1
|
Tümen D, Heumann P, Huber J, Hahn N, Macek C, Ernst M, Kandulski A, Kunst C, Gülow K. Unraveling Cancer's Wnt Signaling: Dynamic Control through Protein Kinase Regulation. Cancers (Basel) 2024; 16:2686. [PMID: 39123414 PMCID: PMC11312265 DOI: 10.3390/cancers16152686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Since the initial identification of oncogenic Wnt in mice and Drosophila, the Wnt signaling pathway has been subjected to thorough and extensive investigation. Persistent activation of Wnt signaling exerts diverse cancer characteristics, encompassing tumor initiation, tumor growth, cell senescence, cell death, differentiation, and metastasis. Here we review the principal signaling mechanisms and the regulatory influence of pathway-intrinsic and extrinsic kinases on cancer progression. Additionally, we underscore the divergences and intricate interplays of the canonical and non-canonical Wnt signaling pathways and their critical influence in cancer pathophysiology, exhibiting both growth-promoting and growth-suppressing roles across diverse cancer types.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Karsten Gülow
- Department of Internal Medicine I Gastroenterology, Hepatology, Endocrinology, Rheumatology, Immunology, and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany; (D.T.); (N.H.)
| |
Collapse
|
2
|
Cheng L, Xu Y, Long Y, Yu F, Gui L, Zhang Q, Lu Y. Liraglutide attenuates palmitate-induced apoptosis via PKA/β-catenin/Bcl-2/Bax pathway in MC3T3-E1 cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:329-341. [PMID: 37439807 DOI: 10.1007/s00210-023-02572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 06/09/2023] [Indexed: 07/14/2023]
Abstract
Liraglutide (LRG), one agonist of glucagon-like peptide-1 receptor (GLP1R), has multiple lipid-lowering effects in type 2 diabetes mellitus, however, studies on the role of LRG in saturated fatty acid-induced bone loss are limited. Therefore, our aim was to investigate whether LRG reduces palmitate (PA)-induced apoptosis and whether the mechanism involves PKA/β-catenin/Bcl-2/Bax in osteoblastic MC3T3-E1 cells. MC3T3-E1 cells were treated with different concentrations of PA, LRG, or pretreated with Exendin 9-39 and H89, cell viability, intracellular reactive oxygen species (ROS), cAMP levels, apoptosis and the expression of protein kinase A (PKA) and phosphorylation of PKA (p-PKA), β-catenin and phosphorylation of β-catenin (Ser675)(p-β-catenin), GLP1R, cleaved-capase 3, Bcl2-Associated X Protein (Bax) and B-cell lymphoma-2 (Bcl-2) along with expression of Osteoprotegerin (OPG) and receptor activator of nuclear factor-κB ligand (RANKL) were evaluated. PA treatment inhibited cell proliferation and cAMP levels, elevated intracellular ROS levels and promoted apoptosis, increased protein expressions of RANKL, Bax and cleaved-caspase3, meanwhile decreased protein expression of OPG and Bcl-2 in a dose-dependent manner. LRG inverted PA-induced apoptosis, increased cAMP levels, promoted expression of p-PKA, p-β-catenin (Ser675) and reversed these gene expressions via increasing GLP1R expression. Pretreatment of the cells with Exendin 9-39 and H89 partially eradicated the protective effect of LRG on PA-induced apoptosis and gene expressions. Therefore, these findings indicated that LRG attenuates PA-induced apoptosis possibly by GLP1R-mediated PKA/β-catenin/Bcl-2/Bax pathway in MC3T3-E1 cells. Our results point to LRG as a new strategy to attenuate bone loss associated with high fat diet beyond its lipid-lowering actions. LRG inhibits PA-mediated apoptosis via GLP1R-mediated PKA/β-catenin/Bcl-2/ Bax pathway, while possibly enhances PA-inhibited differentiation by regulating the expression of OPG and RANKL.
Collapse
Affiliation(s)
- Lanlan Cheng
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yijing Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yueming Long
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Fangmei Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Li Gui
- The Comprehensive Laboratory, School of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Qiu Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Yunxia Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Anhui Medical University, Hefei, China.
- The Comprehensive Laboratory, School of Basic Medical Science, Anhui Medical University, Hefei, China.
| |
Collapse
|
3
|
Baek J, Lopez PA, Lee S, Kim TS, Kumar S, Schaffer DV. Egr1 is a 3D matrix-specific mediator of mechanosensitive stem cell lineage commitment. SCIENCE ADVANCES 2022; 8:eabm4646. [PMID: 35427160 PMCID: PMC9012469 DOI: 10.1126/sciadv.abm4646] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 02/28/2022] [Indexed: 05/31/2023]
Abstract
While extracellular matrix (ECM) mechanics strongly regulate stem cell commitment, the field's mechanistic understanding of this phenomenon largely derives from simplified two-dimensional (2D) culture substrates. Here, we found a 3D matrix-specific mechanoresponsive mechanism for neural stem cell (NSC) differentiation. NSC lineage commitment in 3D is maximally stiffness sensitive in the range of 0.1 to 1.2 kPa, a narrower and more brain-mimetic range than we had previously identified in 2D (0.75 to 75 kPa). Transcriptomics revealed stiffness-dependent up-regulation of early growth response 1 (Egr1) in 3D but not in 2D. Egr1 knockdown enhanced neurogenesis in stiff ECMs by driving β-catenin nuclear localization and activity in 3D, but not in 2D. Mechanical modeling and experimental studies under osmotic pressure indicate that stiff 3D ECMs are likely to stimulate Egr1 via increases in confining stress during cell volumetric growth. To our knowledge, Egr1 represents the first 3D-specific stem cell mechanoregulatory factor.
Collapse
Affiliation(s)
- Jieung Baek
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Paola A. Lopez
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- UC Berkeley–UC San Francisco Graduate Program in Bioengineering, Berkeley, CA 94720, USA
| | - Sangmin Lee
- Department of Mechanical Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Taek-Soo Kim
- Department of Mechanical Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- UC Berkeley–UC San Francisco Graduate Program in Bioengineering, Berkeley, CA 94720, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - David V. Schaffer
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- UC Berkeley–UC San Francisco Graduate Program in Bioengineering, Berkeley, CA 94720, USA
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Helen Wills Neuroscience Institute, Berkeley, CA 94720, USA
| |
Collapse
|
4
|
Daley EJ, Trackman PC. β-Catenin mediates glucose-dependent insulinotropic polypeptide increases in lysyl oxidase expression in osteoblasts. Bone Rep 2021; 14:101063. [PMID: 33981809 PMCID: PMC8081922 DOI: 10.1016/j.bonr.2021.101063] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoblast lysyl oxidase (LOX) is a strongly up-regulated mRNA and protein by the glucose-dependent insulinotropic polypeptide (GIP). LOX is critically required for collagen maturation, and was shown to be dramatically down-regulated in a mouse model of type 1 diabetes, consistent with known low collagen cross-linking and poor bone quality in diabetic bone disease in humans and in mouse models. GIP is a gastric hormone released by the gut upon consumption of nutrients, which then stimulates insulin release from β-cells in the pancreas. GIP is directly anabolic to osteoblasts and to bone, while gut-derived dopamine attenuates effects of GIP on osteoblast anabolic pathways, including LOX expression. GIP-stimulation of LOX expression was shown to be dependent on increased cAMP levels and protein kinase A activity, consistent with the fact that GIP receptors are G protein coupled receptors. Downstream signaling events resulting in increased LOX expression remain, however, unexplored. Here we provide evidence for β-catenin mediation of signaling from GIP to increase LOX expression. Moreover, we have identified a TCF/LEF element in the Lox promoter that is required for GIP-upregulation of LOX. These findings will be of importance in designing potential therapeutic approaches to address deficient LOX production in diabetic bone disease by pointing to the importance of exploring strategies to stimulate β-catenin signaling in osteoblasts under diabetic conditions as potential therapeutic strategies.
Collapse
Affiliation(s)
| | - Philip C. Trackman
- Corresponding author at: Forsyth Institute, Department of Applied Oral Sciences, 250 First Street, Cambridge, MA 02118, United States of America.
| |
Collapse
|
5
|
PKAc-directed interaction and phosphorylation of Ptc is required for Hh signaling inhibition in Drosophila. Cell Discov 2019; 5:44. [PMID: 31636957 PMCID: PMC6796939 DOI: 10.1038/s41421-019-0112-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/04/2019] [Indexed: 01/20/2023] Open
Abstract
Ptc is a gatekeeper to avoid abnormal Hh signaling activation, but the key regulators involved in Ptc-mediated inhibition remain largely unknown. Here, we identify PKAc as a key regulator required for Ptc inhibitory function. In the absence of Hh, PKAc physically interacts with Ptc and phosphorylates Ptc at Ser-1150 and -1183 residues. The presence of Hh unleashes PKAc from Ptc and activates Hh signaling. By combining both in vitro and in vivo functional assays, we demonstrate that such Ptc–PKAc interaction and Ptc phosphorylation are both important for Ptc inhibitory function. Interestingly, we further demonstrate that PKAc is subjected to palmitoylation, contributing to its kinase activity on plasma membrane. Based on those novel findings, we establish a working model on Ptc inhibitory function: In the absence of Hh, PKAc interacts with and phosphorylates Ptc to ensure its inhibitory function; and Hh presence releases PKAc from Ptc, resulting in Hh signaling activation.
Collapse
|
6
|
Chen Z, Xue C. G-Protein-Coupled Receptor 5 (LGR5) Overexpression Activates β-Catenin Signaling in Breast Cancer Cells via Protein Kinase A. Med Sci Monit Basic Res 2019; 25:15-25. [PMID: 30662060 PMCID: PMC6354635 DOI: 10.12659/msmbr.912411] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Targeting cancer stem cells (CSCs) in breast cancer (BrCa) may improve treatment outcome and patient prognosis. Leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5) is a well-recognized adult stem cell and CRC marker, and previous reports have suggested the cancer-promoting role of LGR5 in breast cancer, but the mechanism remains unclear. Material/Methods Potential LGR5-associating genes were explored using STRING database, and LGR5 overexpression and knockdown was constructed in MCF-7 and MDA-MB-453 human BrCa cells, respectively. PKA catalytic subunit activation and PKA kinase activity in human BrCa cells was examined by Western blot and PKA kinase activity assay, respectively. Protein expression level or activation of β-catenin and GSK-3β in human BrCa cells was investigated by Western blot. Cell proliferation, colony formation, Transwell migration, cisplatin sensitivity, and in vivo tumor formation of human BrCa cells were examined. Results LGR5 overexpression increased PKA activation and its kinase activity in human BrCa cells, which was decreased by LGR5 knockdown. LGR5 expression level or PKA kinase activity were correlated with β-catenin Ser 552 phosphorylation but inversely correlated with GSK-3β Ser9 phosphorylation in human BrCa cells in vitro. LGR5/PKA increased cell proliferation, colony formation, Transwell migration, and cisplatin resistance in vitro, as well as tumor formation in vivo, of human BrCa cells. Conclusions LGR5 activates the Wnt/β-catenin signaling pathway in human BrCa cells in vitro via PKA.
Collapse
Affiliation(s)
- Zhishui Chen
- Department of Pathology, Ninety-First Central Hospital of the People's Liberation Army (PLA), Jiaozuo, Henan, China (mainland)
| | - Chengjun Xue
- Department of Pathology, Ninety-First Central Hospital of the People's Liberation Army (PLA), Jiaozuo, Henan, China (mainland)
| |
Collapse
|
7
|
Scott CC, Vossio S, Rougemont J, Gruenberg J. TFAP2 transcription factors are regulators of lipid droplet biogenesis. eLife 2018; 7:36330. [PMID: 30256193 PMCID: PMC6170152 DOI: 10.7554/elife.36330] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 09/26/2018] [Indexed: 12/11/2022] Open
Abstract
How trafficking pathways and organelle abundance adapt in response to metabolic and physiological changes is still mysterious, although a few transcriptional regulators of organellar biogenesis have been identified in recent years. We previously found that the Wnt signaling directly controls lipid droplet formation, linking the cell storage capacity to the established functions of Wnt in development and differentiation. In the present paper, we report that Wnt-induced lipid droplet biogenesis does not depend on the canonical TCF/LEF transcription factors. Instead, we find that TFAP2 family members mediate the pro-lipid droplet signal induced by Wnt3a, leading to the notion that the TFAP2 transcription factor may function as a 'master' regulator of lipid droplet biogenesis.
Collapse
Affiliation(s)
- Cameron C Scott
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Stefania Vossio
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Jacques Rougemont
- Department of Theoretical Physics, University of Geneva, Geneva, Switzerland
| | - Jean Gruenberg
- Department of Biochemistry, University of Geneva, Geneva, Switzerland.,Department of Theoretical Physics, University of Geneva, Geneva, Switzerland
| |
Collapse
|
8
|
Spagnol G, Trease AJ, Zheng L, Gutierrez M, Basu I, Sarmiento C, Moore G, Cervantes M, Sorgen PL. Connexin43 Carboxyl-Terminal Domain Directly Interacts with β-Catenin. Int J Mol Sci 2018; 19:ijms19061562. [PMID: 29882937 PMCID: PMC6032326 DOI: 10.3390/ijms19061562] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 12/13/2022] Open
Abstract
Activation of Wnt signaling induces Connexin43 (Cx43) expression via the transcriptional activity of β-catenin, and results in the enhanced accumulation of the Cx43 protein and the formation of gap junction channels. In response to Wnt signaling, β-catenin co-localizes with the Cx43 protein itself as part of a complex at the gap junction plaque. Work from several labs have also shown indirect evidence of this interaction via reciprocal co-immunoprecipitation. Our goal for the current study was to identify whether β-catenin directly interacts with Cx43, and if so, the location of that direct interaction. Identifying residues involved in direct protein⁻protein interaction is of importance when they are correlated to the phosphorylation of Cx43, as phosphorylation can modify the binding affinities of Cx43 regulatory protein partners. Therefore, combining the location of a protein partner interaction on Cx43 along with the phosphorylation pattern under different homeostatic and pathological conditions will be crucial information for any potential therapeutic intervention. Here, we identified that β-catenin directly interacts with the Cx43 carboxyl-terminal domain, and that this interaction would be inhibited by the Src phosphorylation of Cx43CT residues Y265 and Y313.
Collapse
Affiliation(s)
- Gaelle Spagnol
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Andrew J Trease
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Li Zheng
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Mirtha Gutierrez
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Ishika Basu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Cleofes Sarmiento
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Gabriella Moore
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Matthew Cervantes
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Paul L Sorgen
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
9
|
Park EYJ, Kwak M, Ha K, So I. Identification of clustered phosphorylation sites in PKD2L1: how PKD2L1 channel activation is regulated by cyclic adenosine monophosphate signaling pathway. Pflugers Arch 2017; 470:505-516. [PMID: 29230552 DOI: 10.1007/s00424-017-2095-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/21/2017] [Accepted: 12/01/2017] [Indexed: 01/01/2023]
Abstract
Polycystic kidney disease 2-like-1 (PKD2L1), or polycystin-L or TRPP2, formerly TRPP3, is a transient receptor potential (TRP) superfamily member. It is a calcium-permeable non-selective cation channel that regulates intracellular calcium concentration and thereby calcium signaling. PKD2L1 has been reported to take part in hedgehog signaling in renal primary cilia and sour tasting coupling with PKD1L3. In addition to the previous reports, PKD2L1 is recently found to play a crucial role in localization with β2-adrenergic receptor (β2AR) on the neuronal primary cilia. The disruption of PKD2L1 leads to the loss of β2AR on the primary cilia and reduction in intracellular concentration of cyclic adenosine monophosphate (cAMP). Since the role of cAMP and PKA is frequently mentioned in the studies of PKD diseases, we investigated on the mechanism of cAMP regulation in relation to the function of PKD2L1 channel. In this study, we observed the activity of PKD2L1 channel increased by the downstream cascades of β2AR and found the clustered phosphorylation sites, Ser-682, Ser-685, and Ser-686 that are significant in the channel regulation by phosphorylation.
Collapse
Affiliation(s)
- Eunice Yon June Park
- Department of Physiology, Seoul National University, College of Medicine, Biomedical Science Building 117, 103 Daehakro, Jongro-gu, Seoul, 110-799, South Korea
| | - Misun Kwak
- Department of Physiology, Seoul National University, College of Medicine, Biomedical Science Building 117, 103 Daehakro, Jongro-gu, Seoul, 110-799, South Korea
| | - Kotdaji Ha
- Department of Physiology, University of California, San Francisco, CA, 94158-2517, USA
| | - Insuk So
- Department of Physiology, Seoul National University, College of Medicine, Biomedical Science Building 117, 103 Daehakro, Jongro-gu, Seoul, 110-799, South Korea.
| |
Collapse
|
10
|
Yang L, Zheng T, Wu H, Xin W, Mou X, Lin H, Chen Y, Wu X. Predictive value of apelin-12 in patients with ST-elevation myocardial infarction with different renal function: a prospective observational study. BMJ Open 2017; 7:e018595. [PMID: 29150476 PMCID: PMC5701982 DOI: 10.1136/bmjopen-2017-018595] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVES To investigate factors predicting the onset of major adverse cardiovascular events (MACEs) after primary percutaneous coronary intervention (pPCI) for patients with ST-segment elevation myocardial infarction (STEMI) . BACKGROUND Apelin-12 plays an essential role in cardiovascular homoeostasis. However, current knowledge of its predictive prognostic value is limited. METHODS 464 patients with STEMI (63.0±11.9 years, 355 men) who underwent successful pPCI were enrolled and followed for 2.5 years. Multivariate cox regression analysis and receiver operating characteristic (ROC) curve analysis were performed to determine the factors predicting MACEs. RESULTS 118 patients (25.4%) experienced MACEs in the follow-up period. Multivariate cox regression analysis found low apelin-12 (HR=0.132, 95% CI 0.060 to 0.292, P<0.001), low left ventricular ejection fraction (HR=0.965, 95% CI 0.941 to 0.991, P=0.007), low estimated glomerular filtration rate (eGFR) (HR=0.985, 95% CI 0.977 to 0.993, P<0.001), Killip's classification>I (HR=0.610, 95% CI 0.408 to 0.912, P=0.016) and pathological Q-wave (HR=1.536, 95% CI 1.058 to 2.230, P=0.024) were independent predictors of MACEs in the 2.5 year follow-up period. Low apelin-12 also predicted poorer in-hospital prognosis and MACEs in the 2.5 years follow-up period compared with Δapelin-12 (P=0.0115) and eGFR (P=0.0071) among patients with eGFR>90 mL/min×1.73 m2. Further analysis showed Δapelin-12 <20% was associated with MACEs in patients whose apelin-12 was below 0.76 ng/mL (P=0.0075) on admission. CONCLUSIONS Patients with STEMI receiving pPCI with lower apelin-12 are more likely to suffer MACEs in hospital and 2.5 years postprocedure, particularly in those with normal eGFR levels.
Collapse
Affiliation(s)
- Lingchang Yang
- Department of Emergency Medicine, The First People’s Hospital of Taizhou, Taizhou, Zhejiang, China
| | - Ting Zheng
- Department of Emergency Medicine, The First People’s Hospital of Taizhou, Taizhou, Zhejiang, China
| | - Haopeng Wu
- Department of Emergency Medicine, The First People’s Hospital of Taizhou, Taizhou, Zhejiang, China
| | - Wenwei Xin
- Department of Emergency Medicine, The First People’s Hospital of Taizhou, Taizhou, Zhejiang, China
| | - Xiongneng Mou
- Department of Emergency Medicine, The First People’s Hospital of Taizhou, Taizhou, Zhejiang, China
| | - Hui Lin
- Department of Emergency Medicine, The First People’s Hospital of Taizhou, Taizhou, Zhejiang, China
| | - Yide Chen
- Department of Emergency Medicine, The First People’s Hospital of Taizhou, Taizhou, Zhejiang, China
| | - Xiaoyu Wu
- Department of Emergency Medicine, The First People’s Hospital of Taizhou, Taizhou, Zhejiang, China
| |
Collapse
|
11
|
Ye H, Wang X, Constans MM, Sussman CR, Chebib FT, Irazabal MV, Young WF, Harris PC, Kirschner LS, Torres VE. The regulatory 1α subunit of protein kinase A modulates renal cystogenesis. Am J Physiol Renal Physiol 2017; 313:F677-F686. [PMID: 28615245 DOI: 10.1152/ajprenal.00119.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/09/2017] [Accepted: 06/12/2017] [Indexed: 11/22/2022] Open
Abstract
The failure of the polycystins (PCs) to function in primary cilia is thought to be responsible for autosomal dominant polycystic kidney disease (ADPKD). Primary cilia integrate multiple cellular signaling pathways, including calcium, cAMP, Wnt, and Hedgehog, which control cell proliferation and differentiation. It has been proposed that mutated PCs result in reduced intracellular calcium, which in turn upregulates cAMP, protein kinase A (PKA) signaling, and subsequently other proliferative signaling pathways. However, the role of PKA in ADPKD has not been directly ascertained in vivo, although the expression of the main regulatory subunit of PKA in cilia and other compartments (PKA-RIα, encoded by PRKAR1A) is increased in a mouse model orthologous to ADPKD. Therefore, we generated a kidney-specific knockout of Prkar1a to examine the consequences of constitutive upregulation of PKA on wild-type and Pkd1 hypomorphic (Pkd1RC) backgrounds. Kidney-specific loss of Prkar1a induced renal cystic disease and markedly aggravated cystogenesis in the Pkd1RC models. In both settings, it was accompanied by upregulation of Src, Ras, MAPK/ERK, mTOR, CREB, STAT3, Pax2 and Wnt signaling. On the other hand, Gli3 repressor activity was enhanced, possibly contributing to hydronephrosis and impaired glomerulogenesis in some animals. To assess the relevance of these observations in humans we looked for and found evidence for kidney and liver cystic phenotypes in the Carney complex, a tumoral syndrome caused by mutations in PRKAR1A These observations expand our understanding of the pathogenesis of ADPKD and demonstrate the importance of PRKAR1A highlighting PKA as a therapeutic target in ADPKD.
Collapse
Affiliation(s)
- Hong Ye
- Mayo Clinic, Rochester Minnesota; and
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Gsα Controls Cortical Bone Quality by Regulating Osteoclast Differentiation via cAMP/PKA and β-Catenin Pathways. Sci Rep 2017; 7:45140. [PMID: 28338087 PMCID: PMC5364530 DOI: 10.1038/srep45140] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 02/17/2017] [Indexed: 12/14/2022] Open
Abstract
Skeletal bone formation and maintenance requires coordinate functions of several cell types, including bone forming osteoblasts and bone resorbing osteoclasts. Gsα, the stimulatory subunit of heterotrimeric G proteins, activates downstream signaling through cAMP and plays important roles in skeletal development by regulating osteoblast differentiation. Here, we demonstrate that Gsα signaling also regulates osteoclast differentiation during bone modeling and remodeling. Gnas, the gene encoding Gsα, is imprinted. Mice with paternal allele deletion of Gnas (Gnas+/p-) have defects in cortical bone quality and strength during early development (bone modeling) that persist during adult bone remodeling. Reduced bone quality in Gnas+/p- mice was associated with increased endosteal osteoclast numbers, with no significant effects on osteoblast number and function. Osteoclast differentiation and resorption activity was enhanced in Gnas+/p- cells. During differentiation, Gnas+/p- cells showed diminished pCREB, β-catenin and cyclin D1, and enhanced Nfatc1 levels, conditions favoring osteoclastogenesis. Forskolin treatment increased pCREB and rescued osteoclast differentiation in Gnas+/p- by reducing Nfatc1 levels. Cortical bone of Gnas+/p- mice showed elevated expression of Wnt inhibitors sclerostin and Sfrp4 consistent with reduced Wnt/β-catenin signaling. Our data identify a new role for Gsα signaling in maintaining bone quality by regulating osteoclast differentiation and function through cAMP/PKA and Wnt/β-catenin pathways.
Collapse
|
13
|
Drelon C, Berthon A, Sahut-Barnola I, Mathieu M, Dumontet T, Rodriguez S, Batisse-Lignier M, Tabbal H, Tauveron I, Lefrançois-Martinez AM, Pointud JC, Gomez-Sanchez CE, Vainio S, Shan J, Sacco S, Schedl A, Stratakis CA, Martinez A, Val P. PKA inhibits WNT signalling in adrenal cortex zonation and prevents malignant tumour development. Nat Commun 2016; 7:12751. [PMID: 27624192 PMCID: PMC5027289 DOI: 10.1038/ncomms12751] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 07/28/2016] [Indexed: 01/30/2023] Open
Abstract
Adrenal cortex physiology relies on functional zonation, essential for production of aldosterone by outer zona glomerulosa (ZG) and glucocorticoids by inner zona fasciculata (ZF). The cortex undergoes constant cell renewal, involving recruitment of subcapsular progenitors to ZG fate and subsequent lineage conversion to ZF identity. Here we show that WNT4 is an important driver of WNT pathway activation and subsequent ZG differentiation and demonstrate that PKA activation prevents ZG differentiation through WNT4 repression and WNT pathway inhibition. This suggests that PKA activation in ZF is a key driver of WNT inhibition and lineage conversion. Furthermore, we provide evidence that constitutive PKA activation inhibits, whereas partial inactivation of PKA catalytic activity stimulates β-catenin-induced tumorigenesis. Together, both lower PKA activity and higher WNT pathway activity lead to poorer prognosis in adrenocortical carcinoma (ACC) patients. These observations suggest that PKA acts as a tumour suppressor in the adrenal cortex, through repression of WNT signalling. The adrenal cortex undergoes functional zonation to generate an outer zona glomerulosa (ZG) and inner zona fasciculata (ZF), but how this is regulated at a molecular level is unclear. Here, the authors show that ZG differentiation is stimulated by WNT signalling and that PKA blocks WNT signalling to allow ZF differentiation and also prevents WNT-induced cancer development.
Collapse
Affiliation(s)
- Coralie Drelon
- CNRS, UMR 6293, GReD, Inserm U1103, Clermont Université, F-63171 Aubière Cedex, France
| | - Annabel Berthon
- CNRS, UMR 6293, GReD, Inserm U1103, Clermont Université, F-63171 Aubière Cedex, France.,Developmental Endocrine Oncology and Genetics, Section on Genetics and Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland 20892-1103, USA
| | | | - Mickaël Mathieu
- CNRS, UMR 6293, GReD, Inserm U1103, Clermont Université, F-63171 Aubière Cedex, France
| | - Typhanie Dumontet
- CNRS, UMR 6293, GReD, Inserm U1103, Clermont Université, F-63171 Aubière Cedex, France
| | - Stéphanie Rodriguez
- CNRS, UMR 6293, GReD, Inserm U1103, Clermont Université, F-63171 Aubière Cedex, France
| | - Marie Batisse-Lignier
- CNRS, UMR 6293, GReD, Inserm U1103, Clermont Université, F-63171 Aubière Cedex, France.,Centre Hospitalier Universitaire, Service d'Endocrinologie, Faculté de Médecine, F-63000 Clermont-Ferrand, France
| | - Houda Tabbal
- CNRS, UMR 6293, GReD, Inserm U1103, Clermont Université, F-63171 Aubière Cedex, France
| | - Igor Tauveron
- CNRS, UMR 6293, GReD, Inserm U1103, Clermont Université, F-63171 Aubière Cedex, France.,Centre Hospitalier Universitaire, Service d'Endocrinologie, Faculté de Médecine, F-63000 Clermont-Ferrand, France
| | | | | | - Celso E Gomez-Sanchez
- Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi 39216, USA.,Department of Medicine-Endocrinology, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA
| | - Seppo Vainio
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland
| | - Jingdong Shan
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland
| | - Sonia Sacco
- Inserm UMR1091, CNRS UMR 7277, Institute of Biology Valrose, F-06108 Nice, France
| | - Andreas Schedl
- Inserm UMR1091, CNRS UMR 7277, Institute of Biology Valrose, F-06108 Nice, France
| | - Constantine A Stratakis
- Developmental Endocrine Oncology and Genetics, Section on Genetics and Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland 20892-1103, USA
| | - Antoine Martinez
- CNRS, UMR 6293, GReD, Inserm U1103, Clermont Université, F-63171 Aubière Cedex, France
| | - Pierre Val
- CNRS, UMR 6293, GReD, Inserm U1103, Clermont Université, F-63171 Aubière Cedex, France
| |
Collapse
|
14
|
Gallardo-Montejano VI, Saxena G, Kusminski CM, Yang C, McAfee JL, Hahner L, Hoch K, Dubinsky W, Narkar VA, Bickel PE. Nuclear Perilipin 5 integrates lipid droplet lipolysis with PGC-1α/SIRT1-dependent transcriptional regulation of mitochondrial function. Nat Commun 2016; 7:12723. [PMID: 27554864 PMCID: PMC4999519 DOI: 10.1038/ncomms12723] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 07/27/2016] [Indexed: 12/18/2022] Open
Abstract
Dysfunctional cellular lipid metabolism contributes to common chronic human diseases, including type 2 diabetes, obesity, fatty liver disease and diabetic cardiomyopathy. How cells balance lipid storage and mitochondrial oxidative capacity is poorly understood. Here we identify the lipid droplet protein Perilipin 5 as a catecholamine-triggered interaction partner of PGC-1α. We report that during catecholamine-stimulated lipolysis, Perilipin 5 is phosphorylated by protein kinase A and forms transcriptional complexes with PGC-1α and SIRT1 in the nucleus. Perilipin 5 promotes PGC-1α co-activator function by disinhibiting SIRT1 deacetylase activity. We show by gain-and-loss of function studies in cells that nuclear Perilipin 5 promotes transcription of genes that mediate mitochondrial biogenesis and oxidative function. We propose that Perilipin 5 is an important molecular link that couples the coordinated catecholamine activation of the PKA pathway and of lipid droplet lipolysis with transcriptional regulation to promote efficient fatty acid catabolism and prevent mitochondrial dysfunction.
Collapse
Affiliation(s)
- Violeta I. Gallardo-Montejano
- Division of Endocrinology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Geetu Saxena
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for The Prevention Of Human Diseases, UT Health, Houston, Texas 77030, USA
| | - Christine M. Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Chaofeng Yang
- Division of Endocrinology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - John L. McAfee
- Division of Endocrinology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Lisa Hahner
- Division of Endocrinology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Kathleen Hoch
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for The Prevention Of Human Diseases, UT Health, Houston, Texas 77030, USA
| | - William Dubinsky
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for The Prevention Of Human Diseases, UT Health, Houston, Texas 77030, USA
| | - Vihang A. Narkar
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for The Prevention Of Human Diseases, UT Health, Houston, Texas 77030, USA
| | - Perry E. Bickel
- Division of Endocrinology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
15
|
Rabadán MA, Herrera A, Fanlo L, Usieto S, Carmona-Fontaine C, Barriga EH, Mayor R, Pons S, Martí E. Delamination of neural crest cells requires transient and reversible Wnt inhibition mediated by Dact1/2. Development 2016; 143:2194-205. [PMID: 27122165 DOI: 10.1242/dev.134981] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/18/2016] [Indexed: 02/06/2023]
Abstract
Delamination of neural crest (NC) cells is a bona fide physiological model of epithelial-to-mesenchymal transition (EMT), a process that is influenced by Wnt/β-catenin signalling. Using two in vivo models, we show that Wnt/β-catenin signalling is transiently inhibited at the time of NC delamination. In attempting to define the mechanism underlying this inhibition, we found that the scaffold proteins Dact1 and Dact2, which are expressed in pre-migratory NC cells, are required for NC delamination in Xenopus and chick embryos, whereas they do not affect the motile properties of migratory NC cells. Dact1/2 inhibit Wnt/β-catenin signalling upstream of the transcriptional activity of T cell factor (TCF), which is required for EMT to proceed. Dact1/2 regulate the subcellular distribution of β-catenin, preventing β-catenin from acting as a transcriptional co-activator to TCF, yet without affecting its stability. Together, these data identify a novel yet important regulatory element that inhibits β-catenin signalling, which then affects NC delamination.
Collapse
Affiliation(s)
- M Angeles Rabadán
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri i Reixac 20, Barcelona 08028, Spain
| | - Antonio Herrera
- Department of Cell Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/Baldiri i Reixac 20, Barcelona 08028, Spain
| | - Lucia Fanlo
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri i Reixac 20, Barcelona 08028, Spain
| | - Susana Usieto
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri i Reixac 20, Barcelona 08028, Spain
| | - Carlos Carmona-Fontaine
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Elias H Barriga
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Sebastián Pons
- Department of Cell Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/Baldiri i Reixac 20, Barcelona 08028, Spain
| | - Elisa Martí
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri i Reixac 20, Barcelona 08028, Spain
| |
Collapse
|
16
|
Activation of GLP-1 Receptor Promotes Bone Marrow Stromal Cell Osteogenic Differentiation through β-Catenin. Stem Cell Reports 2016; 6:579-591. [PMID: 26947974 PMCID: PMC4834036 DOI: 10.1016/j.stemcr.2016.02.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 01/30/2016] [Accepted: 02/01/2016] [Indexed: 12/29/2022] Open
Abstract
Glucagon-like peptide 1 (GLP-1) plays an important role in regulating bone remodeling, and GLP-1 receptor agonist shows a positive relationship with osteoblast activity. However, GLP-1 receptor is not found in osteoblast, and the mechanism of GLP-1 receptor agonist on regulating bone remodeling is unclear. Here, we show that the GLP-1 receptor agonist exendin-4 (Ex-4) promoted bone formation and increased bone mass and quality in a rat unloading-induced bone loss model. These functions were accompanied by an increase in osteoblast number and serum bone formation markers, while the adipocyte number was decreased. Furthermore, GLP-1 receptor was detected in bone marrow stromal cells (BMSCs), but not in osteoblast. Activation of GLP-1 receptor by Ex-4 promoted the osteogenic differentiation and inhibited BMSC adipogenic differentiation through regulating PKA/β-catenin and PKA/PI3K/AKT/GSK3β signaling. These findings reveal that GLP-1 receptor regulates BMSC osteogenic differentiation and provide a molecular basis for therapeutic potential of GLP-1 against osteoporosis.
Collapse
|
17
|
Wang J, Han L, Sinnett-Smith J, Han LL, Stevens JV, Rozengurt N, Young SH, Rozengurt E. Positive cross talk between protein kinase D and β-catenin in intestinal epithelial cells: impact on β-catenin nuclear localization and phosphorylation at Ser552. Am J Physiol Cell Physiol 2016; 310:C542-57. [PMID: 26739494 DOI: 10.1152/ajpcell.00302.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/05/2016] [Indexed: 12/17/2022]
Abstract
Given the fundamental role of β-catenin signaling in intestinal epithelial cell proliferation and the growth-promoting function of protein kinase D1 (PKD1) in these cells, we hypothesized that PKDs mediate cross talk with β-catenin signaling. The results presented here provide several lines of evidence supporting this hypothesis. We found that stimulation of intestinal epithelial IEC-18 cells with the G protein-coupled receptor (GPCR) agonist angiotensin II (ANG II), a potent inducer of PKD activation, promoted endogenous β-catenin nuclear localization in a time-dependent manner. A significant increase was evident within 1 h of ANG II stimulation (P< 0.01), peaked at 4 h (P< 0.001), and declined afterwards. GPCR stimulation also induced a marked increase in β-catenin-regulated genes and phosphorylation at Ser(552) in intestinal epithelial cells. Exposure to preferential inhibitors of the PKD family (CRT006610 or kb NB 142-70) or knockdown of the isoforms of the PKD family prevented the increase in β-catenin nuclear localization and phosphorylation at Ser(552) in response to ANG II. GPCR stimulation also induced the formation of a complex between PKD1 and β-catenin, as shown by coimmunoprecipitation that depended on PKD1 catalytic activation, as it was abrogated by cell treatment with PKD family inhibitors. Using transgenic mice that express elevated PKD1 protein in the intestinal epithelium, we detected a marked increase in the localization of β-catenin in the nucleus of crypt epithelial cells in the ileum of PKD1 transgenic mice, compared with nontransgenic littermates. Collectively, our results identify a novel cross talk between PKD and β-catenin in intestinal epithelial cells, both in vitro and in vivo.
Collapse
Affiliation(s)
- Jia Wang
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California
| | - Liang Han
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California
| | - James Sinnett-Smith
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California; CURE, Digestive Diseases Research Center, Los Angeles, California; Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California
| | - Li-Li Han
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California
| | - Jan V Stevens
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California
| | - Nora Rozengurt
- CURE, Digestive Diseases Research Center, Los Angeles, California;
| | - Steven H Young
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California; Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California
| | - Enrique Rozengurt
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California; CURE, Digestive Diseases Research Center, Los Angeles, California; Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California; and Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California
| |
Collapse
|
18
|
Transcriptomic characterization of fibrolamellar hepatocellular carcinoma. Proc Natl Acad Sci U S A 2015; 112:E5916-25. [PMID: 26489647 DOI: 10.1073/pnas.1424894112] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Fibrolamellar hepatocellular carcinoma (FLHCC) tumors all carry a deletion of ∼ 400 kb in chromosome 19, resulting in a fusion of the genes for the heat shock protein, DNAJ (Hsp40) homolog, subfamily B, member 1, DNAJB1, and the catalytic subunit of protein kinase A, PRKACA. The resulting chimeric transcript produces a fusion protein that retains kinase activity. No other recurrent genomic alterations have been identified. Here we characterize the molecular pathogenesis of FLHCC with transcriptome sequencing (RNA sequencing). Differential expression (tumor vs. adjacent normal tissue) was detected for more than 3,500 genes (log2 fold change ≥ 1, false discovery rate ≤ 0.01), many of which were distinct from those found in hepatocellular carcinoma. Expression of several known oncogenes, such as ErbB2 and Aurora Kinase A, was increased in tumor samples. These and other dysregulated genes may serve as potential targets for therapeutic intervention.
Collapse
|
19
|
Ye L, Ding F, Zhang L, Shen A, Yao H, Deng L, Ding Y. Serum apelin is associated with left ventricular hypertrophy in untreated hypertension patients. J Transl Med 2015; 13:290. [PMID: 26342945 PMCID: PMC4560865 DOI: 10.1186/s12967-015-0635-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 08/11/2015] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Apelin is an endogenous ligand for the G protein-coupled receptor APJ. The association between apelin and cardiac modeling has been reported. However, if serum apelin affect the left ventricular hypertrophy (LVH) prevalence in hypertensive patients remains unknown. METHODS We enrolled 344 untreated hypertensive patients. The presence of LVH was determined by echocardiography. The blood was drawn from these patients and serum apelin level was detected. To study the direct effect of apelin on cardiac hypertrophy, cardiomyocytes were cultured and were transfected with apelin gene. Morphometric analysis and measurement of protein contain per cell were then performed. RESULTS We observed a significantly lower serum apelin level in hypertensive patients with LVH compared with those without LVH. Receiver operating characteristic analyses shows that serum apelin level is robust in discriminating patients with LVH from those without. Our in vitro study showed that cellular protein content and cellular size was increased by Ang II treatment, which can be markedly inhibited by the apelin over-expression in cultured cardiomyocytes. CONCLUSION Our clinical date established a link between apelin and LVH, suggesting serum apelin may be used as a predicator for LVH prevalence in hypertensive patients. The direct evidence in vitro suggest apelin pathway is involved in the cardiomyocyte adaption to hypertrophic stimuli.
Collapse
Affiliation(s)
- Lijun Ye
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical College, No. 57 Southern Renmin Avenue, 524023, Zhanjiang, Guangdong, China.
| | - Fenghua Ding
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical College, No. 57 Southern Renmin Avenue, 524023, Zhanjiang, Guangdong, China.
| | - Liang Zhang
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical College, No. 57 Southern Renmin Avenue, 524023, Zhanjiang, Guangdong, China.
| | - Anna Shen
- Department of Cardiology, The Third Affiliated Hospital of Southern Medical University, No.183, West Zhongshan Ave, Guangzhou, Tianhe District, China.
| | - Huaguo Yao
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical College, No. 57 Southern Renmin Avenue, 524023, Zhanjiang, Guangdong, China.
| | - Liehua Deng
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical College, No. 57 Southern Renmin Avenue, 524023, Zhanjiang, Guangdong, China.
| | - Yuanlin Ding
- The Institute of Medical System Biology, School of Public Health, Guangdong Medical College, Dongguan, China.
| |
Collapse
|
20
|
Shi YQ, Yan CC, Zhang X, Yan M, Liu LR, Geng HZ, Lv L, Li BX. Mechanisms underlying probucol-induced hERG-channel deficiency. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:3695-704. [PMID: 26229434 PMCID: PMC4516208 DOI: 10.2147/dddt.s86724] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The hERG gene encodes the pore-forming α-subunit of the rapidly activating delayed rectifier potassium channel (I Kr), which is important for cardiac repolarization. Reduction of I hERG due to genetic mutations or drug interferences causes long QT syndrome, leading to life-threatening cardiac arrhythmias (torsades de pointes) or sudden death. Probucol is a cholesterol-lowering drug that could reduce hERG current by decreasing plasma membrane hERG protein expression and eventually cause long QT syndrome. Here, we investigated the mechanisms of probucol effects on I hERG and hERG-channel expression. Our data demonstrated that probucol reduces SGK1 expression, known as SGK isoform, in a concentration-dependent manner, resulting in downregulation of phosphorylated E3 ubiquitin ligase Nedd4-2 expression, but not the total level of Nedd4-2. As a result, the hERG protein reduces, due to the enhanced ubiquitination level. On the contrary, carbachol could enhance the phosphorylation level of Nedd4-2 as an alternative to SGK1, and thus rescue the ubiquitin-mediated degradation of hERG channels caused by probucol. These discoveries provide a novel mechanism of probucol-induced hERG-channel deficiency, and imply that carbachol or its analog may serve as potential therapeutic compounds for the handling of probucol cardiotoxicity.
Collapse
Affiliation(s)
- Yuan-Qi Shi
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Cai-Chuan Yan
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Xiao Zhang
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Meng Yan
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Li-Rong Liu
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Huai-Ze Geng
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Lin Lv
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Bao-Xin Li
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China ; State-Province Key Laboratory of Biopharmaceutical Engineering, Harbin, Heilongjiang, People's Republic of China
| |
Collapse
|
21
|
Maturana JL, Niechi I, Silva E, Huerta H, Cataldo R, Härtel S, Barros LF, Galindo M, Tapia JC. Transactivation activity and nucleocytoplasmic transport of β-catenin are independently regulated by its C-terminal end. Gene 2015; 573:115-22. [PMID: 26187068 DOI: 10.1016/j.gene.2015.07.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 07/11/2015] [Indexed: 11/20/2022]
Abstract
The key protein in the canonical Wnt pathway is β-catenin, which is phosphorylated both in absence and presence of Wnt signals by different kinases. Upon activation in the cytoplasm, β-catenin can enter into the nucleus to transactivate target gene expression, many of which are cancer-related genes. The mechanism governing β-catenin's nucleocytoplasmic transport has been recently unvealed, although phosphorylation at its C-terminal end and its functional consequences are not completely understood. Serine 646 of β-catenin is a putative CK2 phosphorylation site and lies in a region which has been proposed to be important for its nucleocytoplasmic transport and transactivation activity. This residue was mutated to aspartic acid mimicking CK2-phosphorylation and its effects on β-catenin activity as well as localization were explored. β-Catenin S6464D did not show significant differences in both transcriptional activity and nuclear localization compared to the wild-type form, but displayed a characteristic granular nuclear pattern. Three-dimensional models of nuclei were constructed which showed differences in number and volume of granules, being those from β-catenin S646D more and smaller than the wild-type form. FRAP microscopy was used to compare nuclear export of both proteins which showed a slightly higher but not significant retention of β-catenin S646D. Altogether, these results show that C-terminal phosphorylation of β-catenin seems to be related with its nucleocytoplasmic transport but not transactivation activity.
Collapse
Affiliation(s)
- J L Maturana
- Program of Cellular and Molecular Biology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - I Niechi
- Program of Cellular and Molecular Biology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - E Silva
- Program of Cellular and Molecular Biology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - H Huerta
- Program of Cellular and Molecular Biology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - R Cataldo
- Program of Cellular and Molecular Biology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - S Härtel
- Laboratory for Scientific Image Analysis (SCIAN-Lab), ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - L F Barros
- Centro de Estudios Cientificos, Valdivia, Chile
| | - M Galindo
- Program of Cellular and Molecular Biology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - J C Tapia
- Program of Cellular and Molecular Biology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile.
| |
Collapse
|
22
|
Shin SH, Lee EJ, Chun J, Hyun S, Kang SS. ULK2 Ser 1027 Phosphorylation by PKA Regulates Its Nuclear Localization Occurring through Karyopherin Beta 2 Recognition of a PY-NLS Motif. PLoS One 2015; 10:e0127784. [PMID: 26052940 PMCID: PMC4460075 DOI: 10.1371/journal.pone.0127784] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 04/18/2015] [Indexed: 11/23/2022] Open
Abstract
Uncoordinated 51-like kinase 2 (ULK2), a member of the serine/threonine kinase family, plays an essential role in the regulation of autophagy in mammalian cells. Given the role of autophagy in normal cellular homeostasis and in multiple diseases, improved mechanistic insight into this process may result in the development of novel therapeutic approaches. Here, we present evidence that ULK2 associates with karyopherin beta 2 (Kapβ2) for its transportation into the nucleus. We identify a potential PY-NLS motif (774gpgfgssppGaeaapslRyvPY795) in the S/P space domain of ULK2, which is similar to the consensus PY-NLS motif (R/K/H)X2–5PY. Using a pull-down approach, we observe that ULK2 interacts physically with Kapβ2 both in vitro and in vivo. Confocal microscopy confirmed the co-localization of ULK2 and Kapβ2. Localization of ULK2 to the nuclear region was disrupted by mutations in the putative Kapβ2-binding motif (P794A). Furthermore, in transient transfection assays, the presence of the Kapβ2 binding site mutant (the cytoplasmic localization form) was associated with a substantial increase in autophagy activity (but a decrease in the in vitro serine-phosphorylation) compared with the wild type ULK2. Mutational analysis showed that the phosphorylation on the Ser1027 residue of ULK2 by Protein Kinase A (PKA) is the regulatory point for its functional dissociation from Atg13 and FIP 200, nuclear localization, and autophagy. Taken together, our observations indicate that Kapβ2 interacts with ULK2 through ULK2’s putative PY-NLS motif, and facilitates transport from the cytoplasm to the nucleus, depending on its Ser1027 residue phosphorylation by PKA, thereby reducing autophagic activity.
Collapse
Affiliation(s)
- Sung Hwa Shin
- Department of Biology Education, Chungbuk National University, 1, Chungdae-ro, Seowon-gu, Cheongju, Chungbuk, Republic of Korea
| | - Eun Jeoung Lee
- Department of Internal Medicine, Chungbuk National University, 1, Chungdae-ro, Seowon-gu, Cheongju, Chungbuk, Republic of Korea
| | - Jaesun Chun
- Department of Biology Education, Korea National University of Education, Taeseongtabyeon-ro, Heungdeok-gu, Cheongju, Chungbuk, Republic of Korea
| | - Sunghee Hyun
- Department of Biomedical Laboratory Science, Eulji University, Daejeon, Republic of Korea
| | - Sang Sun Kang
- Department of Biology Education, Chungbuk National University, 1, Chungdae-ro, Seowon-gu, Cheongju, Chungbuk, Republic of Korea
- * E-mail:
| |
Collapse
|
23
|
Bian YY, Guo J, Zhu KX, Guo XN, Peng W, Zhou HM. Resistance investigation of wheat bran polyphenols extracts on HEK293 cells against oxidative damage. RSC Adv 2015. [DOI: 10.1039/c4ra13602k] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Oxidative stress has been considered as a major cause of cellular injury in a variety of clinical abnormalities.
Collapse
Affiliation(s)
- Yuan-Yuan Bian
- State Key Laboratory of Food Science and Technology
- School of Food Science and Technology
- Collaborative Innovation Center for Modern Grain Circulation and Safety
- Jiangnan University
- Wuxi-214122, PR China
| | - Jia Guo
- State Key Laboratory of Food Science and Technology
- School of Food Science and Technology
- Collaborative Innovation Center for Modern Grain Circulation and Safety
- Jiangnan University
- Wuxi-214122, PR China
| | - Ke-Xue Zhu
- State Key Laboratory of Food Science and Technology
- School of Food Science and Technology
- Collaborative Innovation Center for Modern Grain Circulation and Safety
- Jiangnan University
- Wuxi-214122, PR China
| | - Xiao-Na Guo
- State Key Laboratory of Food Science and Technology
- School of Food Science and Technology
- Collaborative Innovation Center for Modern Grain Circulation and Safety
- Jiangnan University
- Wuxi-214122, PR China
| | - Wei Peng
- State Key Laboratory of Food Science and Technology
- School of Food Science and Technology
- Collaborative Innovation Center for Modern Grain Circulation and Safety
- Jiangnan University
- Wuxi-214122, PR China
| | - Hui-Ming Zhou
- State Key Laboratory of Food Science and Technology
- School of Food Science and Technology
- Collaborative Innovation Center for Modern Grain Circulation and Safety
- Jiangnan University
- Wuxi-214122, PR China
| |
Collapse
|
24
|
Liu HT, Chen M, Yu J, Li WJ, Tao L, Li Y, Guo WY, Wang HC. Serum apelin level predicts the major adverse cardiac events in patients with ST elevation myocardial infarction receiving percutaneous coronary intervention. Medicine (Baltimore) 2015; 94:e449. [PMID: 25634182 PMCID: PMC4602953 DOI: 10.1097/md.0000000000000449] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The cardiovascular profile of the apelin makes it a promising therapeutic target for heart failure and ischemic heart disease. However, it remains unknown whether apelin affect the clinical outcome of patients with ST elevation myocardial infarction (STEMI) and received percutaneous coronary intervention (PCI). We enrolled a total of 120 patients with acute STEMI who underwent primary PCI. Serum apelin was detected. After PCI procedure, all patients were followed for 12 months. The follow-up end-point was occurrence of major adverse cardiovascular event (MACE). Lower serum apelin levels (<0.54 ng/mL) was significantly associated with higher serum low density lipoprotein-cholesterol level, higher peak creatine kinase MB fraction (CK-MB) and peak troponin-I (TNI) levels, the number of obstructed vessels, and need for inotropic support. The incidence of MACE was significantly higher in the low apelin group (23 patients out of 67) than in the high apelin group (10 patients out of 75, P < 0.001). Kaplan-Meier analysis revealed that the MACE-free rate was significantly lower in the patients with low apelin than those with high apelin (P < 0.001, log rank test). The multivariate Cox proportional hazard analysis adjusted with the clinical and angiographic characteristic reveals that the serum low apelin is a predictor for MACE incidence (hazard ratio = 2.36, 95% confidence interval: 1.83-3.87, P = 0.004). The finding of this study suggests that the serum apelin may be used as a marker to predict the MACE after PCI in patients with STEMI.
Collapse
Affiliation(s)
- Hai-Tao Liu
- From the Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | |
Collapse
|