom
| | - Sean T May
- Nottingham Arabidopsis Stock Centre, School of Biosciences, The University of Nottingham, Nottingham, United Kingdom
| | - Catharine A Ortori
- Centre for Analytical Bioscience, School of Pharmacy, The University of Nottingham, Nottingham, United Kingdom
| | - David A Barrett
- Centre for Analytical Bioscience, School of Pharmacy, The University of Nottingham, Nottingham, United Kingdom
| | - Viv Perry
- Robinson Research Institute, Medical School, University of Adelaide, Adelaide, Australia
| | | | | | - Eline van der Beek
- Nutricia Research, Utrecht, The Netherlands.,Department of Pediatrics, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Harold Sacks
- VA Endocrinology and Diabetes Division, VA Greater Los Angeles Healthcare System, and Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, California, USA
| | - Helen Budge
- Division of Child Health, Obstetrics & Gynaecology, The University of Nottingham, Nottingham, United Kingdom
| | - Michael E Symonds
- Division of Child Health, Obstetrics & Gynaecology, The University of Nottingham, Nottingham, United Kingdom. .,Nottingham Digestive Disease Centre and Biomedical Research Centre, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham, United Kingdom.
| |
Collapse
|
17
|
Distinct hypoxic regulation of preadipocyte factor-1 (Pref-1) in preadipocytes and mature adipocytes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:334-342. [DOI: 10.1016/j.bbamcr.2017.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/15/2017] [Accepted: 11/10/2017] [Indexed: 01/08/2023]
|
18
|
Zhang L, Qiu B, Wang T, Wang J, Liu M, Xu Y, Wang C, Deng R, Williams K, Yang Z, Liang T, Yong W. Loss of FKBP5 impedes adipocyte differentiation under both normoxia and hypoxic stress. Biochem Biophys Res Commun 2017; 485:761-767. [PMID: 28254433 DOI: 10.1016/j.bbrc.2017.02.126] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 02/25/2017] [Indexed: 12/17/2022]
Abstract
FK506-binding protein 51 (FKBP51) is one of the most important regulators in the GR-mediated stress response, and we previously demonstrated that loss of FKBP5 arrests adipogenesis and renders mice resistant to diet-induced obesity (DIO). However, the exact role of FKBP5 in the process of adipocyte differentiation under hypoxic conditions (the common microenvironment where adipocytes reside in obese individuals) is still unclear. Here, by isolating and culturing WT- and Fkbp5-knockout mouse embryonic fibroblasts (MEFs), and treat them at normal oxygen environment (21% O2, nomorxia) or low oxygen environment (5% O2, hypoxia). Enhanced adipogenesis were observed at hypoxia when compared to normal oxygen environment. The loss of FKBP5 significantly prevents the adipogenesis from KO MEFs under nomorxia condition, with subtle enhancement of adipogenesis at hypoxia condition, which is similar as observed in WT-MEFs at hypoxia condition but with obvious enhancement of adipogenesis. Importantly, the protein level of FKBP5 reduced in undifferentiated MEFs under acute hypoxic stress (24 h), but drastically increased during the mid-late stage of adipocyte (Day 6) differentiation from WT-MEFs under chronic hypoxia. Furthermore, we find under normal and hypoxic conditions that FKBP5 deletion alters the expression profile of adipogenesis-related genes, including those involved in lipogenesis, lipolysis, and energy metabolism, which partially explains the compromised adipocyte differentiation in FKBP51-KO MEFs. Taken together, our findings identify a novel role of FKBP5 in hypoxia-regulated adipogenesis, and provide a candidate for anti-obesity strategies targeting FKBP51.
Collapse
Affiliation(s)
- Lingling Zhang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Bin Qiu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Tingting Wang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Jun Wang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Ming Liu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Yuxue Xu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Chao Wang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Ran Deng
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Kent Williams
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Zhiwei Yang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Tiebing Liang
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Weidong Yong
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
19
|
Long-chain bases from Cucumaria frondosa inhibit adipogenesis and regulate lipid metabolism in 3T3-L1 adipocytes. Food Sci Biotechnol 2016; 25:1753-1760. [PMID: 30263471 DOI: 10.1007/s10068-016-0267-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/29/2016] [Accepted: 10/16/2016] [Indexed: 12/12/2022] Open
Abstract
This study aims to investigate anti-adipogenic effects of long-chain bases from Cucumaria frondosa (Cf-LCBs) in vitro. Results showed that Cf-LCBs inhibited adipocyte differentiation and the expressions of CCAAT/enhancer binding proteins (C/EBPs) and peroxisome proliferators-activated receptor γ (PPARγ). Cf-LCBs increased β-catenin mRNA and nuclear translocation and increased its target genes, cyclin D1 and c-myc. Cf-LCBs enhanced fizzled and lipoprotein-receptor-related protein5/6 (LRP5/6) expressions, whereas wingless-type MMTV integration site10b (WNT10b) and glycogen syntheses kinase 3β (GSK3β) remained unchanged. Cf-LCBs also reduced adipogenesis and recovered WNT/β-catenin signaling in the cells suffering from 21H7, a β-catenin inhibitor. In addition, Cf-LCBs decreased triglyceride content and the expressions of lipogenesis genes. Cf-LCBs increased FFA levels and the expressions of lipidolytic factors. Cf-LCBs promoted the phosphorylation of adenosine-monophosphate-activated protein kinase (AMPK) and acetyl-CoA carboxylase. These findings indicate that Cf-LCBs inhibit adipogenesis through activation of WNT/β-catenin signaling and regulate lipid metabolism via activation of AMPK pathway.
Collapse
|
20
|
Induction of WNT11 by hypoxia and hypoxia-inducible factor-1α regulates cell proliferation, migration and invasion. Sci Rep 2016; 6:21520. [PMID: 26861754 PMCID: PMC4748282 DOI: 10.1038/srep21520] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 01/26/2016] [Indexed: 12/17/2022] Open
Abstract
Changes in cellular oxygen tension play important roles in physiological processes including development and pathological processes such as tumor promotion. The cellular adaptations to sustained hypoxia are mediated by hypoxia-inducible factors (HIFs) to regulate downstream target gene expression. With hypoxia, the stabilized HIF-α and aryl hydrocarbon receptor nuclear translocator (ARNT, also known as HIF-β) heterodimer bind to hypoxia response elements (HREs) and regulate expression of target genes. Here, we report that WNT11 is induced by hypoxia in many cell types, and that transcription of WNT11 is regulated primarily by HIF-1α. We observed induced WNT11 expression in the hypoxic area of allograft tumors. In addition, in mice bearing orthotopic malignant gliomas, inhibition with bevacizumab of vascular endothelial growth factor, which is an important stimulus for angiogenesis, increased nuclear HIF-1α and HIF-2α, and expression of WNT11. Gain- and loss-of-function approaches revealed that WNT11 stimulates proliferation, migration and invasion of cancer-derived cells, and increases activity of matrix metalloproteinase (MMP)-2 and 9. Since tumor hypoxia has been proposed to increase tumor aggressiveness, these data suggest WNT11 as a possible target for cancer therapies, especially for tumors treated with antiangiogenic therapy.
Collapse
|
21
|
Park B, Lee S, Lee B, Kim I, Baek N, Lee TH, Lee SY, Son M, Park H. New ethanol extraction improves the anti-obesity effects of black tea. Arch Pharm Res 2015; 39:310-20. [PMID: 26604105 DOI: 10.1007/s12272-015-0674-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 10/14/2015] [Indexed: 01/09/2023]
Abstract
Black tea has been reported to have anti-obesity effects in both rodents and humans. Gallic acid, an active component of black tea, decomposes quickly into pyrogallol in high-temperature solutions. This study introduced a new, aqueous ethanol extraction of black tea, which resulted in extracts with higher concentrations of gallic acid than conventional black tea extracts prepared by hot-water extraction or hot-ethanol extraction. We confirmed that, compared with the hot-water extract of black tea, the cold-ethanol extract of black tea (CE-BTE) had greater effects on reducing body weight and body fat, improving fatty liver, regulating blood glucose, and reducing blood cholesterol in the high-fat diet-induced obese mouse model. Nonetheless, although CE-BTE significantly reduced fat content, it did not reduce peroxisome proliferator-activated receptor (PPARγ) protein in epididymal fat tissue of HFD mice. We also showed that CE-BTE did not inhibit the function of PPARγ protein to drive adipogenesis of mouse 3T3-L1 preadipocytes. Considering that PPARγ is a master transcription factor not only for adipocyte differentiation, but also for adipose tissue function, such as glucose and lipid metabolism and insulin sensitivity, these results suggest that CE-BTE reduced fat mass and body weight without dampening fat cell homeostasis and insulin sensitivity.
Collapse
Affiliation(s)
- Bongju Park
- Department of Life Science, University of Seoul, Siripdae-gil 13, Dongdaemun-gu, Seoul, 130-743, Korea
| | - Sangjin Lee
- Research Institute, Dong-A ST Co., Ltd., Gyeonggi, 446-905, Korea.,School of Pharmacy, Sungkyunkwan University, Suwon, 440-746, Korea
| | - Bonggyeong Lee
- Research Institute, Dong-A ST Co., Ltd., Gyeonggi, 446-905, Korea
| | - Ingyum Kim
- Department of Life Science, University of Seoul, Siripdae-gil 13, Dongdaemun-gu, Seoul, 130-743, Korea
| | - Namjoon Baek
- Research Institute, Dong-A ST Co., Ltd., Gyeonggi, 446-905, Korea
| | - Tae Ho Lee
- Research Institute, Dong-A ST Co., Ltd., Gyeonggi, 446-905, Korea
| | - Seok-Yong Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 440-746, Korea
| | - Miwon Son
- Research Institute, Dong-A ST Co., Ltd., Gyeonggi, 446-905, Korea.
| | - Hyunsung Park
- Department of Life Science, University of Seoul, Siripdae-gil 13, Dongdaemun-gu, Seoul, 130-743, Korea.
| |
Collapse
|
22
|
Liu F, Dong X, Lv H, Xiu P, Li T, Wang F, Xu Z, Li J. Targeting hypoxia-inducible factor-2α enhances sorafenib antitumor activity via β-catenin/C-Myc-dependent pathways in hepatocellular carcinoma. Oncol Lett 2015; 10:778-784. [PMID: 26622569 DOI: 10.3892/ol.2015.3315] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 05/07/2015] [Indexed: 12/12/2022] Open
Abstract
Sorafenib is a type of multikinase inhibitor that exhibits antiangiogenic and antiproliferative effects; in addition, sorafenib is a unique first-line drug recommended for the treatment of advanced hepatocellular carcinoma (HCC). However, the effectiveness of HCC treatment remains poor due to acquired drug resistance. It has been suggested that hypoxia, induced as a results of the antiangiogenic effects of sustained sorafenib treatment, may be an important factor in sorafenib resistance. The transcription factor hypoxia-inducible factor (HIF)-2α has been reported to be associated with cell proliferation under hypoxic conditions; therefore, it was hypothesized that hypoxia may enhance tumor cell proliferation via this mechanism. The present study aimed to evaluate whether the knock-down of HIF-2α was able to enhance the therapeutic efficacy of sorafenib in order to effectively treat HCC. The results demonstrated that hypoxia protected HCC cells against sorafenib; however, short hairpin RNA-HIF-2α transfection in combination with sorafenib treatment exhibited a significantly synergistic effect against HCC cell proliferation. In addition, HCC cells acquired increased β-catenin/C-Myc expression, which enhanced proliferation under hypoxic conditions; however, targeted knock-down of HIF-2α or C-Myc markedly decreased cell proliferation in HCC cells. In conclusion, the results of the present study indicated that the targeted knock-down of HIF-2α in combination with sorafenib may be a promising strategy for the treatment of HCC.
Collapse
Affiliation(s)
- Feng Liu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Xiaofeng Dong
- Department of Hepatobiliary Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Hong Lv
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Peng Xiu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Tao Li
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Fuhai Wang
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Zongzhen Xu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Jie Li
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
23
|
Guo F, Lang J, Sohn J, Hammond E, Chang M, Pleasure D. Canonical Wnt signaling in the oligodendroglial lineage-puzzles remain. Glia 2015; 63:1671-93. [DOI: 10.1002/glia.22813] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 02/17/2015] [Indexed: 12/17/2022]
Affiliation(s)
- Fuzheng Guo
- Neurology Department; School of Medicine at UC Davis Medical Center; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, Northern California; Sacramento California
| | - Jordan Lang
- Neurology Department; School of Medicine at UC Davis Medical Center; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, Northern California; Sacramento California
| | - Jiho Sohn
- Neurology Department; School of Medicine at UC Davis Medical Center; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, Northern California; Sacramento California
| | - Elizabeth Hammond
- Neurology Department; School of Medicine at UC Davis Medical Center; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, Northern California; Sacramento California
| | - Marcello Chang
- Neurology Department; School of Medicine at UC Davis Medical Center; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, Northern California; Sacramento California
| | - David Pleasure
- Neurology Department; School of Medicine at UC Davis Medical Center; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, Northern California; Sacramento California
| |
Collapse
|
24
|
Sun X, Pang L, Shi M, Huang J, Wang Y. HIF2α induces cardiomyogenesis via Wnt/β-catenin signaling in mouse embryonic stem cells. J Transl Med 2015; 13:88. [PMID: 25889500 PMCID: PMC4399227 DOI: 10.1186/s12967-015-0447-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 02/27/2015] [Indexed: 11/10/2022] Open
Abstract
Background Embryonic stem cells (ESCs) are pluripotent stem cells and can differentiate into cardiomyocytes when cultured in appropriate conditions. The function of hypoxia-inducible factors (HIFs) has been identified in directing the formation of cardiac lineages. The purpose of this study was to investigate the ability of HIF2α to induce differentiation of ESCs into cardiomyocytes and to explore the potential underlying molecular mechanisms. Methods Cardiac differentiation from mouse ESCs was analyzed using the “hanging drop” method, and success was determined by assaying the numbers of beating embryoid bodies and the expression level of cardiac markers. The expression of HIF2α was then manipulated during cardiac differentiation with piggyBac transposon and the lentivirus system. The underlying mechanism was finally examined via administering selective inhibitors of the Wnt/β-catenin signaling pathway. Results Overexpressing HIF2α can significantly drive mouse ESCs to form cardiomyocytes. Contrarily, knockdown of HIF2α inhibits the emergence of cardiac cells. In addition, the cardiomyogenesis-promoting effect of HIF2α occurred by increasing the protein level of β-catenin, an effector that contributes to cardiac differentiation at an early stage of ESC differentiation. Conclusion HIF2α has a cardiomyogenesis-promoting effect in ESCs via enhancing the activation of the Wnt/β-catenin signaling pathway. Our results may be beneficial for generating and applying cardiomyocytes from ESCs safely and effectively in the future. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0447-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaotian Sun
- Department of Cardiothoracic surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Road, Shanghai, 200040, PR, China.
| | - Liewen Pang
- Department of Cardiothoracic surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Road, Shanghai, 200040, PR, China.
| | - Meng Shi
- Department of Cardiothoracic surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Road, Shanghai, 200040, PR, China.
| | - Jiechun Huang
- Department of Cardiothoracic surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Road, Shanghai, 200040, PR, China.
| | - Yiqing Wang
- Department of Cardiothoracic surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Road, Shanghai, 200040, PR, China.
| |
Collapse
|
25
|
Ban JJ, Ruthenborg RJ, Cho KW, Kim JW. Regulation of obesity and insulin resistance by hypoxia-inducible factors. HYPOXIA 2014; 2:171-183. [PMID: 27774475 PMCID: PMC5045065 DOI: 10.2147/hp.s68771] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
In obesity, dysregulated metabolism and aberrant expansion of adipose tissue lead to the development of tissue hypoxia that plays an important role in contributing to obesity-associated metabolic disorders. Recent studies utilizing adipocyte-specific hypoxia-inducible factor-α (HIF-α) gain- or loss-of-function animal models highlight the pivotal involvement of hypoxic responses in the pathogenesis of obesity-associated inflammation and insulin resistance. HIF-1α, a master transcription factor of oxygen homeostasis, induces inflammation and insulin resistance in obesity, whereas its isoform, HIF-2α, exerts opposing functions in these obesity-associated metabolic phenotypes. In this review, recent evidence elucidating functional implications of adipocyte HIFs in obesity and, more importantly, how these regulate obesity-associated inflammation, fibrosis, and insulin resistance will be discussed. Further, we propose that modulation of HIF-1 could be a potential novel therapeutic strategy for antidiabetic treatment.
Collapse
Affiliation(s)
- Jae-Jun Ban
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Robin J Ruthenborg
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Kevin W Cho
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Jung-Whan Kim
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
26
|
Dere R, Perkins AL, Bawa-Khalfe T, Jonasch D, Walker CL. β-catenin links von Hippel-Lindau to aurora kinase A and loss of primary cilia in renal cell carcinoma. J Am Soc Nephrol 2014; 26:553-64. [PMID: 25313256 DOI: 10.1681/asn.2013090984] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
von Hippel-Lindau (VHL) gene mutations are associated with clear cell renal cell carcinoma (ccRCC). A hallmark of ccRCC is loss of the primary cilium. Loss of this key organelle in ccRCC is caused by loss of VHL and associated with increased Aurora kinase A (AURKA) and histone deacetylase 6 (HDAC6) activities, which drive disassembly of the primary cilium. However, the underlying mechanism by which VHL loss increases AURKA levels has not been clearly elucidated, although it has been suggested that hypoxia-inducible factor-1α (HIF-1α) mediates increased AURKA expression in VHL-null cells. By contrast, we found that elevated AURKA expression is not increased by HIF-1α, suggesting an alternate mechanism for AURKA dysregulation in VHL-null cells. We report here that AURKA expression is driven by β-catenin transcription in VHL-null cells. In a panel of RCC cell lines, we observed nuclear accumulation of β-catenin and increased AURKA signaling to HDAC6. Moreover, HIF-1α inhibited AURKA expression by inhibiting β-catenin transcription. VHL knockdown activated β-catenin and elevated AURKA expression, decreased primary cilia formation, and caused significant shortening of cilia length in cells that did form cilia. The β-catenin responsive transcription inhibitor iCRT14 reduced AURKA levels and rescued ciliary defects, inducing a significant increase in primary cilia formation in VHL-deficient cells. These data define a role for β-catenin in regulating AURKA and formation of primary cilia in the setting of VHL deficiency, opening new avenues for treatment with β-catenin inhibitors to rescue ciliogenesis in ccRCC.
Collapse
Affiliation(s)
- Ruhee Dere
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas; and
| | - Ashley Lyn Perkins
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas; and
| | - Tasneem Bawa-Khalfe
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Darius Jonasch
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas; and
| | - Cheryl Lyn Walker
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas; and
| |
Collapse
|
27
|
Abstract
Hypoxia develops in white adipose tissue in obese mice, resulting in changes in adipocyte function that may underpin the dysregulation that leads to obesity-associated disorders. Whether hypoxia occurs in adipose tissue in human obesity is unclear, with recent studies contradicting earlier reports that this was the case. Adipocytes, both murine and human, exhibit extensive functional changes in culture in response to hypoxia, which alters the expression of up to 1,300 genes. These include genes encoding key adipokines such as leptin, interleukin (IL)-6, vascular endothelial growth factor (VEGF), and matrix metalloproteinase-2 (MMP-2), which are upregulated, and adiponectin, which is downregulated. Hypoxia also inhibits the expression of genes linked to oxidative metabolism while stimulating the expression of genes associated with glycolysis. Glucose uptake and lactate release by adipocytes are both stimulated by hypoxia, and insulin sensitivity falls. Preadipocytes and macrophages in adipose tissue also respond to hypoxia. The hypoxia-signaling pathway may provide a new target for the treatment of obesity-associated disorders.
Collapse
Affiliation(s)
- Paul Trayhurn
- Obesity Biology Research Unit, Institute of Ageing and Chronic Diseases, University of Liverpool, Liverpool L69 3GA United Kingdom, and Clore Laboratory, University of Buckingham, Buckingham MK18 1EG, United Kingdom;
| |
Collapse
|
28
|
Lee HY, Choi K, Oh H, Park YK, Park H. HIF-1-dependent induction of Jumonji domain-containing protein (JMJD) 3 under hypoxic conditions. Mol Cells 2014; 37:43-50. [PMID: 24552709 PMCID: PMC3907005 DOI: 10.14348/molcells.2014.2250] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 11/17/2013] [Accepted: 11/18/2013] [Indexed: 01/20/2023] Open
Abstract
Jumonji domain-containing proteins (JMJD) catalyze the oxidative demethylation of a methylated lysine residue of histones by using O2, α-ketoglutarate, vitamin C, and Fe(II). Several JMJDs are induced by hypoxic stress to compensate their presumed reduction in catalytic activity under hypoxia. In this study, we showed that an H3K27me3 specific histone demethylase, JMJD3 was induced by hypoxia-inducible factor (HIF)-1α/β under hypoxia and that treatment with Clioquinol, a HIF-1α activator, increased JMJD3 expression even under normoxia. Chromatin immunoprecipitation (ChIP) analyses showed that both HIF-1α and its dimerization partner HIF-1β/Arnt occupied the first intron region of the mouse JMJD3 gene, whereas the HIF-1α/β heterodimer bound to the upstream region of the human JMJD3, indicating that human and mouse JMJD3 have hypoxia-responsive regulatory regions in different locations. This study shows that both mouse and human JMJD3 are induced by HIF-1.
Collapse
Affiliation(s)
- Ho-Youl Lee
- Department of Life Science, University of Seoul, Seoul 130-743,
Korea
| | - Kang Choi
- Department of Life Science, University of Seoul, Seoul 130-743,
Korea
| | - Hookeun Oh
- Department of Life Science, University of Seoul, Seoul 130-743,
Korea
| | - Young-Kwon Park
- Department of Life Science, University of Seoul, Seoul 130-743,
Korea
| | - Hyunsung Park
- Department of Life Science, University of Seoul, Seoul 130-743,
Korea
| |
Collapse
|